1
|
Hou Y, Ma J, Huang B, Li N, Zhu L, Jia Z, Yang J, Zhang J, Tan W, Xue J. Comparative pathogenicity of vaccinia virus and mpox virus infections in CAST/EiJ mice: Exploring splenomegaly and transcriptomic profiles. Animal Model Exp Med 2025. [PMID: 40275745 DOI: 10.1002/ame2.70026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 04/01/2025] [Accepted: 04/07/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Vaccinia virus (VACV) and mpox virus (MPXV) belong to the orthopoxvirus genus and share high genetic similarity, making VACV widely used in the mpox pandemic. CAST/EiJ mice have been widely used for studying orthopoxvirus infection. However, the histopathological features of CAST/EiJ mice with mpox virus (MPXV) and vaccinia virus (VACV) infections have not been fully elucidated. METHODS Four group of CAST/EiJ mice were challenged with low-dose VACV (103 PFU, VACV-L), high-dose VACV (106 PFU, VACV-H), MPXV (106 PFU) or PBS via intraperitoneal route, and the disease signs and body weight were monitored daily. Subsequently, viral loads and titers in the blood and spleen of CAST/EiJ mice were analyzed via qPCR and TCID50 assay. Finally, the spleen samples were analyzed for histopathological, immunohistochemical and RNA-seq. RESULTS Herein, we found that VACV-L and MPXV caused splenomegaly via the intraperitoneal route, whereas VACV-H caused rapid lethality with limited splenomegaly. Transcriptome analysis from spleen revealed significant differences in gene expression between VACV-L and VACV-H groups, but the differentially expressed genes induced by splenomegaly between VACV-L and MPXV groups were highly similar. Furthermore, pathway enrichment analysis demonstrated that the VACV-L, VACV-H, and MPXV groups were all associated with the calcium, MAPK, and PI3K-Akt signaling pathway. Compared to the lethal infection observed in VACV-H group, the splenomegaly in the VACV-L and MPXV groups was characterized by extramedullary hematopoiesis and increased macrophages infiltration in the red pulp. Transcriptome analysis of the spleen demonstrated that the Wnt, tumor necrosis factor (TNF), and transforming growth factor β (TGF-β) signaling pathways may promote splenomegaly by modulating granulocyte infiltration and inflammatory responses. Compared to VACV-L group, the limited splenomegaly but lethality in VACV-H-infected mice might be associated with extensive splenic necrosis, diffuse congestion, and hemorrhage in the red pulp, as well as changes in the cGMP-PKG, Ras signaling, and Fc gamma R-mediated phagocytosis pathways. CONCLUSIONS Our findings systematically compared the pathogenicity of VACV and MPXV in CAST/EiJ mice, incorporating splenic transcriptome analysis to provide insights into the potential molecular mechanism behind orthopoxvirus-induced splenomegaly in CAST/EiJ mice.
Collapse
Affiliation(s)
- Yongzhi Hou
- NHC Key Laboratory of Human Disease Comparative Medicine, National Center of Technology Innovation for Animal Model, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianrong Ma
- NHC Key Laboratory of Human Disease Comparative Medicine, National Center of Technology Innovation for Animal Model, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Baoying Huang
- NHC Key Laboratory of Biosafety, National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Na Li
- NHC Key Laboratory of Human Disease Comparative Medicine, National Center of Technology Innovation for Animal Model, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lin Zhu
- NHC Key Laboratory of Human Disease Comparative Medicine, National Center of Technology Innovation for Animal Model, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ziqing Jia
- NHC Key Laboratory of Human Disease Comparative Medicine, National Center of Technology Innovation for Animal Model, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiasen Yang
- NHC Key Laboratory of Human Disease Comparative Medicine, National Center of Technology Innovation for Animal Model, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jingjing Zhang
- NHC Key Laboratory of Human Disease Comparative Medicine, National Center of Technology Innovation for Animal Model, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wenjie Tan
- NHC Key Laboratory of Biosafety, National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Jing Xue
- NHC Key Laboratory of Human Disease Comparative Medicine, National Center of Technology Innovation for Animal Model, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Respiratory Health and Multimorbidity, Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
2
|
Zuo J, Wu J, Zhang Z, Long J, Yu C, Liao Y, Zhang H, Yang J. An Mpox Multi-Antigen-Tandem Bivalent mRNA Candidate Vaccine Effectively Protects Mice Against the Vaccinia Virus. Vaccines (Basel) 2025; 13:374. [PMID: 40333228 PMCID: PMC12031407 DOI: 10.3390/vaccines13040374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 03/21/2025] [Accepted: 03/25/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND Since the outbreak of mpox in 2022, the disease has spread rapidly worldwide and garnered significant public attention. Vaccination is regarded as an effective measure to prevent the spread of mpox. The success of the COVID-19 mRNA vaccine demonstrates that mRNA-based vaccines represent a rapid and multifunctional platform with considerable potential, and are expected to be a strategy to address mpox spread. METHODS In this study, we screened an mpox multi-antigen-tandem bivalent mRNA vaccine candidate: a lipid nanoparticle-encapsulated mRNA-1017 and mRNA-1995 (mRNA-3012-LNP). We then evaluated the immunogenicity of the mpox virus (MPXV) bivalent mRNA vaccine candidate and its protective efficacy against the vaccinia virus (VACV) in a mouse model. RESULTS Mice vaccinated with two doses of the mRNA-3012-LNP vaccine exhibited robust binding antibody responses and MPXV-specific Th-1-biased cellular immune responses in vivo. Notably, the boosted immunized mice generated potent neutralizing antibodies against the VACV, effectively protecting them from viral challenge. Additionally, serum transfer protection experiments indicated that serum from mice inoculated with mRNA-3012-LNP was effective in protecting nude mice from VACV challenge. CONCLUSIONS Our results suggest that the mpox bivalent mRNA candidate vaccine mRNA-3012-LNP induces strong immunogenicity and has the potential to serve as a safe and effective vaccine candidate against mpox epidemics.
