153 results on '"Cheng‐Feng Qin"'
Search Results
2. Aerosolized Zika virus infection in Guinea pigs
- Author
-
Hong-Ying, Qiu, Na-Na, Zhang, Qing-Qing, Ma, Rui-Ting, Li, Meng-Yue, Guan, Li-Li, Zhang, Jia, Zhou, Rong-Rong, Zhang, Xing-Yao, Huang, Wen-Hui, Yang, Yong-Qiang, Deng, Cheng-Feng, Qin, and Dong-Sheng, Zhou
- Subjects
Zika Virus Infection ,Epidemiology ,Guinea Pigs ,Immunology ,Zika Virus ,General Medicine ,Microbiology ,Infectious Disease Transmission, Vertical ,Immunity, Humoral ,Infectious Diseases ,Virology ,Drug Discovery ,Animals ,Humans ,Parasitology ,Viremia - Abstract
Zika virus (ZIKV) is primarily transmitted through mosquito bites and sexual contact, and vertical transmission of ZIKV has also been observed in humans. In addition, ZIKV infection via unknown transmission routes has been frequently reported in clinical settings. However, whether ZIKV can be transmitted via aerosol routes remains unknown. In this study, we demonstrated that aerosolized ZIKV is fully infectious in vitro and in vivo. Remarkably, intratracheal (i.t.) inoculation with aerosolized ZIKV led to rapid viremia and viral secretion in saliva, as well as robust humoral and innate immune responses in guinea pigs. Transcriptome analysis further revealed that the expression of genes related to viral processes, biological regulation and the immune response was significantly changed. Together, our results confirm that aerosolized ZIKV can result in systemic infection and induce both innate and adaptive immune responses in guinea pigs, highlighting the possibility of ZIKV transmission via aerosols.
- Published
- 2022
- Full Text
- View/download PDF
3. Comparative characterization of SARS‐CoV‐2 variants of concern and mouse‐adapted strains in mice
- Author
-
Qi Chen, Xing‐Yao Huang, Yu Liu, Meng‐Xu Sun, Bin Ji, Chao Zhou, Hang Chi, Rong‐Rong Zhang, Dan Luo, Ying Tian, Xiao‐Feng Li, Hui Zhao, and Cheng‐Feng Qin
- Subjects
Mice ,Infectious Diseases ,SARS-CoV-2 ,Virology ,Spike Glycoprotein, Coronavirus ,Animals ,COVID-19 ,Humans ,Protein Binding - Abstract
SARS-CoV-2 has evolved into a panel of variants of concern (VOCs) and constituted a sustained threat to global health. The wildtype (WT) SARS-CoV-2 isolates fail to infect mice, while the Beta variant, one of the VOCs, has acquired the capability to infect standard laboratory mice, raising a spreading risk of SARS-CoV-2 from humans to mice. However, the infectivity and pathogenicity of other VOCs in mice remain not fully understood. In this study, we systematically investigated the infectivity and pathogenicity of three VOCs, Alpha, Beta, and Delta, in mice in comparison with two well-understood SARS-CoV-2 mouse-adapted strains, MASCp6 and MASCp36, sharing key mutations in the receptor-binding domain (RBD) with Alpha or Beta, respectively. Our results showed that the Beta variant had the strongest infectivity and pathogenicity among the three VOCs, while the Delta variant only caused limited replication and mild pathogenic changes in the mouse lung, which is much weaker than what the Alpha variant did. Meanwhile, Alpha showed comparable infectivity in lungs in comparison with MASCp6, and Beta only showed slightly lower infectivity in lungs when compared with MASCp36. These results indicated that all three VOCs have acquired the capability to infect mice, highlighting the ongoing spillover risk of SARS-CoV-2 from humans to mice during the continued evolution of SARS-CoV-2, and that the key amino acid mutations in the RBD of mouse-adapted strains may be referenced as an early-warning indicator for predicting the spillover risk of newly emerging SARS-CoV-2 variants.
- Published
- 2022
- Full Text
- View/download PDF
4. Lipid nanoparticle-encapsulated mRNA antibody provides long-term protection against SARS-CoV-2 in mice and hamsters
- Author
-
Yong-Qiang Deng, Na-Na Zhang, Yi-Fei Zhang, Xia Zhong, Sue Xu, Hong-Ying Qiu, Tie-Cheng Wang, Hui Zhao, Chao Zhou, Shu-Long Zu, Qi Chen, Tian-Shu Cao, Qing Ye, Hang Chi, Xiang-Hui Duan, Dan-Dan Lin, Xiao-Jing Zhang, Liang-Zhi Xie, Yu-Wei Gao, Bo Ying, and Cheng-Feng Qin
- Subjects
SARS-CoV-2 ,COVID-19 ,Cell Biology ,Antibodies, Viral ,Antibodies, Neutralizing ,Mice ,Cricetinae ,Liposomes ,Spike Glycoprotein, Coronavirus ,Animals ,Humans ,Nanoparticles ,RNA, Messenger ,Pandemics ,Molecular Biology - Abstract
Monoclonal antibodies represent important weapons in our arsenal to against the COVID-19 pandemic. However, this potential is severely limited by the time-consuming process of developing effective antibodies and the relative high cost of manufacturing. Herein, we present a rapid and cost-effective lipid nanoparticle (LNP) encapsulated-mRNA platform for in vivo delivery of SARS-CoV-2 neutralization antibodies. Two mRNAs encoding the light and heavy chains of a potent SARS-CoV-2 neutralizing antibody HB27, which is currently being evaluated in clinical trials, were encapsulated into clinical grade LNP formulations (named as mRNA-HB27-LNP). In vivo characterization demonstrated that intravenous administration of mRNA-HB27-LNP in mice resulted in a longer circulating half-life compared with the original HB27 antibody in protein format. More importantly, a single prophylactic administration of mRNA-HB27-LNP provided protection against SARS-CoV-2 challenge in mice at 1, 7 and even 63 days post administration. In a close contact transmission model, prophylactic administration of mRNA-HB27-LNP prevented SARS-CoV-2 infection between hamsters in a dose-dependent manner. Overall, our results demonstrate a superior long-term protection against SARS-CoV-2 conferred by a single administration of this unique mRNA antibody, highlighting the potential of this universal platform for antibody-based disease prevention and therapy against COVID-19 as well as a variety of other infectious diseases.
- Published
- 2022
- Full Text
- View/download PDF
5. Memory B cell repertoire from triple vaccinees against diverse SARS-CoV-2 variants
- Author
-
Kang Wang, Zijing Jia, Linilin Bao, Lei Wang, Lei Cao, Hang Chi, Yaling Hu, Qianqian Li, Yunjiao Zhou, Yinan Jiang, Qianhui Zhu, Yongqiang Deng, Pan Liu, Nan Wang, Lin Wang, Min Liu, Yurong Li, Boling Zhu, Kaiyue Fan, Wangjun Fu, Peng Yang, Xinran Pei, Zhen Cui, Lili Qin, Pingju Ge, Jiajing Wu, Shuo Liu, Yiding Chen, Weijin Huang, Qiao Wang, Cheng-Feng Qin, Youchun Wang, Chuan Qin, and Xiangxi Wang
- Subjects
COVID-19 Vaccines ,Multidisciplinary ,SARS-CoV-2 ,Antibodies, Monoclonal ,COVID-19 ,Antibodies, Viral ,Antibodies, Neutralizing ,Disease Models, Animal ,Mice ,Memory B Cells ,Neutralization Tests ,Spike Glycoprotein, Coronavirus ,Animals ,Humans - Abstract
Omicron (B.1.1.529), the most heavily mutated SARS-CoV-2 variant so far, is highly resistant to neutralizing antibodies, raising concerns about the effectiveness of antibody therapies and vaccines1,2. Here we examined whether sera from individuals who received two or three doses of inactivated SARS-CoV-2 vaccine could neutralize authentic Omicron. The seroconversion rates of neutralizing antibodies were 3.3% (2 out of 60) and 95% (57 out of 60) for individuals who had received 2 and 3 doses of vaccine, respectively. For recipients of three vaccine doses, the geometric mean neutralization antibody titre for Omicron was 16.5-fold lower than for the ancestral virus (254). We isolated 323 human monoclonal antibodies derived from memory B cells in triple vaccinees, half of which recognized the receptor-binding domain, and showed that a subset (24 out of 163) potently neutralized all SARS-CoV-2 variants of concern, including Omicron. Therapeutic treatments with representative broadly neutralizing monoclonal antibodies were highly protective against infection of mice with SARS-CoV-2 Beta (B.1.351) and Omicron. Atomic structures of the Omicron spike protein in complex with three classes of antibodies that were active against all five variants of concern defined the binding and neutralizing determinants and revealed a key antibody escape site, G446S, that confers greater resistance to a class of antibodies that bind on the right shoulder of the receptor-binding domain by altering local conformation at the binding interface. Our results rationalize the use of three-dose immunization regimens and suggest that the fundamental epitopes revealed by these broadly ultrapotent antibodies are rational targets for a universal sarbecovirus vaccine.
- Published
- 2022
- Full Text
- View/download PDF
6. Identification of an immunogenic epitope and protective antibody against the furin cleavage site of SARS-CoV-2
- Author
-
Lili, Li, Meiling, Gao, Jie, Li, Xuping, Xie, Hui, Zhao, Yanan, Wang, Xin, Xu, Shulong, Zu, Chunfeng, Chen, Dingyi, Wan, Jing, Duan, Jingfeng, Wang, Saba R, Aliyari, Sarah, Gold, Jicai, Zhang, Cheng-Feng, Qin, Pei-Yong, Shi, Heng, Yang, and Genhong, Cheng
- Subjects
Furin ,Immunogenic epitope ,SARS-CoV-2 ,Prevention ,Mononuclear ,Clinical Sciences ,COVID-19 ,General Medicine ,General Biochemistry, Genetics and Molecular Biology ,Antibodies ,S protein ,Vaccine Related ,Mice ,Epitopes ,Furin site ,Emerging Infectious Diseases ,Infectious Diseases ,Good Health and Well Being ,Biodefense ,Antibody Formation ,Leukocytes ,Pneumonia & Influenza ,Public Health and Health Services ,Animals ,Immunization - Abstract
BackgroundSevere acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of the global coronavirus disease 2019 (COVID-19) pandemic, contains a unique, four amino acid (aa) "PRRA" insertion in the spike (S) protein that creates a transmembrane protease serine 2 (TMPRSS2)/furin cleavage site and enhances viral infectivity. More research into immunogenic epitopes and protective antibodies against this SARS-CoV-2 furin cleavage site is needed.MethodsCombining computational and experimental methods, we identified and characterized an immunogenic epitope overlapping the furin cleavage site that detects antibodies in COVID-19 patients and elicits strong antibody responses in immunized mice. We also identified a high-affinity monoclonal antibody from COVID-19 patient peripheral blood mononuclear cells; the antibody directly binds the furin cleavage site and protects against SARS-CoV-2 infection in a mouse model.FindingsThe presence of "PRRA" amino acids in the S protein of SARS-CoV-2 not only creates a furin cleavage site but also generates an immunogenic epitope that elicits an antibody response in COVID-19 patients. An antibody against this epitope protected against SARS-CoV-2 infection in mice.InterpretationThe immunogenic epitope and protective antibody we have identified may augment our strategy in handling COVID-19 epidemic.FundingThe National Natural Science Foundation of China (82102371, 91542201, 81925025, 82073181, and 81802870), the Chinese Academy of Medical Sciences Initiative for Innovative Medicine (2021-I2M-1-047 and 2022-I2M-2-004), the Non-profit Central Research Institute Fund of the Chinese Academy of Medical Sciences (2020-PT310-006, 2019XK310002, and 2018TX31001), the National Key Research and Development Project of China (2020YFC0841700), US National Institute of Health (NIH) funds grant AI158154, University of California Los Angeles (UCLA) AI and Charity Treks, and UCLA DGSOM BSCRC COVID-19 Award Program. H.Y. is supported by Natural Science Foundation of Jiangsu Province (BK20211554 andBE2022728).
- Published
- 2023
7. Long-term stability and protection efficacy of the RBD-targeting COVID-19 mRNA vaccine in nonhuman primates
- Author
-
Hui Zhao, Tie-Cheng Wang, Xiao-Feng Li, Na-Na Zhang, Liang Li, Chao Zhou, Yong-Qiang Deng, Tian-Shu Cao, Guan Yang, Rui-Ting Li, Yi-Jiao Huang, Yuan-Guo Li, Yi-Ming Zhang, Fang-Xu Li, Yu-Ren Zhou, Yu-Hang Jiang, Xi-Shan Lu, Shi-Hui Sun, Meng-Li Cheng, Kai-Ping Gu, Mei Zhang, Qing-Qing Ma, Xiao Yang, Bo Ying, Yu-Wei Gao, and Cheng-Feng Qin
- Subjects
Cancer Research ,Vaccines ,COVID-19 Vaccines ,SARS-CoV-2 ,QH301-705.5 ,COVID-19 ,Antibodies, Viral ,Antibodies, Neutralizing ,Article ,Macaca fascicularis ,Immunogenicity, Vaccine ,Chlorocebus aethiops ,Spike Glycoprotein, Coronavirus ,Genetics ,Animals ,Humans ,Medicine ,mRNA Vaccines ,Biology (General) ,Vero Cells - Abstract
Messenger RNA (mRNA) vaccine technology has shown its power in preventing the ongoing COVID-19 pandemic. Two mRNA vaccines targeting the full-length S protein of SARS-CoV-2 have been authorized for emergency use. Recently, we have developed a lipid nanoparticle-encapsulated mRNA (mRNA-LNP) encoding the receptor-binding domain (RBD) of SARS-CoV-2 (termed ARCoV), which confers complete protection in mouse model. Herein, we further characterized the protection efficacy of ARCoV in nonhuman primates and the long-term stability under normal refrigerator temperature. Intramuscular immunization of two doses of ARCoV elicited robust neutralizing antibodies as well as cellular response against SARS-CoV-2 in cynomolgus macaques. More importantly, ARCoV vaccination in macaques significantly protected animals from acute lung lesions caused by SARS-CoV-2, and viral replication in lungs and secretion in nasal swabs were completely cleared in all animals immunized with low or high doses of ARCoV. No evidence of antibody-dependent enhancement of infection was observed throughout the study. Finally, extensive stability assays showed that ARCoV can be stored at 2–8 °C for at least 6 months without decrease of immunogenicity. All these promising results strongly support the ongoing clinical trial.
