118 results on '"Cheng‐Feng Qin"'
Search Results
2. Expression pattern and function of SARS-CoV-2 receptor ACE2
- Author
-
Ruiting Li and Cheng-Feng Qin
- Subjects
Microbiology (medical) ,Interaction ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,viruses ,ACE2 ,Disease ,Computational biology ,Infectious and parasitic diseases ,RC109-216 ,Biology ,medicine.disease_cause ,Article ,Expression pattern ,medicine ,Receptor ,skin and connective tissue diseases ,Coronavirus ,Mechanism (biology) ,SARS-CoV-2 ,fungi ,Public Health, Environmental and Occupational Health ,virus diseases ,body regions ,Infectious Diseases ,Adaptation ,Public aspects of medicine ,RA1-1270 ,Function (biology) ,Biotechnology - Abstract
Since the outbreak at the end of 2019, SARS-CoV-2 has been spreading around the world for more than one year. Scientists have been intensely conducting research on this newly emerged coronavirus and the disease caused by it. Angiotensin-converting enzyme 2 (ACE2), as a receptor mediating the cellular entry of SARS-CoV-2, has become a hot spot for researchers. Here, we summarized the recent progresses on the function, expression and distribution characteristics of ACE2 in human body and among populations. We further discussed the interaction mechanism of ACE2 and SARS-CoV-2 S protein, focusing on key residues that effect interaction and binding ability of SARS-CoV-2 variants. This will facilitate researchers to better understand SARS-CoV-2 infection and transmission route, adaptation mechanism, and designing treatment strategies.
- Published
- 2021
3. 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
4. Generation and Characterization of a Nanobody Against SARS-CoV
- Author
-
Cheng-Feng Qin, Qi Shuhui, Jiangfan Li, Yuehong Chen, Ruiwen Fan, Lei He, Yong-Qiang Deng, Xiao-Lu Zhang, Shixiong Hu, and Guangyu Zhao
- Subjects
medicine.drug_class ,viruses ,Immunology ,Antibodies, Viral ,Peripheral blood mononuclear cell ,Neutralization ,law.invention ,Non-competitive inhibition ,Severe acute respiratory syndrome coronavirus (SARS-CoV) ,law ,Virology ,Escherichia coli ,medicine ,Humans ,skin and connective tissue diseases ,Neutralizing antibody ,chemistry.chemical_classification ,biology ,SARS-CoV-2 ,fungi ,COVID-19 ,virus diseases ,Antibodies, Neutralizing ,Receptor-binding domain (RBD) ,body regions ,Enzyme ,Severe acute respiratory syndrome-related coronavirus ,chemistry ,Spike Glycoprotein, Coronavirus ,Nanobody ,Leukocytes, Mononuclear ,biology.protein ,Recombinant DNA ,Molecular Medicine ,Antiviral drug ,Antibody ,Research Article ,Protein Binding - Abstract
The sudden emergence of severe acute respiratory syndrome coronavirus (SARS-CoV) has caused global panic in 2003, and the risk of SARS-CoV outbreak still exists. However, no specific antiviral drug or vaccine is available; thus, the development of therapeutic antibodies against SARS-CoV is needed. In this study, a nanobody phage-displayed library was constructed from peripheral blood mononuclear cells of alpacas immunized with the recombinant receptor-binding domain (RBD) of SARS-CoV. Four positive clones were selected after four rounds of bio-panning and subjected to recombinant expression in E. coli. Further biological identification demonstrated that one of the nanobodies, S14, showed high affinity to SARS-CoV RBD and potent neutralization activity at the picomole level against SARS-CoV pseudovirus. A competitive inhibition assay showed that S14 blocked the binding of SARS-CoV RBD to either soluble or cell-expressed angiotensin-converting enzyme 2 (ACE2). In summary, we developed a novel nanobody targeting SARS-CoV RBD, which might be useful for the development of therapeutics against SARS.
- Published
- 2021
- Full Text
- View/download PDF
5. Recovery and Genetic Characterization of a West Nile Virus Isolate from China
- Author
-
Yong-Qiang Deng, Shihong Fu, Chao Zhou, Songtao Xu, Fan Li, Xiaofeng Li, Hong-Jiang Wang, Hangyu Zhou, Huanyu Wang, Cheng-Feng Qin, Meng-Li Cheng, and Yan Guo
- Subjects
0301 basic medicine ,China ,Gibson assembly ,Lineage (genetic) ,viruses ,030106 microbiology ,Immunology ,Virulence ,Virus ,03 medical and health sciences ,Virology ,Complementary DNA ,Humans ,Phylogeny ,biology ,Phylogenetic tree ,Flavivirus ,Strain (biology) ,biology.organism_classification ,030104 developmental biology ,Molecular Medicine ,West Nile virus ,West Nile Fever ,Research Article - Abstract
West Nile virus (WNV) is an important neurotropic flavivirus that is widely distributed globally. WNV strain XJ11129 was first isolated in Xinjiang, China, and its genetic and biological characteristics remain largely unknown. In this study, phylogenetic and sequence analyses revealed that XJ11129 belongs to lineage 1a and shares high genetic identity with the highly pathogenic strain NY99. Then, the full-length genomic cDNA of XJ11129 was amplified and assembled using a modified Gibson assembly (GA) method. The virus (named rXJ11129) was successfully rescued in days following this method. Compared with other wild-type WNV isolates, rXJ11129 exhibited virulence indistinguishable from that of the NY99 strain in vivo. In summary, the genomic and virulence phenotypes of rXJ11129 were characterized in vivo and in vitro, and these data will improve the understanding of the spread and pathogenesis of this reemerging virus. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (10.1007/s12250-020-00246-x) contains supplementary material, which is available to authorized users.
- Published
- 2020
- Full Text
- View/download PDF
6. 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
7. Vector Competence and Vertical Transmission of Zika Virus in Aedes albopictus (Diptera: Culicidae)
- Author
-
Qin-mei Liu, Chunxiao Li, Cheng-Feng Qin, Yong-Qiang Deng, Tong-Yan Zhao, Dan Xing, Xiaoxia Guo, Ai-Juan Sun, Wu-Chun Cao, Yande Dong, and Yuting Jiang
- Subjects
0301 basic medicine ,Larva ,Aedes albopictus ,biology ,viruses ,fungi ,030231 tropical medicine ,virus diseases ,030108 mycology & parasitology ,biology.organism_classification ,Microbiology ,Virology ,Zika virus ,Family Flaviviridae ,03 medical and health sciences ,0302 clinical medicine ,Infectious Diseases ,Genus Flavivirus ,Instar ,Pathogen ,Extrinsic incubation period - Abstract
Zika virus (ZIKV) is an emerging mosquito-borne pathogen belonging to the genus Flavivirus of the family Flaviviridae. Aedes albopictus is widely distributed in China. However, little is known about the vector competence of Ae. albopictus in China. The present study presents the oral susceptibility and vector competence of Ae. albopictus Guangzhou strain to ZIKV. Additionally, vertical transmission of ZIKV is described. The results demonstrated the susceptibility of local Ae. albopictus mosquitoes to ZIKV with an extrinsic incubation period of 6 days. Disseminated infection was observed in Ae. albopictus starting on day 2 postinfection (PI). Starting on day 6 PI, the saliva of Ae. albopictus exhibited ZIKV infection, and the transmission rate was 36.4%. Vertical transmission was observed during the first gonotrophic cycle. The minimum infection rate was observed in third-to-fourth instar larvae.