Collapse
Affiliation(s)
- Jun Zuo
- College of Chemistry and Life Science, Beijing University of Technology, Pingleyuan 100#, District of Chaoyang, Beijing 100124, China
- Bioinformatics Center of AMMS, Beijing 100850, China
| | - Jiayu Wu
- Bioinformatics Center of AMMS, Beijing 100850, China
| | - Zhen Zhang
- Bioinformatics Center of AMMS, Beijing 100850, China
| | - Jinrong Long
- Bioinformatics Center of AMMS, Beijing 100850, China
| | - Changxiao Yu
- Bioinformatics Center of AMMS, Beijing 100850, China
| | - Yuqin Liao
- Bioinformatics Center of AMMS, Beijing 100850, China
| | - Hongsheng Zhang
- College of Chemistry and Life Science, Beijing University of Technology, Pingleyuan 100#, District of Chaoyang, Beijing 100124, China
| | - Jing Yang
- Bioinformatics Center of AMMS, Beijing 100850, China
| |
Collapse
|
3
|
Zheng X, Huang CH, Yan S, Rong MD. Advances and applications of genome-edited animal models for severe combined immunodeficiency. Zool Res 2025; 46:249-260. [PMID: 39846200 PMCID: PMC11891005 DOI: 10.24272/j.issn.2095-8137.2024.195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 10/29/2024] [Indexed: 01/24/2025] Open
Abstract
Severe combined immunodeficiency disease (SCID), characterized by profound immune system dysfunction, can lead to life-threatening infections and death. Animal models play a pivotal role in elucidating biological processes and advancing therapeutic strategies. Recent advances in gene-editing technologies, including zinc-finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), CRISPR/Cas9, and base editing, have significantly enhanced the generation of SCID models. These models have not only deepened our understanding of disease pathophysiology but have also driven progress in cancer therapy, stem cell transplantation, organ transplantation, and infectious disease management. This review provides a comprehensive overview of current SCID models generated using novel gene-editing approaches, highlighting their potential applications in translational medicine and their role in advancing biomedical research.