- Published
- 2021
8. Enhanced protective immunity against SARS-CoV-2 elicited by a VSV vector expressing a chimeric spike protein
- Author
-
Cheng-Feng Qin, Yongchun Li, Fei Yuan, Jiankai Liu, Jianguo Zhao, Shujun Liu, Aihua Zheng, Junfeng Hao, Chaoyue Zhao, Hongyue Li, Yong-Qiang Deng, Dan Wen, Dong Li, Yuhang Zhang, Hongde Xu, Shuguang Duo, Jiandong Liu, and Yong Wu
- Subjects
Cancer Research ,COVID-19 Vaccines ,QH301-705.5 ,Recombinant Fusion Proteins ,viruses ,medicine.disease_cause ,Antibodies, Viral ,Article ,law.invention ,Chimera (genetics) ,Mice ,Immune system ,Antigen ,law ,Neutralization Tests ,Genetics ,medicine ,Animals ,Humans ,Gene Knock-In Techniques ,Biology (General) ,Neutralizing antibody ,skin and connective tissue diseases ,Coronavirus ,Vaccines ,biology ,SARS-CoV-2 ,fungi ,COVID-19 ,Virology ,Antibodies, Neutralizing ,respiratory tract diseases ,Transplantation ,Mice, Inbred C57BL ,body regions ,Titer ,HEK293 Cells ,Spike Glycoprotein, Coronavirus ,Recombinant DNA ,biology.protein ,Medicine - Abstract
SARS-CoV-2 and SARS-CoV are genetically related coronavirus and share the same cellular receptor ACE2. By replacing the VSV glycoprotein with the spikes (S) of SARS-CoV-2 and SARS-CoV, we generated two replication-competent recombinant viruses, rVSV-SARS-CoV-2 and rVSV-SARS-CoV. Using wild-type and human ACE2 (hACE2) knock-in mouse models, we found a single dose of rVSV-SARS-CoV could elicit strong humoral immune response via both intranasal (i.n.) and intramuscular (i.m.) routes. Despite the high genetic similarity between SARS-CoV-2 and SARS-CoV, no obvious cross-neutralizing activity was observed in the immunized mice sera. In macaques, neutralizing antibody (NAb) titers induced by one i.n. dose of rVSV-SARS-CoV-2 were eight-fold higher than those by a single i.m. dose. Thus, our data indicates that rVSV-SARS-CoV-2 might be suitable for i.n. administration instead of the traditional i.m. immunization in human. Because rVSV-SARS-CoV elicited significantly stronger NAb responses than rVSV-SARS-CoV-2 in a route-independent manner, we generated a chimeric antigen by replacing the receptor binding domain (RBD) of SARS-CoV S with that from the SARS-CoV-2. rVSV expressing the chimera (rVSV-SARS-CoV/2-RBD) induced significantly increased NAbs against SARS-CoV-2 in mice and macaques than rVSV-SARS-CoV-2, with a safe Th1-biased response. Serum immunized with rVSV-SARS-CoV/2-RBD showed no cross-reactivity with SARS-CoV. hACE2 mice receiving a single i.m. dose of either rVSV-SARS-CoV-2 or rVSV-SARS-CoV/2-RBD were fully protected against SARS-CoV-2 challenge without obvious lesions in the lungs. Our results suggest that transplantation of SARS-CoV-2 RBD into the S protein of SARS-CoV might be a promising antigen design for COVID-19 vaccines.
- Published
- 2021
9. Pathogenicity and Structural Basis of Zika Variants with Glycan Loop Deletions in the Envelope Protein
- Author
-
Meng-Li Cheng, Yun-Xiang Yang, Zhong-Yu Liu, Dan Wen, Pan Yang, Xing-Yao Huang, Hao-Long Dong, Yan-Peng Xu, Xiao-Feng Li, Yong-Qiang Deng, Qing Ye, Ling Zhu, Juan Li, Andrew D. Davidson, Ai-Hua Zheng, Wei-Feng Shi, Hui Zhao, Xiang-Xi Wang, and Cheng-Feng Qin
- Subjects
Virulence ,Zika Virus Infection ,Immunology ,Zika Virus ,Virus Replication ,Microbiology ,Mice ,Disease Models, Animal ,Viral Envelope Proteins ,Polysaccharides ,Virology ,Insect Science ,Pathogenesis and Immunity ,Animals - Abstract
The glycan loop of Zika virus (ZIKV) envelope protein (E) contains the glycosylation site and has been well documented to be important for viral pathogenesis and transmission. In the present study, we report that deletions in the E glycan loop, which were recorded in African ZIKV strains previously, have re-emerged in their contemporary Asian lineages. Here, we generated recombinant ZIKV containing specific deletions in the E glycan loop by reverse genetics. Extensive in vitro and in vivo characterization of these deletion mutants demonstrated an attenuated phenotype in an adult A129 mouse model and reduced oral infections in mosquitoes. Surprisingly, these glycan loop deletion mutants exhibited an enhanced neurovirulence phenotype, and resulted in a more severe microcephalic brain in neonatal mouse models. Crystal structures of the ZIKV E protein and a deletion mutant at 2.5 and 2.6 Å, respectively, revealed that deletion of the glycan loop induces encephalitic flavivirus-like conformational alterations, including the appearance of perforations on the surface and a clear change in the topology of the loops. Overall, our results demonstrate that the E glycan loop deletions represent neonatal mouse neurovirulence markers of ZIKV. IMPORTANCE Zika virus (ZIKV) has been identified as a cause of microcephaly and acquired evolutionary mutations since its discovery. Previously deletions in the E glycan loop were recorded in African ZIKV strains, which have re-emerged in the contemporary Asian lineages recently. The glycan loop deletion mutants are not glycosylated, which are attenuated in adult A129 mouse model and reduced oral infections in mosquitoes. More importantly, the glycan loop deletion mutants induce an encephalitic flavivirus-like conformational alteration in the E homodimer, resulting in a significant enhancement of neonatal mouse neurovirulence. This study underscores the critical role of glycan loop deletion mutants in ZIKV pathogenesis, highlighting a need for global virological surveillance for such ZIKV variants.
- Published
- 2022
- Full Text
- View/download PDF
10. The Infection and Pathogenicity of SARS-CoV-2 Variant B.1.351 in hACE2 Mice
- Author
-
Xing-Yao Huang, Meng-Xu Sun, Ruiting Li, Guizhen Wu, Changfa Fan, Ying Tian, Chao Zhou, Cheng-Feng Qin, Qi Chen, and Rong-Rong Zhang
- Subjects
Models, Molecular ,2019-20 coronavirus outbreak ,medicine.medical_specialty ,Letter ,Time Factors ,Coronavirus disease 2019 (COVID-19) ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,DNA Mutational Analysis ,Immunology ,Mice, Transgenic ,Biology ,Evolution, Molecular ,South Africa ,Mice ,Medical microbiology ,Virology ,medicine ,Animals ,Humans ,Selection, Genetic ,Phylogeny ,Immune Evasion ,Virulence ,SARS-CoV-2 ,COVID-19 ,Pathogenicity ,Phylogeography ,Mutation ,Spike Glycoprotein, Coronavirus ,Molecular Medicine ,Genetic Fitness - Abstract
Continued uncontrolled transmission of SARS-CoV-2 in many parts of the world is creating conditions for substantial evolutionary changes to the virus
- Published
- 2021
- Full Text
- View/download PDF
11. Development of a Bicistronic Yellow Fever Live Attenuated Vaccine with Reduced Neurovirulence and Viscerotropism
- Author
-
Hong-Jiang Wang, Yan Guo, Meng-Jiao He, Zhong-Yu Liu, Qing Ye, Xing-Yao Huang, Yong-Qiang Deng, Xiao-Feng Li, and Cheng-Feng Qin
- Subjects
Microbiology (medical) ,Mammals ,General Immunology and Microbiology ,Ecology ,Physiology ,Yellow Fever Vaccine ,Cell Biology ,Internal Ribosome Entry Sites ,Vaccines, Attenuated ,Mice ,Infectious Diseases ,Yellow Fever ,Interferon Type I ,Genetics ,Humans ,Animals ,Yellow fever virus ,Antigens, Viral - Abstract
The yellow fever (YF) live attenuated vaccine strain 17D (termed 17D) has been widely used for the prevention and control of YF disease. However, 17D retains significant neurovirulence and viscerotropism in mice, which is probably linked to the increased occurrences of serious adverse events following 17D vaccination. Thus, the development of an updated version of the YF vaccine with an improved safety profile is of high priority. Here, we generated a viable bicistronic YF virus (YFV) by incorporating the internal ribosome entry site (IRES) from
- Published
- 2022
12. Immune profiles in mouse brain and testes infected by Zika virus with variable pathogenicity
- Author
-
Jingzhe Shang, Chunfeng Li, Zhujia Jin, Shulong Zu, Songjie Chen, Junlan Chen, Ziyi Chen, Hua Tang, Cheng-Feng Qin, Qing Ye, and Aiping Wu
- Subjects
Male ,Microbiology (medical) ,Virulence ,Zika Virus Infection ,Immunology ,Brain ,Zika Virus ,Microbiology ,Mice ,Infectious Diseases ,Testis ,Animals ,Humans ,Child - Abstract
The Zika virus is responsible for neurological diseases such as microcephaly, Guillain-Barré syndrome, neuropathy, and myelitis in human adults and children. Previous studies have shown that the Zika virus can infect nerve progenitor cells and interfere with neural development. However, it is unclear how the immune system responds to infection with Zika viruses with variable pathogenicity. Here, we used two Zika strains with relatively different pathogenicity, the Asian ancestral strain CAM/2010 and the America pandemic strain GZ01/2016, to infect the brains of mice. We found that both strains elicited a strong immune response. Notably, the strain with relatively high pathogenicity, GZ01/2016, caused more intense immune regulation, with stronger CD8+ T cell and macrophage activation at 14 days post infection (dpi), as well as a greater immune gene disturbance. Notably, several TNF family genes were upregulated at 14 dpi, including Tnfrsf9, Tnfsf13, Tnfrsf8, Cd40, and Tnfsf10. It was notable that GZ01/2016 could maintain the survival of nerve cells at 7dpi but caused neurological disorders at 14dpi. These results indicate that Zika viruses with high pathogenicity may induce sustained activation of the immune system leading to nerve tissue damage.
- Published
- 2022
- Full Text
- View/download PDF
13. Different Gene Networks Are Disturbed by Zika Virus Infection in A Mouse Microcephaly Model
- Author
-
Cheng-Feng Qin, Qing-Feng Wu, Yisheng Jiang, Cui Li, Jing Li, Yafei Chang, Qin Wang, Feng Zhang, and Zhiheng Xu
- Subjects
Programmed cell death ,Microcephaly ,Letter ,Gene regulatory network ,Biochemistry ,Zika virus ,Pathogenesis ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Genetics ,medicine ,Animals ,Gene Regulatory Networks ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,biology ,Signaling pathway ,Zika Virus Infection ,Autophagy ,Zika Virus ,Gene network ,biology.organism_classification ,medicine.disease ,Brain development ,Virology ,Disease Models, Animal ,Computational Mathematics ,Microglial cell activation ,030217 neurology & neurosurgery - Abstract
The association of Zika virus (ZIKV) infection with microcephaly has raised alarm worldwide. Their causal link has been confirmed in different animal models infected by ZIKV. However, the molecular mechanisms underlying ZIKV pathogenesis are far from clear. Hence, we performed global gene expression analysis of ZIKV-infected mouse brains to unveil the biological and molecular networks underpinning microcephaly. We found significant dysregulation of the sub-networks associated with brain development, immune response, cell death, microglial cell activation, and autophagy amongst others. We provided detailed analysis of the related complicated gene networks and the links between them. Additionally, we analyzed the signaling pathways that were likely to be involved. This report provides systemic insights into not only the pathogenesis, but also a path to the development of prophylactic and therapeutic strategies against ZIKV infection.
- Published
- 2020
- Full Text
- View/download PDF
14. Adaptation of SARS-CoV-2 in BALB/c mice for testing vaccine efficacy
- Author
-
Zhongpeng Zhao, Shaolong Chen, Jiangfan Li, Yan Guo, Gencheng Han, De Yan Luo, Guan Yang, Yuxian He, Shihui Sun, Yanxiao Wang, Yusen Zhou, Cheng-Feng Qin, Shibo Jiang, Yue Teng, Hang Fan, Xiliang Wang, Lei He, Na Na Zhang, Qi Chen, Yong Qiang Deng, Xiaofeng Li, Xiaolan Yang, Xiao Yang, Shusheng Geng, Li Yuchang, Hui Wang, Xiaoli Sheng, Yan Li, Hongjing Gu, Yujun Cui, Xiaojun Zhou, and Guangyu Zhao
- Subjects
0301 basic medicine ,viruses ,Mice ,Immunogenicity, Vaccine ,0302 clinical medicine ,Medicine ,Lung ,Research Articles ,Infectivity ,Mice, Inbred BALB C ,Vaccines, Synthetic ,Multidisciplinary ,Virulence ,biology ,Immunogenicity ,High-Throughput Nucleotide Sequencing ,Microbio ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Spike Glycoprotein, Coronavirus ,Female ,Angiotensin-Converting Enzyme 2 ,Coronavirus Infections ,Research Article ,COVID-19 Vaccines ,Pneumonia, Viral ,Mice, Transgenic ,Peptidyl-Dipeptidase A ,Deep sequencing ,BALB/c ,Clomiphene ,Betacoronavirus ,03 medical and health sciences ,Animals ,Humans ,Pandemics ,Administration, Intranasal ,business.industry ,SARS-CoV-2 ,R-Articles ,COVID-19 ,Viral Vaccines ,Virus Internalization ,Vaccine efficacy ,medicine.disease ,biology.organism_classification ,Virology ,Disease Models, Animal ,Pneumonia ,Tamoxifen ,030104 developmental biology ,Mutation ,Lung Diseases, Interstitial ,business ,Respiratory tract - Abstract
Modeling SARS-CoV-2 in mice Among the research tools necessary to develop medical interventions to treat severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections, high on the list are informative animal models with which to study viral pathogenesis. Gu et al. developed a mouse model in which a SARS-CoV-2 strain was infectious and could cause an inflammatory response and moderate pneumonia. Adaptation of this viral strain in the mouse appeared to be dependent on a critical amino acid change, Asn501 to Tyr (N501Y), within the receptor-binding domain of the viral spike protein. The new mouse model was used to study neutralizing antibodies and a vaccine candidate against the virus. Science, this issue p. 1603, A SARS-CoV-2 mouse model is used to study viral responses and the development of vaccine candidates., The ongoing coronavirus disease 2019 (COVID-19) pandemic has prioritized the development of small-animal models for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We adapted a clinical isolate of SARS-CoV-2 by serial passaging in the respiratory tract of aged BALB/c mice. The resulting mouse-adapted strain at passage 6 (called MASCp6) showed increased infectivity in mouse lung and led to interstitial pneumonia and inflammatory responses in both young and aged mice after intranasal inoculation. Deep sequencing revealed a panel of adaptive mutations potentially associated with the increased virulence. In particular, the N501Y mutation is located at the receptor binding domain (RBD) of the spike protein. The protective efficacy of a recombinant RBD vaccine candidate was validated by using this model. Thus, this mouse-adapted strain and associated challenge model should be of value in evaluating vaccines and antivirals against SARS-CoV-2.