- Published
- 2020
- Full Text
- View/download PDF
8. The pre-existing cellular immunity to Japanese encephalitis virus heterotypically protects mice from Zika virus infection
- Author
-
Weihong Wang, Hong Tang, Li-Na Shi, Yongfen Xu, Cheng-Feng Qin, Fanfan Zhao, Shengyuan Zhang, Weihong Zhang, Shuru Zhou, Jincun Zhao, Xinwen Lin, Yaling Yang, Marion Tarbe, Jin Zhong, Xiaozhen Liang, Jing Sun, Gang Long, Qibin Leng, Feng Yuan, Shuai Liu, Qiuping Xu, and Wei Zhang
- Subjects
education.field_of_study ,Cellular immunity ,Multidisciplinary ,viruses ,Population ,Biology ,Japanese encephalitis ,010502 geochemistry & geophysics ,Southeast asian ,biology.organism_classification ,medicine.disease ,01 natural sciences ,Virology ,Zika virus ,Vaccination ,Immunization ,medicine ,Antibody-dependent enhancement ,education ,0105 earth and related environmental sciences - Abstract
Zika virus (ZIKV) and Japanese encephalitis virus (JEV) are closely related flaviviruses, ZIKV circulates in the population that has been JEV vaccinated in Southeast Asian countries. This alerts that a pre-existing immunity to JEV would impact ZIKV infection and/or pathogenesis. Herein we showed that the pre-existing immunity to JEV SA14-14-2 vaccination provided an ample protection against non-lethal or lethal dose of ZIKV infection in mice. This was in sharp contrast to the passive immunization of JEV antibodies, which failed to affect ZIKV infection or pathogenesis in mice, albeit these antibodies exhibited cross-reactivity and antibody dependent enhancement (ADE) of ZIKV infection in vitro. Furthermore, we determined that JEV vaccine-elicited CD8+ T cells were required to mediate the heterotypic protection of ZIKV infection, which cross-reacted to ZIKV E and NS5 antigens (E294-302 and NS52839-2848). Adoptive transfer of these CD8+ T cells could partially protect the mice from ZIKV challenge. Therefore, although short of epidemiological evidence, these results suggested that cross-reactive CD8+ T cells activated by JEV vaccination could protect potential ZIKV infection in human populations.
- Published
- 2020
- Full Text
- View/download PDF
9. Rational development of a combined mRNA vaccine against COVID-19 and influenza
- Author
-
Cheng-Feng Qin, Qing Ye, Mei Wu, Chao Zhou, Xi-Shan Lu, Ning Zhang, Hui Zhao, Hang Chi, Xiaojing Zhang, Dandan Ling, Rong-Rong Zhang, Dan Luo, Yi Huang, Hongying Qiu, Ke Xu, Bo Ying, Haifeng Song, and Zhuofan Li
- Subjects
viruses ,virus diseases - Abstract
As the world continues to experience the COVID-19 pandemic, seasonal influenza remain a cause of severe morbidity and mortality globally. Worse yet, coinfection with SARS-CoV-2 and influenza A virus (IAV) leads to more severe clinical outcomes. The development of a combined vaccine against both COVID-19 and influenza is thus of high priority. Based on our established lipid nanoparticle (LNP)-encapsulated mRNA vaccine platform, we developed and characterized a novel mRNA vaccine encoding the HA antigen of influenza A (H1N1) virus, termed ARIAV. Then, ARIAV was combined with our COVID-19 mRNA vaccine ARCoV, which encodes the receptor binding domain (RBD) of the SARS-CoV-2 S protein, to formulate the final combined vaccine, AR-CoV/IAV. Further characterization demonstrated that immunization with two doses of AR-CoV/IAV elicited robust protective antibodies as well as antigen-specific cellular immune responses against SARS-CoV-2 and IAV. More importantly, AR-CoV/IAV immunization protected mice from coinfection with IAV and the SARS-CoV-2 Alpha and Delta variants. Our results highlight the potential of the LNP-mRNA vaccine platform in preventing COVID-19 and influenza, as well as other respiratory diseases.
- Published
- 2022
- Full Text
- View/download PDF
10. Anti-viral memory T cell responses in the absence of IgG production in a COVID-19 convalescent individual
- Author
-
Ling Ni, Fang Ye, Qin Qiao, Yu Feng, Yazheng Yang, Hui Zhao, Li-Nan Zhang, Meng-Li Cheng, Gengzhen Zhu, Xiaoli Li, Xuan Zhong, Ruifeng Li, Cheng-Feng Qin, Fang Chen, and Chen Dong
- Subjects
viruses - Abstract
Cellular and humoral immunity are both important in host defense against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Although defects in SARS-CoV-2-specific T cell immunity have been found in patients with severe lung pathology, it is still largely unclear whether virus-specific T cells are sufficient for host protection. Here, we found that in a previously characterized cohort of convalescent subjects, one individual, though lacking detectable anti-viral neutralizing IgG antibodies, showed virus-specific T cell responses, both in CD4+ and CD8+ T cells. SARS-CoV-2-specific T cells in this and other individuals are maintained for up to 10 months. This study thus further supports a critical role of T cells in host defense against SARS-CoV-2, offering new insights into the design and evaluation of COVID-19 vaccines.
- Published
- 2022
- Full Text
- View/download PDF
11. 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
12. 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
13. 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
14. Electrostatic Interaction Between NS1 and Negatively Charged Lipids Contributes to Flavivirus Replication Organelles Formation
- Author
-
Yali Ci, Yang Yang, Caimin Xu, Cheng-Feng Qin, and Lei Shi
- Subjects
Microbiology (medical) ,viruses ,Phosphatidylinositol Phosphates ,medicine.disease_cause ,replication organelles ,Microbiology ,03 medical and health sciences ,0302 clinical medicine ,flavivirus ,Organelle ,non-structural protein 1 ,medicine ,PI3K/AKT/mTOR pathway ,Original Research ,030304 developmental biology ,Electrostatic interaction ,0303 health sciences ,Mutation ,biology ,Chemistry ,virus diseases ,PIP ,biochemical phenomena, metabolism, and nutrition ,biology.organism_classification ,ER remodeling ,QR1-502 ,Cell biology ,Flavivirus ,Membrane ,030217 neurology & neurosurgery ,Intracellular - Abstract
Flavivirus replication occurs in membranous replication compartments, also known as replication organelles (ROs) derived from the host ER membrane. Our previous study showed that the non-structural (NS) protein 1 (NS1) is the essential factor for RO creation by hydrophobic insertion into the ER membrane. Here, we found that the association of NS1 with the membrane can be facilitated by the electrostatic interaction between NS1 and negatively charged lipids. NS1 binds to a series of negatively charged lipids, including PI4P, and a positively charged residue, R31, located on the membrane-binding face of NS1, plays important roles in this interaction. The NS1 R31E mutation significantly impairs NS1 association with negatively charged membrane and its ER remodeling ability in the cells. To interfere with the electrostatic interaction between NS1 and negatively charged lipids, intracellular phosphatidylinositol phosphates (PIPs) level was downregulated by the overexpression of Sac1 or treatment with PI3K and PI4K inhibitors to attenuate flavivirus replication. Our findings emphasize the importance of electrostatic interaction between NS1 and negatively charged lipids in flavivirus RO formation.
- Published
- 2021
- Full Text
- View/download PDF
15. Nanometer resolution in situ structure of SARS-CoV-2 post-fusion spike
- Author
-
Linhua Tai, Xiaorui Xing, Rao Zihe, Yun Zhu, Fei Sun, Chun Chan, Guoliang Zhu, Minnan Yang, Xiangxi Wang, Guoliang Yin, Lei Cao, and Cheng-Feng Qin
- Subjects
In situ ,Fusion ,Optics ,Materials science ,business.industry ,viruses ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Resolution (electron density) ,Nanometre ,Spike (software development) ,business - Abstract
The spike protein (S) of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) mediates membrane fusion to allow entry of viral genome into host cell. To understand its detailed entry mechanism and develop specific entry inhibitor, the in situ structural information of SARS-CoV-2 spikes in different states are urgently important. Here, by using the cryo-electron microscopic tomograms, we observed spikes of inactivated SARS-CoV-2 virions in both pre-fusion and post-fusion states and solved the nanometer resolution structure of in situ post-fusion spike. With a more complete model compared to previous reports, the relative spatial position between fusion peptide and transmembrane domain was discovered. Novel oligomerizations of spikes on viral membrane were observed, likely suggesting a new mechanism of fusion pore formation.