Collapse
Affiliation(s)
- Xiao Zheng
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Chun-Hui Huang
- Guangdong Key Laboratory of Non-Human Primate Models, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong 510632, China
- School of medicine, Jinan University, Guangzhou, Guangdong 510632, China
| | - Sen Yan
- Guangdong Key Laboratory of Non-Human Primate Models, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong 510632, China
- School of medicine, Jinan University, Guangzhou, Guangdong 510632, China
- Department of Neurology, Guangzhou Red Cross Hospital, Faculty of Medical Science, Jinan University, Guangzhou, Guangdong 510220, China. E-mail:
| | - Ming-Deng Rong
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510280, China. E-mail:
| |
Collapse
|
4
|
Wang J, Zhu F, Jiao D, Yang C, Wang J, Wang F, Zhao H, Wei HJ, Zhao HY. Generation of RAG2 Knockout Immune-Deficient Miniature Pigs. Animals (Basel) 2024; 14:2597. [PMID: 39272382 PMCID: PMC11393836 DOI: 10.3390/ani14172597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/02/2024] [Accepted: 09/04/2024] [Indexed: 09/15/2024] Open
Abstract
Recombination-activating genes (RAGs) play a crucial role in the V(D)J recombination process and the development of immune cells. The development of the immune system and its mechanisms in pigs exhibit greater similarity to those of humans compared to other animals, thus rendering pigs a valuable tool for biomedical research. In this study, we utilized CRISPR/Cas9 gene editing and somatic cell nuclear transfer technology to generate RAG2 knockout (KO) pigs. Furthermore, we evaluated the impact of RAG2 KO on the immune organs and immune cell development through morphological observations, blood analysis and flow cytometry technology. RAG2 KO cell lines were used as donors for cloning. The reconstructed embryos were transplanted into 4 surrogate sows, and after 116 days of gestation, 2 sows gave birth to 12 live piglets, all of which were confirmed to be RAG2 KO. The thymus and spleen sizes of RAG2 KO pigs were significantly smaller than those of wild-type (WT) pigs. Hematoxylin-eosin staining results revealed that the thymus and spleen tissue structures of RAG2 KO pigs were disorganized and lacked the characteristic structures, indicating that RAG2 KO leads to dysplasia of the thymus and spleen. Hematological analysis demonstrated that the total number of white blood cells and lymphocytes in the circulation of RAG2 KO pigs was significantly lower, while the number of eosinophils was higher. Flow cytometry results indicated that the proportions of mature T and B lymphocytes were significantly reduced compared to WT pigs. These findings successfully verified the immunodeficiency phenotype of RAG2 KO pigs. This study may provide experimental animals for the development of tumor models and humanized animals.
Collapse
Affiliation(s)
- Jing Wang
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming 650201, China
- Yunnan Province Xenotransplantation Research Engineering Centre, Yunnan Agricultural University, Kunming 650201, China
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Feiyan Zhu
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming 650201, China
- Yunnan Province Xenotransplantation Research Engineering Centre, Yunnan Agricultural University, Kunming 650201, China
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Deling Jiao
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming 650201, China
- Yunnan Province Xenotransplantation Research Engineering Centre, Yunnan Agricultural University, Kunming 650201, China
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Chang Yang
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming 650201, China
- Yunnan Province Xenotransplantation Research Engineering Centre, Yunnan Agricultural University, Kunming 650201, China
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Junqi Wang
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming 650201, China
- Yunnan Province Xenotransplantation Research Engineering Centre, Yunnan Agricultural University, Kunming 650201, China
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Fengchong Wang
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming 650201, China
- Yunnan Province Xenotransplantation Research Engineering Centre, Yunnan Agricultural University, Kunming 650201, China
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Heng Zhao
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming 650201, China
- Yunnan Province Xenotransplantation Research Engineering Centre, Yunnan Agricultural University, Kunming 650201, China
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Hong-Jiang Wei
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming 650201, China
- Yunnan Province Xenotransplantation Research Engineering Centre, Yunnan Agricultural University, Kunming 650201, China
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Hong-Ye Zhao
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming 650201, China
- Yunnan Province Xenotransplantation Research Engineering Centre, Yunnan Agricultural University, Kunming 650201, China
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| |
Collapse
|
5
|
Carbonaro M, Lee J, Pefanis E, Desclaux M, Wang K, Pennington A, Huang H, Mujica A, Rojas J, Ally R, Kennedy D, Brown M, Rogulin V, Moller-Tank S, Sabin L, Zambrowicz B, Thurston G, Li Z. Efficient engraftment and viral transduction of human hepatocytes in an FRG rat liver humanization model. Sci Rep 2022; 12:14079. [PMID: 35982097 PMCID: PMC9388686 DOI: 10.1038/s41598-022-18119-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 08/05/2022] [Indexed: 11/09/2022] Open
Abstract
Humanized liver rodent models, in which the host liver parenchyma is repopulated by human hepatocytes, have been increasingly used for drug development and disease research. Unlike the leading humanized liver mouse model in which Fumarylacetoacetate Hydrolase (Fah), Recombination Activating Gene (Rag)-2 and Interleukin-2 Receptor Gamma (Il2rg) genes were inactivated simultaneously, generation of similar recipient rats has been challenging. Here, using Velocigene and 1-cell-embryo-targeting technologies, we generated a rat model deficient in Fah, Rag1/2 and Il2rg genes, similar to humanized liver mice. These rats were efficiently engrafted with Fah-expressing hepatocytes from rat, mouse and human. Humanized liver rats expressed human albumin and complement proteins in serum and showed a normal liver zonation pattern. Further, approaches were developed for gene delivery through viral transduction of human hepatocytes either in vivo, or in vitro prior to engraftment, providing a novel platform to study liver disease and hepatocyte-targeted therapies.