- Published
- 2020
15. Nanometer-resolution in situ structure of the SARS-CoV-2 postfusion spike protein
- Author
-
Xiaorui Xing, Yun Zhu, Lei Cao, Chun Chan, Guoliang Zhu, Xiangxi Wang, Minnan Yang, Guoliang Yin, Fei Sun, Linhua Tai, Cheng-Feng Qin, and Zihe Rao
- Subjects
In situ ,Electron Microscope Tomography ,subtomogram analysis ,Glycosylation ,postfusion state ,viruses ,Amino Acid Motifs ,spike protein ,Microbiology ,chemistry.chemical_compound ,Protein Domains ,Chlorocebus aethiops ,medicine ,Animals ,Vero Cells ,Fusion ,Multidisciplinary ,SARS-CoV-2 ,Cryoelectron Microscopy ,Lipid bilayer fusion ,Biological Sciences ,Viral membrane ,cryo-electron tomography ,Entry inhibitor ,Transmembrane domain ,chemistry ,Spike Glycoprotein, Coronavirus ,Biophysics ,Cryo-electron tomography ,Protein Multimerization ,medicine.drug - Abstract
Significance Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a severe threat to public health and the global economy. Its spike protein is responsible for the membrane fusion and is thus a major target for vaccine and drug development. Our study presents the in situ structure of the spike protein in the postfusion state with higher resolution, giving further insights into the design of a viral entry inhibitor. Our observation of the oligomerization states of spikes on the viral membrane implies a possible mechanism of membrane fusion for viral infection., The spike protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) mediates membrane fusion to allow entry of the viral genome into host cells. To understand its detailed entry mechanism and develop a specific entry inhibitor, in situ structural information on the SARS-CoV-2 spike protein in different states is urgent. Here, by using cryo-electron tomography, we observed both prefusion and postfusion spikes in β-propiolactone–inactivated SARS-CoV-2 virions and solved the in situ structure of the postfusion spike at nanometer resolution. Compared to previous reports, the six-helix bundle fusion core, the glycosylation sites, and the location of the transmembrane domain were clearly resolved. We observed oligomerization patterns of the spikes on the viral membrane, likely suggesting a mechanism of fusion pore formation.
- Published
- 2021
- Full Text
- View/download PDF
16. Visualization of yellow fever virus infection in mice using a bioluminescent reporter virus
- Author
-
Hong-Jiang Wang, Zhong-Yu Liu, Yong-Qiang Deng, Cheng-Feng Qin, Tao Jiang, Qing Ye, Xiao-Ling Qin, Dan Li, Hao-Long Dong, and Xiao-Feng Li
- Subjects
Epidemiology ,viruses ,mouse model ,Immunology ,Virus Replication ,Microbiology ,Virus ,Cell Line ,Mice ,Imaging, Three-Dimensional ,Virology ,Drug Discovery ,Yellow Fever ,medicine ,Bioluminescence ,Animals ,Luciferases ,nano-luciferase ,Mice, Inbred BALB C ,biology ,High mortality ,Yellow fever ,food and beverages ,Reporter virus ,General Medicine ,biology.organism_classification ,medicine.disease ,Yellow Fever Virus Infection ,Mice, Inbred C57BL ,Flavivirus ,Infectious Diseases ,Luminescent Measurements ,Parasitology ,Yellow fever virus ,in vivo imaging ,Research Article - Abstract
Yellow fever virus (YFV) is a re-emerging flavivirus, which can lead to severe clinical manifestations and high mortality, with no specific antiviral therapies available. The live-attenuated yellow fever vaccine 17D (YF17D) has been widely used for over eighty years. However, the emergence of yellow fever vaccine-associated viscerotropic disease (YFL-AVD) and yellow fever vaccine-associated neurotropic disease (YFL-AND) raised non-negligible concerns. Additionally, the attenuation mechanism of YF17D is still unclear. Thus, the development of convenient models is crucial to understand the mechanisms behind YF17D attenuation and its adverse effects. In this work, we generated a reporter YF17D expressing nano-luciferase (NLuc). In vitro and in vivo characterization demonstrated that the NLuc-YF17D shared similar biological properties with its parental strain and the NLuc activity can reflect viral infectivity reliably. Combined with in vivo bioluminescence imaging, a series of mice models of YF17D infection was established, which will be useful for the evaluation of antiviral medicines and novel vaccine candidates. Especially, we demonstrated that intraperitoneally (i.p.) infection of NLuc-YF17D in type I interferon receptor-deficient mice A129 resulted in outcomes resembling YEL-AVD and YEL-AND, evidenced by viral replication in multiple organs and invasion of the central neuronal system. Finally, in vitro and in vivo assays based on this reporter virus were established to evaluate the antiviral activities of validated antiviral agents. In conclusion, the bioluminescent reporter virus described herein provides a powerful platform to study YF17D attenuation and vaccine-associated diseases as well as to develop novel countermeasures against YFV.
- Published
- 2021
17. Construction and characterization of UAA-controlled recombinant Zika virus by genetic code expansion
- Author
-
Yan-Peng Xu, Xing-Yao Huang, Xiaofeng Li, Rong-Rong Zhang, Yong-Qiang Deng, Qing Xia, Cheng-Feng Qin, and Qing Ye
- Subjects
DNA, Recombinant ,Computational biology ,Biology ,Virus Replication ,General Biochemistry, Genetics and Molecular Biology ,law.invention ,Zika virus ,Viral Proteins ,law ,Chlorocebus aethiops ,Animals ,Humans ,Amino Acids ,Vero Cells ,General Environmental Science ,Recombination, Genetic ,Base Sequence ,Zika Virus Infection ,Zika Virus ,Genetic code ,biology.organism_classification ,HEK293 Cells ,Genetic Code ,Protein Biosynthesis ,Codon, Terminator ,Recombinant DNA ,General Agricultural and Biological Sciences - Published
- 2020
- Full Text
- View/download PDF
18. Update on the Animal Models and Underlying Mechanisms for ZIKV-Induced Microcephaly
- Author
-
Dan Xu, Cui Li, Zhiheng Xu, and Cheng-Feng Qin
- Subjects
medicine.medical_specialty ,Microcephaly ,Biology ,World health ,Zika virus ,03 medical and health sciences ,0302 clinical medicine ,Virology ,ZikV Infection ,medicine ,Animals ,Humans ,030304 developmental biology ,0303 health sciences ,Zika Virus Infection ,Mechanism (biology) ,Public health ,Brain ,Zika Virus ,Mammalian brain ,biology.organism_classification ,medicine.disease ,Disease Models, Animal ,Public Health ,Neuroscience ,030217 neurology & neurosurgery - Abstract
The circulation of Zika virus (ZIKV) in nearly 80 countries and territories poses a significant global threat to public health. ZIKV is causally linked to severe developmental defects in the brain, recognized as congenital Zika syndrome (CZS), which includes microcephaly and other serious congenital neurological complications. Since the World Health Organization declared the ZIKV outbreak a public health emergency of international concern, remarkable progress has been made in the generation of different ZIKV infection animal models to gain insight into cellular targets and pathogenesis and to explore the associated underlying mechanisms. Here we focus on summarizing our current understanding of the effects of ZIKV on mammalian brain development in different developmental stages and discuss the potential underlying mechanisms of ZIKV-induced CZS, as well as future perspectives.
- Published
- 2019
- Full Text
- View/download PDF
19. Zika virus NS3 is a canonical RNA helicase stimulated by NS5 RNA polymerase
- Author
-
Leijie Wang, Cheng-Feng Qin, Yali Ci, Leiliang Zhang, Lei Shi, Shan Xu, Caimin Xu, and Yang Yang
- Subjects
Gene Expression Regulation, Viral ,Models, Molecular ,Protein Conformation, alpha-Helical ,Base pair ,viruses ,Genetic Vectors ,Gene Expression ,Viral Nonstructural Proteins ,Substrate Specificity ,chemistry.chemical_compound ,Adenosine Triphosphate ,Cricetulus ,RNA polymerase ,Chlorocebus aethiops ,Escherichia coli ,Genetics ,Animals ,Protein Interaction Domains and Motifs ,Cloning, Molecular ,Vero Cells ,RNA, Double-Stranded ,Binding Sites ,biology ,Nucleic Acid Enzymes ,Serine Endopeptidases ,virus diseases ,Helicase ,RNA ,Epithelial Cells ,RNA virus ,Zika Virus ,biochemical phenomena, metabolism, and nutrition ,biology.organism_classification ,RNA Helicase A ,Recombinant Proteins ,digestive system diseases ,Protein Structure, Tertiary ,Cell biology ,Kinetics ,RNA silencing ,Viral replication ,chemistry ,biology.protein ,RNA, Viral ,Protein Conformation, beta-Strand ,RNA Helicases ,Protein Binding - Abstract
Zika virus is a positive single-strand RNA virus whose replication involved RNA unwinding and synthesis. ZIKV NS3 contains a helicase domain, but its enzymatic activity is not fully characterized. Here, we established a dsRNA unwinding assay based on the FRET effect to study the helicase activity of ZIKV NS3, which provided kinetic information in real time. We found that ZIKV NS3 specifically unwound dsRNA/dsDNA with a 3′ overhang in the 3′ to 5′ direction. The RNA unwinding ability of NS3 significantly decreased when the duplex was longer than 18 base pairs. The helicase activity of NS3 depends on ATP hydrolysis and binding to RNA. Mutations in the ATP binding region or the RNA binding region of NS3 impair its helicase activity, thus blocking viral replication in the cell. Furthermore, we showed that ZIKV NS5 interacted with NS3 and stimulated its helicase activity. Disrupting NS3-NS5 interaction resulted in a defect in viral replication, revealing the tight coupling of RNA unwinding and synthesis. We suggest that NS3 helicase activity is stimulated by NS5; thus, viral replication can be carried out efficiently. Our work provides a molecular mechanism of ZIKV NS3 unwinding and novel insights into ZIKV replication.
- Published
- 2019
- Full Text
- View/download PDF
20. Recapitulating Zika Virus Infection in Vagina of Tree Shrew (Tupaia belangeri)
- Author
-
Yong-Qiang Deng, Cheng-Feng Qin, Na-Na Zhang, Xueshan Xia, Yue Feng, Zulqarnain Baloch, Yang Chunguang, Xiaomei Sun, Dao-Qun Li, Li Zhang, Zhili Shen, Jiejie Dai, and Zifeng Yang
- Subjects
0301 basic medicine ,Microbiology (medical) ,Saliva ,Sexual transmission ,Immunology ,Spleen ,Microbiology ,Zika virus ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Cellular and Infection Microbiology ,Zika ,medicine ,Animals ,030212 general & internal medicine ,vaginal douching ,Original Research ,Tupaia ,Fetus ,vaginal infection ,biology ,Zika Virus Infection ,animal model ,Tupaiidae ,Zika Virus ,biology.organism_classification ,Virology ,QR1-502 ,Disease Models, Animal ,030104 developmental biology ,Infectious Diseases ,medicine.anatomical_structure ,Vagina ,Female ,Viral load ,tree shrew - Abstract
Sexual transmission of Zika Virus (ZIKV) elevates the risk of its dissemination in the female reproductive tract and causes a serious threat to the fetus. However, the available animal models are not appropriate to investigate sexual transmission, dynamics of ZIKV infection, replication, and shedding. The use of tree shrew as a small animal model of ZIKV vaginal infection was assessed in this study. A total of 23 sexually mature female tree shrews were infected with ZIKV GZ01viathe intravaginal route. There was no significant difference in change of body weight, and the temperature between ZIKV infected and control animals. Viral RNA loads were detected in blood, saliva, urine, and vaginal douching. ZIKV RNA was readily detected in vaginal lavage of 22 animals (95.65%, 22/23) at 1 dpi, and viral load ranged from 104.46 to 107.35 copies/ml, and the peak of viral load appeared at 1 dpi. The expression of key inflammatory genes, such as IL6, 8, CCL5, TNF-a, and CXCL9, was increased in the spleen of ZIKV infected animals. In the current study, female tree shrews have been successfully infected with ZIKV through the vaginal route for the first time. Interestingly, at first, ZIKV replicates at the local site of infection and then spreads throughout the host body to develop a robust systemic infection and mounted a protective immune response. This small animal model is not only valuable for exploring ZIKV sexual transmission and may also help to explain the cause of debilitating manifestations of the fetusin vivo.
- Published
- 2021
- Full Text
- View/download PDF
21. Convergent evolution of SARS-CoV-2 in human and animals
- Author
-
Cheng-Feng Qin, Xiaofeng Li, Aiping Wu, Hang Fan, Na Han, Hangyu Zhou, and Chengyang Ji
- Subjects
2019-20 coronavirus outbreak ,Coronavirus disease 2019 (COVID-19) ,SARS-CoV-2 ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,COVID-19 ,Cell Biology ,Biology ,Biochemistry ,Virology ,Polymorphism, Single Nucleotide ,Human genetics ,Evolution, Molecular ,Disease Models, Animal ,Convergent evolution ,Drug Discovery ,Spike Glycoprotein, Coronavirus ,Commentary ,Animals ,Humans ,Stem cell ,Developmental biology ,Biotechnology - Published
- 2021
22. Characterization and structural basis of a lethal mouse-adapted SARS-CoV-2
- Author
-
Yong-Qiang Deng, Xiaopeng Song, Meng Shen, Lei Cao, Hui Wang, Xinquan Wang, Si Qin, Yan Guo, Shihui Sun, Yini Qi, Wenjie Tan, Yujun Cui, Min Li, Xiangxi Wang, Ruiting Li, Guan Yang, Lingna Zhao, Hang Fan, Yuehong Chen, Lei Wang, Chao Zhou, Hongjing Gu, Cheng-Feng Qin, Qing Ye, Na Zhu, Xiao Yang, Qi Chen, Rui Feng, Jun Lan, and Yifei Zhang
- Subjects
Male ,viruses ,Science ,Protein domain ,General Physics and Astronomy ,Endogeny ,Plasma protein binding ,Biology ,Severity of Illness Index ,General Biochemistry, Genetics and Molecular Biology ,Deep sequencing ,Virus ,Pathogenesis ,Mice ,Protein Domains ,Animals ,Humans ,Binding site ,Receptor ,Genetics ,Infectivity ,chemistry.chemical_classification ,Multidisciplinary ,Binding Sites ,SARS-CoV-2 ,Strain (biology) ,Lethal dose ,COVID-19 ,General Chemistry ,Virology ,Amino acid ,Disease Models, Animal ,Structural biology ,chemistry ,Amino Acid Substitution ,Spike Glycoprotein, Coronavirus ,Female ,Angiotensin-Converting Enzyme 2 ,Protein Binding - Abstract
The ongoing SARS-CoV-2 pandemic has brought an urgent need for animal models to study the pathogenicity of the virus. Herein, we generated and characterized a novel mouse-adapted SARS-CoV-2 strain, named MASCp36, that causes severe acute respiratory symptoms and mortality in standard laboratory mice. Particularly, this model exhibits age and gender related skewed distribution of mortality akin to severe COVID-19, and the 50% lethal dose (LD50) of MASCp36 was 58 PFU in 9-month-old, male BALB/c mice. Deep sequencing identified three amino acid substitutions, N501Y, Q493H, and K417N, subsequently emerged at the receptor binding domain (RBD) of MASCp36, during in vivo passaging. All three mutations in RBD significantly enhanced the binding affinity to its endogenous receptor, mouse ACE2 (mACE2). Cryo-electron microscopy (cryo-EM) analysis of human ACE2 (hACE2) or mACE2 in complex with the RBD of MASCp36 at 3.1 to 3.7 angstrom resolution elucidates molecular basis for the receptor-binding switch driven by specific amino acid substitutions. Interestingly, N501Y and Q493H enhanced the binding affinity to human ACE2 (hACE2); while triple mutations N501Y/Q493H/K417N decreased affinity to hACE2, thus led to the reduced infectivity of MASCp36 to human cells. Our study not only provides a robust platform for studying the pathogenesis of severe COVID-19 and rapid evaluation of coutermeasures against SARS-CoV-2, but also unveils the molecular mechanism for the rapid adaption and evolution of SARS-CoV-2 in human and animals.One sentence summaryA mouse adapted SARS-CoV-2 strain that harbored specific amino acid substitutions in the RBD of S protein showed 100% mortality in aged, male BALB/c mice.