- Published
- 2021
- Full Text
- View/download PDF
16. Structure and function analysis of a potent human neutralizing antibody CA521LALA against SARS-CoV-2
- Author
-
Changlin Dou, Cheng-Feng Qin, Lan Wang, Deyong Song, Yong-Qiang Deng, Wenbo Wang, Chuangchuang Dong, Zhenfei Ning, Xiu Liu, Chuan Liu, Guangying Du, Chunjie Sha, Kailin Wang, Jun Lu, Baiping Sun, Yanyan Zhao, Qiaoping Wang, Hongguang Xu, Ying Li, Zhenduo Shen, Jie Jiao, Ruiying Wang, Jingwei Tian, and Wanhui Liu
- Subjects
viruses - Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of the ongoing coronavirus disease 2019 (COVID-19) pandemic, which has resulted in ~1,119,431 deaths. There is currently no approved vaccines or therapeutics for treating COVID-19. The SARS-CoV-2 Spike protein promotes entry into host cells and is considered a key therapeutic target by many researchers. Here we describe the identification of several monoclonal antibodies that target the SARS-CoV-2 Spike protein. One human antibody, CA521LALA, demonstrated neutralization potential by immunizing human antibody transgenic mice. CA521LALA showed potent SARS-CoV-2-specific neutralization activity against SARS-CoV-2 pseudovirus and authentic SARS-CoV-2 infection in vitro. The LALA mutation introduced to CA521 abrogates the binding with Fc receptors or complement receptors reducing antibody-dependent enhancement seen with anti-SARS-CoV antibodies. CA521LALA also demonstrated having a long half-life of 9.5 days in mice and 9.3 days in rhesus monkeys. CA521LALA inhibited SARS-CoV-2 infection in SARS-CoV-2 susceptible mice at a therapeutic setting with the virus titer of the lung reduced by 4.5 logs. Structural analysis by cryo-EM revealed that CA521LALA recognizes an epitope overlapping with angiotensin converting enzyme 2 (ACE2)-binding sites in SARS-CoV-2 receptor binding domain (RBD) in the Spike protein. CA521LALA blocks the interaction by binding all three RBDs of one SARS CoV-2 spike trimer simultaneously. These results demonstrate the importance for antibody-based therapeutic interventions in the treatment of COVID-19 and identifies CA521LALA a promising antibody that reacts with SARS-CoV-2 Spike protein to strongly neutralize its activity.
- Published
- 2021
- Full Text
- View/download PDF
17. Double lock of a potent human therapeutic monoclonal antibody against SARS-CoV-2
- Author
-
Liangzhi Xie, Desheng Kong, Yao Sun, Xiaofeng Li, Xiaorui Xing, Ling Zhu, Qi Chen, Junjie Hu, Yong Qiang Deng, Lei Cao, Chunxia Luo, Zhen Cui, Zihe Rao, Weijin Huang, Jianhui Nie, Xiangxi Wang, Lei Wang, Neil Shaw, Youchun Wang, Na Na Zhang, Zhe Lv, Chun Yun Sun, Chang Fa Fan, Tian Shu Cao, Cheng-Feng Qin, Qing Ye, Yanjing Zhang, Huiyu Wang, Rong Rong Zhang, Nan Wang, Qianqian Li, and Juan Ma
- Subjects
Coronavirus disease 2019 (COVID-19) ,AcademicSubjects/SCI00010 ,medicine.drug_class ,viruses ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Monoclonal antibody ,Cryo-EM structure ,03 medical and health sciences ,0302 clinical medicine ,Fab Fragments ,medicine ,skin and connective tissue diseases ,Receptor ,030304 developmental biology ,0303 health sciences ,in vivo protection ,Multidisciplinary ,biology ,SARS-CoV-2 ,Chemistry ,preclinical safety evaluation ,fungi ,COVID-19 ,Lipid bilayer fusion ,immuno-therapy ,Effective dose (pharmacology) ,Cell biology ,biology.protein ,human neutralizing antibody ,Antibody ,AcademicSubjects/MED00010 ,030217 neurology & neurosurgery ,Research Article - Abstract
Receptor recognition and subsequent membrane fusion are essential for the establishment of successful infection by SARS-CoV-2. Halting these steps can cure COVID-19. Here we have identified and characterized a potent human monoclonal antibody, HB27, that blocks SARS-CoV-2 attachment to its cellular receptor at sub-nM concentrations. Remarkably, HB27 can also prevent SARS-CoV-2 membrane fusion. Consequently, a single dose of HB27 conferred effective protection against SARS-CoV-2 in two established mouse models. Rhesus macaques showed no obvious adverse events when administrated with 10 times the effective dose of HB27. Cryo-EM studies on complex of SARS-CoV-2 trimeric S with HB27 Fab reveal that three Fab fragments work synergistically to occlude SARS-CoV-2 from binding to the ACE2 receptor. Binding of the antibody also restrains any further conformational changes of the receptor binding domain, possibly interfering with progression from the prefusion to the postfusion stage. These results suggest that HB27 is a promising candidate for immuno-therapies against COVID-19.
- Published
- 2020
- Full Text
- View/download PDF
18. 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
19. Establishment of replication-competent vesicular stomatitis virus-based recombinant viruses suitable for SARS-CoV-2 entry and neutralization assays
- Author
-
Xuanling Shi, Yuhang Zhang, Qi Zhang, Linqi Zhang, Cheng-Feng Qin, Fei Yuan, Hongyue Li, Chaoyue Zhao, and Aihua Zheng
- Subjects
0301 basic medicine ,Epidemiology ,viruses ,030106 microbiology ,Immunology ,Pneumonia, Viral ,Virus Replication ,Microbiology ,Neutralization ,Vesicular stomatitis Indiana virus ,law.invention ,Cell Line ,03 medical and health sciences ,Betacoronavirus ,Viral envelope ,Viral Envelope Proteins ,law ,neutralization assay ,Virology ,Drug Discovery ,Humans ,skin and connective tissue diseases ,Pandemics ,Tropism ,replication-competent ,biology ,SARS-CoV-2 ,fungi ,virus diseases ,COVID-19 ,General Medicine ,Virus Internalization ,biology.organism_classification ,body regions ,030104 developmental biology ,Infectious Diseases ,Viral replication ,Vesicular stomatitis virus ,VSV ,Recombinant DNA ,biology.protein ,entry ,Parasitology ,Antibody ,Coronavirus Infections ,Research Article - Abstract
Replication-competent vesicular stomatitis virus (VSV)-based recombinant viruses are useful tools for studying emerging and highly pathogenic enveloped viruses in level 2 biosafety facilities. Here, we used a replication-competent recombinant VSVs (rVSVs) encoding the spike (S) protein of SARS-CoV-2 in place of the original G glycoprotein (rVSV-eGFP-SARS-CoV-2) to develop a high-throughput entry assay for SARS-CoV-2. The S protein was incorporated into the recovered rVSV-eGFP-SARS-CoV-2 particles, which could be neutralized by sera from convalescent COVID-19 patients. The recombinant SARS-CoV-2 also displayed entry characteristics similar to the wild type virus, such as cell tropism and pH-dependence. The neutralizing titers of antibodies and sera measured by rVSV-eGFP-SARS-CoV-2 were highly correlated with those measured by wild-type viruses or pseudoviruses. Therefore, this is a safe and convenient screening tool for SARS-CoV-2, and it may promote the development of COVID-19 vaccines and therapeutics.
- Published
- 2020
20. Persistent Viral Presence Determines the Clinical Course of the Disease in COVID-19
- Author
-
Xiaohua Peng, Dawei Zhang, Jing-Hui Dong, Zhu Chen, Cheng-Feng Qin, Peng Zhao, De Chang, Bo-An Li, Lokesh Sharma, Charles S. Dela Cruz, Fu-Sheng Wang, Enqiang Qin, Lixin Xie, Bo-Yu Li, Hongxia Liu, Zhe Xu, and Guang Yang
- Subjects
Male ,viruses ,Disease ,Comorbidity ,Host recovery ,Severity of Illness Index ,Immunology and Allergy ,Child ,COVID-19, Coronavirus disease 2019 ,biology ,Viral culture ,Middle Aged ,C-Reactive Protein ,Viral persistence ,Child, Preschool ,CRP, C-reactive protein ,Female ,medicine.symptom ,Inflammation Mediators ,Coronavirus Infections ,Adult ,Adolescent ,Pneumonia, Viral ,Viral clearance ,Inflammation ,Real-Time Polymerase Chain Reaction ,Virus ,Article ,AMMS, Academy of Military Medical Sciences ,Betacoronavirus ,Young Adult ,Severity of illness ,medicine ,Humans ,Interleukin 6 ,Glucocorticoids ,Pandemics ,Disease severity ,Aged ,SARS-CoV-2, Severe acute respiratory syndrome coronavirus 2 ,business.industry ,Interleukin-6 ,SARS-CoV-2 ,C-reactive protein ,COVID-19 ,Infant ,medicine.disease ,Respiration, Artificial ,Immunology ,biology.protein ,business - Abstract
Background The clinical management of coronavirus disease 2019 (COVID-19) is dependent on understanding the underlying factors that contribute to the disease severity. In the absence of effective antiviral therapies, other host immunomodulatory therapies such as targeting inflammatory response are currently being used without clear evidence of their effectiveness. Because inflammation is an essential component of host antiviral mechanisms, therapies targeting inflammation may adversely affect viral clearance and disease outcome. Objective To understand whether the persistent presence of the virus is a key determinant in the disease severity during COVID-19 and to determine whether the viral reactivation in some patients is associated with infectious viral particles. Methods The data for patients were available including the onset of the disease, duration of viral persistence, measurements of inflammatory markers such as IL-6 and C-reactive protein, chest imaging, disease symptoms, and their durations among others. Follow-up tests were performed to determine whether the viral negative status persists after their recovery. Results Our data show that patients with persistent viral presence (>16 days) have more severe disease outcomes including extensive lung involvement and requirement of respiratory support. Two patients who died of COVID-19 were virus-positive at the time of their death. Four patients demonstrated virus-positive status on the follow-up tests, and these patient samples were sent to viral culture facility where virus culture could not be established. Conclusions These data suggest that viral persistence is the key determining factor of the disease severity. Therapies that may impair the viral clearance may impair the host recovery from COVID-19.