Collapse
Affiliation(s)
| | - Jeffrey Lee
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | | | | | - Kehui Wang
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | | | - Hui Huang
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | - Alejo Mujica
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | - Jose Rojas
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | - Roxanne Ally
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | | | | | | | | | - Leah Sabin
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | | | | | - Zhe Li
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA.
| |
Collapse
|
6
|
Xiong R, Fu R, Wu Y, Wu X, Cao Y, Qu Z, Yang Y, Liu S, Huo G, Wang S, Huang W, Lyu J, Zhu X, Liang C, Peng Y, Wang Y, Fan C. Long-Term Infection and Pathogenesis in a Novel Mouse Model of Human Respiratory Syncytial Virus. Viruses 2022; 14:1740. [PMID: 36016362 PMCID: PMC9415064 DOI: 10.3390/v14081740] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [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
Affiliation(s)
- Rui Xiong
- National Rodent Laboratory Animal Resources Center, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China
- Department of Microbiology & Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100083, China
| | - Rui Fu
- National Rodent Laboratory Animal Resources Center, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China
| | - Yong Wu
- National Rodent Laboratory Animal Resources Center, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China
| | - Xi Wu
- National Rodent Laboratory Animal Resources Center, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China
| | - Yuan Cao
- National Rodent Laboratory Animal Resources Center, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China
| | - Zhe Qu
- National Center for Safety Evaluation of Drugs, Institute for Food and Drug Safety Evaluation, National Institutes for Food and Drug Control (NIFDC), 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 (NIFDC), Beijing 100176, China
| | - Susu Liu
- National Rodent Laboratory Animal Resources Center, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China
| | - Guitao Huo
- National Center for Safety Evaluation of Drugs, Institute for Food and Drug Safety Evaluation, National Institutes for Food and Drug Control (NIFDC), Beijing 100176, China
| | - Sanlong Wang
- National Center for Safety Evaluation of Drugs, Institute for Food and Drug Safety Evaluation, National Institutes for Food and Drug Control (NIFDC), Beijing 100176, China
| | - Weijin Huang
- Division of HIV/AIDS and Sexually Transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China
| | - Jianjun Lyu
- Department of Pathology, InnoStar Bio-Tech Nantong Co., Ltd., Nantong 226133, China
| | - Xiang Zhu
- Department of Microbiology & Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100083, China
| | - Chunnan Liang
- National Rodent Laboratory Animal Resources Center, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China
| | - Yihong Peng
- Department of Microbiology & Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100083, China
| | - Youchun Wang
- Division of HIV/AIDS and Sexually Transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China
| | - Changfa Fan
- National Rodent Laboratory Animal Resources Center, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China
| |
Collapse
|
7
|
Sheean ME, Malikova E, Duarte D, Capovilla G, Fregonese L, Hofer MP, Magrelli A, Mariz S, Mendez-Hermida F, Nistico R, Leest T, Sipsas NV, Tsigkos S, Vitezic D, Larsson K, Sepodes B, Stoyanova-Beninska V. Nonclinical data supporting orphan medicinal product designations in the area of rare infectious diseases. Drug Discov Today 2019; 25:274-291. [PMID: 31704277 DOI: 10.1016/j.drudis.2019.10.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 10/15/2019] [Accepted: 10/30/2019] [Indexed: 01/13/2023]
Abstract
This review provides an overview of nonclinical in vivo models that can be used to support orphan designation in selected rare infectious diseases in Europe, with the aim to inform and stimulate the planning of nonclinical development in this area of often neglected diseases.
Collapse
Affiliation(s)
- Maria E Sheean
- Orphan Medicines Office, European Medicines Agency, Amsterdam, The Netherlands; Max-Delbrück Center for Molecular Medicine in Helmholz Association, Berlin, Germany.
