1
|
Huang X, Cheng Z, Lv Y, Li W, Liu X, Huang W, Zhao C. Neutralization potency of the 2023-24 seasonal influenza vaccine against circulating influenza H3N2 strains. Hum Vaccin Immunother 2024; 20:2380111. [PMID: 39205645 PMCID: PMC11364067 DOI: 10.1080/21645515.2024.2380111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/07/2024] [Revised: 06/27/2024] [Accepted: 07/11/2024] [Indexed: 09/04/2024] Open
Abstract
Seasonal influenza is a severe disease that significantly impacts public health, causing millions of infections and hundreds of thousands of deaths each year. Seasonal influenza viruses, particularly the H3N2 subtype, exhibit high antigenic variability, often leading to mismatch between vaccine strains and circulating strains. Therefore, rapidly assessing the alignment between existing seasonal influenza vaccine and circulating strains is crucial for enhancing vaccine efficacy. This study, based on a pseudovirus platform, evaluated the match between current influenza H3N2 vaccine strains and circulating strains through cross-neutralization assays using clinical human immune sera against globally circulating influenza virus strains. The research results show that although mutations are present in the circulating strains, the current H3N2 vaccine strain still imparting effective protection, providing a scientific basis for encouraging influenza vaccination. This research methodology can be sustainably applied for the neutralization potency assessment of subsequent circulating strains, establishing a persistent methodological framework.
Collapse
Affiliation(s)
- Xiande Huang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Ziqi Cheng
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Yake Lv
- Center of Vaccine Clinical Evaluation, Institute for Immunization Program, Shaanxi Provincial Centre for Disease Control and Prevention, Xi’an, Shaanxi Province, China
| | - Weixuan Li
- Center of Vaccine Clinical Evaluation, Institute for Immunization Program, Shaanxi Provincial Centre for Disease Control and Prevention, Xi’an, Shaanxi Province, China
| | - Xiaoyu Liu
- Center of Vaccine Clinical Evaluation, Institute for Immunization Program, Shaanxi Provincial Centre for Disease Control and Prevention, Xi’an, Shaanxi Province, China
| | - Weijin Huang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Chenyan Zhao
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| |
Collapse
|
2
|
Santosh AK, Kumar D, Kaur C, Gupta P, Jasmeen P, Dilip L, Kavitha G, Basagoudanavar S, Hosamani M, Balamurugan V, Sharada R, Rathnamma D, Sunil KM, Hegde NR, Isloor S. Evaluation of the immune status of dogs vaccinated against rabies by an enzyme-linked immunosorbent assay using crude preparations of insect cells infected with a recombinant baculovirus encoding the rabies virus glycoprotein gene. PLoS One 2024; 19:e0314516. [PMID: 39625902 PMCID: PMC11614288 DOI: 10.1371/journal.pone.0314516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/19/2024] [Accepted: 11/11/2024] [Indexed: 12/06/2024] Open
Abstract
Evaluation of the effectiveness of vaccination of animals against rabies is not routinely implemented. In cases where it is carried out, the rapid fluorescent focus inhibition test (RFFIT) or the fluorescent antibody virus neutralization (FAVN) test are the recommended tests. However, both of these tests require handling of live rabies virus (RABV), and are cumbersome to perform. In view of this, the enzyme-linked immunosorbent assay (ELISA) has been proposed as a surrogate test; however, availability of appropriate antigen is a major impediment for the development of ELISAs to detect anti-rabies antibodies. The most widely used antigen is the RABV glycoprotein (G) purified from cell culture-propagated virus, which requires a biosafety level 3 containment. The alternative is to use recombinantly expressed G, which needs to be to be properly glycosylated and folded to serve as the best antigen. The most suitable system for its production is the baculovirus expression system (BVES). However, purification of RABV G is challenging. We therefore tested partially purified preparations in the form of extracts of insect cells infected with baculovirus expressing RABV G, against sera from vaccinated dogs in an indirect ELISA. The results showed good concordance against RFFIT, with sensitivity and specificity of 90.48% and 80.00%, respectively. The system may be used for quick screening to determine the presence and an approximate level of antibodies, and can be modified to enable monitoring of mass dog vaccination programs, as well as to facilitate certification of dogs intended for international travel and transportation.
Collapse
Affiliation(s)
- A. K. Santosh
- KVAFSU-CVA Rabies Diagnostic Laboratory, Department of Veterinary Microbiology, Veterinary College, Karnataka Veterinary Animal and Fisheries Sciences University, Bengaluru, India
| | - Deepak Kumar
- Ella Foundation, Genome Valley, Turkapally, Shameerpet Mandal, Hyderabad, India
| | - Charanpreet Kaur
- National Institute of Animal Biotechnology, Gachibowli, Hyderabad, India
| | - Priya Gupta
- National Institute of Animal Biotechnology, Gachibowli, Hyderabad, India
- Regional Centre for Biotechnology, Faridabad, India
| | - Pagala Jasmeen
- National Institute of Animal Biotechnology, Gachibowli, Hyderabad, India
- Regional Centre for Biotechnology, Faridabad, India
| | - L. Dilip
- KVAFSU-CVA Rabies Diagnostic Laboratory, Department of Veterinary Microbiology, Veterinary College, Karnataka Veterinary Animal and Fisheries Sciences University, Bengaluru, India
| | - G. Kavitha
- KVAFSU-CVA Rabies Diagnostic Laboratory, Department of Veterinary Microbiology, Veterinary College, Karnataka Veterinary Animal and Fisheries Sciences University, Bengaluru, India
| | | | | | - V. Balamurugan
- ICAR – National Institute of Veterinary Epidemiology and Disease Informatics, Bengaluru, India
| | - R. Sharada
- KVAFSU-CVA Rabies Diagnostic Laboratory, Department of Veterinary Microbiology, Veterinary College, Karnataka Veterinary Animal and Fisheries Sciences University, Bengaluru, India
| | - D. Rathnamma
- KVAFSU-CVA Rabies Diagnostic Laboratory, Department of Veterinary Microbiology, Veterinary College, Karnataka Veterinary Animal and Fisheries Sciences University, Bengaluru, India
| | - K. M. Sunil
- KVAFSU-CVA Rabies Diagnostic Laboratory, Department of Veterinary Microbiology, Veterinary College, Karnataka Veterinary Animal and Fisheries Sciences University, Bengaluru, India
| | - Nagendra R. Hegde
- National Institute of Animal Biotechnology, Gachibowli, Hyderabad, India
- Regional Centre for Biotechnology, Faridabad, India
| | - Shrikrishna Isloor
- KVAFSU-CVA Rabies Diagnostic Laboratory, Department of Veterinary Microbiology, Veterinary College, Karnataka Veterinary Animal and Fisheries Sciences University, Bengaluru, India
| |
Collapse
|
3
|
Long C, Wang W, Du J, Xu G, Yu C, Wang L. Developing a human monoclonal antibody combination CRM25 to prevent rabies after exposure. Int J Antimicrob Agents 2024; 64:107383. [PMID: 39542064 DOI: 10.1016/j.ijantimicag.2024.107383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/12/2024] [Revised: 10/24/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024]
Abstract
OBJECTIVE Immunization against rabies post-exposure prophylaxis requires passive immunization with either monoclonal antibody (mAb) or blood-derived rabies immunoglobin (RIG). Currently, replacing traditional RIG with emerging mAb or mAb combinations is highly recommended due to the limited supply and potential safety risks of RIG. METHODS We developed a mAb combination named CRM25 by combining two human mAbs, RM02 and RM05, at a 1:1 mass ratio. RESULTS RM02 and RM05 were non-competing and non-overlapping mAbs targeting epitopes I and III, respectively. K226 and G229 were found to be the critical amino acid sites for RM02 neutralization, but the mutant I338T displayed decreased susceptibility to RM05 neutralization. Notably, CRM25 was capable of cross-neutralizing rabies virus (RABV) strains containing K226M or I338T mutations. CRM25 additionally showed an inhibitory effect on the infection of all tested common RABVs and non-RABV phylogroup I lyssaviruses. CRM25 not only exhibited neutralizing activity but also exhibited antiviral effects via Fc-mediated effector functions. Importantly, CRM25 was comparable to human RIG in terms of its capacity to protect Syrian golden hamsters from lethal RABV challenges. CONCLUSIONS These findings promote more thorough research on CRM25's antiviral properties in cells and in vivo to enhance its clinical applicability and suggest that it may be a viable candidate medication for rabies post-exposure prophylaxis.
Collapse
Affiliation(s)
- Caifeng Long
- Division of Monoclonal Antibody Products, National Institutes for Food and Drug Control, State Key Laboratory of Drug Regulatory Science, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, China
| | - Wenbo Wang
- Center for Drug Evaluation, National Medical Products Administration, Beijing, China
| | - Jialiang Du
- Division of Monoclonal Antibody Products, National Institutes for Food and Drug Control, State Key Laboratory of Drug Regulatory Science, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, China
| | - Gangling Xu
- Division of Monoclonal Antibody Products, National Institutes for Food and Drug Control, State Key Laboratory of Drug Regulatory Science, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, China
| | - Chuanfei Yu
- Division of Monoclonal Antibody Products, National Institutes for Food and Drug Control, State Key Laboratory of Drug Regulatory Science, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, China.
| | - Lan Wang
- Division of Monoclonal Antibody Products, National Institutes for Food and Drug Control, State Key Laboratory of Drug Regulatory Science, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, China.
| |
Collapse
|
4
|
Sharma N, Jamwal VL, Nagial S, Ranjan M, Rath D, Gandhi SG. Current status of diagnostic assays for emerging zoonotic viruses: Nipah and Hendra. Expert Rev Mol Diagn 2024; 24:473-485. [PMID: 38924448 DOI: 10.1080/14737159.2024.2368591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/29/2023] [Accepted: 06/12/2024] [Indexed: 06/28/2024]
Abstract
INTRODUCTION Nipah and Hendra viruses belong to the Paramyxoviridae family, which pose a significant threat to human health, with sporadic outbreaks causing severe morbidity and mortality. Early symptoms include fever, cough, sore throat, and headache, which offer little in terms of differential diagnosis. There are no specific therapeutics and vaccines for these viruses. AREAS COVERED This review comprehensively covers a spectrum of diagnostic techniques for Nipah and Hendra virus infections, discussed in conjunction with appropriate type of samples during the progression of infection. Serological assays, reverse transcriptase Real-Time PCR assays, and isothermal amplification assays are discussed in detail, along with a listing of few commercially available detection kits. Patents protecting inventions in Nipah and Hendra virus detection are also covered. EXPERT OPINION Despite several outbreaks of Nipah and Hendra infections in the past decade, in-depth research into their pathogenesis, Point-of-Care diagnostics, specific therapies, and human vaccines is lacking. A prompt and accurate diagnosis is pivotal for efficient outbreak management, patient treatment, and the adoption of preventative measures. The emergence of rapid point-of-care tests holds promise in enhancing diagnostic capabilities in real-world settings. The patent landscape emphasizes the importance of innovation and collaboration within the legal and business realms.
Collapse
Affiliation(s)
- Nancy Sharma
- Infectious Diseases Division, CSIR-Indian Institute of Integrative Medicine (CSIR-IIIM), Jammu, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Vijay Lakshmi Jamwal
- Microfluidics Design and Bioengineering Lab, Chemical Engineering Department, Indian Institute of Technology Jammu (IIT), Jammu, India
| | - Sakshi Nagial
- Infectious Diseases Division, CSIR-Indian Institute of Integrative Medicine (CSIR-IIIM), Jammu, India
| | - Manish Ranjan
- Department of Microbiology, All India Institute of Medical Sciences Jammu (AIIMS), Jammu, India
| | - Dharitri Rath
- Microfluidics Design and Bioengineering Lab, Chemical Engineering Department, Indian Institute of Technology Jammu (IIT), Jammu, India
| | - Sumit G Gandhi
- Infectious Diseases Division, CSIR-Indian Institute of Integrative Medicine (CSIR-IIIM), Jammu, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
5
|
Trischitta P, Tamburello MP, Venuti A, Pennisi R. Pseudovirus-Based Systems for Screening Natural Antiviral Agents: A Comprehensive Review. Int J Mol Sci 2024; 25:5188. [PMID: 38791226 PMCID: PMC11121416 DOI: 10.3390/ijms25105188] [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] [Academic Contribution Register] [Received: 03/27/2024] [Revised: 05/03/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
Since the outbreak of COVID-19, researchers have been working tirelessly to discover effective ways to combat coronavirus infection. The use of computational drug repurposing methods and molecular docking has been instrumental in identifying compounds that have the potential to disrupt the binding between the spike glycoprotein of SARS-CoV-2 and human ACE2 (hACE2). Moreover, the pseudovirus approach has emerged as a robust technique for investigating the mechanism of virus attachment to cellular receptors and for screening targeted small molecule drugs. Pseudoviruses are viral particles containing envelope proteins, which mediate the virus's entry with the same efficiency as that of live viruses but lacking pathogenic genes. Therefore, they represent a safe alternative to screen potential drugs inhibiting viral entry, especially for highly pathogenic enveloped viruses. In this review, we have compiled a list of antiviral plant extracts and natural products that have been extensively studied against enveloped emerging and re-emerging viruses by pseudovirus technology. The review is organized into three parts: (1) construction of pseudoviruses based on different packaging systems and applications; (2) knowledge of emerging and re-emerging viruses; (3) natural products active against pseudovirus-mediated entry. One of the most crucial stages in the life cycle of a virus is its penetration into host cells. Therefore, the discovery of viral entry inhibitors represents a promising therapeutic option in fighting against emerging viruses.
Collapse
Affiliation(s)
- Paola Trischitta
- Department of Chemical, Biological, Pharmaceutical and Environmental Science, University of Messina, Viale Ferdinando Stagno d’Alcontres 31, 98166 Messina, Italy; (P.T.); (M.P.T.)
- Department of Chemistry, Biology, and Biotechnology, University of Perugia, Via Elce di Sotto 8, 06123 Perugia, Italy
| | - Maria Pia Tamburello
- Department of Chemical, Biological, Pharmaceutical and Environmental Science, University of Messina, Viale Ferdinando Stagno d’Alcontres 31, 98166 Messina, Italy; (P.T.); (M.P.T.)
| | - Assunta Venuti
- International Agency for Research on Cancer (IARC), World Health Organization, 69366 Lyon, CEDEX 07, France;
| | - Rosamaria Pennisi
- Department of Chemical, Biological, Pharmaceutical and Environmental Science, University of Messina, Viale Ferdinando Stagno d’Alcontres 31, 98166 Messina, Italy; (P.T.); (M.P.T.)
| |
Collapse
|
6
|
Liang Z, Tong J, Wu X, Liu S, Wu J, Yu Y, Zhang L, Zhao C, Lu Q, Nie J, Huang W, Wang Y. Development of a SARS-CoV-2 neutralization assay based on a pseudotyped virus using a HIV system. MedComm (Beijing) 2024; 5:e517. [PMID: 38525106 PMCID: PMC10959455 DOI: 10.1002/mco2.517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/12/2023] [Revised: 02/06/2024] [Accepted: 02/25/2024] [Indexed: 03/26/2024] Open
Abstract
Regarding the extensive global attention to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that constitutes an international public health emergency, pseudovirus neutralization assays have been widely applied due to their advantages of being able to be conducted in biosafety level 2 laboratories and having a high safety factor. In this study, by adding a blue fluorescent protein (AmCyan) gene to the HIV system pSG3-△env backbone plasmid HpaI and truncating the C-terminal 21 amino acids of the SARS-CoV-2 spike protein (S), high-titer SARS-CoV-2-Sdel21-AmCyan fluorescent pseudovirus was successfully packaged. The fluorescent pseudovirus was used to establish a neutralization assay in a 96-well plate using 293T cells stably transfected with the AF cells. Then, parameters such as the ratio of backbone and membrane plasmid, sensitive cells, inoculation of cells and virus, as well as incubation and detection time were optimized. The pseudovirus neutralization assay demonstrated high accuracy, sensitivity, repeatability, and a strong correlation with the luminescent pseudovirus neutralization assay. Additionally, we scaled up the neutralizing antibody determination method by increasing the plate size from 96 wells to 384 wells. We have established a robust fluorescent pseudotyped virus neutralization assay for SARS-CoV-2 using the HIV system, providing a foundation for serum neutralization antibody detection, monoclonal antibody screening, and vaccine development.