- Published
- 2020
- Full Text
- View/download PDF
21. 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
22. 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
23. 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
24. HDL-scavenger receptor B type 1 facilitates SARS-CoV-2 entry
- Author
-
Cheng-Feng Qin, Xiaoli Yang, Jin Sun, Xuejun Wang, Hui Zhong, Rui Zhang, Luming Wan, Wei Chen, Jianmin Li, Feng Yin, Zhe Zhang, Nan Wang, Yanhong Zhang, Yumeng Peng, Yong-Qiang Deng, Rong Wang, Jun Zhang, Huilong Li, Chen Fan, Yufei Wang, Xiaolin Wang, Haotian Lin, Jiangyue Feng, Huan Yang, Yuan Cao, Peng Du, Xiaopan Yang, Qi Gao, Congwen Wei, Yulong Zong, Jing Gong, Wanchuan Zhang, Dongyu Li, and Qiulin Yan
- Subjects
viruses ,Endocrinology, Diabetes and Metabolism ,Virus Attachment ,Plasma protein binding ,Biology ,medicine.disease_cause ,Virus ,Cell Line ,Physiology (medical) ,medicine ,Internal Medicine ,Humans ,Scavenger receptor ,skin and connective tissue diseases ,Receptor ,Coronavirus ,Host factor ,SARS-CoV-2 ,fungi ,COVID-19 ,Cell Biology ,Scavenger Receptors, Class B ,Virus Internalization ,Virology ,body regions ,Viral Tropism ,Cholesterol ,Cell culture ,Host-Pathogen Interactions ,Spike Glycoprotein, Coronavirus ,Tissue tropism ,Receptors, Virus ,Disease Susceptibility ,Lipoproteins, HDL ,Protein Binding - Abstract
Responsible for the ongoing coronavirus disease 19 (COVID-19) pandemic, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infects host cells through binding of the viral spike protein (SARS-2-S) to the cell-surface receptor angiotensin-converting enzyme 2 (ACE2). Here we show that the high-density lipoprotein (HDL) scavenger receptor B type 1 (SR-B1) facilitates ACE2-dependent entry of SARS-CoV-2. We find that the S1 subunit of SARS-2-S binds to cholesterol and possibly to HDL components to enhance viral uptake in vitro. SR-B1 expression facilitates SARS-CoV-2 entry into ACE2-expressing cells by augmenting virus attachment. Blockade of the cholesterol-binding site on SARS-2-S1 with a monoclonal antibody, or treatment of cultured cells with pharmacological SR-B1 antagonists, inhibits HDL-enhanced SARS-CoV-2 infection. We further show that SR-B1 is coexpressed with ACE2 in human pulmonary tissue and in several extrapulmonary tissues. Our findings reveal that SR-B1 acts as a host factor that promotes SARS-CoV-2 entry and may help explain viral tropism, identify a possible molecular connection between COVID-19 and lipoprotein metabolism, and highlight SR-B1 as a potential therapeutic target to interfere with SARS-CoV-2 infection.
- Published
- 2020
25. Detection of SARS-CoV-2-Specific Humoral and Cellular Immunity in COVID-19 Convalescent Individuals
- Author
-
Xinquan Wang, Peng Wei, Chao Zhou, Mengting Gou, Ling Ni, Tian-Shu Cao, Jiwan Ge, Yong Qiang Deng, Han Guo, Rong-Rong Zhang, Xiaoli Li, Cheng-Feng Qin, Yu Feng, Fang Ye, Fang Chen, Meng Li Cheng, Chen Dong, Pengzhi Wang, Lin Sun, Peng Liang, and Hui Zhao
- Subjects
0301 basic medicine ,Adult ,Male ,Cellular immunity ,viruses ,Pneumonia, Viral ,Immunology ,Biology ,Antibodies, Viral ,Neutralization ,Immunoglobulin G ,Virus ,Article ,03 medical and health sciences ,Betacoronavirus ,0302 clinical medicine ,Immune system ,Humans ,Immunology and Allergy ,Pandemics ,Immunity, Cellular ,SARS-CoV-2 ,Antibody titer ,COVID-19 ,Convalescence ,adaptive immunity ,biochemical phenomena, metabolism, and nutrition ,Middle Aged ,Acquired immune system ,Antibodies, Neutralizing ,COVID-19 patients ,SARS-CoV-2-specific antibody ,Immunity, Humoral ,030104 developmental biology ,SARS-CoV-2-specific T cells ,Infectious Diseases ,030220 oncology & carcinogenesis ,Spike Glycoprotein, Coronavirus ,biology.protein ,Female ,Antibody ,Coronavirus Infections - Abstract
Summary The World Health Organization has declared SARS-CoV-2 virus outbreak a world-wide pandemic. However, there is very limited understanding on the immune responses, especially adaptive immune responses to SARS-CoV-2 infection. Here, we collected blood from COVID-19 patients who have recently become virus-free and therefore were discharged, and detected SARS-CoV-2-specific humoral and cellular immunity in 8 newly discharged patients. Follow-up analysis on another cohort of 6 patients 2 weeks post discharge also revealed high titers of IgG antibodies. In all 14 patients tested, 13 displayed serum neutralizing activities in a pseudotype entry assay. Notably, there was a strong correlation between neutralization antibody titers and the numbers of virus-specific T cells. Our work provides a basis for further analysis of protective immunity to SARS-CoV-2, and understanding the pathogenesis of COVID-19, especially in the severe cases. It has also implications in developing an effective vaccine to SARS-CoV-2 infection., Highlights 1. SARS-CoV-2-specific antibodies are detected in COVID-19 convalescent subjects. 2. Most COVID-19 convalescent individuals have detectable neutralizing antibodies. 3. Cellular immune responses to SARS-CoV-2 are found in COVID-19 convalescent subjects 4. Neutralization antibody titers correlate with the numbers of virus-specific T cells., In blood samples from COVID-19 convalescent subjects, Ni et al. have detected SARS-CoV-2-specific humoral and cellular immunity. Most subjects display serum neutralizing activities, which correlate with the numbers of virus-specific T cells.
- Published
- 2020
- Full Text
- View/download PDF
26. Broad-spectrum virucidal activity of bacterial secreted lipases against flaviviruses, SARS-CoV-2 and other enveloped viruses
- Author
-
Yibin Zhu, Xi Yu, Lei Zhang, Liangqin Tong, Xiaoping Xiao, Penghua Wang, Yun Yang, Mingyu Shi, Han Wang, Cheng-Feng Qin, Nana Zhang, Qiang Ding, Linqi Zhang, and Gong Cheng
- Subjects
Infectivity ,biology ,medicine.drug_class ,viruses ,Dengue virus ,biology.organism_classification ,medicine.disease_cause ,Virology ,Zika virus ,Herpes simplex virus ,Viral envelope ,Chromobacterium ,biology.protein ,medicine ,Lipase ,Antiviral drug - Abstract
Viruses are the major aetiological agents of acute and chronic severe human diseases that place a tremendous burden on global public health and economy; however, for most viruses, effective prophylactics and therapeutics are lacking, in particular, broad-spectrum antiviral agents. Herein, we identified 2 secreted bacterial lipases from aChromobacteriumbacterium, namedChromobacteriumantiviral effector-1 (CbAE-1) andCbAE-2, with a broad-spectrum virucidal activity against dengue virus (DENV), Zika virus (ZIKV), severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), human immunodeficiency virus (HIV) and herpes simplex virus (HSV). TheCbAEs potently blocked viral infection in the extracellular milieu through their lipase activity. Mechanistic studies showed that this lipase activity directly disrupted the viral envelope structure, thus inactivating infectivity. A mutation ofCbAE-1 in its lipase motif fully abrogated the virucidal ability. Furthermore,CbAE-2 presented low toxicityin vivoandin vitro, highlighting its potential as a broad-spectrum antiviral drug.