| | - Eva Malikova
- Committee of Orphan Medicinal Products, European Medicines Agency, Amsterdam, The Netherlands; State Institute for Drug Control, Bratislava, Slovak Republic; Comenius University, Department of Pharmacology and Toxicology, Bratislava, Slovak Republic
| | - Dinah Duarte
- Committee of Orphan Medicinal Products, European Medicines Agency, Amsterdam, The Netherlands; INFARMED - Autoridade Nacional do Medicamento, Lisbon, Portugal
| | - Giuseppe Capovilla
- Committee of Orphan Medicinal Products, European Medicines Agency, Amsterdam, The Netherlands; C. Poma Hospital, Mantova, Italy; Fondazione Poliambulanza, Brescia, Italy
| | - Laura Fregonese
- Orphan Medicines Office, European Medicines Agency, Amsterdam, The Netherlands
| | - Matthias P Hofer
- Orphan Medicines Office, European Medicines Agency, Amsterdam, The Netherlands
| | - Armando Magrelli
- Committee of Orphan Medicinal Products, European Medicines Agency, Amsterdam, The Netherlands; National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Segundo Mariz
- Orphan Medicines Office, European Medicines Agency, Amsterdam, The Netherlands
| | - Fernando Mendez-Hermida
- Committee of Orphan Medicinal Products, European Medicines Agency, Amsterdam, The Netherlands; Agencia Española de Medicamentos y Productos Sanitarios, Madrid, Spain
| | - Robert Nistico
- Committee of Orphan Medicinal Products, European Medicines Agency, Amsterdam, The Netherlands; Malta Medicines Authority, San Ġwann, Malta
| | - Tim Leest
- Committee of Orphan Medicinal Products, European Medicines Agency, Amsterdam, The Netherlands; The Federal Agency for Medicines and Health Products, Brussels, Belgium
| | - Nikolaos V Sipsas
- Committee of Orphan Medicinal Products, European Medicines Agency, Amsterdam, The Netherlands; Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Stelios Tsigkos
- Orphan Medicines Office, European Medicines Agency, Amsterdam, The Netherlands
| | - Dinko Vitezic
- Committee of Orphan Medicinal Products, European Medicines Agency, Amsterdam, The Netherlands; University of Rijeka Medical School and University Hospital Centre Rijeka, Rijeka, Croatia
| | - Kristina Larsson
- Orphan Medicines Office, European Medicines Agency, Amsterdam, The Netherlands
| | - Bruno Sepodes
- Committee of Orphan Medicinal Products, European Medicines Agency, Amsterdam, The Netherlands; INFARMED - Autoridade Nacional do Medicamento, Lisbon, Portugal; Universidade de Lisboa - Faculdade de Farmácia, Lisbon, Portugal
| | - Violeta Stoyanova-Beninska
- Committee of Orphan Medicinal Products, European Medicines Agency, Amsterdam, The Netherlands; Medicines Evaluation Board, Utrecht, The Netherlands
| |
Collapse
|
8
|
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] [Scholar 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
|
9
|
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] [Scholar 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
|
10
|
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 PMCID: PMC6816429 DOI: 10.1080/21645515.2019.1627820] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 05/15/2019] [Accepted: 06/02/2019] [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
|
11
|
Li Q, Qin Z, Wang Q, Xu T, Yang Y, He Z. Applications of Genome Editing Technology in Animal Disease Modeling and Gene Therapy. Comput Struct Biotechnol J 2019; 17:689-698. [PMID: 31303973 PMCID: PMC6603303 DOI: 10.1016/j.csbj.2019.05.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 05/24/2019] [Accepted: 05/26/2019] [Indexed: 02/05/2023] Open
Abstract
Genome editing technology is a technique for targeted genetic modifications, enabling the knockout and addition of specific DNA fragments. This technology has been widely used in various types of biomedical research, clinics and agriculture. In terms of disease research, constructing appropriate animal models is necessary. Combining reproductive technology with genome editing, many animal disease models have been generated for basic and clinical research. In addition, precisely targeted modifications allow genome editing to flourish in the field of gene therapy. Many mutations refractory to traditional gene therapy could be permanently corrected at the DNA level. Thus, genome editing is undoubtedly a promising technology for gene therapy. In this review, we mainly introduce the applications of genome editing in constructing animal disease models and gene therapies, as well as its future prospects and challenges.
Collapse
Affiliation(s)
- Qian Li
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Zhou Qin
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Qingnan Wang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Ting Xu
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Yang Yang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Zhiyao He
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| |
Collapse
|
12
|
Perdiguero B, Gómez CE, Esteban M. Bioluminescence Imaging as a Tool for Poxvirus Biology. Methods Mol Biol 2019; 2023:269-285. [PMID: 31240684 DOI: 10.1007/978-1-4939-9593-6_17] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Bioluminescence imaging, with luciferase as a reporter-encoding gene, has been successfully and widely used for studies to follow viral infection in an organism and to measure therapeutic efficacy of antiviral agents in small animal models. Bioluminescence is produced by the reaction of a luciferase enzyme stably inserted into the viral genome with a defined substrate systemically delivered into the animal. The light emitted is captured allowing the detection of viral infection sites and the quantification of viral replication in the context of tissues of a living animal. The goal of this chapter is to provide a technical background for the evaluation of poxvirus infection in cells and animals through bioluminescence imaging technology using luciferase-expressing recombinant poxviruses.