Collapse
Affiliation(s)
- Ziteng Liang
- Chinese Academy of Medical Sciences & Peking Union Medical CollegeDongcheng District, BeijingChina
- Division of HIV/AIDS and Sex‐transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), WHO Collaborating Center for Standardization and Evaluation of BiologicalsNHC Key Laboratory of Research on Quality and Standardization of Biotech Products and NMPA Key Laboratory for Quality Research and Evaluation of Biological ProductsBeijingChina
| | - Jincheng Tong
- Division of HIV/AIDS and Sex‐transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), WHO Collaborating Center for Standardization and Evaluation of BiologicalsNHC Key Laboratory of Research on Quality and Standardization of Biotech Products and NMPA Key Laboratory for Quality Research and Evaluation of Biological ProductsBeijingChina
| | - Xi Wu
- Division of HIV/AIDS and Sex‐transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), WHO Collaborating Center for Standardization and Evaluation of BiologicalsNHC Key Laboratory of Research on Quality and Standardization of Biotech Products and NMPA Key Laboratory for Quality Research and Evaluation of Biological ProductsBeijingChina
| | - Shuo Liu
- Changping LaboratoryChangping District, BeijingChina
| | - Jiajing Wu
- Beijing Yunling Biotechnology Co., Ltd.BeijingChina
| | - Yuanling Yu
- Changping LaboratoryChangping District, BeijingChina
| | - Li Zhang
- Division of HIV/AIDS and Sex‐transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), WHO Collaborating Center for Standardization and Evaluation of BiologicalsNHC Key Laboratory of Research on Quality and Standardization of Biotech Products and NMPA Key Laboratory for Quality Research and Evaluation of Biological ProductsBeijingChina
| | - Chenyan Zhao
- Division of HIV/AIDS and Sex‐transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), WHO Collaborating Center for Standardization and Evaluation of BiologicalsNHC Key Laboratory of Research on Quality and Standardization of Biotech Products and NMPA Key Laboratory for Quality Research and Evaluation of Biological ProductsBeijingChina
| | - Qiong Lu
- Division of HIV/AIDS and Sex‐transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), WHO Collaborating Center for Standardization and Evaluation of BiologicalsNHC Key Laboratory of Research on Quality and Standardization of Biotech Products and NMPA Key Laboratory for Quality Research and Evaluation of Biological ProductsBeijingChina
| | - Jianhui Nie
- Division of HIV/AIDS and Sex‐transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), WHO Collaborating Center for Standardization and Evaluation of BiologicalsNHC Key Laboratory of Research on Quality and Standardization of Biotech Products and NMPA Key Laboratory for Quality Research and Evaluation of Biological ProductsBeijingChina
| | - Weijin Huang
- Division of HIV/AIDS and Sex‐transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), WHO Collaborating Center for Standardization and Evaluation of BiologicalsNHC Key Laboratory of Research on Quality and Standardization of Biotech Products and NMPA Key Laboratory for Quality Research and Evaluation of Biological ProductsBeijingChina
| | - Youchun Wang
- Chinese Academy of Medical Sciences & Peking Union Medical CollegeDongcheng District, BeijingChina
- Division of HIV/AIDS and Sex‐transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), WHO Collaborating Center for Standardization and Evaluation of BiologicalsNHC Key Laboratory of Research on Quality and Standardization of Biotech Products and NMPA Key Laboratory for Quality Research and Evaluation of Biological ProductsBeijingChina
- Changping LaboratoryChangping District, BeijingChina
| |
Collapse
|
7
|
Thimmiraju SR, Kimata JT, Pollet J. Pseudoviruses, a safer toolbox for vaccine development against enveloped viruses. Expert Rev Vaccines 2024; 23:174-185. [PMID: 38164690 DOI: 10.1080/14760584.2023.2299380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/13/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024]
Abstract
INTRODUCTION Pseudoviruses are recombinant, replication-incompetent, viral particles designed to mimic the surface characteristics of native enveloped viruses. They are a safer, and cost-effective research alternative to live viruses. With the potential emergence of the next major infectious disease, more vaccine scientists must become familiar with the pseudovirus platform as a vaccine development tool to mitigate future outbreaks. AREAS COVERED This review aims at vaccine developers to provide a basic understanding of pseudoviruses, list their production methods, and discuss their utility to assess vaccine efficacy against enveloped viral pathogens. We further illustrate their usefulness as wet-lab simulators for emerging mutant variants, and new viruses to help prepare for current and future viral outbreaks, minimizing the need for gain-of-function experiments with highly infectious or lethal enveloped viruses. EXPERT OPINION With this platform, researchers can better understand the role of virus-receptor interactions and entry in infections, prepare for dangerous mutations, and develop effective vaccines.
Collapse
Affiliation(s)
- Syamala R Thimmiraju
- Department of Pediatrics, Section of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
- Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, USA
| | - Jason T Kimata
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Jeroen Pollet
- Department of Pediatrics, Section of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
- Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
8
|
Jiang H, Jin P, Guo X, Zhu J, Wang X, Wan P, Wan J, Liu J, Li J, Zhu F. The 6-Month Antibody Durability of Heterologous Convidecia Plus CoronaVac and Homologous CoronaVac Immunizations in People Aged 18-59 Years and over 60 Years Based on Two Randomized Controlled Trials in China. Vaccines (Basel) 2023; 11:1815. [PMID: 38140219 PMCID: PMC10747853 DOI: 10.3390/vaccines11121815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/07/2023] [Revised: 11/09/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023] Open
Abstract
Previous reports have shown that heterologous boosting with the AD5-vectored COVID-19 vaccine Convidecia based on a primary series of two doses of inactivated vaccine induces increasing immune responses. However, the immune persistence until 6 months after the heterologous prime-boost immunization was limited. Participants were from two single-center, randomized, controlled, observer-blinded trials, which involved individuals of 18-59 years of age and over 60 years of age. Eligible participants who previously primed with one dose or two doses of CoronaVac were stratified and randomly assigned to inoculate a booster dose of Convidecia or CoronaVac. Neutralizing antibodies against a live SARS-CoV-2 prototype virus and Delta and Omicron (B.1.1.529) variants, pseudovirus neutralizing antibodies against Omicron BA.4/5 variants, and anti-SARS-CoV-2 RBD antibodies at month 6 were detected, and the fold decreases and rate difference were calculated by comparing the levels of antibodies at month 6 with the peak levels at month 1. The neutralizing antibody titers against prototype SARS-CoV-2, RBD-specific IgG antibodies, and the Delta variant in the heterologous regimen of the CoronaVac plus Convidecia groups were significantly higher than those of the homologous prime-boost groups. In three-dose regimen groups, the geometric mean titers (GMTs) of neutralizing antibodies against prototype SARS-CoV-2 were 30.6 (95% CI: 25.1; 37.2) in the heterologous boosting group versus 6.9 (95% CI: 5.6; 8.6) in the homologous boosting group (p < 0.001) at month 6 in participants aged 18-59 years, and in the two-dose regimen, the neutralizing antibody GMTs were 8.5 (95% CI: 6.2; 11.7) and 2.7 (2.3 to 3.1) (heterologous regimen group versus CoronaVac regimen group, p < 0.001). Participants aged over 60 years had similar levels of neutralizing antibodies against the prototype, with GMTs of 49.1 (38.0 to 63.6) in the group receiving two doses of CoronaVac plus one dose of Convidecia versus 9.4 (7.7 to 11.4) in the group receiving three doses of CoronaVac (p < 0.001) and 11.6 (8.4 to 16.0) in the group receiving one dose of CoronaVac and one dose of Convidecia versus 3.3 (2.7 to 4.0) in the group receiving two doses of CoronaVac (p < 0.001). Compared with day 14, over sixfold decreases in neutralizing antibody GMTs were observed in the heterologous groups of the three- or two-dose regimen groups of younger and elderly participants, while in the homologous regimen groups, the GMTs of neutralizing antibodies decreased about fivefold in the two age groups. The heterologous prime-boost regimen with two doses of CoronaVac and one dose of Convidecia was persistently more immunogenic than the regimen of the homologous prime-boost with three doses of CoronaVac.
Collapse
Affiliation(s)
- Hudachuan Jiang
- School of Public Health, Southeast University, 87 Dingjiaqiao Avenue, Nanjing 210009, China;
| | - Pengfei Jin
- NHC Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, 172 Jiangsu Avenue, Nanjing 210009, China; (P.J.); (X.G.); (J.L.)
- School of Science, China Pharmaceutical University, Nanjing 210009, China
| | - Xiling Guo
- NHC Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, 172 Jiangsu Avenue, Nanjing 210009, China; (P.J.); (X.G.); (J.L.)
| | - Jiahong Zhu
- Lianshui County Center for Disease Control and Prevention, Huaian 223400, China;
| | - Xue Wang
- CanSino Biologics Inc., Tianjin 300450, China; (X.W.); (P.W.); (J.W.)
| | - Peng Wan
- CanSino Biologics Inc., Tianjin 300450, China; (X.W.); (P.W.); (J.W.)
| | - Jingxuan Wan
- CanSino Biologics Inc., Tianjin 300450, China; (X.W.); (P.W.); (J.W.)
| | - Jingxian Liu
- NHC Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, 172 Jiangsu Avenue, Nanjing 210009, China; (P.J.); (X.G.); (J.L.)
| | - Jingxin Li
- School of Public Health, Southeast University, 87 Dingjiaqiao Avenue, Nanjing 210009, China;
- NHC Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, 172 Jiangsu Avenue, Nanjing 210009, China; (P.J.); (X.G.); (J.L.)
- School of Public Health, National Vaccine Innovation Platform, Nanjing Medical University, Nanjing 210009, China
| | - Fengcai Zhu
- School of Public Health, Southeast University, 87 Dingjiaqiao Avenue, Nanjing 210009, China;
- NHC Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, 172 Jiangsu Avenue, Nanjing 210009, China; (P.J.); (X.G.); (J.L.)
- School of Public Health, National Vaccine Innovation Platform, Nanjing Medical University, Nanjing 210009, China
| |
Collapse
|
9
|
Jin L, Tang R, Wu S, Guo X, Huang H, Hou L, Chen X, Zhu T, Gou J, Zhong J, Pan H, Cui L, Chen Y, Xia X, Feng J, Wang X, Zhao Q, Xu X, Li Z, Zhang X, Chen W, Li J, Zhu F. Antibody persistence and safety after heterologous boosting with orally aerosolised Ad5-nCoV in individuals primed with two-dose CoronaVac previously: 12-month analyses of a randomized controlled trial. Emerg Microbes Infect 2023; 12:2155251. [PMID: 36503413 PMCID: PMC10519268 DOI: 10.1080/22221751.2022.2155251] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/27/2022] [Accepted: 11/30/2022] [Indexed: 12/14/2022]
Abstract
Antibody persistence and safety up to 12 months of heterologous orally administered adenovirus type-5 vector-based COVID-19 vaccine (Ad5-nCoV) in individuals who were primed with two-dose inactivated SARS-CoV-2 vaccine (CoronaVac) previously, has not been reported yet. This randomized, open-label, single-centre trial included Chinese adults who have received two-dose CoronaVac randomized to low-dose or high-dose aerosolised Ad5-nCoV group, or CoronaVac group. In this report, we mainly evaluated the geometric mean titres (GMTs) of neutralizing antibodies (NAbs) against live wild-type SARS-CoV-2 virus and omicron BA.4/5 pseudovirus at 12 months after the booster dose and the incidence of serious adverse events (SAEs) till month 12. Of 419 participants, all were included in the safety analysis and 120 (28.64%) were included in the immunogenicity analysis. Serum NAb GMT against live wild-type SARS-CoV-2 was 204.36 (95% CI 152.91, 273.14) in the low-dose group and 171.38 (95% CI 121.27, 242.19) in the high-dose group at month 12, significantly higher than the GMT in the CoronaVac group (8.00 [95% CI 4.22, 15.17], p < 0.0001). Serum NAb GMT against omicron BA.4/5 pseudovirus was 40.97 (95% CI 30.15, 55.67) in the low-dose group and 35.08 (95% CI 26.31, 46.77) in the high-dose group at month 12, whereas the GMT in the CoronaVac group was below the lower limit of detection. No vaccine-related SAEs were observed. Orally administered aerosolised Ad5-nCoV following two-dose CoronaVac priming has a good safety profile and is persistently more immunogenic than three-dose CoronaVac within 12 months after the booster dose.Trial registration: ClinicalTrials.gov identifier: NCT05043259..