- Published
- 2020
23. 25-Hydroxycholesterol is a potent SARS-CoV-2 inhibitor
- Author
-
Lili Li, Heng Yang, Chao Zhou, Sarah Gold, Cheng-Feng Qin, Jie Li, Genhong Cheng, Changqing Zhang, Jun He, Xiaofeng Li, Xiang Li, Shulong Zu, Qi Chen, Hui Zhao, and Yong-Qiang Deng
- Subjects
2019-20 coronavirus outbreak ,Fatal outcome ,Coronavirus disease 2019 (COVID-19) ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Clinical Sciences ,Biology ,Peptidyl-Dipeptidase A ,Virus Replication ,Transgenic ,Mice ,Betacoronavirus ,Fatal Outcome ,Chlorocebus aethiops ,Animals ,Humans ,Viral ,Letter to the Editor ,Molecular Biology ,Vero Cells ,Pandemics ,Inbred BALB C ,Aged ,Innate immunity ,Extramural ,Animal ,Reverse Transcriptase Polymerase Chain Reaction ,SARS-CoV-2 ,COVID-19 ,Cell Biology ,Pneumonia ,Molecular biology ,Hydroxycholesterols ,Mice transgenic ,Mechanisms of disease ,Disease Models ,Female ,Angiotensin-Converting Enzyme 2 ,Biochemistry and Cell Biology ,Coronavirus Infections ,Developmental Biology - Abstract
Author(s): Zu, Shulong; Deng, Yong-Qiang; Zhou, Chao; Li, Jie; Li, Lili; Chen, Qi; Li, Xiao-Feng; Zhao, Hui; Gold, Sarah; He, Jun; Li, Xiang; Zhang, Changqing; Yang, Heng; Cheng, Genhong; Qin, Cheng-Feng
- Published
- 2020
24. Type-IInterferon-Inducible SERTAD3 Inhibits Influenza A Virus Replication by Blocking the Assembly of Viral RNA Polymerase Complex
- Author
-
Cheng-Feng Qin, Xiaofeng Li, Na-Na Zhang, Ning Lu, Sarah Gold, Ping Liu, Nina N. Sun, Peishuang Du, Lili Li, Shulong Zu, Rong-Rong Zhang, Chunfeng Li, Yong-Qiang Deng, Jingfeng Wang, Genhong Cheng, Xiang Chen, and Tao Jiang
- Subjects
0301 basic medicine ,RNA-dependent RNA polymerase ,Peptide ,Biology ,IFN-stimulated genes ,Virus Replication ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,Madin Darby Canine Kidney Cells ,03 medical and health sciences ,chemistry.chemical_compound ,Dogs ,0302 clinical medicine ,SERTAD3 ,Interferon ,RNA polymerase ,Influenza A virus ,medicine ,Animals ,Humans ,Viral Replicase Complex Proteins ,Viral rna ,Gene ,lcsh:QH301-705.5 ,Polymerase ,Mice, Knockout ,chemistry.chemical_classification ,virus diseases ,Virology ,Mice, Inbred C57BL ,Protein Subunits ,IAV ,HEK293 Cells ,030104 developmental biology ,chemistry ,lcsh:Biology (General) ,A549 Cells ,Interferon Type I ,Trans-Activators ,biology.protein ,type I interferon ,Peptides ,030217 neurology & neurosurgery ,Protein Binding ,medicine.drug - Abstract
Summary Influenza A virus (IAV) infection stimulates a type I interferon (IFN-I) response in host cells that exerts antiviral effects by inducing the expression of hundreds of IFN-stimulated genes (ISGs). However, most ISGs are poorly studied for their roles in the infection of IAV. Herein, we demonstrate that SERTA domain containing 3 (SERTAD3) has a significant inhibitory effect on IAV replication in vitro. More importantly, Sertad3−/− mice develop more severe symptoms upon IAV infection. Mechanistically, we find SERTAD3 reduces IAV replication through interacting with viral polymerase basic protein 2 (PB2), polymerase basic protein 1 (PB1), and polymerase acidic protein (PA) to disrupt the formation of the RNA-dependent RNA polymerase (RdRp) complex. We further identify an 8-amino-acid peptide of SERTAD3 as a minimum interacting motif that can disrupt RdRp complex formation and inhibit IAV replication. Thus, our studies not only identify SERTAD3 as an antiviral ISG, but also provide the mechanism of potential application of SERTAD3-derived peptide in suppressing influenza replication.
- Published
- 2020
25. Axl Deficiency Promotes the Neuroinvasion of Japanese Encephalitis Virus by Enhancing IL-1α Production from Pyroptotic Macrophages
- Author
-
Dai-Shu Han, Hongning Zhou, Cheng-Feng Qin, Zhao-Yang Wang, Dongying Fan, Jing An, Zi-Da Zhen, and Peigang Wang
- Subjects
Programmed cell death ,interleukin-1α ,viruses ,Immunology ,Biology ,Microbiology ,Receptor tyrosine kinase ,Virus ,Pathogenesis ,Mice ,Phosphatidylinositol 3-Kinases ,03 medical and health sciences ,0302 clinical medicine ,Interleukin-1alpha ,Proto-Oncogene Proteins ,Virology ,medicine ,Animals ,Encephalitis, Viral ,Encephalitis, Japanese ,030304 developmental biology ,Encephalitis Virus, Japanese ,Mice, Knockout ,Mice, Inbred BALB C ,0303 health sciences ,Macrophages ,pyroptosis ,Viral encephalitis ,Pyroptosis ,Brain ,Receptor Protein-Tyrosine Kinases ,Axl ,Japanese encephalitis ,biology.organism_classification ,medicine.disease ,Axl Receptor Tyrosine Kinase ,Disease Models, Animal ,Japanese encephalitis virus ,Flavivirus ,Blood-Brain Barrier ,Insect Science ,biology.protein ,Pathogenesis and Immunity ,Female ,030217 neurology & neurosurgery - Abstract
Japanese encephalitis virus (JEV) is a mosquito-borne flavivirus that causes Japanese encephalitis (JE), the most commonly diagnosed viral encephalitis worldwide. The fatality rate of JE is 20%, and nearly half of the surviving patients develop neuropsychiatric sequelae. Axl is a receptor tyrosine kinase that plays multiple roles in flaviviral infections. Currently, the involvement of Axl in JEV infection remains enigmatic. In this study, we demonstrate that Axl impedes the pathogenesis of severe JE in mice by maintaining blood-brain-barrier (BBB) integrity and restricting viral neuroinvasion. Furthermore, serum IL-1α is a key mediator of this process and is primarily released by JEV-infected pyroptotic macrophages to elicit BBB breakdown, while an IL-1α antagonist can effectively reduce the incidence of severe JE. Our work uncovers the protective role of Axl in antagonizing severe JE and shows that the use of an IL-1α antagonist may be a promising tactic to prevent severe JE., Japanese encephalitis virus (JEV) is a flavivirus that causes Japanese encephalitis (JE), which has an unclear pathogenesis. Despite vaccination, thousands of deaths attributed to JE are reported annually. In this study, we report that mice deficient for Axl, a receptor tyrosine kinase that plays multiple roles in flaviviral infection, displayed greater mortality upon JEV infection. The effect of Axl deficiency on JEV infection was mediated by markedly elevated serum interleukin-1α (IL-1α) levels, which devastated the blood-brain-barrier and promoted viral neuroinvasion within 24 h postinfection. Using an in situ infection model, we showed that dead macrophages were the primary source of observed increased serum IL-1α levels. Axl deficiency enhanced cell death and caused pyroptosis in 80% of JEV-infected macrophages by disrupting phosphatidylinositol 3-kinase (PI3K)-Akt signaling. Intriguingly, the primary effector released by pyroptotic macrophages in our model was IL-1α rather than IL-1β. Finally, we assessed the effect of an IL-1α antagonist and demonstrated that it effectively prevented the incidence of JE. Our results indicate that Axl plays a protective role in JEV infection, identify IL-1α released by pyroptotic macrophages as a crucial factor promoting JEV neuroinvasion, and suggest that an IL-1α antagonist may be a candidate for JE therapy. IMPORTANCE Japanese encephalitis virus (JEV) is a mosquito-borne flavivirus that causes Japanese encephalitis (JE), the most commonly diagnosed viral encephalitis worldwide. The fatality rate of JE is 20%, and nearly half of the surviving patients develop neuropsychiatric sequelae. Axl is a receptor tyrosine kinase that plays multiple roles in flaviviral infections. Currently, the involvement of Axl in JEV infection remains enigmatic. In this study, we demonstrate that Axl impedes the pathogenesis of severe JE in mice by maintaining blood-brain-barrier (BBB) integrity and restricting viral neuroinvasion. Furthermore, serum IL-1α is a key mediator of this process and is primarily released by JEV-infected pyroptotic macrophages to elicit BBB breakdown, while an IL-1α antagonist can effectively reduce the incidence of severe JE. Our work uncovers the protective role of Axl in antagonizing severe JE and shows that the use of an IL-1α antagonist may be a promising tactic to prevent severe JE.
- Published
- 2020
- Full Text
- View/download PDF
26. The m
- Author
-
Jun'e, Liu, Yan-Peng, Xu, Kai, Li, Qing, Ye, Hang-Yu, Zhou, Hanxiao, Sun, Xiaoyu, Li, Liu, Yu, Yong-Qiang, Deng, Rui-Ting, Li, Meng-Li, Cheng, Bo, He, Jia, Zhou, Xiao-Feng, Li, Aiping, Wu, Chengqi, Yi, and Cheng-Feng, Qin
- Subjects
Adenosine ,SARS-CoV-2 ,COVID-19 ,Genome, Viral ,DNA Methylation ,Virus Replication ,RNA modification ,Article ,Cell Line ,Mechanisms of disease ,Gene Expression Regulation ,Chlorocebus aethiops ,Host-Pathogen Interactions ,Mutagenesis, Site-Directed ,Animals ,Humans ,RNA, Messenger ,Vero Cells ,Phylogeny - Abstract
The newly identified Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has resulted in a global health emergency because of its rapid spread and high mortality. The molecular mechanism of interaction between host and viral genomic RNA is yet unclear. We demonstrate herein that SARS-CoV-2 genomic RNA, as well as the negative-sense RNA, is dynamically N6-methyladenosine (m6A)-modified in human and monkey cells. Combined RIP-seq and miCLIP analyses identified a total of 8 m6A sites at single-base resolution in the genome. Especially, epidemic strains with mutations at these identified m6A sites have emerged worldwide, and formed a unique cluster in the US as indicated by phylogenetic analysis. Further functional experiments showed that m6A methylation negatively regulates SARS-CoV-2 infection. SARS-CoV-2 infection also triggered a global increase in host m6A methylome, exhibiting altered localization and motifs of m6A methylation in mRNAs. Altogether, our results identify m6A as a dynamic epitranscriptomic mark mediating the virus–host interaction.
- Published
- 2020
27. Rational development of a human antibody cocktail that deploys multiple functions to confer Pan-SARS-CoVs protection
- Author
-
Lei Wang, Lei Cao, Tianhao Weng, Yan Run, Yao Sun, Sun Xinglu, Keda Chen, Tian-Shu Cao, Na-Na Zhang, Zhe Lv, Weidong Jiang, Yan Xintian, Lang Guojun, Hu Yuhao, Hangping Yao, Danrong Shi, Jie Zhang, Tao Jiang, Xiangyun Lu, Rui Feng, Shihui Sun, Yan-Peng Xu, Yunhua Zhou, Yong-Qiang Deng, Dandan Zhu, Kong Chao, Lanjuan Li, Tan Yongcong, Rong-Rong Zhang, Guan Yang, Linfang Cheng, Xiaofeng Li, Qingyu Lv, Nan Wang, Shao Junbin, Qi Chen, Nanping Wu, Hui Zhao, Xing-Yao Huang, Liu Chanjuan, Zhang Wenhai, Xiangxi Wang, Hong-Ying Qiu, and Cheng-Feng Qin
- Subjects
medicine.drug_class ,viruses ,Immunology ,Cooperativity ,Monoclonal antibody ,Antibodies, Viral ,Epitope ,Neutralization ,Article ,Epitopes ,Chlorocebus aethiops ,medicine ,Animals ,Humans ,Neutralizing antibody ,Molecular Biology ,Vero Cells ,biology ,SARS-CoV-2 ,fungi ,virus diseases ,COVID-19 ,Cell Biology ,Virology ,Antibodies, Neutralizing ,Disease Models, Animal ,biology.protein ,Vero cell ,Antibody ,Structural biology ,Single-Chain Antibodies - Abstract
Structural principles underlying the composition and synergistic mechanisms of protective monoclonal antibody cocktails are poorly defined. Here, we exploited antibody cooperativity to develop a therapeutic antibody cocktail against SARS-CoV-2. On the basis of our previously identified humanized cross-neutralizing antibody H014, we systematically analyzed a fully human naive antibody library and rationally identified a potent neutralizing antibody partner, P17, which confers effective protection in animal model. Cryo-EM studies dissected the nature of the P17 epitope, which is SARS-CoV-2 specific and distinctly different from that of H014. High-resolution structure of the SARS-CoV-2 spike in complex with H014 and P17, together with functional investigations revealed that in a two-antibody cocktail, synergistic neutralization was achieved by S1 shielding and conformational locking, thereby blocking receptor attachment and viral membrane fusion, conferring high potency as well as robustness against viral mutation escape. Furthermore, cluster analysis identified a hypothetical 3rd antibody partner for further reinforcing the cocktail as pan-SARS-CoVs therapeutics.