Collapse
Affiliation(s)
- Beatriz Perdiguero
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, CSIC, Madrid, Spain
| | - Carmen Elena Gómez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, CSIC, Madrid, Spain
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, CSIC, Madrid, Spain.
| |
Collapse
|
13
|
Noto FK, Adjan-Steffey V, Tong M, Ravichandran K, Zhang W, Arey A, McClain CB, Ostertag E, Mazhar S, Sangodkar J, DiFeo A, Crawford J, Narla G, Jamling TY. Sprague Dawley Rag2-Null Rats Created from Engineered Spermatogonial Stem Cells Are Immunodeficient and Permissive to Human Xenografts. Mol Cancer Ther 2018; 17:2481-2489. [PMID: 30206106 DOI: 10.1158/1535-7163.mct-18-0156] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 07/10/2018] [Accepted: 09/05/2018] [Indexed: 12/22/2022]
Abstract
The rat is the preferred model for toxicology studies, and it offers distinctive advantages over the mouse as a preclinical research model including larger sample size collection, lower rates of drug clearance, and relative ease of surgical manipulation. An immunodeficient rat would allow for larger tumor size development, prolonged dosing and drug efficacy studies, and preliminary toxicologic testing and pharmacokinetic/pharmacodynamic studies in the same model animal. Here, we created an immunodeficient rat with a functional deletion of the Recombination Activating Gene 2 (Rag2) gene, using genetically modified spermatogonial stem cells (SSC). We targeted the Rag2 gene in rat SSCs with TALENs and transplanted these Rag2-deficient SSCs into sterile recipients. Offspring were genotyped, and a founder with a 27 bp deletion mutation was identified and bred to homozygosity to produce the Sprague-Dawley Rag2 - Rag2 tm1Hera (SDR) knockout rat. We demonstrated that SDR rat lacks mature B and T cells. Furthermore, the SDR rat model was permissive to growth of human glioblastoma cell line subcutaneously resulting in successful growth of tumors. In addition, a human KRAS-mutant non-small cell lung cancer cell line (H358), a patient-derived high-grade serous ovarian cancer cell line (OV81), and a patient-derived recurrent endometrial cancer cell line (OV185) were transplanted subcutaneously to test the ability of the SDR rat to accommodate human xenografts from multiple tissue types. All human cancer cell lines showed efficient tumor uptake and growth kinetics indicating that the SDR rat is a viable host for a range of xenograft studies. Mol Cancer Ther; 17(11); 2481-9. ©2018 AACR.
Collapse
Affiliation(s)
| | | | - Min Tong
- Poseida Therapeutics Inc., San Diego, California
| | | | - Wei Zhang
- Hera BioLabs Inc., Lexington, Kentucky
| | | | | | - Eric Ostertag
- Transposagen Biopharmaceuticals Inc., Lexington, Kentucky
| | - Sahar Mazhar
- Case Western Reserve University, Cleveland, Ohio
| | | | | | - Jack Crawford
- Hera BioLabs Inc., Lexington, Kentucky.,Transposagen Biopharmaceuticals Inc., Lexington, Kentucky
| | - Goutham Narla
- Hera BioLabs Inc., Lexington, Kentucky.,The University of Michigan, Ann Arbor, Michigan
| | - Tseten Y Jamling
- Hera BioLabs Inc., Lexington, Kentucky. .,Transposagen Biopharmaceuticals Inc., Lexington, Kentucky
| |
Collapse
|
14
|
He D, Zhang J, Wu W, Yi N, He W, Lu P, Li B, Yang N, Wang D, Xue Z, Zhang P, Fan G, Zhu X. A novel immunodeficient rat model supports human lung cancer xenografts. FASEB J 2018; 33:140-150. [PMID: 29944447 DOI: 10.1096/fj.201800102rr] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Patient-derived xenograft (PDX) animal models allow the exogenous growth of human tumors, offering an irreplaceable preclinical tool for oncology research. Mice are the most commonly used host for human PDX models, however their small body size limits the xenograft growth, sample collection, and drug evaluation. Therefore, we sought to develop a novel rat model that could overcome many of these limitations. We knocked out Rag1, Rag2, and Il2rg in Sprague Dawley (SD) rats by clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated protein 9 technology. The development of lymphoid organs is significantly impaired in Rag1-/-Rag2-/-Il2rg-/Y (designated as SD-RG) rats. Consequently, SD-RG rats are severely immunodeficient with an absence of mature T, B, and NK cells in the immune system. After subcutaneous injection of tumor cell lines of different origin, such as NCI-H460, U-87MG, and MDA-MB-231, the tumors grow significantly faster and larger in SD-RG rats than in nonobese diabetic- Cg-Prkdcscid Il2rgtm1Wjl/SzJ mice. Most important of all, we successfully established a PDX model of lung squamous cell carcinoma in which the grafts recapitulate the histopathologic features of the primary tumor for several passages. In conclusion, the severely immunodeficient SD-RG rats support fast growth of PDX compared with mice, thus holding great potential to serve as a new model for oncology research.-He, D., Zhang, J., Wu, W., Yi, N., He, W., Lu, P., Li, B., Yang, N., Wang, D., Xue, Z., Zhang, P., Fan, G., Zhu, X. A novel immunodeficient rat model supports human lung cancer xenografts.