Collapse
Affiliation(s)
- Lairun Jin
- School of Public Health, Southeast University, Nanjing, People’s Republic of China
| | - Rong Tang
- National Health Commission (NHC) Key Laboratory of Enteric Pathogenic Microbiology (Jiangsu Provincial Center for Disease Control and Prevention), Nanjing, People’s Republic of China
| | - Shipo Wu
- Beijing Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, People’s Republic of China
| | - Xiling Guo
- National Health Commission (NHC) Key Laboratory of Enteric Pathogenic Microbiology (Jiangsu Provincial Center for Disease Control and Prevention), Nanjing, People’s Republic of China
| | - Haitao Huang
- Cansino Biologics Inc., Tianjin, People’s Republic of China
| | - Lihua Hou
- Beijing Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, People’s Republic of China
| | - Xiaoqin Chen
- Donghai County Center for Disease Control and Prevention, Lianyungang, Jiangsu, People’s Republic of China
| | - Tao Zhu
- Cansino Biologics Inc., Tianjin, People’s Republic of China
| | - Jinbo Gou
- Cansino Biologics Inc., Tianjin, People’s Republic of China
| | - Jin Zhong
- Donghai County Center for Disease Control and Prevention, Lianyungang, Jiangsu, People’s Republic of China
| | - Hongxing Pan
- National Health Commission (NHC) Key Laboratory of Enteric Pathogenic Microbiology (Jiangsu Provincial Center for Disease Control and Prevention), Nanjing, People’s Republic of China
| | - Lunbiao Cui
- National Health Commission (NHC) Key Laboratory of Enteric Pathogenic Microbiology (Jiangsu Provincial Center for Disease Control and Prevention), Nanjing, People’s Republic of China
| | - Yin Chen
- National Health Commission (NHC) Key Laboratory of Enteric Pathogenic Microbiology (Jiangsu Provincial Center for Disease Control and Prevention), Nanjing, People’s Republic of China
| | - Xin Xia
- School of Public Health, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Jialu Feng
- School of Public Health, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Xue Wang
- Cansino Biologics Inc., Tianjin, People’s Republic of China
| | - Qi Zhao
- Cansino Biologics Inc., Tianjin, People’s Republic of China
| | - XiaoYu Xu
- Vazyme Biotech Co., Ltd, Nanjing, People’s Republic of China
| | - Zhuopei Li
- School of Public Health, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Xiaoyin Zhang
- School of Public Health, Southeast University, Nanjing, People’s Republic of China
| | - Wei Chen
- Beijing Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, People’s Republic of China
| | - Jingxin Li
- National Health Commission (NHC) Key Laboratory of Enteric Pathogenic Microbiology (Jiangsu Provincial Center for Disease Control and Prevention), Nanjing, People’s Republic of China
- School of Public Health, Nanjing Medical University, Nanjing, People’s Republic of China
- Institute of Global Public Health and Emergency Pharmacy, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Fengcai Zhu
- School of Public Health, Southeast University, Nanjing, People’s Republic of China
- National Health Commission (NHC) Key Laboratory of Enteric Pathogenic Microbiology (Jiangsu Provincial Center for Disease Control and Prevention), Nanjing, People’s Republic of China
- School of Public Health, Nanjing Medical University, Nanjing, People’s Republic of China
- Institute of Global Public Health and Emergency Pharmacy, China Pharmaceutical University, Nanjing, People’s Republic of China
| |
Collapse
|
10
|
Cantoni D, Wilkie C, Bentley EM, Mayora-Neto M, Wright E, Scott S, Ray S, Castillo-Olivares J, Heeney JL, Mattiuzzo G, Temperton NJ. Correlation between pseudotyped virus and authentic virus neutralisation assays, a systematic review and meta-analysis of the literature. Front Immunol 2023; 14:1184362. [PMID: 37790941 PMCID: PMC10544934 DOI: 10.3389/fimmu.2023.1184362] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/11/2023] [Accepted: 08/28/2023] [Indexed: 10/05/2023] Open
Abstract
Background The virus neutralization assay is a principal method to assess the efficacy of antibodies in blocking viral entry. Due to biosafety handling requirements of viruses classified as hazard group 3 or 4, pseudotyped viruses can be used as a safer alternative. However, it is often queried how well the results derived from pseudotyped viruses correlate with authentic virus. This systematic review and meta-analysis was designed to comprehensively evaluate the correlation between the two assays. Methods Using PubMed and Google Scholar, reports that incorporated neutralisation assays with both pseudotyped virus, authentic virus, and the application of a mathematical formula to assess the relationship between the results, were selected for review. Our searches identified 67 reports, of which 22 underwent a three-level meta-analysis. Results The three-level meta-analysis revealed a high level of correlation between pseudotyped viruses and authentic viruses when used in an neutralisation assay. Reports that were not included in the meta-analysis also showed a high degree of correlation, with the exception of lentiviral-based pseudotyped Ebola viruses. Conclusion Pseudotyped viruses identified in this report can be used as a surrogate for authentic virus, though care must be taken in considering which pseudotype core to use when generating new uncharacterised pseudotyped viruses.
Collapse
Affiliation(s)
- Diego Cantoni
- MRC-University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow, United Kingdom
| | - Craig Wilkie
- School of Mathematics & Statistics, University of Glasgow, Glasgow, United Kingdom
| | - Emma M. Bentley
- Medicines and Healthcare Products Regulatory Agency, South Mimms, United Kingdom
| | - Martin Mayora-Neto
- Viral Pseudotype Unit, Medway School of Pharmacy, The Universities of Greenwich and Kent at Medway, Chatham, United Kingdom
| | - Edward Wright
- Viral Pseudotype Unit, School of Life Sciences, University of Sussex, Brighton, United Kingdom
| | - Simon Scott
- Viral Pseudotype Unit, Medway School of Pharmacy, The Universities of Greenwich and Kent at Medway, Chatham, United Kingdom
| | - Surajit Ray
- School of Mathematics & Statistics, University of Glasgow, Glasgow, United Kingdom
| | - Javier Castillo-Olivares
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge University, Cambridge, United Kingdom
| | - Jonathan Luke Heeney
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge University, Cambridge, United Kingdom
- DIOSynVax, University of Cambridge, Cambridge, United Kingdom
| | - Giada Mattiuzzo
- Medicines and Healthcare Products Regulatory Agency, South Mimms, United Kingdom
| | - Nigel James Temperton
- Viral Pseudotype Unit, Medway School of Pharmacy, The Universities of Greenwich and Kent at Medway, Chatham, United Kingdom
| |
Collapse
|
11
|
Huang S, Wu Z, Zhou B, Jiang X, Lavillette D, Fan G. Heat-Denatured Lysozyme is a Novel Potential Non-alcoholic Disinfectant Against Respiratory Virus. FOOD AND ENVIRONMENTAL VIROLOGY 2023; 15:212-223. [PMID: 37155116 PMCID: PMC10166042 DOI: 10.1007/s12560-023-09556-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 03/23/2022] [Accepted: 04/20/2023] [Indexed: 05/10/2023]
Abstract
Respiratory diseases are significant recurrent threats to global public health. Since the 1918 Spanish flu pandemic, seasonal influenza viruses continue to cause epidemics around the world each year. More recently, the COVID-19 global pandemic conducted a public health crisis with more than 6 million deaths and it also severely affected the global economy. Due to the phenomenon that people get infection from objects carrying viruses, it has aroused people's attention to home disinfection. As there is no ideal existing common domestic disinfectant, new and safer antiviral disinfectants are urgently needed. Lysozyme is a natural antibacterial agent widespread in nature and widely used in healthcare and food industry because of is recognized safety. Recently, it has been shown that thermally denatured lysozyme has the ability to kill murine norovirus and hepatitis A virus. In our study, we also demonstrated that heat-denatured lysozyme (HDLz) had an antiviral effect against H1N1 influenza A virus, and we optimized its antiviral activities by testing different heating denaturation conditions, to generalize this property, using pseudotype virus neutralization assay, we found that HDLz can also inhibit the entry of H5N1, H5N6, and H7N1 avian influenza viruses as well as SARS-CoV and SARS-CoV-2 particles in cell with IC50 at the ng/mL range. Finally, using western blot analysis, we provide evidence that HDLz polymerization correlates with antiviral effect, which may be a precious possible quality control test. Altogether, our data support HDLz as a powerful anti-respiratory virus disinfectant as a sole or additive of current disinfectants to reduce concentration of toxic component.
Collapse
Affiliation(s)
- Suqiong Huang
- Chongqing Research Center for Pharmaceutical Engineering, School of Pharmacy, Chongqing Medical University, Yuzhong District, No. 1 Yixueyuan Road, Chongqing, 400016 People’s Republic of China
- Department of Pharmacy, Sichuan Provincial People’s Hospital Qionglai Hospital, Medical Center Hospital of Qionglai City, No. 172 Xinglin Road, Qionglai City, Chengdu, Sichuan Province 611530 People’s Republic of China
| | - Zhenghua Wu
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080 People’s Republic of China
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240 People’s Republic of China
| | - Bingjie Zhou
- University of CAS, Beijing, 101408 China
- CAS Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai CAS, 320 Yueyang Road, Shanghai, 200031 China
| | - Xinhui Jiang
- Chongqing Research Center for Pharmaceutical Engineering, School of Pharmacy, Chongqing Medical University, Yuzhong District, No. 1 Yixueyuan Road, Chongqing, 400016 People’s Republic of China
| | - Dimitri Lavillette
- CAS Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai CAS, 320 Yueyang Road, Shanghai, 200031 China
- Pasteurien College, Soochow University, Jiangsu, 215006 China
| | - Guorong Fan
- Chongqing Research Center for Pharmaceutical Engineering, School of Pharmacy, Chongqing Medical University, Yuzhong District, No. 1 Yixueyuan Road, Chongqing, 400016 People’s Republic of China
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080 People’s Republic of China
| |
Collapse
|
12
|
Wu G, Lorraine MM, Goharriz H, Amaya-Cuesta J, Fooks AR, Banyard AC. A simplified method for measuring neutralising antibodies against rabies virus. J Virol Methods 2023:114769. [PMID: 37391076 DOI: 10.1016/j.jviromet.2023.114769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/28/2023] [Revised: 06/21/2023] [Accepted: 06/26/2023] [Indexed: 07/02/2023]
Abstract
Rabies virus (RABV) causes a fatal encephalitis that can be prevented through timely vaccination. The levels of virus neutralising antibodies against rabies virus induced by vaccination can be measured using the fluorescent antibody virus neutralisation (FAVN) test. Following incubation of live virus with sera, this method involves the fixation of cell monolayers and staining of rabies virus-specific antigen using fluorescein isothiocyanate (FITC) -conjugated antibody to enable visualisation of rabies virus antigen using a fluorescence microscope. To simplify this procedure, a fluorescent recombinant rabies virus was constructed using reverse genetics by inserting the gene for the mCherry fluorescent protein in front of the ribonucleoprotein gene of the SAD B-19 genome and replacing its glycoprotein with that of the Challenge Virus Standard (CVS)-11 RABV strain to ensure antigenic authenticity with the FAVN. This new recombinant virus (termed mCCCG) expressed the mCherry protein to high levels enabling direct observation of infected cells. In vitro growth kinetics of mCCCG were indistinguishable from that of CVS-11. The stability of the recombinant virus was assessed by sequencing several passages of the rescued virus and only minor changes were detected. Comparative assessment of the virus neutralisation test using mCherry producing virus (NTmCV) against the FAVN demonstrated that test results were equivalent to each other; therefore, mCCCG can be used as an alternative to CVS-11 for measuring antibody titres against the rabies virus. The use of NTmCV removes the need for expensive antibody conjugates and significantly reduces assay time. This would be particularly beneficial for RABV serological assessment in resource limited settings. Moreover, the reading of the plates can be automatically using a cell imaging reader.
Collapse
Affiliation(s)
- Guanghui Wu
- Viral Zoonoses Group, Animal and Plant Health Agency (APHA), Weybridge, Surrey, KT15 3NB, UK.
| | - M McElhinney Lorraine
- Viral Zoonoses Group, Animal and Plant Health Agency (APHA), Weybridge, Surrey, KT15 3NB, UK
| | - Hooman Goharriz
- Viral Zoonoses Group, Animal and Plant Health Agency (APHA), Weybridge, Surrey, KT15 3NB, UK
| | - Joan Amaya-Cuesta
- Viral Zoonoses Group, Animal and Plant Health Agency (APHA), Weybridge, Surrey, KT15 3NB, UK
| | - Anthony R Fooks
- Viral Zoonoses Group, Animal and Plant Health Agency (APHA), Weybridge, Surrey, KT15 3NB, UK
| | - Ashley C Banyard
- Viral Zoonoses Group, Animal and Plant Health Agency (APHA), Weybridge, Surrey, KT15 3NB, UK
| |
Collapse
|
13
|
Chen C, Song X, Yu Y, Wang X, Xu H, Ji W, Ma J, Zhao C, Feng S, Wang Y, Su XD, Wang W. Aptamer-based nanointerferometer enables amplification-free ultrasensitive detection and differentiation of SARS-CoV-2 variants. Anal Chim Acta 2023; 1260:341207. [PMID: 37121656 PMCID: PMC10085716 DOI: 10.1016/j.aca.2023.341207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/01/2022] [Revised: 03/07/2023] [Accepted: 04/10/2023] [Indexed: 05/02/2023]
Abstract
The state-of-the-art SARS-CoV-2 detection methods include qRT-PCR and antibody-based lateral flow assay (LFA) point-of-care tests. Despite the high sensitivity and selectivity, qRT-PCR is slow, expensive and needs well-trained operators. On the other extreme, LFA suffers from low sensitivity albeit its fast detection speed, low detection cost and ease of use. Therefore, the continuing COVID-19 pandemic calls for a SARS-CoV-2 detection method that is rapid, convenient and cost-effective without compromise in sensitivity. Here we provide a proof-of-principle demonstration of an optimized aptamer-based nanointerferometer that enables rapid and amplification-free detection of SARS-CoV-2 spike protein-coated pseudovirus directly from human saliva with the limit of detection (LOD) of about 400 copies per mL. This LOD is on par with that of qRT-PCR, making it 1000 to 100,000-fold more sensitive than commercial LFA tests. Using various combinations of negative selections during the screens for the aptamer targeting the receptor binding domain of the spike protein of SARS-CoV-2, we isolated two aptamers that can distinguish the Omicron and Delta variants. Integrating these two aptamers with LFA strips or the nanointerferometer sensors allows both detection and differentiation of the Omicron and Delta variants which has the potential to realize rapid triage of patients infected different SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Changtian Chen
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, 100871, China; Center for Life Sciences, Beijing, 100871, China
| | - Xiaohui Song
- State Key Laboratory of Protein and Plant Gene Research, And Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, 100871, China
| | - Yuanling Yu
- Changping Laboratory, Yard 28, Science Park Road, Changping District, Beijing, China
| | - Xingwei Wang
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, 100871, China
| | - Hua Xu
- State Key Laboratory of Protein and Plant Gene Research, And Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, 100871, China
| | - Weiwei Ji
- College of Physics and Electronic Science, Hubei Normal University, Hubei, 435002, China
| | - Jingchen Ma
- School of Materials and Energy, Guangdong University of Technology, Guangzhou, 510006, China
| | - Chenyan Zhao
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, 102629, China
| | - Silu Feng
- School of Integrated Circuits, Guangdong University of Technology, Guangzhou, 510006, China.
| | - Youchun Wang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, 102629, China; Changping Laboratory, Yard 28, Science Park Road, Changping District, Beijing, China.
| | - Xiao-Dong Su
- State Key Laboratory of Protein and Plant Gene Research, And Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, 100871, China.
| | - Wei Wang
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, 100871, China; Center for Life Sciences, Beijing, 100871, China.
| |
Collapse
|
14
|
Luo X, Wang C, Huang Y, Cong S, Tan J, Hou W, Ma F, Zheng L. Establishment of a neutralization assay for Nipah virus using a high-titer pseudovirus system. Biotechnol Lett 2023; 45:489-498. [PMID: 36680637 PMCID: PMC9860241 DOI: 10.1007/s10529-023-03351-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/22/2022] [Revised: 12/22/2022] [Accepted: 01/05/2023] [Indexed: 01/22/2023]
Abstract
OBJECTIVE To construct a high-titer Nipah pseudovirus packaging system using the HIV lentivirus backbone vector and establish a safe neutralization assay for Nipah pseudovirus in biosafety level 2 facilities. METHODS Nipah virus (NiV) fusion protein (F) and glycoprotein (G) recombinant expression plasmids, psPAX2, and pLenti CMV Puro LUC (w168-1) were transiently transfected into 293T cells for 72 h for the generation of a NiV pseudovirus. The neutralization ability of Nipah virus F and G protein antibodies was assessed using the pseudovirus. RESULTS A NiV pseudovirus was constructed using 293T cells. The ideal mass ratio of plasmid psPAX2: w168-1: F: G for transfection was determined to be 4:4:1:1. The specificity of recombinant F and G protein expression was indicated by indirect immunofluorescence and western blotting. The pseudovirus particles showed obvious spikes under a transmission electron microscope. The NiV pseudovirus titer was 4.73 × 105 median tissue culture infective dose per mL, and the pseudovirus could be effectively neutralized by polyclonal antibodies specifically targeting the F and G proteins respectively. CONCLUSIONS A NiV pseudovirus was successfully generated using HIV vector systems, and was used as a platform for a safe and reliable pseudovirus-based neutralizing assay that can be performed in biosafety level 2 facilities.