- Published
- 2020
- Full Text
- View/download PDF
27. Rapid adaptation of SARS-CoV-2 in BALB/c mice: Novel mouse model for vaccine efficacy
- Author
-
Yujun Cui, Na-Na Zhang, Xiliang Wang, Cheng-Feng Qin, Qi Chen, Yong-Qiang Deng, Shaolong Chen, Hongjing Gu, Hang Fan, Xiaofeng Li, Yuchang Li, Bojiang Shi, Jiangfan Li, Xiaolan Yang, Yanxiao Wang, Xiaojun Zhou, Shihui Sun, Xiaoli Sheng, Yue Teng, Guangyu Zhao, Lei He, Xiao Yang, Deyan Luo, Shusheng Geng, Yan Li, Zhou Yusen, Zhongpeng Zhao, Guan Yang, Yuxian He, Hui Wang, and Gencheng Han
- Subjects
Infectivity ,biology ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Protein subunit ,viruses ,fungi ,Vaccine efficacy ,biology.organism_classification ,Virology ,BALB/c ,respiratory tract diseases ,Animal model ,In vivo ,biology.protein ,Antibody ,skin and connective tissue diseases - Abstract
Coronavirus disease 2019 (COVID-19) threatens global public health and economy. In order to develop safe and effective vaccines, suitable animal models must be established. Here we report the rapid adaption of SARS-CoV-2 in BALB/c mice, based on which a convenient, economical and effective animal model was developed. Specifically, we found that mouse-adapted SARS-CoV-2 at passage 6 (MACSp6) efficiently infected both aged and young wild-type BALB/c mice, resulting in moderate pneumonia as well as inflammatory responses. The elevated infectivity of MACSp6 in mice could be attributed to the substitution of a key residue (N501Y) in the receptorbinding domain (RBD). Using this novel animal model, we further evaluated the in vivo protective efficacy of an RBD-based SARS-CoV-2 subunit vaccine, which elicited highly potent neutralizing antibodies and conferred full protection against SARS-CoV-2 MACSp6 challenge. This novel mouse model is convenient and effective in evaluating the in vivo protective efficacy of SARS-CoV-2 vaccine.SummaryThis study describes a unique mouse model for SARS-CoV-2 infection and confirms protective efficacy of a SARS-CoV-2 RBD subunit vaccine.
- Published
- 2020
- Full Text
- View/download PDF
28. Rapid development of an inactivated vaccine for SARS-CoV-2
- Author
-
Wen Shi, Ge Xiaoqin, Deyu Jiang, Jiangning Liu, Xiuyu Lou, Linlin Bao, Fang Cai, Ling Zhu, Minnan Yang, Biao Kan, Li Yurong, Qiang Gao, Dongdong Wu, Cheng-Feng Qin, Chuan Qin, Nan Wang, Li Yajing, Yaling Hu, Xuejie Gong, Yanhui Yin, Changgui Li, Weidong Yin, Hong Gao, Jinxing Lu, Jing Li, Wei Deng, Lin Wang, Kangwei Xu, Yanjun Zhang, Hengming Zhang, Haiyan Mao, Zhu Lang, Xiangxi Wang, and Zhe Lv
- Subjects
Coronavirus disease 2019 (COVID-19) ,biology ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,viruses ,fungi ,Virology ,Virus ,respiratory tract diseases ,body regions ,Immunization ,Immunopathology ,Inactivated vaccine ,Pandemic ,biology.protein ,Antibody ,skin and connective tissue diseases - Abstract
The COVID-19 pandemic caused by SARS-CoV-2 has brought about an unprecedented crisis, taking a heavy toll on human health, lives as well as the global economy. There are no SARS-CoV-2-specific treatments or vaccines available due to the novelty of this virus. Hence, rapid development of effective vaccines against SARS-CoV-2 is urgently needed. Here we developed a pilot-scale production of a purified inactivated SARS-CoV-2 virus vaccine candidate (PiCoVacc), which induced SARS-CoV-2-specific neutralizing antibodies in mice, rats and non-human primates. These antibodies potently neutralized 10 representative SARS-CoV-2 strains, indicative of a possible broader neutralizing ability against SARS-CoV-2 strains circulating worldwide. Immunization with two different doses (3μg or 6 μg per dose) provided partial or complete protection in macaques against SARS-CoV-2 challenge, respectively, without any antibody-dependent enhancement of infection. Systematic evaluation of PiCoVacc via monitoring clinical signs, hematological and biochemical index, and histophathological analysis in macaques suggests that it is safe. These data support the rapid clinical development of SARS-CoV-2 vaccines for humans.One Sentence SummaryA purified inactivated SARS-CoV-2 virus vaccine candidate (PiCoVacc) confers complete protection in non-human primates against SARS-CoV-2 strains circulating worldwide by eliciting potent humoral responses devoid of immunopathology
- Published
- 2020
- Full Text
- View/download PDF
29. Rational Design of a Replication-Competent and Inheritable Magnetic Viruses for Targeting Biomedical Applications
- Author
-
Tianxu Yang, Cheng-Feng Qin, Ruiting Li, Ying Zhang, Yan Guo, Xiaoyu Wang, Guangchuan Wang, Xiaofeng Li, Rong-Rong Zhang, Hangyu Zhou, Ruikang Tang, Yan-Peng Xu, and Yueqi Zhao
- Subjects
inorganic chemicals ,Picornavirus ,viruses ,02 engineering and technology ,010402 general chemistry ,01 natural sciences ,Virus ,Biomaterials ,In vivo ,Humans ,General Materials Science ,Tropism ,Enterovirus ,Infectivity ,biology ,Chemistry ,Magnetic Phenomena ,Rational design ,General Chemistry ,021001 nanoscience & nanotechnology ,biology.organism_classification ,In vitro ,0104 chemical sciences ,Cell biology ,Nanoparticles ,0210 nano-technology ,Surface protein ,Biotechnology - Abstract
Infection with live-attenuated vaccines always inevitably induces side effects that reduce their safety. This study suggests a concept of magnetic virus produced by genetically modifying viral surfaces with Fe3 O4 nanoparticles (NPs) to control their tropisms. An iron-affinity peptide is designed to be displayed on the viral surface protein (VP1) of human enterovirus type 71 (EV71), a typical nonenveloped picornavirus, as the model. The modified EV71 can self-bind with Fe3 O4 NPs under physiological conditions, resulting in novel EV71-Fe3 O4 hybrid materials. This rationally engineered EV71 with Fe3 O4 retains its original biological infectivity, but its tropism can be precisely controlled by magnetism. Both in vitro and in vivo experiments demonstrate that EV71-Fe3 O4 can infect only a desired area within the limit of the applied magnetic field, which effectively reduces its pathological damage. More importantly, this characteristic of EV71 can be inherited due to the gene-induced coassembly of viruses and NPs. This achievement provides a proof of concept in virus vaccine improvement by a combination of gene modification and material incorporation, leading to great potential for biomedical developments.