Collapse
Affiliation(s)
- Di He
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Junhui Zhang
- Department of Regenerative Medicine, Translational Center for Stem Cell Research, Tongji Hospital, Tongji University Suzhou Institute, Tongji University School of Medicine, Shanghai, China
| | - Wanwan Wu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Ning Yi
- Department of Regenerative Medicine, Translational Center for Stem Cell Research, Tongji Hospital, Tongji University Suzhou Institute, Tongji University School of Medicine, Shanghai, China
| | - Wen He
- Department of Regenerative Medicine, Translational Center for Stem Cell Research, Tongji Hospital, Tongji University Suzhou Institute, Tongji University School of Medicine, Shanghai, China
| | - Ping Lu
- Department of Regenerative Medicine, Translational Center for Stem Cell Research, Tongji Hospital, Tongji University Suzhou Institute, Tongji University School of Medicine, Shanghai, China
| | - Bin Li
- Alphacait AL Biotech Company, Hangzhou, China
| | - Nan Yang
- PharmaLegacy Laboratories Company, Shanghai, China; and
| | - Di Wang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Zhigang Xue
- Department of Regenerative Medicine, Translational Center for Stem Cell Research, Tongji Hospital, Tongji University Suzhou Institute, Tongji University School of Medicine, Shanghai, China
| | - Peng Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Guoping Fan
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China.,Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Xianmin Zhu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| |
Collapse
|
15
|
Liu Q, Zhou S, Fan C, Huang W, Li Q, Liu S, Wu X, Li B, Wang Y. Biodistribution and residence time of adenovector serotype 5 in normal and immunodeficient mice and rats detected with bioluminescent imaging. Sci Rep 2017; 7:3597. [PMID: 28620164 PMCID: PMC5472566 DOI: 10.1038/s41598-017-03852-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 05/08/2017] [Indexed: 12/03/2022] Open
Abstract
As concerns increase about adenovirus type 5 (Ad5) being a safe gene transfer vector, it is important to evaluate its distribution, residence time, and possible toxicity in immunodeficient populations. To characterize the potential risk associated with different Ad5 vector delivery modes, we used immunocompetent and immunodeficient Rag2 -/- animals to establish mouse and rat models that could be monitored with bioluminescent imaging following intramuscular or intravascular infection with an engineered replication-incompetent Ad5 virus carrying the firefly luciferase gene (Ad5-Fluc). The Ad5 vector was less well-tolerated by Rag2 -/- animals than by wildtype ones, with delayed residence time, wider virus dissemination, less weight gain, and relatively severe pathological changes. In intravascularly Ad5-Fluc-infected Rag2 -/- mice, systemic virus dissemination extended from the abdomen to the limbs and head on day 9 post-infection. Additionally, significant increases in plasma TNF-α and IFN-γ, which may be important factors in the heightened immunopathology in the liver and brain, were detected in the Rag2 -/- mice 30 days after intravascular delivery. The Ad5 vector was better tolerated after intramuscular delivery than after intravascular delivery. Ad5-Fluc/Rag2 -/- mice and rats can be used as reliable models of an immunodeficient population in which to evaluate the safety of Ad5-vectored vaccines or gene therapy products.
Collapse
Affiliation(s)
- Qiang Liu
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Shuya Zhou
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Changfa Fan
- 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
| | - 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
| | - Xi Wu
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Baowen Li
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Youchun Wang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control, Beijing, 100050, China.