Collapse
Affiliation(s)
- Xiaoyi Luo
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, China CDC, Beijing, 100052, China
- Department of Toxicology, School of Public Health, China Medical University, Shenyang, 110122, China
| | - Chao Wang
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, China CDC, Beijing, 100052, China
| | - Yiman Huang
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, China CDC, Beijing, 100052, China
| | - Shanshan Cong
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, China CDC, Beijing, 100052, China
- Department of Toxicology, School of Public Health, China Medical University, Shenyang, 110122, China
| | - Jingjing Tan
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, China CDC, Beijing, 100052, China
- Department of Toxicology, School of Public Health, China Medical University, Shenyang, 110122, China
| | - Wenzhe Hou
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, China CDC, Beijing, 100052, China
| | - Fenlian Ma
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, China CDC, Beijing, 100052, China.
| | - Lishu Zheng
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, China CDC, Beijing, 100052, China.
| |
Collapse
|
15
|
Pseudotyped Viruses for Lyssavirus. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1407:191-208. [PMID: 36920698 DOI: 10.1007/978-981-99-0113-5_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 03/16/2023]
Abstract
Lyssaviruses, which belong to the family Rhabdoviridae, are enveloped and bullet-shaped ssRNA viruses with genetic diversity. All members of Lyssavirus genus are known to infect warm-blooded animals and cause the fatal disease rabies. The rabies virus (RABV) in lyssavirus is the major pathogen to cause fatal rabies. The pseudotyped RABV is constructed to study the biological functions of G protein and evaluation of anti-RABV products including vaccine-induced antisera, rabies immunoglobulins (RIG), neutralizing mAbs, and other antiviral inhibitors. In this chapter, we focus on RABV as a representative and describe the construction of RABV G protein bearing pseudotyped virus and its applications. Other non-RABV lyssaviruses are also included.
Collapse
|
16
|
Assays Based on Pseudotyped Viruses. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1407:29-44. [PMID: 36920690 DOI: 10.1007/978-981-99-0113-5_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 03/16/2023]
Abstract
Pseudotyped viruses are more and more widely used in virus research and the evaluation of antiviral products because of their high safety, simple operation, high accessibility, ease in achieving standardization, and high throughput. The development of measures based on pseudotyped virus is closely related to the characteristics of viruses, and it is also necessary to follow the principles of assay development. Only in the process of method development, where the key parameters that affect the results are systematically optimized and the preliminary established method is fully validated, can the accuracy, reliability, and repeatability of the test results be ensured. Only the method established on this basis can be transferred to different laboratories and make the results of different laboratories comparable. This paper summarizes the specific aspects and general principles in the development of assays based on pseudotyped virus, which is of reference value for the development of similar methods.
Collapse
|
17
|
Application of Pseudotyped Viruses. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1407:45-60. [PMID: 36920691 DOI: 10.1007/978-981-99-0113-5_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 03/16/2023]
Abstract
Highly pathogenic emerging and reemerging viruses have serious public health and socioeconomic implications. Although conventional live virus research methods can more reliably investigate disease pathogenicity and evaluate antiviral products, they usually depend on high-level biosafety laboratories and skilled researchers; these requirements hinder in vitro assessments of efficacy, as well as efforts to test vaccines and antibody drugs. In contrast, pseudotyped viruses (i.e., single-round infectious viruses that mimic the membrane structures of various live viruses) are widely used in studies of highly pathogenic viruses because they can be handled in biosafety level 2 facilities. This chapter provides a concise overview of various aspects of pseudotyped virus technologies, including (1) exploration of the mechanisms of viral infection; (2) evaluation of the efficacies of vaccines and monoclonal antibodies based on pseudovirion-based neutralization assay; (3) assessment of antiviral agents (i.e., antibody-based drugs and inhibitors); (4) establishment of animal models of pseudotyped virus infection in vivo; (5) investigation of the evolution, infectivity, and antigenicity of viral variants and viral glycosylation; and (6) prediction of antibody-dependent cell-mediated cytotoxic activity.
Collapse
|
18
|
Wang Y, Zhou Z, Wu X, Li T, Wu J, Cai M, Nie J, Wang W, Cui Z. Pseudotyped Viruses. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1407:1-27. [PMID: 36920689 DOI: 10.1007/978-981-99-0113-5_1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 03/16/2023]
Abstract
Pseudotyped viruses have been constructed for many viruses. They can mimic the authentic virus and have many advantages compared to authentic viruses. Thus, they have been widely used as a surrogate of authentic virus for viral function analysis, detection of neutralizing antibodies, screening viral entry inhibitors, and others. This chapter reviewed the progress in the field of pseudotyped viruses in general, including the definition and the advantages of pseudotyped viruses, their potential usage, different strategies or vectors used for the construction of pseudotyped viruses, and factors that affect the construction of pseudotyped viruses.
Collapse
Affiliation(s)
- Youchun Wang
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
- Institute of Medical Biology, Chinese Academy of Medicine Sciences & Peking Union Medical College, Kunming, China.
| | - Zehua Zhou
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC) and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Beijing, China
| | - Xi Wu
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC) and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Beijing, China
| | - Tao Li
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC) and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Beijing, China
| | - Jiajing Wu
- Beijing Yunling Biotechnology Co., Ltd., Beijing, China
| | - Meina Cai
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC) and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Beijing, China
| | - Jianhui Nie
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC) and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Beijing, China
| | - Wenbo Wang
- Division of Monoclonal Antibody Products, National Institutes for Food and Drug Control (NIFDC) and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Beijing, China
| | - Zhimin Cui
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC) and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Beijing, China
| |
Collapse
|
19
|
Gao Z, Li T, Han J, Feng S, Li L, Jiang Y, Xu Z, Hao P, Chen J, Hao J, Xu P, Tian M, Jin N, Huang W, Li C. Assessment of the immunogenicity and protection of a Nipah virus soluble G vaccine candidate in mice and pigs. Front Microbiol 2022; 13:1031523. [PMID: 36274696 PMCID: PMC9583134 DOI: 10.3389/fmicb.2022.1031523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/30/2022] [Accepted: 09/21/2022] [Indexed: 11/13/2022] Open
Abstract
Nipah virus (NiV) is a newly emerged extremely dangerous zoonotic pathogen highly fatal to humans. Currently, no approved vaccine is available against NiV. This study employed a mammalian eukaryotic system to express NiV soluble G glycoprotein (NiV-sG), using CpG oligodeoxynucleotides (CpG)/Aluminum salt (Alum) as adjuvants to obtain a recombinant subunit vaccine candidate. We also evaluated the immunogenicity and efficacy of the protein in mice and pigs. The results showed that humoral and cellular immune responses were induced in all the vaccination groups in two animal models. The levels of specific and neutralizing antibodies and the proliferation levels of T helper(Th) cells were significantly higher than those in the control group. The protective efficacy of the subunit vaccines evaluated in the pseudovirus in vivo infection mouse model strongly suggested that this vaccine could provide protective immunity against NiV. A neoadjuvant (HTa) based on liposomes and cholera toxin combined with CpG/Alum was exploited and evaluated in mice. The neoadjuvant group showed a more protective efficacy than the CpG/Alum group. The aforementioned results indicated that the subunit vaccine could be used as a promising candidate vaccine for preventing Nipah virus infection.
Collapse
Affiliation(s)
- Zihan Gao
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Institute of Veterinary Medicine, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Tao Li
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC) and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Beijing, China
| | - Jicheng Han
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Institute of Veterinary Medicine, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Sheng Feng
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Institute of Veterinary Medicine, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Letian Li
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Institute of Veterinary Medicine, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Yuhang Jiang
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Institute of Veterinary Medicine, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Zhiqiang Xu
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Institute of Veterinary Medicine, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Pengfei Hao
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Institute of Veterinary Medicine, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Jing Chen
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Institute of Veterinary Medicine, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Jiayi Hao
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Institute of Veterinary Medicine, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Peng Xu
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Institute of Veterinary Medicine, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Mingyao Tian
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Institute of Veterinary Medicine, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Ningyi Jin
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Institute of Veterinary Medicine, Chinese Academy of Agricultural Sciences, Changchun, China
- *Correspondence: Ningyi Jin,
| | - Weijin Huang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC) and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Beijing, China
- Weijin Huang,
| | - Chang Li
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Institute of Veterinary Medicine, Chinese Academy of Agricultural Sciences, Changchun, China
- Chang Li,
| |
Collapse
|
20
|
Long-Term Infection and Pathogenesis in a Novel Mouse Model of Human Respiratory Syncytial Virus. Viruses 2022; 14:v14081740. [PMID: 36016362 PMCID: PMC9415064 DOI: 10.3390/v14081740] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/05/2022] [Revised: 07/30/2022] [Accepted: 08/03/2022] [Indexed: 11/24/2022] Open
Abstract
Intensive efforts have been made to develop models of hRSV infection or disease using various animals. However, the limitations such as semi-permissiveness and short duration of infection have impeded their applications in both the pathogenesis of hRSV and therapeutics development. Here, we present a mouse model based on a Rag2 gene knockout using CRISPR/Cas9 technology. Rag2−/− mice sustained high viral loads upon intranasal inoculation with hRSV. The average peak titer rapidly reached 1 × 109.8 copies/g and 1c106 TCID50 in nasal cavity, as well as 1 × 108 copies/g and 1 × 105 TCID50 in the lungs up to 5 weeks. Mild interstitial pneumonia, severe bronchopneumonia, elevated cytokines and NK cells were seen in Rag2−/− mice. A humanized monoclonal antibody showed strong antiviral activity in this animal model, implying that Rag2−/− mice that support long-term stable infection are a useful tool for studying the transmission and pathogenesis of human RSV, as well as evaluating therapeutics.
Collapse
|
21
|
Zhang H, Deng T, Fang Q, Li S, Gao S, Jiang W, Chen G, Yu K, Zhou L, Li T, Zheng Q, Yu H, Li S, Xia N, Gu Y. Endodomain truncation of the HIV-1 envelope protein improves the packaging efficiency of pseudoviruses. Virology 2022; 574:1-8. [PMID: 35858511 DOI: 10.1016/j.virol.2022.07.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/27/2021] [Revised: 07/03/2022] [Accepted: 07/08/2022] [Indexed: 10/17/2022]
Abstract
HIV-1 remains one of the most devastating infectious pathogens without available vaccines. A valid neutralization assay using multiple representative virus strains is prerequisite for antibody response analysis in HIV-1 vaccine development, where HIV pseudoviruses (PsVs) commonly serve as surrogate agents for the authentic HIV, offering a safer manipulation in Biosafety Level 2+. However, PsV production is of low efficiency and is unstable in this field. Here, we optimize PsV production conditions via the use of alternative host cells, packaging ratios and gene truncation. We show that a 153-aa truncation of the endodomain substantially enhances the packaging efficiency of HIV PsVs, providing 4 to 25 times higher infection titers than the full-length Env. Further, we obtained a robust HIV-1 PsV panel covering 12 representative global strains for neutralization assay testing. This work sheds light on how to optimize HIV PsV packaging, and provides functional insight into the cytoplasmic domain of HIV-1.
Collapse
Affiliation(s)
- Hui Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, Fujian, 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, Fujian, 361102, China
| | - Tingting Deng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, Fujian, 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, Fujian, 361102, China
| | - Qianjiao Fang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, Fujian, 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, Fujian, 361102, China
| | - Shaoyong Li
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, Fujian, 361102, China
| | - Shuangquan Gao
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, Fujian, 361102, China
| | - Wenling Jiang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, Fujian, 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, Fujian, 361102, China
| | - Gege Chen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, Fujian, 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, Fujian, 361102, China
| | - Kunyu Yu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, Fujian, 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, Fujian, 361102, China
| | - Lizhi Zhou
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, Fujian, 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, Fujian, 361102, China
| | - Tingting Li
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, Fujian, 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, Fujian, 361102, China
| | - Qingbing Zheng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, Fujian, 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, Fujian, 361102, China
| | - Hai Yu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, Fujian, 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, Fujian, 361102, China
| | - Shaowei Li
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, Fujian, 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, Fujian, 361102, China
| | - Ningshao Xia
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, Fujian, 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, Fujian, 361102, China; The Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen, Fujian, 361102, China
| | - Ying Gu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, Fujian, 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, Fujian, 361102, China.
| |
Collapse
|
22
|
Schöler L, Le-Trilling VTK, Dittmer U, Fiedler M, Trilling M. Establishment and clinical validation of an in-cell-ELISA-based assay for the rapid quantification of Rabies lyssavirus neutralizing antibodies. PLoS Negl Trop Dis 2022; 16:e0010425. [PMID: 35536867 PMCID: PMC9159627 DOI: 10.1371/journal.pntd.0010425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/08/2021] [Revised: 06/01/2022] [Accepted: 04/18/2022] [Indexed: 11/19/2022] Open
Abstract
Neutralizing antibodies (nAbs) prevent the entry of viruses into permissive cells. Since nAbs represent correlates of protection against the Rabies lyssavirus, the presence of sufficient nAbs indicates effective vaccination. Accordingly, Rabies lyssavirus-specific nAb titers need to be determined in routine diagnostics to identify individuals being at risk of Rabies lyssavirus infections due to insufficient immunity. The current gold standard for the quantification of Rabies lyssavirus-specific nAbs is the rapid fluorescent focus inhibition test (RFFIT). However, RFFITs are expensive and labor-intensive since multiple microplate wells must be evaluated one-by-one by trained personnel through microscopic inspection, which limits the number of samples that can be processed. To overcome this disadvantage, we established a novel assay for Rabies lyssavirus-specific nAbs relying on an in-cell-ELISA (icELISA)-based neutralization test (icNT). The icNT differs from the RFFIT in the readout phase, and can be automatically quantified in minutes using broadly available microplate readers. During the establishment, icNT parameters such as antibody concentrations, permeabilization procedures, blocking reagents, infectious doses, and the duration of infection were optimized. Afterwards, a dose-dependent detection of Rabies lyssavirus neutralization was demonstrated using the WHO Standard Rabies Immunoglobulin reference. A panel of 200 sera with known RFFIT titers revealed very good sensitivity and specificity of the icNT. Furthermore, the icNT showed very good intra- and inter-assay precision. By recognizing Rabies lyssavirus-specific antigens, the assay can be applied immediately to automatically quantify the concentration of Rabies lyssavirus nAbs in routine diagnostics or for various basic research questions such as screening for antiviral compounds.
Collapse
Affiliation(s)
- Lara Schöler
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | | | - Ulf Dittmer
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Melanie Fiedler
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Mirko Trilling
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
23
|
Zhang S, Yan F, Liu D, Li E, Feng N, Xu S, Wang H, Gao Y, Yang S, Zhao Y, Xia X. Bacterium-Like Particles Displaying the Rift Valley Fever Virus Gn Head Protein Induces Efficacious Immune Responses in Immunized Mice. Front Microbiol 2022; 13:799942. [PMID: 35369468 PMCID: PMC8969503 DOI: 10.3389/fmicb.2022.799942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/22/2021] [Accepted: 02/03/2022] [Indexed: 11/13/2022] Open
Abstract
Rift Valley fever virus (RVFV), a mosquito-borne zoonotic phlebovirus, causes serious disease in humans and ruminants. According to the World Health Organization, Rift Valley fever is classified as a priority disease, and as such, vaccine development is of high priority due to the lack of licensed vaccines. In this study, a bacterium-like particle vaccine (BLP), RVFV-BLPs, is constructed. A novel display system is described, which is based on non-living and non-genetically modified Gram-positive bacterial cells, designated as Gram-positive enhancer matrix (GEM). The RVFV Gn head protein was displayed on the surface of GEM by co-expression with the peptidoglycan-binding domain (protein anchor) at the C-terminus. We determined that the RVFV Gn head-PA fusion protein was successfully displayed on the GEM. Mice immunized with RVFV-BLPs produced humoral and cellular immunity. Interestingly, comparing the production of RVFV Gn head-specific IgG and its subtype by vaccinating with different antigen doses of the RVFV-BLPs determined that the RVFV-BLPs (50 μg) group showed a greater effect than the other two groups. More importantly, antibodies produced by mice immunized with RVFV-BLPs (50 μg) exhibited potent neutralizing activity against RVFV pseudovirus. RVFV-BLPs (50 μg) also could induce IFN-γ and IL-4 in immunized mice; these mice generated memory cells among the proliferating T cell population after immunization with RVFV-BLPs with effector memory T cells as the major population, which means that RVFV-BLPs is an effective vaccine to establish a long-lived population of memory T cells. The findings suggest that the novel RVFV-BLPs subunit vaccine has the potential to be considered a safe and effective candidate vaccine against RVFV infection.