- Published
- 2020
28. Structural basis for neutralization of SARS-CoV-2 and SARS-CoV by a potent therapeutic antibody
- Author
-
Qing Ye, Qi Chen, Shihui Sun, Zhe Lv, Nan Wang, Youchun Wang, Zhen Cui, Changfa Fan, Dandan Zhu, Weijin Huang, Jianhui Nie, Zihe Rao, Neil Shaw, Chun Yun Sun, Yao Sun, Qianqian Li, Xiaofeng Li, Liangzhi Xie, Yong-Qiang Deng, Lei Cao, Xiangxi Wang, Ling Zhu, and Cheng-Feng Qin
- Subjects
medicine.drug_class ,viruses ,Pneumonia, Viral ,Peptidyl-Dipeptidase A ,Monoclonal antibody ,Antibodies, Monoclonal, Humanized ,Virus ,Epitope ,Neutralization ,Betacoronavirus ,Immunoglobulin Fab Fragments ,Mice ,Protein Domains ,Report ,Chlorocebus aethiops ,medicine ,Animals ,Humans ,Neutralizing antibody ,skin and connective tissue diseases ,Lung ,Pandemics ,Vero Cells ,Multidisciplinary ,biology ,Chemistry ,SARS-CoV-2 ,fungi ,Biochem ,COVID-19 ,Microbio ,Virology ,Antibodies, Neutralizing ,respiratory tract diseases ,body regions ,Epitope mapping ,Severe acute respiratory syndrome-related coronavirus ,Monoclonal ,Humanized mouse ,biology.protein ,Receptors, Virus ,Angiotensin-Converting Enzyme 2 ,Antibody ,Protein Multimerization ,Coronavirus Infections ,Epitope Mapping ,Conformational epitope ,Reports - Abstract
The COVID-19 pandemic caused by the SARS-CoV-2 virus has resulted in an unprecedented public health crisis. There are no approved vaccines or therapeutics for treating COVID-19. Here we reported a humanized monoclonal antibody, H014, efficiently neutralizes SARS-CoV-2 and SARS-CoV pseudoviruses as well as authentic SARS-CoV-2 at nM level by engaging the S receptor binding domain (RBD). Importantly, H014 administration reduced SARS-CoV-2 titers in the infected lungs and prevented pulmonary pathology in hACE2 mouse model. Cryo-EM characterization of the SARS-CoV-2 S trimer in complex with the H014 Fab fragment unveiled a novel conformational epitope, which is only accessible when the RBD is in open conformation. Biochemical, cellular, virological and structural studies demonstrated that H014 prevents attachment of SARS-CoV-2 to its host cell receptors. Epitope analysis of available neutralizing antibodies against SARS-CoV and SARS-CoV-2 uncover broad cross-protective epitopes. Our results highlight a key role for antibody-based therapeutic interventions in the treatment of COVID-19.One sentence summaryA potent neutralizing antibody conferred protection against SARS-CoV-2 in an hACE2 humanized mouse model by sterically blocking the interaction of the virus with its receptor.
- Published
- 2020
29. A Thermostable mRNA Vaccine against COVID-19
- Author
-
Hua Yuan Yan, Cheng-Feng Qin, Xing Yao Huang, Shu Long Zu, Qing Ye, Tian Shu Cao, Xiaofeng Li, Hang Fan, Chao Zhou, Yi Jiao Huang, Peng Gao, Yan Guo, Xia Zhong, Yuhang Jiang, Qi He, Zhen Yu Zha, Na Na Zhang, Dongsheng Zhou, Rong Rong Zhang, Weijin Huang, Shi Hui Sun, Yong Qiang Deng, You Chun Wang, Xiao Yang, Guan Yang, Qi Chen, Hui Zhao, Hong Ying Qiu, Bo Ying, Xia Lin Xue, and Jian Hui Nie
- Subjects
Male ,Mice ,0302 clinical medicine ,Immunogenicity, Vaccine ,Chlorocebus aethiops ,0303 health sciences ,Mice, Inbred ICR ,Vaccines, Synthetic ,Protection ,biology ,Viral Vaccine ,Spike Glycoprotein, Coronavirus ,RNA, Viral ,Female ,Antibody ,Coronavirus Infections ,COVID-19 Vaccines ,Lipid nanoparticle ,Injections, Intramuscular ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Animals ,Humans ,RNA, Messenger ,Binding site ,Gene ,Vaccine Potency ,Vero Cells ,030304 developmental biology ,Messenger RNA ,Binding Sites ,SARS-CoV-2 ,COVID-19 ,Viral Vaccines ,Th1 Cells ,Non-human primate ,Virology ,Antibodies, Neutralizing ,Macaca fascicularis ,mRNA vaccine ,HEK293 Cells ,Immunization ,Mouse adapted strain ,Vero cell ,biology.protein ,Nanoparticles ,030217 neurology & neurosurgery ,HeLa Cells - Abstract
Summary There has been an urgent need of vaccines against coronavirus disease 2019 (COVID-19) due to the ongoing SARS-CoV-2 pandemic. Among all approaches, messenger RNA (mRNA) -based vaccine has emerged as a rapid and versatile platform to quickly respond to such a challenge. Here, we developed a lipid-nanoparticle-encapsulated mRNA (mRNA-LNP) encoding the receptor binding domain (RBD) of SARS-CoV-2 as a vaccine candidate (termed ARCoV). Intramuscular immunization of ARCoV mRNA-LNPs elicited robust neutralizing antibodies against SARS-CoV-2 as well as Th1-biased cellular response in mice and non-human primates. Two doses of ARCoV immunization in mice conferred complete protection against the challenge of a SARS-CoV-2 mouse adapted strain. Additionally, ARCoV was manufactured in liquid formulation and can be stored at room temperature for at least one week. This novel COVID-19 mRNA vaccine, ARCoV, is currently being evaluated in phase 1 clinical trials., Highlights Development of LNP-encapsulated mRNA vaccine (ARCoV) targeting the RBD of SARS-CoV-2 ARCoV induces neutralizing antibodies and T-cell immunity in mice and NHPs. ARCoV vaccination confers full protection against SARS-CoV-2 challenge in mice. ARCoV is a thermostable vaccine candidate for phase I studies, ARCoV is an LNP-encapsulated mRNA vaccine platform that is highly immunogenic and safe in mice and non-human primates, conferring protection against the challenge a SARS-CoV-2 mouse adapted strain.
- Published
- 2020
30. Development of an inactivated vaccine candidate for SARS-CoV-2
- Author
-
Jing Li, Xiuyu Lou, Cheng-Feng Qin, Yanhui Yin, Chuan Qin, Deyu Jiang, Wei Deng, Fang Cai, Xiangxi Wang, Zhe Lv, Minnan Yang, Linlin Bao, Yanjun Zhang, Weidong Yin, Li Yajing, Jiangning Liu, Haiyan Mao, Zhu Lang, Hong Gao, Ge Xiaoqin, Li Yurong, Dongdong Wu, Jinxing Lu, Qiang Gao, Nan Wang, Kangwei Xu, Wen Shi, Ling Zhu, Lin Wang, Changgui Li, Hengming Zhang, Yaling Hu, Xuejie Gong, and Biao Kan
- Subjects
0301 basic medicine ,Male ,medicine.medical_specialty ,COVID-19 Vaccines ,Pneumonia, Viral ,Dose-Response Relationship, Immunologic ,Pilot Projects ,Antibodies, Viral ,Virus ,Immunoglobulin G ,03 medical and health sciences ,Betacoronavirus ,Mice ,0302 clinical medicine ,Medical microbiology ,Immunogenicity, Vaccine ,Chlorocebus aethiops ,Medicine ,Animals ,Rats, Wistar ,Pandemics ,Vero Cells ,Mice, Inbred BALB C ,Multidisciplinary ,biology ,business.industry ,SARS-CoV-2 ,Immunogenicity ,Viral Vaccine ,COVID-19 ,Viral Vaccines ,Viral Load ,Virology ,Antibodies, Neutralizing ,Macaca mulatta ,Rats ,030104 developmental biology ,Vaccines, Inactivated ,030220 oncology & carcinogenesis ,Inactivated vaccine ,biology.protein ,Female ,Antibody ,business ,Coronavirus Infections ,Viral load - Abstract
Vaccine candidate tested in monkeys Global spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has led to an urgent race to develop a vaccine. Gao et al. report preclinical results of an early vaccine candidate called PiCoVacc, which protected rhesus macaque monkeys against SARS-CoV-2 infection when analyzed in short-term studies. The researchers obtained multiple SARS-CoV-2 strains from 11 hospitalized patients across the world and then chemically inactivated the harmful properties of the virus. Animals were immunized with one of two vaccine doses and then inoculated with SARS-CoV-2. Those that received the lowest dose showed signs of controlling the infection, and those receiving the highest dose appeared more protected and did not have detectable viral loads in the pharynx or lungs at 7 days after infection. The next steps will be testing for safety and efficacy in humans. Science , this issue p. 77
- Published
- 2020
- Full Text
- View/download PDF
31. A Mouse Model of SARS-CoV-2 Infection and Pathogenesis
- Author
-
Cheng-Feng Qin, Yu Sen Zhou, Quan Ming Liu, Xiaofeng Li, Chang Fa Fan, Hong Jing Gu, Tong Yan Zhao, Shi Hui Sun, Xing Yao Huang, You Chun Wang, Yan Guo, Xiao Yang, Su Su Liu, Qi Chen, Quan Liu, Yue Lei Shen, Rui Xiong, Weijin Huang, Yan Xiao Wang, Na Na Zhang, Yong Zhou, Yong Qiang Deng, and Guan Yang
- Subjects
Aging ,Pneumonia, Viral ,Pathogenesis ,Biology ,Nose ,Peptidyl-Dipeptidase A ,medicine.disease_cause ,Virus Replication ,Microbiology ,Article ,Mouse model ,Betacoronavirus ,Virology ,medicine ,CRISPR ,Animals ,Gene Knock-In Techniques ,Lung ,Pandemics ,Coronavirus ,Transmission (medicine) ,SARS-CoV-2 ,Stomach ,Angiotensin converting enzyme II ,Brain ,COVID-19 ,Viral Load ,Mice, Inbred C57BL ,Trachea ,Disease Models, Animal ,medicine.anatomical_structure ,Viral replication ,Immunology ,Cytokines ,RNA, Viral ,Parasitology ,Nasal administration ,Angiotensin-Converting Enzyme 2 ,CRISPR-Cas Systems ,Coronavirus Infections ,Lung Diseases, Interstitial ,Viral load - Abstract
Summary Since December 2019, a novel coronavirus SARS-CoV-2 has emerged and rapidly spread throughout the world, resulting in a global public health emergency. The lack of vaccine and antivirals has brought an urgent need for animal model. Human Angiotensin converting enzyme II (ACE2) has been identified as a functional receptor for SARS-CoV-2. In this study, we generated a mouse model expressing human ACE2 (hACE2) using CRISPR/Cas9 knock-in technology. Compared with wild-type C57BL/6 mice, both young and aged hACE2 mice sustained high viral loads in lung, trachea and brain upon intranasal infection. Although fatalities were not observed, interstitial pneumonia and elevated cytokines were seen in SARS-CoV-2 infected- aged hACE2 mice. Interestingly, intragastric inoculation of SARS-CoV-2 was evidenced to cause productive infection and lead to pulmonary pathological changes in hACE2 mice. Overall, this animal model described here provides a useful tool for studying SARS-CoV-2 transmission and pathogenesis, and evaluating COVID-19 vaccines and therapeutics., Graphical Abstract, Highlights ● Human ACE2 knock-in mice were generated using CRISPR/Cas9 technology ● SARS-CoV-2 leads to robust replication in lung, trachea and brain ● SARS-CoV-2 causes interstitial pneumonia and elevated cytokine in aged hACE2 mice ● High dose of SARS-CoV-2 can establish infection via intragastric route in hACE2 mice, The COVID-19 pandemic has brought an urgent need for small animal models. Here, Sun et al. established an ACE2 humanized mouse by CRISPR/Cas9 knock-in technology. This hACE2 mice are susceptible to SARS-CoV-2 infection upon intranasal inoculation, and the resulting pulmonary infection and pathological changes resemble those observed in COVID-19 patients.
- Published
- 2020
32. Potential Vector Competence of Mosquitoes to Transmit Baiyangdian Virus, a New Tembusu-Related Virus in China
- Author
-
Tao Jiang, Xiaoxia Guo, Cheng-Feng Qin, Dan Xing, Yuting Jiang, Teng Zhao, Yande Dong, Tong-Yan Zhao, and Chunxiao Li
- Subjects
0301 basic medicine ,China ,animal structures ,animal diseases ,viruses ,030231 tropical medicine ,Mosquito Vectors ,Viral Plaque Assay ,Microbiology ,Virus ,Flavivirus Infections ,03 medical and health sciences ,0302 clinical medicine ,Aedes ,Virology ,Animals ,biology ,Tembusu ,Bird Diseases ,Flavivirus ,virus diseases ,030108 mycology & parasitology ,biology.organism_classification ,Culex ,Infectious Diseases ,Ducks ,Animals, Zoo - Abstract
A new duck Tembusu-related flavivirus, Baiyangdian virus (BYDV), caused duck egg-drop syndrome in China. The rapid spread, unknown transmission routes, and zoonotic nature, raise serious concern about BYDV as a potential threat to human health. The study provides the first evaluation on the vector competence of
- Published
- 2020
33. Short Direct Repeats in the 3′ Untranslated Region Are Involved in Subgenomic Flaviviral RNA Production
- Author
-
Zhiming Yuan, Xiao-Dan Li, Han-Qing Ye, Xing Yao Huang, Cheng-Feng Qin, Xiaolin Niu, Na Li, Yan Peng Xu, Hao Long Dong, Bo Zhang, Qiu Yan Zhang, Xiaofeng Li, Hong Jiang Wang, Xianyang Fang, Peng Gong, Pei Yong Shi, Cheng Lin Deng, Qi Chen, and Hui Zhao
- Subjects
Untranslated region ,Immunology ,Genome, Viral ,Biology ,Microbiology ,Genome ,Conserved sequence ,Cricetinae ,Virology ,Exoribonuclease ,Chlorocebus aethiops ,Animals ,Humans ,Direct repeat ,3' Untranslated Regions ,Vero Cells ,Subgenomic mRNA ,Genetics ,Three prime untranslated region ,Flavivirus ,RNA ,Genome Replication and Regulation of Viral Gene Expression ,Culicidae ,A549 Cells ,Tandem Repeat Sequences ,Insect Science ,Nucleic Acid Conformation ,RNA, Viral - Abstract
Mosquito-borne flaviviruses consist of a positive-sense genome RNA flanked by the untranslated regions (UTRs). There is a panel of highly complex RNA structures in the UTRs with critical functions. For instance, Xrn1-resistant RNAs (xrRNAs) halt Xrn1 digestion, leading to the production of subgenomic flaviviral RNA (sfRNA). Conserved short direct repeats (DRs), also known as conserved sequences (CS) and repeated conserved sequences (RCS), have been identified as being among the RNA elements locating downstream of xrRNAs, but their biological function remains unknown. In this study, we revealed that the specific DRs are involved in the production of specific sfRNAs in both mammalian and mosquito cells. Biochemical assays and structural remodeling demonstrate that the base pairings in the stem of these DRs control sfRNA formation by maintaining the binding affinity of the corresponding xrRNAs to Xrn1. On the basis of these findings, we propose that DRs functions like a bracket holding the Xrn1-xrRNA complex for sfRNA formation. IMPORTANCE Flaviviruses include many important human pathogens. The production of subgenomic flaviviral RNAs (sfRNAs) is important for viral pathogenicity as a common feature of flaviviruses. sfRNAs are formed through the incomplete degradation of viral genomic RNA by the cytoplasmic 5ʹ–3ʹ exoribonuclease Xrn1 halted at the Xrn1-resistant RNA (xrRNA) structures within the 3ʹ-UTR. The 3ʹ-UTRs of the flavivirus genome also contain distinct short direct repeats (DRs), such as RCS3, CS3, RCS2, and CS2. However, the biological functions of these ancient primary DR sequences remain largely unknown. Here, we found that DR sequences are involved in sfRNA formation and viral virulence and provide novel targets for the rational design of live attenuated flavivirus vaccine.