| |
Collapse
|
16
|
Liu Q, Fan C, Li Q, Zhou S, Huang W, Wang L, Sun C, Wang M, Wu X, Ma J, Li B, Xie L, Wang Y. Antibody-dependent-cellular-cytotoxicity-inducing antibodies significantly affect the post-exposure treatment of Ebola virus infection. Sci Rep 2017; 7:45552. [PMID: 28358050 PMCID: PMC5372081 DOI: 10.1038/srep45552] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 02/28/2017] [Indexed: 01/11/2023] Open
Abstract
Passive immunotherapy with monoclonal antibodies (mAbs) is an efficacious treatment for Ebola virus (EBOV) infections in animal models and humans. Understanding what constitutes a protective response is critical for the development of novel therapeutic strategies. We generated an EBOV-glycoprotein-pseudotyped Human immunodeficiency virus to develop sensitive neutralizing and antibody-dependent cellular cytotoxicity (ADCC) assays as well as a bioluminescent-imaging-based mouse infection model that does not require biosafety level 4 containment. The in vivo treatment efficiencies of three novel anti-EBOV mAbs at 12 h post-infection correlated with their in vitro anti-EBOV ADCC activities, without neutralizing activity. When they were treated with these mAbs, natural killer cell (NK)-deficient mice had lower viral clearance than WT mice, indicating that the anti-EBOV mechanism of the ADCC activity of these mAbs is predominantly mediated by NK cells. One potent anti-EBOV mAb (M318) displayed unprecedented neutralizing and ADCC activities (neutralization IC50, 0.018 μg/ml; ADCC EC50, 0.095 μg/ml). These results have important implications for the efficacy of antiviral drugs and vaccines as well as for pathogenicity studies of EBOV.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/biosynthesis
- Antibodies, Monoclonal/immunology
- Antibodies, Neutralizing/administration & dosage
- Antibodies, Viral/administration & dosage
- Antibody-Dependent Cell Cytotoxicity
- Disease Models, Animal
- Female
- Hemorrhagic Fever, Ebola/drug therapy
- Hemorrhagic Fever, Ebola/immunology
- Humans
- Killer Cells, Natural/immunology
- Mice, Inbred BALB C
- Mice, Inbred C57BL
Collapse
Affiliation(s)
- Qiang Liu
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control, Beijing 100050, China
| | - Changfa Fan
- Division of Animal Model Research, Institute for Laboratory Animal Resources, 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
| | - Shuya Zhou
- 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
| | - Lan Wang
- Division of Monoclonal Antibody, National Institutes for Food and Drug Control, Beijing 100050, China
| | | | - Meng Wang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, 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
| | - Jian Ma
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control, Beijing 100050, China
| | - Baowen Li
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control, Beijing 100050, China
| | | | - Youchun Wang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control, Beijing 100050, China
| |
Collapse
|
17
|
Nie J, Wu X, Ma J, Cao S, Huang W, Liu Q, Li X, Li Y, Wang Y. Development of in vitro and in vivo rabies virus neutralization assays based on a high-titer pseudovirus system. Sci Rep 2017; 7:42769. [PMID: 28218278 PMCID: PMC5316940 DOI: 10.1038/srep42769] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 01/13/2017] [Indexed: 12/25/2022] Open
Abstract
Pseudoviruses are useful virological tools because of their safety and versatility; however the low titer of these viruses substantially limits their wider applications. We developed a highly efficient pseudovirus production system capable of yielding 100 times more rabies pseudovirus than the traditional method. Employing the high-titer pseudoviruses, we have developed robust in vitro and in vivo neutralization assays for the evaluation of rabies vaccine, which traditionally relies on live-virus based assays. Compared with current rapid fluorescent focus inhibition test (RFFIT), our in vitro pseudovirus-based neutralization assay (PBNA) is much less labor-intensive while demonstrating better reproducibility. Moreover, the in vivo PBNA assay was also found to be superior to the live virus based assay. Following intravenous administration, the pseudovirus effectively infected the mice, with dynamic viral distributions being sequentially observed in spleen, liver and brain. Furthermore, data from in vivo PBNA showed great agreement with those generated from the live virus model but with the experimental time significantly reduced from 2 weeks to 3 days. Taken together, the effective pseudovirus production system facilitated the development of novel PBNA assays which could replace live virus-based traditional assays due to its safety, rapidity, reproducibility and high throughput capacity.
Collapse
Affiliation(s)
- Jianhui Nie
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC), No. 2 Tiantanxili, Beijing 100050, China
| | - Xiaohong Wu
- Division of Arboviral Vaccines, National Institutes for Food and Drug Control (NIFDC), No. 2 Tiantanxili, Beijing 100050, China
| | - Jian Ma
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC), No. 2 Tiantanxili, Beijing 100050, China
| | - Shouchun Cao
- Division of Arboviral Vaccines, National Institutes for Food and Drug Control (NIFDC), No. 2 Tiantanxili, Beijing 100050, China
| | - Weijin Huang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC), No. 2 Tiantanxili, Beijing 100050, China
| | - Qiang Liu
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC), No. 2 Tiantanxili, Beijing 100050, China
| | - Xuguang Li
- Centre for Biologics Evaluation, Biologics and Genetic Therapies Directorate, Health Canada, Ottawa, On K1A 0K9, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, On, Canada
| | - Yuhua Li
- Division of Arboviral Vaccines, National Institutes for Food and Drug Control (NIFDC), No. 2 Tiantanxili, Beijing 100050, China
| | - Youchun Wang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC), No. 2 Tiantanxili, Beijing 100050, China
| |
Collapse
|