Collapse
Affiliation(s)
- Shengnan Zhang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Feihu Yan
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Dongping Liu
- The Nanjing Unicorn Academy of Innovation, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Nanjing, China
| | - Entao Li
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Na Feng
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Shengnan Xu
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Hualei Wang
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yuwei Gao
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Songtao Yang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
- *Correspondence: Songtao Yang,
| | - Yongkun Zhao
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
- Yongkun Zhao,
| | - Xianzhu Xia
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
- Xianzhu Xia,
| |
Collapse
|
24
|
Palmer P, Del Rosario JMM, da Costa KAS, Carnell GW, Huang CQ, Heeney JL, Temperton NJ, Wells DA. AutoPlate: Rapid Dose-Response Curve Analysis for Biological Assays. Front Immunol 2022; 12:681636. [PMID: 35222351 PMCID: PMC8866857 DOI: 10.3389/fimmu.2021.681636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/16/2021] [Accepted: 12/29/2021] [Indexed: 11/13/2022] Open
Abstract
The emergence of COVID-19 has emphasised that biological assay data must be analysed quickly to develop safe, effective and timely vaccines/therapeutics. For viruses such as SARS-CoV-2, the primary way of measuring immune correlates of protection is through assays such as the pseudotype microneutralisation (pMN) assay, thanks to its safety and versatility. However, despite the presence of existing tools for data analysis such as PRISM and R the analysis of these assays remains cumbersome and time-consuming. We introduce an open-source R Shiny web application and R library (AutoPlate) to accelerate data analysis of dose-response curve immunoassays. Using example data from influenza studies, we show that AutoPlate improves on available analysis software in terms of ease of use, flexibility and speed. AutoPlate (https://philpalmer.shinyapps.io/AutoPlate/) is a tool for the use of laboratories and wider scientific community to accelerate the analysis of biological assays in the development of viral vaccines and therapeutics.
Collapse
Affiliation(s)
- Phil Palmer
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Joanne Marie M Del Rosario
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent, Chatham, United Kingdom.,Department of Physical Sciences and Mathematics, College of Arts and Sciences, University of the Philippines Manila, Manila, Philippines
| | - Kelly A S da Costa
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent, Chatham, United Kingdom
| | - George W Carnell
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Chloe Q Huang
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Jonathan L Heeney
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom.,DIOSynVax, University of Cambridge, Cambridge, United Kingdom
| | - Nigel J Temperton
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent, Chatham, United Kingdom
| | - David A Wells
- DIOSynVax, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
25
|
Su K, Xue J, Shan X, Ye H, Zhang L, Tan S, Shao J, Shi Y, Wang Z, Zhang L. Review of Detection and Quantification of Rabies Virus Antibodies. Viral Immunol 2021; 34:522-530. [PMID: 34550784 DOI: 10.1089/vim.2020.0317] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 02/07/2023] Open
Abstract
Rabies is an almost invariably fatal disease. According to the World Health Organization (WHO), rabies virus neutralizing antibody (RVNA) titers of ≥0.5 IU/mL are considered adequate for rabies protection. Therefore, detection and quantification of RABV antibodies are important. Many methods have been developed for detecting RABV antibodies. In the present study, we reviewed several methods of detecting RABV antibodies in human and animal samples and evaluated and compared their performance. Of 34 methods, 5 demonstrated unsatisfactory sensitivity or specificity. The others exhibited sensitivity and specificity of ≥75%. The correlation coefficient for five of eight methods was >0.8. The Bland-Altman mean bias of five of five methods was <±2.0. The kappa values of 25 of 28 methods were higher than 0.4, demonstrating at least moderate agreement. Analysis of the performance of these methods emphasized that any new technology should be considered carefully and objectively before being used as an appropriate and applicable alternative.
Collapse
Affiliation(s)
- Kewen Su
- Department of Sanitary Analysis, Hangzhou Hospital for the Prevention and Treatment of Occupational Disease, Hangzhou, China
| | - Jian Xue
- School of Public Health and Management, Chongqing Medical University, Chongqing, China
| | - Xiaoyue Shan
- Department of Sanitary Analysis, Hangzhou Hospital for the Prevention and Treatment of Occupational Disease, Hangzhou, China
| | - Haipeng Ye
- Department of Sanitary Analysis, Hangzhou Hospital for the Prevention and Treatment of Occupational Disease, Hangzhou, China
| | - Ling Zhang
- Department of Sanitary Analysis, Hangzhou Hospital for the Prevention and Treatment of Occupational Disease, Hangzhou, China
| | - Siwei Tan
- Department of Sanitary Analysis, Hangzhou Hospital for the Prevention and Treatment of Occupational Disease, Hangzhou, China
| | - Ji Shao
- Department of Sanitary Analysis, Hangzhou Hospital for the Prevention and Treatment of Occupational Disease, Hangzhou, China
| | - Yanpeng Shi
- Department of Sanitary Analysis, Hangzhou Hospital for the Prevention and Treatment of Occupational Disease, Hangzhou, China
| | - Zhe Wang
- Institute for Communicable Disease Control and Prevention, Hangzhou Center for Disease Control and Prevention, Hangzhou, China
| | - Lei Zhang
- Department of Sanitary Analysis, Hangzhou Hospital for the Prevention and Treatment of Occupational Disease, Hangzhou, China
| |
Collapse
|
26
|
Rodriguez MC, Fontana D, Garay E, Prieto C. Detection and quantification of anti-rabies glycoprotein antibodies: current state and perspectives. Appl Microbiol Biotechnol 2021; 105:6547-6557. [PMID: 34448897 PMCID: PMC8390338 DOI: 10.1007/s00253-021-11515-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/26/2021] [Revised: 08/06/2021] [Accepted: 08/10/2021] [Indexed: 12/25/2022]
Abstract
Rabies is an ancient fatal disease with no other available treatment than post-exposure vaccination, where the bite of infected animals, mainly dogs, is the leading cause of its transmission to human beings. In this context, global vaccination campaigns of companion animals, as well as wildlife reservoirs vaccination, are key factors to achieve the "Zero by 30" plan that pursues the eradication of dog-mediated human rabies by 2030. Rabies virus-neutralizing antibodies (VNAs) play an essential role in the disease protection, as it correlates with an adequate immune response and allows evaluating pre- or post-exposure prophylaxis efficacy. Hence, counting with reliable, accurate, and robust serological tests is of paramount importance. Currently, RFFIT and FAVN are the gold standard VNAs tests recommended by both the WHO and the OIE. Despite these methodologies are efficient and widely used, they present several drawbacks, as they are less easily to standardize and require the use of live rabies virus, containment facilities, and skilled professionals. Thus, in this review, we describe the state-of-the-art of alternative analytical methodologies currently available for rabies serology, with novel approaches based on pseudotyped recombinant viruses and emphasizing in the antigen binding methodologies that detect and quantify antibodies against the rabies glycoprotein. We discussed the wide range of assays that are interesting tools for a faster measurement of anti-rabies glycoprotein antibodies and, in some cases, less complex and more versatile than the gold standard methods. Finally, we discussed the key issues during the design and optimization steps of ELISA assays, highlighting the importance of validation and standardization procedures to improve rabies serology tests and, as a consequence, their results. KEY POINTS: • An exhaustive revision of rabies serology testing was made. • No rabies serology assay can be thought as better than others for all intents and purposes. • The validation procedure guarantees reliable and consistent results among the globe.
Collapse
Affiliation(s)
- Maria Celeste Rodriguez
- UNL, CONICET, FBCB (School of Biochemistry and Biological Sciences), CBL (Biotechnological Center of Litoral), Cell Culture Laboratory, Ciudad Universitaria, Ruta Nacional 168 - Km 472.4 - C.C. 242 - (S3000ZAA), Santa Fe, Argentina
| | - Diego Fontana
- UNL, CONICET, FBCB (School of Biochemistry and Biological Sciences), CBL (Biotechnological Center of Litoral), Cell Culture Laboratory, Ciudad Universitaria, Ruta Nacional 168 - Km 472.4 - C.C. 242 - (S3000ZAA), Santa Fe, Argentina.
| | - Ernesto Garay
- UNL, CONICET, FBCB (School of Biochemistry and Biological Sciences), CBL (Biotechnological Center of Litoral), Cell Culture Laboratory, Ciudad Universitaria, Ruta Nacional 168 - Km 472.4 - C.C. 242 - (S3000ZAA), Santa Fe, Argentina
| | - Claudio Prieto
- UNL, FBCB (School of Biochemistry and Biological Sciences), CBL (Biotechnological Center of Litoral), Biotechnological Development Laboratory, Ciudad Universitaria, Ruta Nacional 168 - Km 472.4 - C.C. 242 - (S3000ZAA), Santa Fe, Argentina
| |
Collapse
|
27
|
Kalkeri R, Cai Z, Lin S, Farmer J, Kuzmichev YV, Koide F. SARS-CoV-2 Spike Pseudoviruses: A Useful Tool to Study Virus Entry and Address Emerging Neutralization Escape Phenotypes. Microorganisms 2021; 9:1744. [PMID: 34442823 PMCID: PMC8398529 DOI: 10.3390/microorganisms9081744] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/15/2021] [Revised: 08/06/2021] [Accepted: 08/06/2021] [Indexed: 12/12/2022] Open
Abstract
SARS-CoV-2 genetic variants are emerging around the globe. Unfortunately, several SARS-CoV-2 variants, especially variants of concern (VOCs), are less susceptible to neutralization by the convalescent and post-vaccination sera, raising concerns of increased disease transmissibility and severity. Recent data suggests that SARS-CoV-2 neutralizing antibody levels are a reliable correlate of vaccine-mediated protection. However, currently used BSL3-based virus micro-neutralization (MN) assays are more laborious, time-consuming, and expensive, underscoring the need for BSL2-based, cost-effective neutralization assays against SARS-CoV-2 variants. In light of this unmet need, we have developed a BSL-2 pseudovirus-based neutralization assay (PBNA) in cells expressing the human angiotensin-converting enzyme-2 (hACE2) receptor for SARS-CoV-2. The assay is reproducible (R2 = 0.96), demonstrates a good dynamic range and high sensitivity. Our data suggest that the biological Anti-SARS-CoV-2 research reagents such as NIBSC 20/130 show lower neutralization against B.1.351 SA (South Africa) and B.1.1.7 UK (United Kingdom) VOC, whereas a commercially available monoclonal antibody MM43 retains activity against both these variants. SARS-CoV-2 spike PBNAs for VOCs would be useful tools to measure the neutralization ability of candidate vaccines in both preclinical models and clinical trials and would further help develop effective prophylactic countermeasures against emerging neutralization escape phenotypes.
Collapse
Affiliation(s)
- Raj Kalkeri
- Department of Infectious Disease Research, Drug Development Division, Southern Research, 431 Aviation Way, Frederick, MD 21701, USA; (Z.C.); (S.L.); (Y.V.K.)
| | - Zhaohui Cai
- Department of Infectious Disease Research, Drug Development Division, Southern Research, 431 Aviation Way, Frederick, MD 21701, USA; (Z.C.); (S.L.); (Y.V.K.)
| | - Shuling Lin
- Department of Infectious Disease Research, Drug Development Division, Southern Research, 431 Aviation Way, Frederick, MD 21701, USA; (Z.C.); (S.L.); (Y.V.K.)
| | - John Farmer
- Department of Infectious Disease Research, Drug Development Division, Southern Research, 2000 Ninth Avenue South, Birmingham, AL 35205, USA;
| | - Yury V. Kuzmichev
- Department of Infectious Disease Research, Drug Development Division, Southern Research, 431 Aviation Way, Frederick, MD 21701, USA; (Z.C.); (S.L.); (Y.V.K.)
| | - Fusataka Koide
- Department of Infectious Disease Research, Drug Development Division, Southern Research, 431 Aviation Way, Frederick, MD 21701, USA; (Z.C.); (S.L.); (Y.V.K.)
| |
Collapse
|
28
|
Chen M, Zhang XE. Construction and applications of SARS-CoV-2 pseudoviruses: a mini review. Int J Biol Sci 2021; 17:1574-1580. [PMID: 33907521 PMCID: PMC8071765 DOI: 10.7150/ijbs.59184] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/08/2021] [Accepted: 02/24/2021] [Indexed: 12/20/2022] Open
Abstract
The ongoing coronavirus disease 2019 pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has posed a serious threat to global public health and social stability. There is an urgent need for understanding the nature and infection mechanism of the virus. Owing to its high infectivity and pathogenicity and lack of effective treatments, live SARS-CoV-2 has to be handled in biosafety level 3 laboratories, which has impeded research into SARS-CoV-2 and the development of vaccines and therapeutics. Pseudotyped viruses that lack certain gene sequences of the virulent virus are safer and can be investigated in biosafety level 2 laboratories, providing a useful virological tool for the study of SARS-CoV-2. In this review, we will discuss the construction of SARS-CoV-2 pseudoviruses based on different packaging systems, current applications, limitations, and further explorations.
Collapse
Affiliation(s)
- Minghai Chen
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Xian-En Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.,CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| |
Collapse
|
29
|
Wu J, Cao S, Lei S, Liu Q, Li Y, Yu Y, Xie H, Li Q, Zhao X, Chen R, Huang W, Xiao X, Yu Y, Song D, Li Y, Wang Y. Clofazimine: A Promising Inhibitor of Rabies Virus. Front Pharmacol 2021; 12:598241. [PMID: 33815101 PMCID: PMC8012719 DOI: 10.3389/fphar.2021.598241] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/24/2020] [Accepted: 02/09/2021] [Indexed: 12/25/2022] Open
Abstract
With an almost 100% mortality rate, rabies virus (RABV) infection is a global concern. Limited post-exposure prophylaxis and lack of an effective treatment necessitate novel antiviral therapies against RABV. Here, using a high-throughput screening (HTS) method developed in our lab, 11 candidates with anti-RABV activity were identified from a library of 767 clinical drugs. Clofazimine (CFZ), an anti-leprosy drug, displayed an EC50 of 2.28 μM, and SI over 967 against RABV. Investigations into the underlying mechanisms revealed that CFZ targeted viral membrane fusion at the early stages of virus replication. Moreover, CFZ and Clofazimine salicylates (CFZS) exhibited elevated survival rates in vivo, compared with the positive control T-705. Thus, this study revealed CFZ as a promising drug against RABV infection.