- Published
- 2020
30. 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
31. Characterization of anti-viral immunity in recovered individuals infected by SARS-CoV-2
- Author
-
Meng-Li Chen, Ling Ni, Cheng-Feng Qin, Jiwan Ge, Xinquan Wang, Xiaoli Li, Yong-Qiang Deng, Yu Feng, Hui Zhao, Chen Dong, Peng Wei, Peng Liang, Lin Sun, Han Guo, Fang Ye, Fang Chen, and Pengzhi Wang
- Subjects
Cellular immunity ,biology ,business.industry ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,T cell ,viruses ,fungi ,Outbreak ,biochemical phenomena, metabolism, and nutrition ,Pathogenesis ,Titer ,Immune system ,medicine.anatomical_structure ,Immunology ,biology.protein ,Medicine ,Antibody ,business ,skin and connective tissue diseases - Abstract
The WHO has declared SARS-CoV-2 outbreak a public health emergency of international concern. However, to date, there was hardly any study in characterizing the immune responses, especially adaptive immune responses to SARS-CoV-2 infection. In this study, we collected blood from COVID-19 patients who have recently become virus-free and therefore were discharged, and analyzed their SARS-CoV-2-specific antibody and T cell responses. We observed SARS-CoV-2-specific humoral and cellular immunity in the patients. Both were detected in newly discharged patients, suggesting both participate in immune-mediated protection to viral infection. However, follow-up patients (2 weeks post discharge) exhibited high titers of IgG antibodies, but with low levels of virus-specific T cells, suggesting that they may enter a quiescent state. Our work has thus provided a basis for further analysis of protective immunity to SARS-CoV-2, and understanding the pathogenesis of COVID-19, especially in the severe cases. It has also implications in designing an effective vaccine to protect and treat SARS-CoV-2 infection.
- Published
- 2020
- Full Text
- View/download PDF
32. A single nonsynonymous mutation on gene encoding E protein of Zika virus leads to increased neurovirulence in vivo
- Author
-
Ge N, Zunpeng Liu, Cheng-Feng Qin, Yan Zhang, Jin Xin, Cheng M, Yigang Tong, Shu J, and Ziying Xu
- Subjects
Nonsynonymous substitution ,0303 health sciences ,Microcephaly ,biology ,Viral pathogenesis ,viruses ,Virulence ,medicine.disease ,biology.organism_classification ,Virology ,3. Good health ,Zika virus ,03 medical and health sciences ,0302 clinical medicine ,Viral envelope ,medicine ,Missense mutation ,Gene ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
Zika virus can infect a wide range of tissues including the developmental brain of human fetuses, causing from mild to severe clinical diseases. Whether its genetic characteristics impacts on viral pathogenesis is incompletely understood. We have obtained viral variants through serially passage of a clinical Zika virus isolate (SW01) in neonatal mice in vivo and found some of which exhibited markedly increased virulence and neurotropism. By deep sequencing analysis, the more pathogenic viral variants were found to contain four dominant nonsynonymous nucleotide mutations on genes encoding E and NS2A proteins. Further investigation using molecularly cloned viruses revealed that a single 67D (Aspatic acid) to N (Asparagine) substitution on E protein is sufficient to confer the increased virulence and neurotropism. These findings provide new insight into Zika virus pathogenesis and suggest novel targets for the development of therapeutics.Author SummaryRecent large outbreaks of Zika virus infection worldwide have revealed an association between the viral infection and increased cases of specific neurological problems including Congenital Zika Syndrome (including microcephaly) and adult Guillain–Barré Syndrome. However, the determinants of the increased neurovirulence of Zika virus remain uncertain. One hypothesis is that some unique changes across the Zika viral genome have led to the occurrence of these neurological diseases. To test this hypothesis, we continuously propagated a clinical isolate of contemporary Zika virus (SW01) in neonatal mice brain for 11 times to obtain an mouse central nervous system (CNS) adapted Zika virus (MA-SW01) that showed significantly increased neurovirulence in vivo. We then discovered that a single G to A nucleotide substitution at the 1069 site of Zika virus open reading frame leading to a D (aspartic acid) to N (asparagine) in viral Envelope protein is responsible for the increased neurovirulence. These findings improve our understanding of the neurological pathogenesis of Zika virus and provide clues for the development of antiviral strategy.
- Published
- 2020
- Full Text
- View/download PDF
33. Biomimetic inorganic camouflage circumvents antibody-dependent enhancement of infection
- Author
-
Yong-Qiang Deng, Cheng-Feng Qin, Qing-Gong Nian, Xiaoyu Wang, Xurong Xu, Xiaofeng Li, Ruikang Tang, Yun Xiao, Hui Zhao, and Dong Yang
- Subjects
0301 basic medicine ,viruses ,Immunogenicity ,General Chemistry ,Dengue virus ,Biology ,medicine.disease_cause ,Virology ,Virus ,In vitro ,03 medical and health sciences ,030104 developmental biology ,Immune system ,In vivo ,medicine ,biology.protein ,Antibody-dependent enhancement ,Antibody - Abstract
Pre-existing antibodies can aggravate disease during subsequent infection or vaccination via the mechanism of antibody-dependent enhancement (ADE) of infection. Herein, using dengue virus (DENV) as a model, we present a versatile surface-camouflage strategy to obtain a virus core-calcium phosphate shell hybrid by self-templated biomineralization. The shelled DENV stealthily avoids recognition by pre-existing antibodies under extracellular conditions, resulting in the efficient abrogation of the ADE of infection both in vitro and in vivo. Moreover, the nanoshell can spontaneously degrade under intracellular conditions to restore the virus activity and immunogenicity due to its pH-sensitive behaviour. This work demonstrates that the biomimetic material shell can significantly improve the administration safety and potency of the DENV vaccine, which provides the promising prospect of chemically designed virus-material hybrids for immune evasion.
- Published
- 2017
- Full Text
- View/download PDF
34. Zika NS1–induced ER remodeling is essential for viral replication
- Author
-
Cheng-Feng Qin, Na-Na Zhang, Caimin Xu, Yuqiang Niu, Zhong-Yu Liu, Lei Shi, Yali Ci, Wei Yang, and Yang Yang
- Subjects
viruses ,Tissue membrane ,Viral Nonstructural Proteins ,Endoplasmic Reticulum ,Virus Replication ,Article ,Zika virus ,03 medical and health sciences ,Replication compartment ,Humans ,Research Articles ,030304 developmental biology ,0303 health sciences ,biology ,Zika Virus Infection ,Vesicle ,030302 biochemistry & molecular biology ,virus diseases ,Zika Virus ,Cell Biology ,biochemical phenomena, metabolism, and nutrition ,biology.organism_classification ,Cell biology ,Flavivirus ,Viral replication ,Ultrastructure ,Biogenesis - Abstract
Zika virus induces the formation of vesicles from ER membrane that form viral replication factories in the ER lumen. Ci et al. show that the Zika NS1 protein plays a key role in this remodeling of the ER as the insertion of the hydrophobic regions of NS1 into the inner leaflet of the ER membrane creates the compartments essential for viral replication., Zika virus (ZIKV), a recently emerged member of the flavivirus family, forms replication compartments at the ER during its lifecycle. The proteins that are responsible for the biogenesis of replication compartments are not well defined. Here, we show that Zika nonstructural protein 1 (NS1)–induced ER remodeling is essential for viral replication. NS1 expressed in the ER lumen induced ER perinuclear aggregation with an ultrastructure resembling that of the replication compartment. Data from model membrane system indicated that the membrane-binding and membrane-remodeling properties of NS1 depend on its hydrophobic insertion into the membrane. These findings demonstrate that NS1 plays a crucial role in flavivirus replication compartment formation by remodeling the ER structure., Graphical Abstract
- Published
- 2019
- Full Text
- View/download PDF
35. 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
36. 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
37. Zika NS2B is a crucial factor recruiting NS3 to the ER and activating its protease activity
- Author
-
Shan Xu, Lei Shi, Caimin Xu, Fangfei Jia, Yang Yang, Cheng-Feng Qin, and Huaipeng Xing
- Subjects
Cancer Research ,viruses ,medicine.medical_treatment ,Biology ,Viral Nonstructural Proteins ,medicine.disease_cause ,Endoplasmic Reticulum ,Virus Replication ,Zika virus ,03 medical and health sciences ,Viral Proteins ,Virology ,medicine ,Humans ,030304 developmental biology ,0303 health sciences ,Mutation ,NS3 ,Protease ,030306 microbiology ,Endoplasmic reticulum ,Serine Endopeptidases ,virus diseases ,Zika Virus ,biochemical phenomena, metabolism, and nutrition ,biology.organism_classification ,digestive system diseases ,Mitochondria ,Flavivirus ,Infectious Diseases ,HEK293 Cells ,Viral protease ,Function (biology) ,HeLa Cells ,Peptide Hydrolases ,Protein Binding - Abstract
Zika virus (ZIKV) is an emergent flavivirus associated with severe neurological disorders. ZIKV NS3 protein is a viral protease that cleaves the ZIKV polyprotein precursor into individual viral proteins. In this study, we found that ZIKV NS3 by itself exhibited mitochondrial localization, which was quite different from its endoplasmic reticulum (ER) localization in ZIKV-infected cells. We screened viral proteins and identified NS2B as the bona fide recruiter of NS3 to the ER. The NS2B C-terminal tail interacted with NS3 protease domain to retain NS3 on the ER. β-Sheet motifs that formed between NS2B and the NS3 protease domain played important roles in their interaction, while mutation in the β-strand of NS2B attenuated NS2B-NS3 interaction and impaired the ability of NS3 protease to cleave the polyprotein precursor into multiple viral proteins. Consequently, NS2B mutations led to severe inhibition of ZIKV replication and production due to insufficient NS3 protease activity. In summary, our study reveals the critical role of NS2B in NS3 recruitment and protease function and provides mechanistic insight into ZIKV replication.