- Published
- 2020
- Full Text
- View/download PDF
34. Salivary factor LTRIN from Aedes aegypti facilitates the transmission of Zika virus by interfering with the lymphotoxin-β receptor
- Author
-
Xue Hao, Xiaomin Guo, Hongning Zhou, Cheng-Feng Qin, Tao Xu, Lin Jin, Pengpeng Li, Mingdong Yang, Peng Sun, Xiaopeng Qi, Mingzhao Zhu, Hua Peng, Chuanbin Shen, Chunmiao Hu, Ombati Rose, Ren Lai, Gong Cheng, and Jingya Guo
- Subjects
0301 basic medicine ,030106 microbiology ,Immunology ,Mosquito Vectors ,Aedes aegypti ,Proinflammatory cytokine ,Zika virus ,Mice ,03 medical and health sciences ,Immune system ,Aedes ,Lymphotoxin beta Receptor ,Animals ,Humans ,Immunology and Allergy ,Saliva ,Infectivity ,biology ,Zika Virus Infection ,Zika Virus ,biology.organism_classification ,Virology ,030104 developmental biology ,Lymphotoxin ,Insect Proteins ,Lymphotoxin beta receptor - Abstract
Pathogens have co-evolved with mosquitoes to optimize transmission to hosts. Mosquito salivary-gland extract is known to modulate host immune responses and facilitate pathogen transmission, but the underlying molecular mechanisms of this have remained unknown. In this study, we identified and characterized a prominent 15-kilodalton protein, LTRIN, obtained from the salivary glands of the mosquito Aedes aegypti. LTRIN expression was upregulated in blood-fed mosquitoes, and LTRIN facilitated the transmission of Zika virus (ZIKV) and exacerbated its pathogenicity by interfering with signaling through the lymphotoxin-β receptor (LTβR). Mechanically, LTRIN bound to LTβR and 'preferentially' inhibited signaling via the transcription factor NF-κB and the production of inflammatory cytokines by interfering with the dimerization of LTβR during infection with ZIKV. Furthermore, treatment with antibody to LTRIN inhibited mosquito-mediated infection with ZIKV, and abolishing LTβR potentiated the infectivity of ZIKV both in vitro and in vivo. This study provides deeper insight into the transmission of mosquito-borne diseases in nature and supports the therapeutic potential of inhibiting the action of LTRIN to disrupt ZIKV transmission.
- Published
- 2018
- Full Text
- View/download PDF
35. American Strain of Zika Virus Causes More Severe Microcephaly Than an Old Asian Strain in Neonatal Mice
- Author
-
Guanghui Li, Yong Qiang Deng, Hong Jiang Wang, Feng Zhang, Xing Yao Huang, Haozhen Yang, Ling Yuan, Zhiheng Xu, Zhong-Yu Liu, Qin Wang, Lei Shi, Cheng-Feng Qin, and Qing Ye
- Subjects
0301 basic medicine ,Microcephaly ,lcsh:Medicine ,Brain damage ,Severity of Illness Index ,General Biochemistry, Genetics and Molecular Biology ,American strain ,Zika virus ,Asian strain ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,medicine ,Animals ,Humans ,Progenitor cell ,Neurons ,lcsh:R5-920 ,biology ,Zika Virus Infection ,Strain (biology) ,lcsh:R ,Brain ,General Medicine ,Zika Virus ,biology.organism_classification ,medicine.disease ,Venezuela ,Virology ,Oligodendrocyte ,United States ,Flavivirus ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Immunology ,medicine.symptom ,lcsh:Medicine (General) ,030217 neurology & neurosurgery ,Research Paper - Abstract
Zika virus (ZIKV) has evolved from an overlooked mosquito-borne flavivirus into a global health threat due to its astonishing causal link to microcephaly and other disorders. ZIKV has been shown to infect neuronal progenitor cells of the fetal mouse brain, which is comparable to the first-trimester human fetal brain, and result in microcephaly. However, whether there are different effects between the contemporary ZIKV strain and its ancestral strain in the neonatal mouse brain, which is comparable with the second-trimester human fetal brain, is unclear. Here we adopted a mouse model which enables us to study the postnatal effect of ZIKV infection. We show that even 100 pfu of ZIKV can replicate and infect neurons and oligodendrocytes in most parts of the brain. Compared with the ancestral strain from Cambodia (CAM/2010), infection of the ZIKV strain from Venezuela (VEN/2016) leads to much more severe microcephaly, accompanied by more neuronal cell death, abolishment of oligodendrocyte development, and a more dramatic immune response. The serious brain damage caused by VEN/2016 infection would be helpful to elucidate why the American strain resulted in severe neurovirulence in infants and will provide clinical guidance for the diagnosis and treatment of infection by different ZIKV strains., Highlights • The infection of an American strain of ZIKV leads to more severe microcephaly than the ancestral Asian strain. • American strain infects more cells, and induces more dramatic immune response and cell death than ancestral Asian strain. World attention has been drawn to a global Zika virus (ZIKV) outbreak due to its unexpected causal link to congenital brain abnormalities, especially microcephaly. Infection of pregnant women with the American Zika strain, but not the ancestral Asian strain, can result in microcephaly in infants. However, the phenotypic difference between the contemporary American strain and ancestral Asian strain of ZIKV is still unclear. We employed the ZIKV infection model of a neonatal mouse brain to compare the difference between these two strains. We find that infection by the American strain leads to more severe microcephaly than the ancestral Asian strain.
- Published
- 2017
36. A peptide-based viral inactivator inhibits Zika virus infection in pregnant mice and fetuses
- Author
-
Kwok-Yung Yuen, Shibo Jiang, Xiaohui Zhou, Na-Na Zhang, Yufeng Yu, Yanyan Dai, Yu Meng, Lanying Du, Yuan Li, Lu Lu, Jia-Nan Hao, Qian Wang, Peng Zou, Min Tian, Cheng-Feng Qin, Fei Yu, Yong-Qiang Deng, and Jasper Fuk-Woo Chan
- Subjects
0301 basic medicine ,General Physics and Astronomy ,Antibodies, Viral ,Virus Replication ,Zika virus ,Mice ,Viral Envelope Proteins ,Pregnancy ,Interferon ,Cricetinae ,Chlorocebus aethiops ,reproductive and urinary physiology ,Mice, Inbred ICR ,Multidisciplinary ,Zika Virus Infection ,Transmission (medicine) ,Flow Cytometry ,Flavivirus ,Female ,Antibody ,medicine.drug ,Science ,030106 microbiology ,Biology ,Antiviral Agents ,Article ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,03 medical and health sciences ,medicine ,Animals ,Humans ,Vero Cells ,Fetus ,Virion ,Viral Vaccines ,Zika Virus ,General Chemistry ,Viral membrane ,biology.organism_classification ,Antibodies, Neutralizing ,Virology ,Mice, Inbred C57BL ,Disease Models, Animal ,HEK293 Cells ,030104 developmental biology ,Microscopy, Fluorescence ,Drug Design ,Vero cell ,biology.protein ,Pregnancy, Animal ,Peptides - Abstract
Zika virus (ZIKV), a re-emerging flavivirus associated with neurological disorders, has spread rapidly to more than 70 countries and territories. However, no specific vaccines or antiviral drugs are currently available to prevent or treat ZIKV infection. Here we report that a synthetic peptide derived from the stem region of ZIKV envelope protein, designated Z2, potently inhibits infection of ZIKV and other flaviviruses in vitro. We show that Z2 interacts with ZIKV surface protein and disrupts the integrity of the viral membrane. Z2 can penetrate the placental barrier to enter fetal tissues and is safe for use in pregnant mice. Intraperitoneal administration of Z2 inhibits vertical transmission of ZIKV in pregnant C57BL/6 mice and protects type I or type I/II interferon receptor-deficient mice against lethal ZIKV challenge. Thus, Z2 has potential to be further developed as an antiviral treatment against ZIKV infection in high-risk populations, particularly pregnant women., Zika virus (ZIKV) has spread rapidly in recent years and there is a need for antiviral treatments. Here, the authors develop an antiviral peptide, based on the stem region of ZIKV envelope protein, and show that it is safe in pregnant mice and inhibits ZIKV infection in pregnant mice and fetuses.
- Published
- 2017
37. Recovery of the Zika virus through an in vitro ligation approach
- Author
-
Cheng-Feng Qin, Qiu-Yan Zhang, Cheng-Lin Deng, Han-Qing Ye, Bo Zhang, Si-Qing Liu, and Dong-Dong Chen
- Subjects
0301 basic medicine ,Adenosine ,DNA, Complementary ,Genome, Viral ,Recombinant virus ,Antiviral Agents ,Virus ,Cell Line ,Zika virus ,03 medical and health sciences ,Aedes ,Cricetinae ,Virology ,Complementary DNA ,Chlorocebus aethiops ,Animals ,Cloning, Molecular ,Deoxyribonucleases, Type II Site-Specific ,Vero Cells ,biology ,Zika Virus ,biology.organism_classification ,Molecular biology ,Flavivirus ,Restriction site ,030104 developmental biology ,Viral replication ,DNA, Viral ,Vero cell ,RNA, Viral - Abstract
In this study, an in vitro ligation method was developed to assemble a full-length infectious cDNA clone of the Zika virus (ZIKV). Four contiguous cDNA subclones covering the complete ZIKV genome were constructed with unique BglI restriction sites at the ends of each fragment. The BglI restriction sites only allow in vitro ligation to happen between interconnecting restriction sites from adjacent cDNA fragments, resulting in an intact full-length cDNA of ZIKV. RNA transcripts derived from the full-length cDNA were infectious. The recombinant virus replicated as efficiently as the wild-type virus with similar growth kinetics and plaque morphologies in Vero and C6/36 cells. Both viruses were inhibited by NITD008 treatment. This in vitro ligation method will facilitate manipulation of the viral genome through genetic modifications of four separated subclones of ZIKV for the rapid and rational development of candidate vaccines and viral replication study.
- Published
- 2017
- Full Text
- View/download PDF
38. Zika virus directly infects peripheral neurons and induces cell death
- Author
-
Ronghua Li, Feiran Zhang, Zhiheng Xu, In Young Choi, Luoxiu Huang, Tianlei Xu, Hotae Lim, Cheng-Feng Qin, Qing-Feng Wu, Fahimeh Mirakhori, Yohan Oh, Peng Jin, Hengli Tang, Yaqing Wang, Hongjun Song, Hao Wu, Zhexing Wen, Gabsang Lee, Cui Li, Emily M. Lee, and Guo Li Ming
- Subjects
0301 basic medicine ,Programmed cell death ,Mice, 129 Strain ,Global Health ,Article ,Zika virus ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Neural Stem Cells ,In vivo ,Chlorocebus aethiops ,Animals ,Humans ,Vero Cells ,Cells, Cultured ,Neurons ,Mice, Inbred ICR ,biology ,Cell Death ,Molecular pathology ,Zika Virus Infection ,General Neuroscience ,Peripheral Nervous System Diseases ,Neural crest ,Zika Virus ,biology.organism_classification ,Virology ,Neural stem cell ,In vitro ,3. Good health ,030104 developmental biology ,nervous system ,Immunology ,Vero cell ,030217 neurology & neurosurgery - Abstract
Zika virus (ZIKV)-infection is associated with neurological disorders of both the central and peripheral nervous systems (PNS), yet few studies have directly examined PNS-infection. Here we show that intraperitoneally or intraventricularly-injected ZIKV in the mouse could infect and impact peripheral neurons in vivo. Moreover, ZIKV productively infects stem cell-derived human neural crest cells and peripheral neurons in vitro, leading to increased cell death, transcriptional dysregulation and cell-type specific molecular pathology.
- Published
- 2017
39. Structure and function ofcis‐acting RNA elements of flavivirus
- Author
-
Cheng-Feng Qin and Zhong-Yu Liu
- Subjects
Gene Expression Regulation, Viral ,0301 basic medicine ,Untranslated region ,viruses ,030106 microbiology ,Genome, Viral ,Dengue virus ,Virus Replication ,medicine.disease_cause ,Genome ,Virus ,Flavivirus Infections ,Zika virus ,Structure-Activity Relationship ,03 medical and health sciences ,Virology ,medicine ,Animals ,Humans ,Nucleotide Motifs ,Nucleic acid structure ,Phylogeny ,Repetitive Sequences, Nucleic Acid ,Genetics ,biology ,Flavivirus ,virus diseases ,RNA ,biology.organism_classification ,030104 developmental biology ,Infectious Diseases ,Nucleic Acid Conformation ,RNA, Viral - Abstract
The genus Flavivirus is a group of single-stranded, positive-sense RNA viruses that includes numerous human pathogens with global impact, such as dengue virus (DENV), yellow fever virus (YFV), West Nile virus (WNV), and Zika virus (ZIKV). The approximately 11-kilobase genome is flanked by highly structured untranslated regions (UTRs), which contain various cis-acting RNA elements with unique structures and functions. Moreover, local RNA elements circularize the genome non-covalently through long-range interactions. Interestingly, many flavivirus cis-acting RNA elements contain group-specific motifs or are specific for the given phylogenetic groups, suggesting their potential association with flavivirus evolution and diversification. In this review, we summarize recent advances about the structure and function of cis-acting RNA elements in flavivirus genomes and highlight the potential implications for flavivirus evolution. Finally, the scientific questions remained to be answered in the field are also discussed.