Collapse
Affiliation(s)
- Jiajing Wu
- Department of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control, Beijing, China.,Wuhan Institute of Biological Products, Hubei, China
| | - Shouchun Cao
- Department of Arboviral Vaccine, National Institutes for Food and Drug Control, Beijing, China
| | - Shan Lei
- Department of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control, Beijing, China
| | - Qiang Liu
- Department of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control, Beijing, China
| | - Yinghong Li
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yueyang Yu
- Department of Arboviral Vaccine, National Institutes for Food and Drug Control, Beijing, China
| | - Hui Xie
- Department of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control, Beijing, China
| | - Qianqian Li
- Department of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control, Beijing, China
| | - Xiaoqiang Zhao
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Ruifeng Chen
- Department of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control, Beijing, China
| | - Weijin Huang
- Department of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control, Beijing, China
| | - Xinyue Xiao
- Institute for Reference Standards and Standardization, National Institutes for Food and Drug Control, Beijing, China
| | - Yongxin Yu
- Department of Arboviral Vaccine, National Institutes for Food and Drug Control, Beijing, China
| | - Danqing Song
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yuhua Li
- Department of Arboviral Vaccine, National Institutes for Food and Drug Control, Beijing, China
| | - Youchun Wang
- Department of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control, Beijing, China
| |
Collapse
|
30
|
Pseudotyping of VSV with Ebola virus glycoprotein is superior to HIV-1 for the assessment of neutralising antibodies. Sci Rep 2020; 10:14289. [PMID: 32868837 PMCID: PMC7459353 DOI: 10.1038/s41598-020-71225-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/02/2020] [Accepted: 07/31/2020] [Indexed: 11/17/2022] Open
Abstract
Ebola virus (EBOV) is an enveloped, single-stranded RNA virus that can cause Ebola virus disease (EVD). It is thought that EVD survivors are protected against subsequent infection with EBOV and that neutralising antibodies to the viral surface glycoprotein (GP) are potential correlates of protection. Serological studies are vital to assess neutralising antibodies targeted to EBOV GP; however, handling of EBOV is limited to containment level 4 laboratories. Pseudotyped viruses can be used as alternatives to live viruses, which require high levels of bio-containment, in serological and viral entry assays. However, neutralisation capacity can differ among pseudotyped virus platforms. We evaluated the suitability of EBOV GP pseudotyped human immunodeficiency virus type 1 (HIV-1) and vesicular stomatitis virus (VSV) to measure the neutralising ability of plasma from EVD survivors, when compared to results from a live EBOV neutralisation assay. The sensitivity, specificity and correlation with live EBOV neutralisation were greater for the VSV-based pseudotyped virus system, which is particularly important when evaluating EBOV vaccine responses and immuno-therapeutics. Therefore, the EBOV GP pseudotyped VSV neutralisation assay reported here could be used to provide a better understanding of the putative correlates of protection against EBOV.
Collapse
|
31
|
Younes N, Al-Sadeq DW, AL-Jighefee H, Younes S, Al-Jamal O, Daas HI, Yassine HM, Nasrallah GK. Challenges in Laboratory Diagnosis of the Novel Coronavirus SARS-CoV-2. Viruses 2020; 12:E582. [PMID: 32466458 PMCID: PMC7354519 DOI: 10.3390/v12060582] [Citation(s) in RCA: 254] [Impact Index Per Article: 50.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/27/2020] [Revised: 05/19/2020] [Accepted: 05/20/2020] [Indexed: 12/13/2022] Open
Abstract
The recent outbreak of the Coronavirus disease 2019 (COVID-19) has quickly spread worldwide since its discovery in Wuhan city, China in December 2019. A comprehensive strategy, including surveillance, diagnostics, research, clinical treatment, and development of vaccines, is urgently needed to win the battle against COVID-19. The past three unprecedented outbreaks of emerging human coronavirus infections at the beginning of the 21st century have highlighted the importance of readily available, accurate, and rapid diagnostic technologies to contain emerging and re-emerging pandemics. Real-time reverse transcriptase-polymerase chain reaction (rRT-PCR) based assays performed on respiratory specimens remain the gold standard for COVID-19 diagnostics. However, point-of-care technologies and serologic immunoassays are rapidly emerging with high sensitivity and specificity as well. Even though excellent techniques are available for the diagnosis of symptomatic patients with COVID-19 in well-equipped laboratories; critical gaps still remain in screening asymptomatic people who are in the incubation phase of the virus, as well as in the accurate determination of live viral shedding during convalescence to inform decisions for ending isolation. This review article aims to discuss the currently available laboratory methods and surveillance technologies available for the detection of COVID-19, their performance characteristics and highlight the gaps in current diagnostic capacity, and finally, propose potential solutions. We also summarize the specifications of the majority of the available commercial kits (PCR, EIA, and POC) for laboratory diagnosis of COVID-19.
Collapse
Affiliation(s)
- Nadin Younes
- Biomedical Research Center, Qatar University, P.O. Box 2713 Doha, Qatar; (N.Y.); (D.W.A.-S.); (H.A.-J.); (O.A.-J.); (H.M.Y.)
| | - Duaa W. Al-Sadeq
- Biomedical Research Center, Qatar University, P.O. Box 2713 Doha, Qatar; (N.Y.); (D.W.A.-S.); (H.A.-J.); (O.A.-J.); (H.M.Y.)
- College of Medicine, Member of QU Health, Qatar University, P.O. Box 2713 Doha, Qatar
| | - Hadeel AL-Jighefee
- Biomedical Research Center, Qatar University, P.O. Box 2713 Doha, Qatar; (N.Y.); (D.W.A.-S.); (H.A.-J.); (O.A.-J.); (H.M.Y.)
| | - Salma Younes
- Department of Biomedical Science, College of Health Sciences, Member of QU Health, Qatar University, P.O. Box 2713 Doha, Qatar;
| | - Ola Al-Jamal
- Biomedical Research Center, Qatar University, P.O. Box 2713 Doha, Qatar; (N.Y.); (D.W.A.-S.); (H.A.-J.); (O.A.-J.); (H.M.Y.)
| | - Hanin I. Daas
- College of Dental Medicine, Member of QU Health, Qatar University, P.O. Box 2713 Doha, Qatar;
| | - Hadi. M. Yassine
- Biomedical Research Center, Qatar University, P.O. Box 2713 Doha, Qatar; (N.Y.); (D.W.A.-S.); (H.A.-J.); (O.A.-J.); (H.M.Y.)
- Department of Biomedical Science, College of Health Sciences, Member of QU Health, Qatar University, P.O. Box 2713 Doha, Qatar;
| | - Gheyath K. Nasrallah
- Biomedical Research Center, Qatar University, P.O. Box 2713 Doha, Qatar; (N.Y.); (D.W.A.-S.); (H.A.-J.); (O.A.-J.); (H.M.Y.)
- Department of Biomedical Science, College of Health Sciences, Member of QU Health, Qatar University, P.O. Box 2713 Doha, Qatar;
| |
Collapse
|
32
|
Nie J, Li Q, Wu J, Zhao C, Hao H, Liu H, Zhang L, Nie L, Qin H, Wang M, Lu Q, Li X, Sun Q, Liu J, Fan C, Huang W, Xu M, Wang Y. Establishment and validation of a pseudovirus neutralization assay for SARS-CoV-2. Emerg Microbes Infect 2020; 9:680-686. [PMID: 32207377 PMCID: PMC7144318 DOI: 10.1080/22221751.2020.1743767] [Citation(s) in RCA: 547] [Impact Index Per Article: 109.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/24/2022]
Abstract
Pseudoviruses are useful virological tools because of their safety and versatility, especially for emerging and re-emerging viruses. Due to its high pathogenicity and infectivity and the lack of effective vaccines and therapeutics, live SARS-CoV-2 has to be handled under biosafety level 3 conditions, which has hindered the development of vaccines and therapeutics. Based on a VSV pseudovirus production system, a pseudovirus-based neutralization assay has been developed for evaluating neutralizing antibodies against SARS-CoV-2 in biosafety level 2 facilities. The key parameters for this assay were optimized, including cell types, cell numbers, virus inoculum. When tested against the SARS-CoV-2 pseudovirus, SARS-CoV-2 convalescent patient sera showed high neutralizing potency, which underscore its potential as therapeutics. The limit of detection for this assay was determined as 22.1 and 43.2 for human and mouse serum samples respectively using a panel of 120 negative samples. The cutoff values were set as 30 and 50 for human and mouse serum samples, respectively. This assay showed relatively low coefficient of variations with 15.9% and 16.2% for the intra- and inter-assay analyses respectively. Taken together, we established a robust pseudovirus-based neutralization assay for SARS-CoV-2 and are glad to share pseudoviruses and related protocols with the developers of vaccines or therapeutics to fight against this lethal virus.
Collapse
Affiliation(s)
- Jianhui Nie
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, People's Republic of China
| | - Qianqian Li
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, People's Republic of China.,Graduate School of Peking Union Medical College, Beijing, People's Republic of China
| | - Jiajing Wu
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, People's Republic of China.,Wuhan Institute of Biological Products, Wuhan, People's Republic of China
| | - Chenyan Zhao
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, People's Republic of China
| | - Huan Hao
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, People's Republic of China
| | - Huan Liu
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, People's Republic of China
| | - Li Zhang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, People's Republic of China
| | - Lingling Nie
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, People's Republic of China
| | - Haiyang Qin
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, People's Republic of China
| | - Meng Wang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, People's Republic of China
| | - Qiong Lu
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, People's Republic of China
| | - Xiaoyu Li
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, People's Republic of China
| | - Qiyu Sun
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, People's Republic of China
| | - Junkai Liu
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, People's Republic of China
| | - Changfa Fan
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control, Beijing, People's Republic of China
| | - Weijin Huang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, People's Republic of China
| | - Miao Xu
- Institute for Biological Product Control, National Institutes for Food and Drug Control, Beijing, People's Republic of China
| | - Youchun Wang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, People's Republic of China.,Graduate School of Peking Union Medical College, Beijing, People's Republic of China
| |
Collapse
|
33
|
Wang W, Ma J, Nie J, Li J, Cao S, Wang L, Yu C, Huang W, Li Y, Yu Y, Liang M, Zirkle B, Chen XS, Li X, Kong W, Wang Y. Antigenic variations of recent street rabies virus. Emerg Microbes Infect 2020; 8:1584-1592. [PMID: 31682199 PMCID: PMC6844422 DOI: 10.1080/22221751.2019.1683436] [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] [Academic Contribution Register] [Indexed: 11/04/2022]
Abstract
The genetic and/or antigenic differences between street rabies virus (RABV) and vaccine strains could potentially affect effectiveness of rabies vaccines. As such, it is important to continue monitoring the glycoprotein (G) of the street isolates. All RABVG sequences in public database were retrieved and analysed. Using a pseudovirus system, we investigated 99 naturally occurring mutants for their reactivities to well-characterized neutralizing monoclonal antibodies (mAbs) and vaccine-induced antisera. A divergence in G sequences was found between vaccine strains and recent street isolates, with mutants demonstrating resistance to neutralizing mAbs and vaccine-induced antibodies. Moreover, antigenic variants were observed in a wide range of animal hosts and geographic locations, with most of them emerging since 2010. As the number of antigenic variants has increased in recent years, close monitoring on street isolates should be strengthened.
Collapse
Affiliation(s)
- Wenbo Wang
- College of Life Science, Jilin University, Changchun, People's Republic of China.,Division of Monoclonal Antibody Products, National Institutes for Food and Drug Control (NIFDC), Beijing, People's Republic of China
| | - Jian Ma
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC), Beijing, People's Republic of China
| | - Jianhui Nie
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC), Beijing, People's Republic of China
| | - Jia Li
- Division of Arboviral Vaccine, National Institutes for Food and Drug Control (NIFDC), Beijing, People's Republic of China
| | - Shouchun Cao
- Division of Arboviral Vaccine, National Institutes for Food and Drug Control (NIFDC), Beijing, People's Republic of China
| | - Lan Wang
- Division of Monoclonal Antibody Products, National Institutes for Food and Drug Control (NIFDC), Beijing, People's Republic of China
| | - Chuanfei Yu
- Division of Monoclonal Antibody Products, National Institutes for Food and Drug Control (NIFDC), Beijing, People's Republic of China
| | - Weijin Huang
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC), Beijing, People's Republic of China
| | - Yuhua Li
- Division of Arboviral Vaccine, National Institutes for Food and Drug Control (NIFDC), Beijing, People's Republic of China
| | - Yongxin Yu
- Division of Arboviral Vaccine, National Institutes for Food and Drug Control (NIFDC), Beijing, People's Republic of China
| | - Mifang Liang
- Key Laboratory for Medical Virology, NHFPC, National Institute for Viral Disease Control and Prevention, China CDC, Beijing, People's Republic of China
| | - Brett Zirkle
- Department of Molecular and Computational Biology/Chemistry Department/Norris Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Xiaojiang S Chen
- Department of Molecular and Computational Biology/Chemistry Department/Norris Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Xuguang Li
- Centre for Biologics Evaluation, Biologics and Genetic Therapies Directorate, Health Canada, Canada
| | - Wei Kong
- College of Life Science, Jilin University, Changchun, People's Republic of China
| | - Youchun Wang
- College of Life Science, Jilin University, Changchun, People's Republic of China.,Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC), Beijing, People's Republic of China
| |
Collapse
|
34
|
Abstract
To better evaluate HIV-1 vaccines and therapeutics, the National Institutes for Food and Drug Control of China developed a panel of HIV-1 pseudoviruses including 462 viral strains derived from China, covering the majority of contemporaneous subtypes and circulating recombinant forms. Compared with the standard pseudovirus panels derived from other countries/regions, the Chinese isolates are more susceptible to neutralization by the sera obtained in China, revealing the strain/subtype specificity. Some of these pseudoviruses have already been used for the evaluation of HIV vaccines and drug candidates in Chinese clinical trials. The pseudoviruses panel is widely shared with interested scientists involved in the research and development of vaccines and antiviral drugs against HIV-1 strains prevalent in China.
Collapse
Affiliation(s)
- Jianhui Nie
- Division of HIV/AIDS and Sexually Transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC), Beijing, People's Republic of China
| | - Weijin Huang
- Division of HIV/AIDS and Sexually Transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC), Beijing, People's Republic of China
| | - Qiang Liu
- Division of HIV/AIDS and Sexually Transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC), Beijing, People's Republic of China
| | - Youchun Wang
- Division of HIV/AIDS and Sexually Transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC), Beijing, People's Republic of China
| |
Collapse
|
35
|
Genetically Modified Rabies Virus Vector-Based Rift Valley Fever Virus Vaccine is Safe and Induces Efficacious Immune Responses in Mice. Viruses 2019; 11:v11100919. [PMID: 31597372 PMCID: PMC6832564 DOI: 10.3390/v11100919] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/08/2019] [Revised: 09/29/2019] [Accepted: 09/30/2019] [Indexed: 12/17/2022] Open
Abstract
Rift Valley fever virus (RVFV), which causes Rift Valley fever (RVF), is a mosquito-borne zoonotic pathogen that causes serious morbidity and mortality in livestock and humans. RVF is a World Health Organization (WHO) priority disease and, together with rabies, is a major health burden in Africa. Here, we present the development and characterization of an inactivated recombinant RVFV and rabies virus (RABV) vaccine candidate (rSRV9-eGn). Immunization with rSRV9-eGn stimulated the production of RVFV-specific IgG antibodies and induced humoral and cellular immunity in mice but did not induce the production of neutralizing antibodies. IgG1 and IgG2a were the main isotypes observed by IgG subtype detection, and IgG3 antibodies were not detected. The ratios of IgG1/IgG2a > 1 indicated a Type 2 humoral immune response. An effective vaccine is intended to establish a long-lived population of memory T cells, and mice generated memory cells among the proliferating T cell population after immunization with rSRV9-eGn, with effector memory T cells (TEM) as the major population. Due to the lack of prophylactic treatment experiments, it is impossible to predict whether this vaccine can protect animals from RVFV infection with only high titres of anti-RVFV IgG antibodies and no neutralizing antibodies induced, and thus, protection confirmation needs further verification. However, this RVFV vaccine designed with RABV as the vector provides ideas for the development of vaccines that prevent RVFV and RABV infections.