- Published
- 2019
38. 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
39. 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
40. 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
41. Vector competence of Aedes albopictus and Aedes aegypti (Diptera: Culicidae) for the DEN2-FJ10 and DEN2-FJ11 strains of the dengue 2 virus in Fujian, China
- Author
-
Cheng-Feng Qin, Xiaoxia Guo, Yingmei Zhang, Dan Xing, Zhang Hengduan, Yande Dong, Tong-Yan Zhao, and Chunxiao Li
- Subjects
0301 basic medicine ,China ,Aedes albopictus ,viruses ,Veterinary (miscellaneous) ,education ,030231 tropical medicine ,Virulence ,Aedes aegypti ,Dengue virus ,medicine.disease_cause ,Virus ,Dengue fever ,Dengue ,03 medical and health sciences ,0302 clinical medicine ,Aedes ,medicine ,Animals ,Humans ,biology ,fungi ,Genetic Variation ,Dengue Virus ,biology.organism_classification ,medicine.disease ,Virology ,Insect Vectors ,Rats ,030104 developmental biology ,Infectious Diseases ,Infectious disease (medical specialty) ,Insect Science ,Vector (epidemiology) ,Parasitology - Abstract
Dengue is an acute, emerging, infectious disease transmitted by Aedes mosquitoes that has become a serious global public health problem. The DEN2-FJ10 and DEN2-FJ11 strains of the dengue 2 virus were originally isolated from the serum of a patient with dengue fever in Fujian Province, China, in 1999. Our data provide the first assessment of the vector competence of Aedes mosquitoes with respect to the DEN2-FJ10 and DEN2-FJ11 strains of the dengue virus. There were significant differences in the replication rates of these two viral strains in Aedes albopictus and Aedes aegypti (P0.05); replication of the DEN2-FJ10 strain was greater in Ae. aegypti than in Ae. albopictus 5 days post infection whereas replication of the DEN2-FJ11 was greater in Ae. albopictus than in Ae. aegypti 7 days post infection. The replicative ability of the DEN2-FJ11 strain was greater than that of the DEN2-FJ10 strain in infected Ae. albopictus. In infected Ae. aegypti, rapid proliferation of the DEN2-FJ10 strain occurred earlier than in the DEN2-FJ11 strain. There were no significant differences in the midgut and salivary gland infection rates of Ae. albopictus and Ae. aegypti with respect to either viral strain. Although the DEN2-FJ10 and DEN2-FJ11 strains differ in their virulence to neonatal rats, there was no significant difference in the ability of either Ae. albopictus or Ae. aegypti to transmit the DEN2-FJ10 and DEN2-FJ10 strains of the dengue 2 virus (P0.05). In summary, our results indicate that Ae. albopictus and Ae. aegypti mosquitoes are moderately competent vectors of the DEN2-FJ10 and DEN2-FJ11 strains of the dengue virus and provide the first evidence of the effect of these two viral strains on the vector competence of mosquitoes in China.
- Published
- 2016
- Full Text
- View/download PDF
42. Structures of the Zika Virus Envelope Protein and Its Complex with a Flavivirus Broadly Protective Antibody
- Author
-
Cheng-Feng Qin, Yuan Yuan, Jinghua Yan, Yong-Qiang Deng, Yi Shi, Haixia Xiao, Huijun Cheng, Jianxun Qi, Yanfang Zhang, Hao Song, Lianpan Dai, Xishan Lu, Jian Song, Joel Haywood, Abednego Moki Musyoki, and George F. Gao
- Subjects
Male ,Models, Molecular ,0301 basic medicine ,Protein Conformation ,Viral protein ,viruses ,030106 microbiology ,Antibodies, Viral ,medicine.disease_cause ,Microbiology ,Epitope ,Cell Line ,Flavivirus Infections ,Zika virus ,Epitopes ,Mice ,03 medical and health sciences ,Viral Envelope Proteins ,Viral entry ,Virology ,medicine ,Animals ,chemistry.chemical_classification ,biology ,Zika Virus Infection ,Flavivirus ,Zika Virus ,Virus Internalization ,biology.organism_classification ,Antibodies, Neutralizing ,Disease Models, Animal ,030104 developmental biology ,chemistry ,biology.protein ,Parasitology ,Antibody ,Crystallization ,Glycoprotein - Abstract
Zika virus (ZIKV), a mosquito-borne flavivirus, is a current global public health concern. The flavivirus envelope (E) glycoprotein is responsible for virus entry and represents a major target of neutralizing antibodies for other flaviviruses. Here, we report the structures of ZIKV E protein at 2.0 Å and in complex with a flavivirus broadly neutralizing murine antibody 2A10G6 at 3.0 Å. ZIKV-E resembles all the known flavivirus E structures but contains a unique, positively charged patch adjacent to the fusion loop region of the juxtaposed monomer, which may influence host attachment. The ZIKV-E-2A10G6 complex structure reveals antibody recognition of a highly conserved fusion loop. 2A10G6 binds to ZIKV-E with high affinity in vitro and neutralizes currently circulating ZIKV strains in vitro and in mice. The E protein fusion loop epitope represents a potential candidate for therapeutic antibodies against ZIKV.
- Published
- 2016
- Full Text
- View/download PDF
43. A bispecific antibody effectively neutralizes all four serotypes of dengue virus by simultaneous blocking virus attachment and fusion
- Author
-
Cheng-Feng Qin, Chao Wang, Xiaojie Yu, E-De Qin, Xiaofeng Li, Guanghui Ji, Jianxin Dai, Yajun Guo, Tian Xia, Huajing Wang, Bohua Li, Tao Jiang, Yang Yang, Yong-Qiang Deng, Hui Zhao, Xin Shi, Yanchun Meng, and Wenlong Tan
- Subjects
0301 basic medicine ,medicine.drug_class ,viruses ,030106 microbiology ,Immunology ,Virus Attachment ,CHO Cells ,Dengue virus ,Antibodies, Viral ,Serogroup ,medicine.disease_cause ,Monoclonal antibody ,Epitope ,Virus ,Dengue ,Mice ,03 medical and health sciences ,Cricetulus ,In vivo ,Cricetinae ,Antibodies, Bispecific ,medicine ,Animals ,Humans ,Immunology and Allergy ,Antibody-dependent enhancement ,biology ,virus diseases ,Dengue Virus ,biology.organism_classification ,Antibodies, Neutralizing ,Virology ,Flavivirus ,030104 developmental biology ,biology.protein ,Antibody ,Reports - Abstract
Although dengue virus (DENV) infection severely threatens the health of humans, no specific antiviral drugs are currently approved for clinical use against DENV infection. Attachment and fusion are 2 critical steps for the flavivirus infection, and the corresponding functional epitopes are located at E protein domain III (E-DIII) and domain II (E-DII), respectively. Here, we constructed a bispecific antibody (DVD-1A1D-2A10) based on the 2 well-characterized anti-DENV monoclonal antibodies 1A1D-2 (1A1D) and 2A10G6 (2A10). The 1A1D antibody binds E-DIII and can block the virus attaching to the cell surface, while the 2A10 antibody binds E-DII and is able to prevent the virus from fusing with the endosomal membrane. Our data showed that DVD-1A1D-2A10 retained the antigen-binding activity of both parental antibodies. Importantly, it was demonstrated to be significantly more effective at neutralizing DENV than its parental antibodies both in vitro and in vivo, even better than the combination of them. To eliminate the potential antibody-dependent enhancement (ADE) effect, this bispecific antibody was successfully engineered to prevent Fc-γ-R interaction. Overall, we generated a bispecific anti-DENV antibody targeting both attachment and fusion stages, and this bispecific antibody broadly neutralized all 4 serotypes of DENV without risk of ADE, suggesting that it has great potential as a novel antiviral strategy against DENV.