- Published
- 2019
- Full Text
- View/download PDF
40. Zika virus degrades the ω-3 fatty acid transporter Mfsd2a in brain microvascular endothelial cells and impairs lipid homeostasis
- Author
-
Xingyao Huang, Xiaojing Chi, Tianli Lin, Lei Shi, Xiuying Liu, Cheng-Feng Qin, Jia Zhou, Min Cheng, Wei Yang, Hua Xu, and Jingjing Fan
- Subjects
Microcephaly ,Docosahexaenoic Acids ,Biology ,Microbiology ,Pathogenesis ,03 medical and health sciences ,0302 clinical medicine ,Virology ,Fatty Acids, Omega-3 ,Human Umbilical Vein Endothelial Cells ,medicine ,Animals ,Homeostasis ,Humans ,Research Articles ,030304 developmental biology ,Mice, Knockout ,0303 health sciences ,Multidisciplinary ,Symporters ,Zika Virus Infection ,HEK 293 cells ,SciAdv r-articles ,Brain ,Endothelial Cells ,Lipid metabolism ,Zika Virus ,Human brain ,Lipid Metabolism ,medicine.disease ,Cell biology ,HEK293 Cells ,Phenotype ,medicine.anatomical_structure ,Docosahexaenoic acid ,Microvessels ,Proteolysis ,Ectopic expression ,030217 neurology & neurosurgery ,Research Article - Abstract
Both the endothelial transporter Mfsd2a and docosahexaenoic acid uptake are disrupted in Zika virus–induced microcephaly models., Zika virus (ZIKV) infection during pregnancy increases the risk of postnatal microcephaly. Neurovascular function provides a homeostatic environment for proper brain development. The major facilitator superfamily domain-containing protein 2 (Mfsd2a) is selectively expressed in human brain microvascular endothelial cells (hBMECs) and is the major transporter mediating the brain uptake of docosahexaenoic acid (DHA). We have discovered a pivotal role for Mfsd2a in the pathogenesis of ZIKV. ZIKV disrupted Mfsd2a both in cultured primary hBMECs and in the neonatal mouse brain. ZIKV envelope (E) protein specifically interacted with Mfsd2a and promoted Mfsd2a polyubiquitination for proteasome-dependent degradation. Infection with ZIKV or ectopic expression of ZIKV E impaired Mfsd2a-mediated DHA uptake. Lipidomic analysis revealed obvious differences in DHA-containing lipids after ZIKV infection. Supplementation with DHA rescued ZIKV-caused growth restriction and microcephaly. Our findings suggest endothelial Mfsd2a as an important pathogenic mediator and supplementation with DHA as a potential therapeutic option for ZIKV infection.
- Published
- 2019
- Full Text
- View/download PDF
41. Long non‐coding subgenomic flavivirus RNAs have extended 3D structures and are flexible in solution
- Author
-
Yupeng Zhang, Zhong-Yu Liu, Xianyang Fang, Yikan Zhang, Yan Wang, Cheng-Feng Qin, Meng-Li Cheng, and Junfeng Ma
- Subjects
computational modeling ,Models, Molecular ,small angle X‐ray scattering ,West Nile virus ,Genome, Viral ,Computational biology ,Dengue virus ,3D structure ,subgenomic flavivirus RNA ,medicine.disease_cause ,Biochemistry ,Article ,Zika virus ,03 medical and health sciences ,0302 clinical medicine ,X-Ray Diffraction ,Structural Biology ,Genetics ,medicine ,Animals ,Humans ,Molecular Biology ,030304 developmental biology ,Subgenomic mRNA ,0303 health sciences ,Base Sequence ,biology ,Flavivirus ,Articles ,Zika Virus ,biology.organism_classification ,Pathogenicity ,Microbiology, Virology & Host Pathogen Interaction ,Long non-coding RNA ,Solutions ,long non‐coding RNA ,Nucleic Acid Conformation ,RNA, Viral ,RNA, Long Noncoding ,030217 neurology & neurosurgery - Abstract
Most mosquito‐borne flaviviruses, including Zika virus (ZIKV), Dengue virus (DENV), and West Nile virus (WNV), produce long non‐coding subgenomic RNAs (sfRNAs) in infected cells that link to pathogenicity and immune evasion. Until now, the structural characterization of these lncRNAs remains limited. Here, we studied the 3D structures of individual and combined subdomains of sfRNAs, and visualized the accessible 3D conformational spaces of complete sfRNAs from DENV2, ZIKV, and WNV by small angle X‐ray scattering (SAXS) and computational modeling. The individual xrRNA1s and xrRNA2s adopt similar structures in solution as the crystal structure of ZIKV xrRNA1, and all xrRNA1‐2s form compact structures with reduced flexibility. While the DB12 of DENV2 is extended, the DB12s of ZIKV and WNV are compact due to the formation of intertwined double pseudoknots. All 3′ stem‐loops (3′SLs) share similar rod‐like structures. Complete sfRNAs are extended and sample a large conformational space in solution. Our work not only provides structural insight into the function of flavivirus sfRNAs, but also highlights strategies of visualizing other lncRNAs in solution by SAXS and computational methods.
- Published
- 2019
- Full Text
- View/download PDF
42. A broadly neutralizing germline-like human monoclonal antibody against dengue virus envelope domain III
- Author
-
Na-Na Zhang, Cheng Lei, Peng Zou, Vinita Puri, Xiaolong Tian, Yong-Qiang Deng, Yang Feng, Shun Li, Ponraj Prabakaran, Wei Li, Dan Hu, Chunyu Wang, Zhongyu Zhu, Yanling Wu, Xiaohui Zhou, Jane Cardosa, Qi Zhao, Yulu Wang, Tianlei Ying, Cheng-Feng Qin, and Dimiter S. Dimitrov
- Subjects
RNA viruses ,Physiology ,viruses ,Dengue virus ,Pathology and Laboratory Medicine ,Antibodies, Viral ,medicine.disease_cause ,Biochemistry ,Epitope ,Germline ,Dengue fever ,Dengue ,Epitopes ,Viral Envelope Proteins ,Immune Physiology ,Cricetinae ,Medicine and Health Sciences ,Public and Occupational Health ,Biology (General) ,Vaccines ,0303 health sciences ,Immune System Proteins ,030302 biochemistry & molecular biology ,Antibodies, Monoclonal ,virus diseases ,Animal Models ,Vaccination and Immunization ,Infectious Diseases ,Experimental Organism Systems ,Medical Microbiology ,Viral Pathogens ,Viruses ,Pathogens ,Antibody ,Research Article ,Infectious Disease Control ,medicine.drug_class ,QH301-705.5 ,Immunology ,Mouse Models ,Library Screening ,Biology ,Research and Analysis Methods ,Monoclonal antibody ,Microbiology ,Antibodies ,Cell Line ,03 medical and health sciences ,Model Organisms ,Antigen ,Virology ,Vaccine Development ,medicine ,Genetics ,Animals ,Humans ,Animal Models of Disease ,Antigens ,Molecular Biology Techniques ,Microbial Pathogens ,Molecular Biology ,Dengue vaccine ,030304 developmental biology ,Molecular Biology Assays and Analysis Techniques ,Flaviviruses ,Organisms ,Biology and Life Sciences ,Proteins ,Dengue Virus ,biochemical phenomena, metabolism, and nutrition ,RC581-607 ,medicine.disease ,Antibodies, Neutralizing ,Animal Models of Infection ,Animal Studies ,biology.protein ,Parasitology ,Preventive Medicine ,Immunologic diseases. Allergy - Abstract
Dengue is the most widespread vector-borne viral disease caused by dengue virus (DENV) for which there are no safe, effective drugs approved for clinical use. Here, by using sequential antigen panning of a yeast antibody library derived from healthy donors against the DENV envelop protein domain III (DIII) combined with depletion by an entry defective DIII mutant, we identified a cross-reactive human monoclonal antibody (mAb), m366.6, which bound with high affinity to DENV DIII from all four DENV serotypes. Immunogenetic analysis indicated that m366.6 is a germline-like mAb with very few somatic mutations from the closest VH and Vλ germline genes. Importantly, we demonstrated that it potently neutralized DENV both in vitro and in the mouse models of DENV infection without detectable antibody-dependent enhancement (ADE) effect. The epitope of m366.6 was mapped to the highly conserved regions on DIII, which may guide the design of effective dengue vaccine immunogens. Furthermore, as the first germline-like mAb derived from a naïve antibody library that could neutralize all four DENV serotypes, the m366.6 can be a tool for exploring mechanisms of DENV infection, and is a promising therapeutic candidate., Author summary Dengue virus infects 50–100 million people each year. Infection is initiated by entry of the virus into cells mediated by the viral envelope glycoproteins. There are four closely related DENV serotypes, but they all are antigenically distinct, with each comprising several genotypes that exhibit differences in their infection characteristics in both the mosquito vector and in the human host. One of the confounding problems that has faced vaccine and biological drugs development for decades is the inability of antibodies to one serotype to protect against infection by another one. Instead, the induced humoral immune response to one dengue virus infection can enhance the infection and disease processes brought by a subsequent infection with another dengue serotype. In this study, by using a competitive sorting strategy to interrogate a human naïve antibody library, we identified a cross-reactive mAb, designated as m366.6, against the four DENV serotypes. The mAb m366.6 possesses only few somatic mutations from the closest VH and Vλ germline genes and high affinity to DIII. Most importantly, the germline-like m366.6 demonstrated a broad spectrum of neutralization against the four DENV serotypes. Thus, m366.6 is a promising candidate therapeutics and its epitope may imply on the design of effective vaccine immunogens to elicit m366.6-like antibodies in vivo.
- Published
- 2019
- Full Text
- View/download PDF
43. Erratum for Chen et al., 'Treatment of Human Glioblastoma with a Live Attenuated Zika Virus Vaccine Candidate'
- Author
-
Pei Yong Shi, Jianghong Man, Jin Wu, Chao Shan, Xiaofeng Li, Tongyan Zhao, Feng Ma, Cheng-Feng Qin, Dapei Li, Yan Wu, Qi Chen, Qing Ye, Xuping Xie, Chunfeng Li, Haitao Wu, Qian Zhu, Xiaoling Qin, and Xiaoyan Zhan
- Subjects
Mice, Nude ,Apoptosis ,Vaccines, Attenuated ,Microbiology ,Zika virus ,Mice ,03 medical and health sciences ,Virology ,Chlorocebus aethiops ,Animals ,Humans ,Medicine ,Vero Cells ,030304 developmental biology ,Inflammation ,Oncolytic Virotherapy ,Mice, Inbred BALB C ,0303 health sciences ,biology ,Brain Neoplasms ,030306 microbiology ,business.industry ,Published Erratum ,Zika Virus ,biology.organism_classification ,medicine.disease ,Xenograft Model Antitumor Assays ,QR1-502 ,Mice, Inbred C57BL ,Oncolytic Viruses ,Viral Tropism ,Neoplastic Stem Cells ,Female ,Erratum ,Glioblastoma ,business - Abstract
Glioblastoma (GBM) is the deadliest type of brain tumor, and glioma stem cells (GSCs) contribute to tumor recurrence and therapeutic resistance. Thus, an oncolytic virus targeting GSCs may be useful for improving GBM treatment. Because Zika virus (ZIKV) has an oncolytic tropism for infecting GSCs, we investigated the safety and efficacy of a live attenuated ZIKV vaccine candidate (ZIKV-LAV) for the treatment of human GBM in a GSC-derived orthotopic model. Intracerebral injection of ZIKV-LAV into mice caused no neurological symptoms or behavioral abnormalities. The neurovirulence of ZIKV-LAV was more attenuated than that of the licensed Japanese encephalitis virus LAV 14-14-2, underlining the superior safety of ZIKV-LAV for potential GBM treatment. Importantly, ZIKV-LAV significantly reduced intracerebral tumor growth and prolonged animal survival by selectively killing GSCs within the tumor. Mechanistically, ZIKV infection elicited antiviral immunity, inflammation, and GSC apoptosis. Together, these results further support the clinical development of ZIKV-LAV for GBM therapy.
- Published
- 2019
- Full Text
- View/download PDF
44. Flavivirus induces and antagonizes antiviral RNA interference in both mammals and mosquitoes
- Author
-
Yan-Peng Xu, Ruiting Li, Jiuyue Xu, Yan Guo, Jie Cui, Jing Kong, Cheng-Feng Qin, Hui Zhou, Yang Qiu, Miao Wang, Da Zheng, Xiaofeng Li, Xi Zhou, Rong-Rong Zhang, and Meng Miao
- Subjects
Ribonuclease III ,Small interfering RNA ,viruses ,Mutant ,Immunology ,Mosquito Vectors ,Viral Nonstructural Proteins ,Dengue fever ,law.invention ,Flavivirus Infections ,03 medical and health sciences ,law ,RNA interference ,Virology ,parasitic diseases ,medicine ,Animals ,Research Articles ,030304 developmental biology ,Regulation of gene expression ,0303 health sciences ,Multidisciplinary ,biology ,Flavivirus ,030302 biochemistry & molecular biology ,fungi ,virus diseases ,SciAdv r-articles ,biochemical phenomena, metabolism, and nutrition ,medicine.disease ,biology.organism_classification ,In vitro ,Culicidae ,Gene Expression Regulation ,Host-Pathogen Interactions ,Suppressor ,RNA Interference ,Research Article - Abstract
Flaviviruses such as dengue use the same strategy to evade RNAi-based intrinsic antiviral defenses in both mammals and mosquitoes., Mosquito-borne flaviviruses infect both mammals and mosquitoes. RNA interference (RNAi) has been demonstrated as an anti-flavivirus mechanism in mosquitoes; however, whether and how flaviviruses induce and antagonize RNAi-mediated antiviral immunity in mammals remains unknown. We show that the nonstructural protein NS2A of dengue virus-2 (DENV2) act as a viral suppressor of RNAi (VSR). When NS2A-mediated RNAi suppression was disabled, the resulting mutant DENV2 induced Dicer-dependent production of abundant DENV2-derived siRNAs in differentiated mammalian cells. VSR-disabled DENV2 showed severe replication defects in mosquito and mammalian cells and in mice that were rescued by RNAi deficiency. Moreover, NS2As of multiple flaviviruses act as VSRs in vitro and during viral infection in both organisms. Overall, our findings demonstrate that antiviral RNAi can be induced by flavivirus, while flavivirus uses NS2A as a bona fide VSR to evade RNAi in mammals and mosquitoes, highlighting the importance of RNAi in flaviviral vector-host life cycles.