Collapse
|
36
|
Nie J, Liu L, Wang Q, Chen R, Ning T, Liu Q, Huang W, Wang Y. Nipah pseudovirus system enables evaluation of vaccines in vitro and in vivo using non-BSL-4 facilities. Emerg Microbes Infect 2019; 8:272-281. [PMID: 30866781 PMCID: PMC6455126 DOI: 10.1080/22221751.2019.1571871] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/05/2023]
Abstract
Because of its high infectivity in humans and the lack of effective vaccines, Nipah virus is classified as a category C agent and handling has to be performed under biosafety level 4 conditions in non-endemic countries, which has hindered the development of vaccines. Based on a highly efficient pseudovirus production system using a modified HIV backbone vector, a pseudovirus-based mouse model has been developed for evaluating the efficacy of Nipah vaccines in biosafety level 2 facilities. For the first time, the correlates of protection have been identified in a mouse model. The limited levels of neutralizing antibodies against immunogens fusion protein (F), glycoprotein (G), and combination of F and G (FG) were found to be 148, 275, and 115, respectively, in passive immunization. Relatively lower limited levels of protection of 52, and 170 were observed for immunogens F, and G, respectively, in an active immunization model. Although the minimal levels for protection of neutralizing antibody in passive immunization were slightly higher than those in active immunization, neutralizing antibody played a key role in protection against Nipah virus infection. The immunogens F and G provided similar protection, and the combination of these immunogens did not provide better outcomes. Either immunogen F or G would provide sufficient protection for Nipah vaccine. The Nipah pseudovirus mouse model, which does not involve highly pathogenic virus, has the potential to greatly facilitate the standardization and implementation of an assay to propel the development of NiV vaccines.
Collapse
Affiliation(s)
- Jianhui Nie
- a Division of HIV/AIDS and Sexually Transmitted Virus Vaccines , National Institutes for Food and Drug Control (NIFDC) , Beijing , People's Republic of China
| | - Lin Liu
- a Division of HIV/AIDS and Sexually Transmitted Virus Vaccines , National Institutes for Food and Drug Control (NIFDC) , Beijing , People's Republic of China
| | - Qing Wang
- a Division of HIV/AIDS and Sexually Transmitted Virus Vaccines , National Institutes for Food and Drug Control (NIFDC) , Beijing , People's Republic of China
| | - Ruifeng Chen
- a Division of HIV/AIDS and Sexually Transmitted Virus Vaccines , National Institutes for Food and Drug Control (NIFDC) , Beijing , People's Republic of China
| | - Tingting Ning
- a Division of HIV/AIDS and Sexually Transmitted Virus Vaccines , National Institutes for Food and Drug Control (NIFDC) , Beijing , People's Republic of China
| | - Qiang Liu
- a Division of HIV/AIDS and Sexually Transmitted Virus Vaccines , National Institutes for Food and Drug Control (NIFDC) , Beijing , People's Republic of China
| | - Weijin Huang
- a Division of HIV/AIDS and Sexually Transmitted Virus Vaccines , National Institutes for Food and Drug Control (NIFDC) , Beijing , People's Republic of China
| | - Youchun Wang
- a Division of HIV/AIDS and Sexually Transmitted Virus Vaccines , National Institutes for Food and Drug Control (NIFDC) , Beijing , People's Republic of China
| |
Collapse
|
37
|
Timiryasova TM, Luo P, Zheng L, Singer A, Zedar R, Garg S, Petit C, Moore S, Hu BT, Brown M. Rapid fluorescent focus inhibition test optimization and validation: Improved detection of neutralizing antibodies to rabies virus. J Immunol Methods 2019; 474:112626. [PMID: 31228423 DOI: 10.1016/j.jim.2019.06.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/03/2019] [Revised: 06/18/2019] [Accepted: 06/18/2019] [Indexed: 11/18/2022]
Abstract
The rabies rapid fluorescent focus inhibition test (RFFIT) is the most widely used cell-based assay for detecting and quantitating rabies virus neutralizing antibodies (RVNA) in human serum. However, it is a complex, labor intensive, and somewhat subjective manual assay, the performance of which may be affected by a number of factors including the quality of cells and virus, variability of assay reagents and the skill and expertise of analysts. This study sought to identify and evaluate conditions that may impact RFFIT performance and RVNA detection by evaluating assay parameters including: different serial dilution scheme of serum samples in a 96-well microplate using semi-automated pipetting systems, the range of dose of challenge virus standard (CVS-11) strain of rabies virus, the effect of complement (C'), the effect of cell seeding density and passage number, the effect of diethylaminoethyl (DEAE) dextran concentration on virus infectivity, and the assay incubation period prior to immunostaining. In addition the evaluation of counting fluorescent foci using a microscope versus using scanned images from a cell imaging reader was performed in an effort to ease the reading of slides and have permanent records of the raw data. The results from optimization of each parameter are presented along with subsequent assay validation in accordance with the International Conference on Harmonization (ICH) guidelines. The improved and optimized RFFIT accuracy, linearity and sensitivity was demonstrated by testing World Health Organization (WHO)-1 and WHO-2 Standard Rabies Immune Globulins (SRIGs) and complete assay development and validation was performed in compliance with Good Clinical Laboratory Practice (GCLP) guidelines.
Collapse
Affiliation(s)
| | - Ping Luo
- Sanofi Pasteur, Discovery Drive, Swiftwater, PA 18370, USA.
| | - Lingyi Zheng
- Sanofi Pasteur, Discovery Drive, Swiftwater, PA 18370, USA.
| | - Amy Singer
- Sanofi Pasteur, Discovery Drive, Swiftwater, PA 18370, USA.
| | - Rebecca Zedar
- Sanofi Pasteur, Discovery Drive, Swiftwater, PA 18370, USA.
| | - Sanjay Garg
- Sanofi Pasteur, Discovery Drive, Swiftwater, PA 18370, USA.
| | - Celine Petit
- Sanofi Pasteur, Discovery Drive, Swiftwater, PA 18370, USA.
| | - Susan Moore
- Kansas State Veterinary Diagnostic Laboratory at Kansas State University College of Veterinary Medicine, Manhattan, KS 66502, USA.
| | - Branda T Hu
- Sanofi Pasteur, Discovery Drive, Swiftwater, PA 18370, USA.
| | - Monique Brown
- Sanofi Pasteur, Discovery Drive, Swiftwater, PA 18370, USA.
| |
Collapse
|
38
|
Ma J, Chen R, Huang W, Nie J, Liu Q, Wang Y, Yang X. In vitro and in vivo efficacy of a Rift Valley fever virus vaccine based on pseudovirus. Hum Vaccin Immunother 2019; 15:2286-2294. [PMID: 31170027 DOI: 10.1080/21645515.2019.1627820] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 02/07/2023] Open
Abstract
Rift Valley fever virus (RVFV), a recognized category A priority pathogen, causes large outbreaks of Rift Valley fever with some fatalities in humans in humans and huge economic losses in livestock. As wild-type RVFV must be handled in BSL-3 or BSL-4 laboratories, we constructed a high-titer vesicular stomatitis virus (VSV) pseudotype bearing RVFV envelope glycoproteins to detect neutralizing antibodies in vitro under BSL-2 conditions. The neutralizing properties of 39 amino acid mutant sites that have occurred naturally over time in the RVFV envelope glycoproteins were analyzed with their corresponding pseudoviral mutants separately. Compared with the results in the primary strain, the variants showed no statistically significant differences. We next established a Balb/c mouse pseudovirus infection model for detecting neutralizing antibodies against pseudovirus. Five immunizations with pseudoviral DNA protected the mice from infection with the pseudovirus. Bioluminescence imaging, which we used to evaluate viral dissemination and distribution in the mice, showed a good relationship between the neutralizing antibodies titers in vitro. These pseudovirus methods will allow for the safe determination of neutralizing antibodies in vivo and in vitro, and will assist with studies on vaccines and drugs against RVFV with the long term objective of Rift Valley fever prevention.
Collapse
Affiliation(s)
- Jian Ma
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC) , Beijing , China.,National Engineering Technology Research Center of Combination Vaccines , Wuhan , China
| | - Ruifeng Chen
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC) , Beijing , China
| | - Weijin Huang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC) , Beijing , China
| | - Jianhui Nie
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC) , Beijing , China
| | - Qiang Liu
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC) , Beijing , China
| | - Youchun Wang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC) , Beijing , China
| | - Xiaoming Yang
- National Engineering Technology Research Center of Combination Vaccines , Wuhan , China.,China National Biotec Group Company Limited , Beijing , China
| |
Collapse
|
39
|
Jalal PJ, Urbanowicz RA, Horncastle E, Pathak M, Goddard C, Saeed A, Mason CP, Ball JK, Irving WL, McClure CP, King BJ, Tarr AW. Expression of human ficolin-2 in hepatocytes confers resistance to infection by diverse hepatotropic viruses. J Med Microbiol 2019; 68:642-648. [PMID: 30747617 DOI: 10.1099/jmm.0.000935] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/17/2022] Open
Abstract
The liver-expressed pattern recognition receptors mannose-binding lectin (MBL), ficolin-2 and ficolin-3 contribute to the innate immune response by activating complement. Binding of soluble ficolin-2 to viral pathogens can directly neutralize virus entry. We observed that the human hepatoma cell line HuH7.5, which is routinely used for the study of hepatotropic viruses, is deficient in expression of MBL, ficolin-2 and ficolin-3. We generated a cell line that expressed and secreted ficolin-2. This cell line (HuH7.5 [FCN2]) was more resistant to infection with hepatitis C virus (HCV), ebolavirus and vesicular stomatitis virus, but surprisingly was more susceptible to infection with rabies virus. Cell-to-cell spread of HCV was also inhibited in ficolin-2 expressing cells. This illustrates that ficolin-2 expression in hepatocytes contributes to innate resistance to virus infection, but some viruses might utilize ficolin-2 to facilitate entry.
Collapse
Affiliation(s)
- Paywast J Jalal
- School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
- Biology Department, Faculty of Science, University of Sulaimani, Sulaimani, Iraq
| | - Richard A Urbanowicz
- School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
| | - Emma Horncastle
- School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
| | - Monika Pathak
- School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
| | - Chun Goddard
- School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
| | - Amanj Saeed
- Biology Department, Faculty of Science, University of Sulaimani, Sulaimani, Iraq
| | - Christopher P Mason
- School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
| | - Jonathan K Ball
- School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, UK
| | - William L Irving
- School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, UK
| | - C Patrick McClure
- School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, UK
| | - Barnabas J King
- School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
| | - Alexander W Tarr
- School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, UK
| |
Collapse
|
40
|
Porta C, Marino A, Consonni FM, Bleve A, Mola S, Storto M, Riboldi E, Sica A. Metabolic influence on the differentiation of suppressive myeloid cells in cancer. Carcinogenesis 2019; 39:1095-1104. [PMID: 29982315 DOI: 10.1093/carcin/bgy088] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/11/2018] [Accepted: 06/27/2018] [Indexed: 12/11/2022] Open
Abstract
New evidences indicate that the metabolic instruction of immunity (immune metabolism) results from the integration of cell metabolism and whole-body metabolism, which are both influenced by nutrition, microbiome metabolites and disease-driven metabolism (e.g. cancer metabolism). Cancer metabolism influences the immunological homeostasis and promotes immune alterations that support disease progression, hence influencing the clinical outcome. Cancer cells display increased glucose uptake and fermentation of glucose to lactate, even in the presence of completely functioning mitochondria. A major side effect of this event is immunosuppression, characterized by limited immunogenicity of cancer cells and restriction of the therapeutic efficacy of anticancer immunotherapy. Here, we discuss how the metabolism of myeloid cells associated with cancer contributes to the differentiation of their suppressive phenotype and therefore to cancer immune evasion.
Collapse
Affiliation(s)
- Chiara Porta
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Novara
| | - Arianna Marino
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Novara
| | | | - Augusto Bleve
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Novara
| | - Silvia Mola
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Novara
| | - Mariangela Storto
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Novara
| | - Elena Riboldi
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Novara
| | - Antonio Sica
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Novara.,Humanitas Clinical and Research Center, Rozzano, Italy
| |
Collapse
|
41
|
Fan C, Wu X, Liu Q, Li Q, Liu S, Lu J, Yang Y, Cao Y, Huang W, Liang C, Ying T, Jiang S, Wang Y. A Human DPP4-Knockin Mouse's Susceptibility to Infection by Authentic and Pseudotyped MERS-CoV. Viruses 2018; 10:v10090448. [PMID: 30142928 PMCID: PMC6164841 DOI: 10.3390/v10090448] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/24/2018] [Revised: 08/13/2018] [Accepted: 08/17/2018] [Indexed: 01/01/2023] Open
Abstract
Infection by the Middle East respiratory syndrome coronavirus (MERS-CoV) causes respiratory illness and has a high mortality rate (~35%). The requirement for the virus to be manipulated in a biosafety level three (BSL-3) facility has impeded development of urgently-needed antiviral agents. Here, we established anovel mouse model by inserting human dipeptidyl peptidase 4 (hDPP4) into the Rosa26 locus using CRISPR/Cas9, resulting in global expression of the transgene in a genetically stable mouse line. The mice were highly susceptible to infection by MERS-CoV clinical strain hCoV-EMC, which induced severe diffuse pulmonary disease in the animals, and could also be infected by an optimized pseudotyped MERS-CoV. Administration of the neutralizing monoclonal antibodies, H111-1 and m336, as well as a fusion inhibitor peptide, HR2P-M2, protected mice from challenge with authentic and pseudotyped MERS-CoV. These results confirmed that the hDPP4-knockin mouse is a novel model for studies of MERS-CoV pathogenesis and anti-MERS-CoV antiviral agents in BSL-3 and BSL-2facilities, respectively.
Collapse
Affiliation(s)
- Changfa Fan
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control, Beijing 100050, China.
| | - Xi Wu
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control, Beijing 100050, China.
| | - Qiang Liu
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, National Institutes for Food and Drug Control, Beijing 100050, China.
| | - Qianqian Li
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, National Institutes for Food and Drug Control, Beijing 100050, China.
| | - Susu Liu
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control, Beijing 100050, China.
| | - Jianjun Lu
- National Center for Safety Evaluation of Drugs, Institute for Food and Drug Safety Evaluation, National Institutes for Food and Drug Control, A8 Hongda Middle Street, Beijing Economic-Technological Development Area, Beijing 100176, China.
| | - Yanwei Yang
- National Center for Safety Evaluation of Drugs, Institute for Food and Drug Safety Evaluation, National Institutes for Food and Drug Control, A8 Hongda Middle Street, Beijing Economic-Technological Development Area, Beijing 100176, China.
| | - Yuan Cao
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control, Beijing 100050, China.
| | - Weijin Huang
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, National Institutes for Food and Drug Control, Beijing 100050, China.
| | - Chunnan Liang
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control, Beijing 100050, China.
| | - Tianlei Ying
- Key Laboratory of Medical Molecular Virology of the Ministries of Education and Health, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology of the Ministries of Education and Health, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Youchun Wang
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, National Institutes for Food and Drug Control, Beijing 100050, China.
| |
Collapse
|
42
|
Synthesis and biological evaluation of novel tricyclic matrinic derivatives as potential anti-filovirus agents. Acta Pharm Sin B 2018; 8:629-638. [PMID: 30109186 PMCID: PMC6089848 DOI: 10.1016/j.apsb.2018.01.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/10/2017] [Revised: 12/15/2017] [Accepted: 01/04/2018] [Indexed: 12/12/2022] Open
Abstract
Twenty-six novel tricyclic sophoridinic and matrinic derivatives containing a common chlorinated benzene fragment were designed, synthesized and evaluated for their anti-ebolavirus (EBOV) activities. Structure-activity relationship analysis indicated: (i) 12N-dichlorobenzyl motif was beneficial for the activity; (ii) the chiral configuration at C5 atom might not affect the activity much. Among the target compounds, compound 7d exhibited the most potent potency against EBOV with an IC50 value of 5.29 μmol/L and an SI value of over 37.8. Further in vivo anti-EBOV assay of 7d identified its high effectiveness, and in vivo anti-MARV assay of 7d suggested its inspiring broad-spectrum anti-filovirus activity. The results provided powerful information on further strategic optimization and development of this kind of compounds against filoviruses.
Collapse
|
43
|
Development of in vitro and in vivo neutralization assays based on the pseudotyped H7N9 virus. Sci Rep 2018; 8:8484. [PMID: 29855533 PMCID: PMC5981435 DOI: 10.1038/s41598-018-26822-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/04/2018] [Accepted: 05/15/2018] [Indexed: 11/08/2022] Open
Abstract
H7N9 viral infections pose a great threat to both animal and human health. This avian virus cannot be handled in level 2 biocontainment laboratories, substantially hindering evaluation of prophylactic vaccines and therapeutic agents. Here, we report a high-titer pseudoviral system with a bioluminescent reporter gene, enabling us to visually and quantitatively conduct analyses of virus replications in both tissue cultures and animals. For evaluation of immunogenicity of H7N9 vaccines, we developed an in vitro assay for neutralizing antibody measurement based on the pseudoviral system; results generated by the in vitro assay were found to be strongly correlated with those by either hemagglutination inhibition (HI) or micro-neutralization (MN) assay. Furthermore, we injected the viruses into Balb/c mice and observed dynamic distributions of the viruses in the animals, which provides an ideal imaging model for quantitative analyses of prophylactic and therapeutic monoclonal antibodies. Taken together, the pseudoviral systems reported here could be of great value for both in vitro and in vivo evaluations of vaccines and antiviral agents without the need of wild type H7N9 virus.
Collapse
|
44
|
Discovery and evolution of aloperine derivatives as novel anti-filovirus agents through targeting entry stage. Eur J Med Chem 2018; 149:45-55. [DOI: 10.1016/j.ejmech.2018.02.061] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/18/2017] [Revised: 02/18/2018] [Accepted: 02/19/2018] [Indexed: 11/19/2022]
|
45
|
Li Q, Liu Q, Huang W, Li X, Wang Y. Current status on the development of pseudoviruses for enveloped viruses. Rev Med Virol 2017; 28. [PMID: 29218769 PMCID: PMC7169153 DOI: 10.1002/rmv.1963] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/24/2017] [Revised: 11/01/2017] [Accepted: 11/02/2017] [Indexed: 12/21/2022]
Abstract
Emerging and reemerging infectious diseases have a strong negative impact on public health. However, because many of these pathogens must be handled in biosafety level, 3 or 4 containment laboratories, research and development of antivirals or vaccines against these diseases are often impeded. Alternative approaches to address this issue have been vigorously pursued, particularly the use of pseudoviruses in place of wild‐type viruses. As pseudoviruses have been deprived of certain gene sequences of the virulent virus, they can be handled in biosafety level 2 laboratories. Importantly, the envelopes of these viral particles may have similar conformational structures to those of the wild‐type viruses, making it feasible to conduct mechanistic investigation on viral entry and to evaluate potential neutralizing antibodies. However, a variety of challenging issues remain, including the production of a sufficient pseudovirus yield and the inability to produce an appropriate pseudotype of certain viruses. This review discusses current progress in the development of pseudoviruses and dissects the factors that contribute to low viral yields.
Collapse
Affiliation(s)
- Qianqian Li
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, National Institutes for Food and Drug Control, Beijing, China
| | - Qiang Liu
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, National Institutes for Food and Drug Control, Beijing, China
| | - Weijin Huang
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, National Institutes for Food and Drug Control, Beijing, China
| | - Xuguang Li
- Division of Regulatory Research, Centre for Biologics Evaluation, Biologics and Genetic Therapies Directorate, Health Canada, Ottawa, Canada
| | - Youchun Wang
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, National Institutes for Food and Drug Control, Beijing, China
| |
Collapse
|
46
|
Chen Q, Nie J, Huang W, Jiao Y, Li L, Zhang T, Zhao J, Wu H, Wang Y. Development and optimization of a sensitive pseudovirus-based assay for HIV-1 neutralizing antibodies detection using A3R5 cells. Hum Vaccin Immunother 2017; 14:199-208. [PMID: 28933644 DOI: 10.1080/21645515.2017.1373922] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 10/18/2022] Open
Abstract
Sensitive assays for HIV-1 neutralizing antibody detection are urgently needed for vaccine immunogen optimization and identification of protective immune response levels. In this study, we developed an easy-to-use HIV-1 pseudovirus neutralization assay based on a human CD4+ lymphoblastoid cell line A3R5 by employing a high efficient pseudovirus production system. Optimal conditions for cell counts, infection time, virus dose and concentration of DEAE-dextran were tested and identified. For T-cell line-adapted tier 1 virus strains, significantly higher inhibitory efficiency was observed for both monoclonal neutralizing antibody (4 fold) and plasma (2 fold) samples in A3R5 than those in TZM-bl assay. For circulating tier 2 strains, the A3R5 pseudovirus assay showed even much higher sensitivity for both neutralizing antibody (10 fold) and plasma (9 fold) samples. When sequential neutralizing antibody seroconverting samples were tested in both A3R5 and TZM-bl assays, the seroconverting points could be detected earlier for tier 1 (15.7 weeks) and tier 2 (68.3 weeks) strains in A3R5 assay respectively. The high sensitive pseudovirus assay using more physiological target cells could serve as an alternative to the TZM-bl assay for evaluation of vaccine-induced neutralizing antibodies and identification of the correlates of protection.
Collapse
Affiliation(s)
- Qingqing Chen
- a Division of HIV/AIDS and Sex-transmitted Virus Vaccines , National Institutes for Food and Drug Control (NIFDC) , Beijing , China
| | - Jianhui Nie
- a Division of HIV/AIDS and Sex-transmitted Virus Vaccines , National Institutes for Food and Drug Control (NIFDC) , Beijing , China
| | - Weijin Huang
- a Division of HIV/AIDS and Sex-transmitted Virus Vaccines , National Institutes for Food and Drug Control (NIFDC) , Beijing , China
| | - Yanmei Jiao
- b Beijing You'an Hospital, Capital Medical University , Beijing , China
| | - Lan Li
- b Beijing You'an Hospital, Capital Medical University , Beijing , China
| | - Tong Zhang
- b Beijing You'an Hospital, Capital Medical University , Beijing , China
| | - Juan Zhao
- a Division of HIV/AIDS and Sex-transmitted Virus Vaccines , National Institutes for Food and Drug Control (NIFDC) , Beijing , China
| | - Hao Wu
- b Beijing You'an Hospital, Capital Medical University , Beijing , China
| | - Youchun Wang
- a Division of HIV/AIDS and Sex-transmitted Virus Vaccines , National Institutes for Food and Drug Control (NIFDC) , Beijing , China
| |
Collapse
|
47
|
Wu J, Zhao C, Liu Q, Huang W, Wang Y. Development and application of a bioluminescent imaging mouse model for Chikungunya virus based on pseudovirus system. Vaccine 2017; 35:6387-6394. [PMID: 29031692 DOI: 10.1016/j.vaccine.2017.10.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/16/2017] [Revised: 09/09/2017] [Accepted: 10/03/2017] [Indexed: 01/28/2023]
Abstract
Chikungunya virus (CHIKV) is an arthropod-borne virus that is transmitted to humans primarily via the bite of an infected mosquito. Infection of humans by CHIKV can cause chikungunya fever which is an acute febrile illness associated with severe, often debilitating polyarthralgias. Since a re-emergence of CHIKV in 2004, the virus has spread into novel locations in nearly 40 countries including non-endemic regions and has led to millions of cases of disease throughout countries. Handling of CHIKV is restricted to the high-containment Biosafety Level 3 (BSL-3) facilities, which greatly impede the research progress of this virus. In this study, an envelope-pseudotyped virus expressing the firefly luciferase reporter protein (pHIV-CHIKV-Fluc) was generated. An in vitro sensitive neutralizing assay and an in vivo bioluminescent-imaging-based mouse infection model had been developed based on the CHIKV pseudovirus. Utilizing the platform, protection effect of DNA vaccine was evaluated. Therefore, this study provides a safe, sensitive and visualizing model for evaluating vaccines and antiviral therapies against CHIKV in low containment BSL-2 laboratories.
Collapse
Affiliation(s)
- Jiajing Wu
- Division of HIV/AIDS and Sexually Transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China
| | - Chenyan Zhao
- Division of HIV/AIDS and Sexually Transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China
| | - Qiang Liu
- Division of HIV/AIDS and Sexually Transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China
| | - Weijin Huang
- Division of HIV/AIDS and Sexually Transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China
| | - Youchun Wang
- Division of HIV/AIDS and Sexually Transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China.
| |
Collapse
|
48
|
Chen Q, Tang K, Zhang X, Chen P, Guo Y. Establishment of pseudovirus infection mouse models for in vivo pharmacodynamics evaluation of filovirus entry inhibitors. Acta Pharm Sin B 2017; 8:200-208. [PMID: 29719780 PMCID: PMC5925413 DOI: 10.1016/j.apsb.2017.08.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/26/2017] [Revised: 08/23/2017] [Accepted: 08/28/2017] [Indexed: 10/25/2022] Open
Abstract
Filoviruses cause severe and fatal viral hemorrhagic fever in humans. Filovirus research has been extensive since the 2014 Ebola outbreak. Due to their high pathogenicity and mortality, live filoviruses require Biosafety Level-4 (BSL-4) facilities, which have restricted the development of anti-filovirus vaccines and drugs. An HIV-based pseudovirus cell infection assay is widely used for viral entry studies in BSL-2 conditions. Here, we successfully constructed nine in vitro pseudo-filovirus models covering all filovirus genera and three in vivo pseudo-filovirus-infection mouse models using Ebola virus, Marburg virus, and Lloviu virus as representative viruses. The pseudo-filovirus-infected mice showed visualizing bioluminescence in a dose-dependent manner. A bioluminescence peak in mice was reached on day 5 post-infection for Ebola virus and Marburg virus and on day 4 post-infection for Lloviu virus. Two known filovirus entry inhibitors, clomiphene and toremiphene, were used to validate the model. Collectively, our study shows that all genera of filoviruses can be well-pseudotyped and are infectious in vitro. The pseudo-filovirus-infection mouse models can be used for in vivo activity evaluation of anti-filovirus drugs. This sequential in vitro and in vivo evaluation system of filovirus entry inhibitors provides a secure and efficient platform for screening and assessing anti-filovirus agents in BSL-2 facilities.
Collapse
Key Words
- 3D, 3-dimensional
- BDBV, Bundibugyo virus
- BSL, Biosafety Level
- CLO, clomiphene
- DLIT, Diffuse Luminescence Imaging Tomography
- EBOV, Ebola virus
- Ebola
- Entry inhibitor
- Filovirus
- GP, glycoprotein
- IC50, the 50% inhibitory concentration
- LLOV, Lloviu virus
- MARV, Marburg virus
- Marburg
- Mouse model
- Pseudovirus
- RAVV, Ravn virus
- RESTV, Reston virus
- ROI, region of interest
- SD, standard deviation
- SEM, standard error of the mean
- SUDV, Sudan virus
- TAFV, Taï forest virus
- TORE, toremiphene
- VSV-G, vesicular stomatitis virus glycoprotein
- d.p.i., day post-infection
- h.p.i., hour post-infection
- i.p., intraperitoneally
- lg, logarithm
Collapse
|
49
|
An LASV GPC pseudotyped virus based reporter system enables evaluation of vaccines in mice under non-BSL-4 conditions. Vaccine 2017; 35:5172-5178. [PMID: 28797730 DOI: 10.1016/j.vaccine.2017.07.101] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/08/2017] [Revised: 07/16/2017] [Accepted: 07/27/2017] [Indexed: 12/23/2022]
Abstract
Lassa virus (LASV) causes a severe hemorrhagic fever endemic throughout western Africa. Because of the ability to cause lethal disease in humans, limited treatment options, and potential as a bioweapon, the need for vaccines to prevent LASV epidemic is urgent. However, LASV vaccine development has been hindered by the lack of appropriate small animal models for efficacy evaluation independent of biosafety level four (BSL-4) facilities. Here we generated an LASV-glycoprotein precursor (GPC)-pseudotyped Human immunodeficiency virus containing firefly luciferase (Fluc) reporter gene as surrogate to develop a bioluminescent-imaging-based BALB/c mouse model for one-round infection under non-BSL-4 conditions, in which the bioluminescent intensity of Fluc was utilized as endpoint when evaluating vaccine efficacy. Electron microscopy analysis demonstrated that LASV GPC pseudotyped virus appeared structurally similar to native virion. Meanwhile, we constructed DNA vaccine (pSV1.0-LASVGPC) and pseudoparticle-based vaccine (LASVpp) that displayed conformational GPC protein of LASV strain Josiah to vaccinate BALB/c mice using intramuscular electroporation and by intraperitoneal routes, respectively. Vaccinated mice in LASVpp alone and DNA prime+LASVpp boost schedules were protected against 100 AID50 of LASV pseudovirus challenge, and it was found that in vivo efficiencies correlated with their anti-LASV neutralizing activities and MCP-1 cytokine levels in serum sampled before infection. The bioluminescence pseudovirus infection model can be useful tool for the preliminary evaluation of immunogenicity and efficacy of vaccine candidates against LASV outside of BSL-4 containments, and the results with pseudoparticle-based vaccine provided very helpful information for LASV vaccine design.
Collapse
|
50
|
Zhang L, Li Q, Liu Q, Huang W, Nie J, Wang Y. A bioluminescent imaging mouse model for Marburg virus based on a pseudovirus system. Hum Vaccin Immunother 2017; 13:1811-1817. [PMID: 28481728 DOI: 10.1080/21645515.2017.1325050] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 10/19/2022] Open
Abstract
Marburg virus (MARV) can cause lethal hemorrhagic fever in humans. Handling of MARV is restricted to high-containment biosafety level 4 (BSL-4) facilities, which greatly impedes research into this virus. In this study, a high titer of MARV pseudovirus was generated through optimization of the HIV backbone vectors, the ratio of backbone vector to MARV glycoprotein expression vector, and the transfection reagents. An in vitro neutralization assay and an in vivo bioluminescent imaging mouse model for MARV were developed based on the pseudovirus. Protective serum against MARV was successfully induced in guinea pigs, which showed high neutralization activity in vitro and could also protect Balb/c mice from MARV pseudovirus infection in vivo. This system could be a convenient tool to enable the evaluation of vaccines and therapeutic drugs against MARV in non-BSL-4 laboratories.
Collapse
Affiliation(s)
- Li Zhang
- a Division of HIV/AIDS and Sexually-transmitted Virus Vaccines , National Institutes for Food and Drug Control , Beijing , China
| | - Qianqian Li
- a Division of HIV/AIDS and Sexually-transmitted Virus Vaccines , National Institutes for Food and Drug Control , Beijing , China
| | - Qiang Liu
- a Division of HIV/AIDS and Sexually-transmitted Virus Vaccines , National Institutes for Food and Drug Control , Beijing , China
| | - Weijin Huang
- a Division of HIV/AIDS and Sexually-transmitted Virus Vaccines , National Institutes for Food and Drug Control , Beijing , China
| | - Jianhui Nie
- a Division of HIV/AIDS and Sexually-transmitted Virus Vaccines , National Institutes for Food and Drug Control , Beijing , China
| | - Youchun Wang
- a Division of HIV/AIDS and Sexually-transmitted Virus Vaccines , National Institutes for Food and Drug Control , Beijing , China
| |
Collapse
|