- Published
- 2019
45. Zika Virus Infection in Tupaia belangeri Causes Dermatological Manifestations and Confers Protection against Secondary Infection
- Author
-
Na-Na Zhang, Qi Wang, Xiao-Peng Qi, Yong-Qiang Deng, Xueshan Xia, Guoqing Wang, Xiaomei Sun, Li Zhang, Fuchun Zhang, Cheng-Feng Qin, Yuanyuan Han, Yue Feng, Jiejie Dai, Qing Ye, and Feng Ma
- Subjects
Male ,Secondary infection ,Immunology ,Viremia ,Biology ,Microbiology ,Zika virus ,Cell Line ,Pathogenesis ,03 medical and health sciences ,Tupaia belangeri ,0302 clinical medicine ,Immune system ,Virology ,Cricetinae ,medicine ,Animals ,Humans ,Saliva ,030304 developmental biology ,Inflammation ,Tupaia ,0303 health sciences ,Zika Virus Infection ,Zika Virus ,biology.organism_classification ,medicine.disease ,Rash ,Disease Models, Animal ,030220 oncology & carcinogenesis ,Insect Science ,Skin Diseases, Viral ,biology.protein ,Pathogenesis and Immunity ,Female ,Antibody ,medicine.symptom - Abstract
Animal models of Zika virus (ZIKV) infection have recently been established in mice, guinea pigs, and nonhuman primates. Tree shrews (Tupaia belangeri) are an emerging experimental animal in biomedical applications, but their susceptibility to ZIKV infection has not been explored. In the present study, we show that subcutaneous inoculation of ZIKV led to rapid viremia and viral secretion in saliva, as well as to typical dermatological manifestations characterized by massive diffuse skin rash on the trunk. Global transcriptomic sequencing of peripheral blood mononuclear cells isolated from ZIKV-infected animals revealed systematic gene expression changes related to the inflammatory response and dermatological manifestations. Importantly, ZIKV infection readily triggered the production of high-titer neutralizing antibodies, thus preventing secondary homologous infection in tree shrews. However, neonatal tree shrews succumbed to ZIKV challenge upon intracerebral infection. The tree shrew model described here recapitulates the most common dermatological manifestations observed in ZIKV-infected patients and may greatly facilitate the elucidation of ZIKV pathogenesis and the development of novel vaccines and therapeutics. IMPORTANCE The reemergence of Zika virus (ZIKV) has caused a global public health crisis since 2016, and there are currently no vaccines or antiviral drugs to prevent or treat ZIKV infection. However, considerable advances have been made in understanding the biology and pathogenesis of ZIKV infection. In particular, various animal models have been successfully established to mimic ZIKV infection and its associated neurological diseases and to evaluate potential countermeasures. However, the clinical symptoms in these mouse and nonhuman primate models are different from the common clinical manifestations seen in human ZIKV patients; in particular, dermatological manifestations are rarely recapitulated in these animal models. Here, we developed a new animal model of ZIKV infection in tree shrews, a rat-sized, primate-related mammal. In vitro and in vivo characterization of ZIKV infection in tree shrews established a direct link between ZIKV infection and the immune responses and dermatological manifestations. The tree shrew model described here, as well as other available animal models, provides a valuable platform to study ZIKV pathogenesis and to evaluate vaccines and therapeutics.
- Published
- 2019
46. Aedes aegypti HPX8C modulates immune responses against viral infection
- Author
-
Xiao Feng Li, Cheng-Feng Qin, Aihua Zheng, Yong Qiang Deng, Yang Cheng, Ju Mei Wang, Zhen Zou, Hong Jiang, Long Sheng Xing, Dan Wen, and Zuo Kun Shi
- Subjects
0301 basic medicine ,lcsh:Arctic medicine. Tropical medicine ,lcsh:RC955-962 ,030231 tropical medicine ,Heme ,Aedes aegypti ,Dengue virus ,Real-Time Polymerase Chain Reaction ,medicine.disease_cause ,Dengue fever ,Dengue ,Transcriptome ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Aedes ,Immunity ,medicine ,Animals ,Vector (molecular biology) ,Peroxidase ,Innate immune system ,Host Microbial Interactions ,biology ,Gene Expression Profiling ,lcsh:Public aspects of medicine ,Toll-Like Receptors ,Public Health, Environmental and Occupational Health ,lcsh:RA1-1270 ,Dengue Virus ,medicine.disease ,biology.organism_classification ,Virology ,Immunity, Innate ,Specific Pathogen-Free Organisms ,030104 developmental biology ,Infectious Diseases ,Gene Expression Regulation ,RNA Interference ,Reactive Oxygen Species ,Digestive System ,Antimicrobial Cationic Peptides ,Signal Transduction - Abstract
Mosquitoes act as vectors of numerous pathogens that cause human diseases. Dengue virus (DENV) transmitted by mosquito, Aedes aegypti, is responsible for dengue fever epidemics worldwide with a serious impact on human health. Currently, disease control mainly relies on vector targeted intervention strategies. Therefore, it is imperative to understand the molecular mechanisms underlying the innate immune response of mosquitoes against pathogens. In the present study, the expression profiles of immunity-related genes in the midgut responding to DENV infection by feeding were analyzed by transcriptome and quantitative real-time PCR. The level of Antimicrobial peptides (AMPs) increased seven days post-infection (d.p.i.), which could be induced by the Toll immune pathway. The expression of reactive oxygen species (ROS) genes, including antioxidant genes, such as HPX7, HPX8A, HPX8B, HPX8C were induced at one d.p.i. and peaked again at ten d.p.i. in the midgut. Interestingly, down-regulation of the antioxidant gene HPX8C by RNA interference led to reduction in the virus titer in the mosquito, probably due to the elevated levels of ROS. Application of a ROS inhibitor and scavenger molecules further established the role of oxygen free radicals in the modulation of the immune response to DENV infection. Overall, our comparative transcriptome analyses provide valuable information about the regulation of immunity related genes in the transmission vector in response to DENV infection. It further allows us to identify novel molecular mechanisms underlying the host-virus interaction, which might aid in the development of novel strategies to control mosquito-borne diseases.
- Published
- 2019
47. Aedes mosquitoes acquire and transmit Zika virus by breeding in contaminated aquatic environments
- Author
-
Pei Yong Shi, Fuchun Zhang, Bernard Cazelles, Cheng-Feng Qin, Senyan Du, Clara Champagne, Liangqin Tong, Chun-Hong Chen, Renli Zhang, Qiyong Liu, Jianying Liu, Ping Ma, Gong Cheng, Jie Zhao, Yang Liu, Guodong Liang, Huaiyu Tian, and Penghua Wang
- Subjects
0301 basic medicine ,Science ,General Physics and Astronomy ,02 engineering and technology ,Aedes aegypti ,Breeding ,Article ,General Biochemistry, Genetics and Molecular Biology ,Zika virus ,03 medical and health sciences ,Aedes ,Mosquito larvae ,parasitic diseases ,Animals ,Humans ,In patient ,lcsh:Science ,Multidisciplinary ,Sewage ,biology ,Zika Virus Infection ,Transmission (medicine) ,Aquatic ecosystem ,Water Pollution ,fungi ,Virion ,food and beverages ,Zika Virus ,General Chemistry ,Hydrogen-Ion Concentration ,021001 nanoscience & nanotechnology ,biology.organism_classification ,Virology ,6. Clean water ,3. Good health ,Mice, Inbred C57BL ,Flavivirus ,030104 developmental biology ,lcsh:Q ,0210 nano-technology - Abstract
Zika virus (ZIKV) is a mosquito-borne flavivirus that predominantly circulates between humans and Aedes mosquitoes. Clinical studies have shown that Zika viruria in patients persists for an extended period, and results in infectious virions being excreted. Here, we demonstrate that Aedes mosquitoes are permissive to ZIKV infection when breeding in urine or sewage containing low concentrations of ZIKV. Mosquito larvae and pupae, including from field Aedes aegypti can acquire ZIKV from contaminated aquatic systems, resulting in ZIKV infection of adult females. Adult mosquitoes can transmit infectious virions to susceptible type I/II interferon receptor-deficient (ifnagr-/-) C57BL/6 (AG6) mice. Furthermore, ZIKV viruria from infected AG6 mice can causes mosquito infection during the aquatic life stages. Our studies suggest that infectious urine could be a natural ZIKV source, which is potentially transmissible to mosquitoes when breeding in an aquatic environment., Here the authors show that Aedes mosquitoes can acquire ZIKV by breeding in contaminated aquatic systems, and that these infected mosquitoes can transmit ZIKV to susceptible mice. This suggests that human urine containing aquatic environments could contribute to ZIKV transmission.
- Published
- 2019
- Full Text
- View/download PDF
48. Infectivity of Zika virus on primary cells support tree shrew as animal model
- Author
-
Xiao-Peng Qi, Na-Na Zhang, Chunguang Yang, Zhili Shen, Xiaomei Sun, Xueshan Xia, Jiejie Dai, Cheng-Feng Qin, Zifeng Yang, Li Zhang, Yong-Qiang Deng, Dao-Qun Li, and Yue Feng
- Subjects
0301 basic medicine ,Epidemiology ,viruses ,030106 microbiology ,Immunology ,primary cells ,Biology ,Kidney ,Virus Replication ,Microbiology ,Article ,Zika virus ,Tree shrew ,03 medical and health sciences ,Animal model ,Virology ,Small animal ,Drug Discovery ,Chlorocebus aethiops ,ZikV Infection ,Human Umbilical Vein Endothelial Cells ,Animals ,Humans ,Lung ,Vero Cells ,Tropism ,Aorta ,Cells, Cultured ,Skin ,Infectivity ,Zika Virus Infection ,infectivity ,tropism ,Tupaiidae ,General Medicine ,Zika Virus ,biology.organism_classification ,Flavivirus ,Disease Models, Animal ,030104 developmental biology ,Infectious Diseases ,HEK293 Cells ,Liver ,Parasitology ,tree shrew - Abstract
Zika virus (ZIKV) is a mosquito-borne flavivirus that caused the public health emergency. Recently, we have proved a novel small animal tree shrew was susceptive to ZIKV infection and presented the most common rash symptoms as ZIKV patients. Here we further cultured the primary cells from different tissues of this animal to determine the tissue tropism of ZIKV infection in vitro. The results showed that the primary cells from tree shrew kidney, lung, liver, skin and aorta were permissive to ZIKV infection and could support viral replication by the detection of viral specific RNA intra- and extra-cells. In comparing, the skin fibroblast and vascular endothelial cells were highly permissive to ZIKV infection with high releasing of active virus particles in supernatants proved by its infectivity in established neonatal mouse model. The expressions of ZIKV envelop and nonstructural protein-1, and the effects and strong immune response of primary tree shrew cells were also detected followed by ZIKV infection. These findings provide powerful in vitro cell-level evidence to support tree shrew as animal model of ZIKV infection and may help to explain the rash manifestations in vivo.
- Published
- 2019
49. The evolution of Zika virus from Asia to the Americas
- Author
-
Wei-Feng Shi, Zhong-Yu Liu, and Cheng-Feng Qin
- Subjects
0303 health sciences ,Asia ,General Immunology and Microbiology ,biology ,030306 microbiology ,Zika Virus Infection ,Amino acid substitution ,Zika Virus ,biology.organism_classification ,Global Health ,Microbiology ,Virology ,Zika virus ,Evolution, Molecular ,03 medical and health sciences ,Infectious Diseases ,Culicidae ,Amino Acid Substitution ,Mutation ,Global health ,Animals ,Humans ,Americas ,Phylogeny - Abstract
Zika virus (ZIKV) was once considered an obscure member of the large and diverse family of mosquito-borne flaviviruses, and human infections with ZIKV were thought to be sporadic, with mild and self-limiting symptoms. The large-scale ZIKV epidemics in the Americas and the unexpected uncovering of a link to congenital birth defects escalated ZIKV infections to the status of a global public health emergency. Recent studies that combined reverse genetics with modelling in multiple systems have provided evidence that ZIKV has acquired additional amino acid substitutions at the same time as congenital Zika syndrome and other birth defects were detected. In this Progress article, we summarize the evolution of ZIKV during its spread from Asia to the Americas and discuss potential links to pathogenesis.
- Published
- 2019
50. Human MxB Inhibits the Replication of Hepatitis C Virus
- Author
-
Cheng-Feng Qin, Dongrong Yi, Jing An, Leiliang Zhang, Fengwen Xu, Yongxin Zhang, Jinming Zhou, Zhenlong Liu, Fei Guo, Kavita Raniga, Shan Cen, Xiaoyu Li, Ni An, Chen Liang, Jing Wang, Quanjie Li, and Rui Zhou
- Subjects
Myxovirus Resistance Proteins ,viruses ,Hepatitis C virus ,Immunology ,Cypa ,Hepacivirus ,Viral Nonstructural Proteins ,Biology ,Dengue virus ,Endoplasmic Reticulum ,Virus Replication ,medicine.disease_cause ,Microbiology ,Virus ,Cell Line ,03 medical and health sciences ,Flaviviridae ,Interferon ,Virology ,Chlorocebus aethiops ,medicine ,Animals ,Humans ,NS5A ,Vero Cells ,030304 developmental biology ,0303 health sciences ,030306 microbiology ,biology.organism_classification ,Virus-Cell Interactions ,3. Good health ,HEK293 Cells ,Viral replication ,Gene Knockdown Techniques ,Insect Science ,Cyclosporine ,Interferons ,Cyclophilin A ,Protein Binding ,medicine.drug - Abstract
Type I interferon (IFN) inhibits viruses by inducing the expression of antiviral proteins. The IFN-induced myxovirus resistance B (MxB) protein has been reported to inhibit a limited number of viruses, including HIV-1 and herpesviruses, but its antiviral coverage remains to be explored further. Here we show that MxB interferes with RNA replication of hepatitis C virus (HCV) and significantly inhibits viral replication in a cyclophilin A (CypA)-dependent manner. Our data further show that MxB interacts with the HCV protein NS5A, thereby impairing NS5A interaction with CypA and NS5A localization to the endoplasmic reticulum, two events essential for HCV RNA replication. Interestingly, we found that MxB significantly inhibits two additional CypA-dependent viruses of the Flaviviridae family, namely, Japanese encephalitis virus and dengue virus, suggesting a potential link between virus dependence on CypA and virus susceptibility to MxB inhibition. Collectively, these data have identified MxB as a key factor behind IFN-mediated suppression of HCV infection, and they suggest that other CypA-dependent viruses may also be subjected to MxB restriction. IMPORTANCE Viruses of the Flaviviridae family cause major illness and death around the world and thus pose a great threat to human health. Here we show that IFN-inducible MxB restricts several members of the Flaviviridae, including HCV, Japanese encephalitis virus, and dengue virus. This finding not only suggests an active role of MxB in combating these major pathogenic human viruses but also significantly expands the antiviral spectrum of MxB. Our study further strengthens the link between virus dependence on CypA and susceptibility to MxB restriction and also suggests that MxB may employ a common mechanism to inhibit different viruses. Elucidating the antiviral functions of MxB advances our understanding of IFN-mediated host antiviral defense and may open new avenues to the development of novel antiviral therapeutics.
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.