- Published
- 2016
- Full Text
- View/download PDF
44. Generation of a recombinant West Nile virus stably expressing the Gaussia luciferase for neutralization assay
- Author
-
Pan Tao Zhang, Cheng Lin Deng, Bei Fen Shen, Si Qing Liu, Han-Qing Ye, Bao Di Shang, Ming Lv, Bo Zhang, Chao Shan, Cheng-Feng Qin, Xiao-Dan Li, and Pei Yong Shi
- Subjects
0301 basic medicine ,Cancer Research ,Vaccine evaluation ,medicine.drug_class ,animal diseases ,viruses ,Antibodies, Viral ,Neutralization ,Virus ,Copepoda ,03 medical and health sciences ,Gaussia ,Genes, Reporter ,Neutralization Tests ,Virology ,Complementary DNA ,medicine ,Animals ,Humans ,Luciferase ,Luciferases ,Neutralizing antibody ,biology ,virus diseases ,biology.organism_classification ,Antibodies, Neutralizing ,nervous system diseases ,030104 developmental biology ,Infectious Diseases ,biology.protein ,Antiviral drug ,West Nile virus ,West Nile Fever - Abstract
West Nile virus (WNV) is a neurotropic human pathogen that has caused increasing infected cases over recent years. There is currently no licensed vaccine or effective drug for prevention and treatment of WNV infection in humans. To facilitate antiviral drug discovery and neutralizing antibody detection, a WNV cDNA clone containing a luciferase reporter gene was constructed through incorporating Gaussia luciferase (Gluc) gene within the capsid-coding region of WNV genome. Transfection of BHK-21 cells with the cDNA clone-derived RNA generated luciferase reporter WNV (WNV-Gluc) and the stable WNV-Gluc with high titers (>10(7)PFU/ml) was obtained through plaque purification. Luciferase activity was used to effectively quantify the viral production of WNV-Gluc. Using the reporter virus WNV-Gluc, we developed a luciferase based assay in a 12-well format for evaluating neutralizing antibodies. The reporter virus could be a powerful tool for epidemiological investigation of WNV, vaccine evaluation, antiviral drug screening, and the study of WNV replication and pathogenesis.
- Published
- 2016
- Full Text
- View/download PDF
45. Intracellular delivery of biomineralized monoclonal antibodies to combat viral infection
- Author
-
Xurong Xu, Shun-Ya Zhu, Qing-Gong Nian, Xiaofeng Li, Zhiyong Song, Long Liu, Tao Jiang, Xiaoyu Wang, Cheng-Feng Qin, Ruikang Tang, Hangyu Zhou, Yong-Qiang Deng, and Guangchuan Wang
- Subjects
Calcium Phosphates ,medicine.drug_class ,viruses ,Enzyme-Linked Immunosorbent Assay ,02 engineering and technology ,Biology ,010402 general chemistry ,Monoclonal antibody ,01 natural sciences ,Viral infection ,Catalysis ,Materials Chemistry ,medicine ,Humans ,Metals and Alloys ,Antibodies, Monoclonal ,General Chemistry ,021001 nanoscience & nanotechnology ,Virology ,0104 chemical sciences ,Surfaces, Coatings and Films ,Electronic, Optical and Magnetic Materials ,Viral replication ,Virus Diseases ,Ceramics and Composites ,biology.protein ,Antibody ,0210 nano-technology ,Intracellular - Abstract
Conventional therapeutic monoclonal antibodies (mAbs) are invalid for intracellular viruses but by using in situ biomineralization treatment, they can be successfully delivered into cells to inhibit intracellular viral replication. This achievement significantly expands the applications of mAbs and provides a new intracellular strategy to control viral infections.
- Published
- 2016
- Full Text
- View/download PDF
46. Susceptibility of Armigeres subalbatus Coquillett (Diptera: Culicidae) to Zika virus through oral and urine infection
- Author
-
Dan Xing, Zhang Hengduan, Qin-mei Liu, Qun Wu, Yingmei Zhang, Ai-Juan Sun, Chunxiao Li, Cheng-Feng Qin, Wu-Chun Cao, Xiaoxia Guo, Tong-Yan Zhao, Yong-Qiang Deng, and Yande Dong
- Subjects
RNA viruses ,0301 basic medicine ,Life Cycles ,Saliva ,Physiology ,RC955-962 ,Urine ,Disease Vectors ,Pathology and Laboratory Medicine ,Mosquitoes ,Salivary Glands ,Zika virus ,Mice ,Medical Conditions ,Larvae ,0302 clinical medicine ,Arctic medicine. Tropical medicine ,Medicine and Health Sciences ,Pathogen ,biology ,Zika Virus Infection ,Transmission (medicine) ,Eukaryota ,Body Fluids ,Insects ,Ovaries ,Blood ,Infectious Diseases ,Medical Microbiology ,Viral Pathogens ,Larva ,Viruses ,Female ,Public aspects of medicine ,RA1-1270 ,Pathogens ,Anatomy ,Research Article ,Arthropoda ,030231 tropical medicine ,Mosquito Vectors ,Microbiology ,Virus ,03 medical and health sciences ,Exocrine Glands ,parasitic diseases ,Animals ,Humans ,Microbial Pathogens ,Aedes ,Biology and life sciences ,Flaviviruses ,fungi ,Organisms ,Reproductive System ,Public Health, Environmental and Occupational Health ,Outbreak ,Zika Virus ,biology.organism_classification ,Invertebrates ,Virology ,Insect Vectors ,Species Interactions ,Culicidae ,030104 developmental biology ,Zoology ,Entomology ,Digestive System ,Developmental Biology - Abstract
Background Zika virus (ZIKV) disease outbreaks have been occurring in South America since 2015, and has spread to North America. Because birth defects and cases of Guillain Barré have been associated with infection with ZIKV, this has drawn global attention. ZIKV is generally considered an Aedes-transmitted pathogen. The transmission of ZIKV through blood by Aedes mosquito bites has been recognized as the major transmission route. However, it is not clear whether there are other transmission routes that can cause viral infection in mosquitos. The aim of the present study is to describe the susceptibility of Armigeres subalbatus, which often develop in human waste lagoons, to ZIKV, through oral infection in adult mosquitoes and urine infection in larvae. Methodology/Principal findings Five-day-old female Ar. subalbatus ingested infectious blood meals containing ZIKV. After 4, 7, and 10 days of ingesting infectious blood meals, ZIKV could be detected in the midguts, salivary glands, ovaries, and collected saliva of mosquitoes. The ZIKV infection rate (IR) on day 10 reached 40% in salivary glands and 13% in saliva, indicating that these mosquitoes were able to transmit ZIKV. In addition, ZIKV infection was also discovered in mosquito ovaries, suggesting the possibility of vertical transmission of virus. Moreover, Ar. subalbatus transmitted ZIKV to infant mice bitten by infectious mosquitoes. In a second experiment, 1st-instar larvae of Ar. subalbatus were reared in water containing ZIKV and human urine. After pupation, pupae were placed in clean water and transferred to a mosquito cage for emergence. Although ZIKV RNA was detected in all of the larvae tested, ZIKV was not detected in the saliva of any adult Ar. subalbatus. Considering that there are more uncontrollable factors in nature than in the laboratory environment, the possibility that the virus is transmitted to adult mosquitoes via larvae is very small period. Conclusions/Significance Adult Ar. subalbatus could be infected with ZIKV and transmit ZIKV through mosquito bites. Therefore, in many rural areas in China and in undeveloped areas of other Asian countries, the management of human waste lagoons in the prevention and control of Zika disease should be considered. Corresponding adjustments and modifications should also be made in prevention and control strategies against ZIKV., Author summary Zika is a serious threat to global health since the outbreak in Brazil in 2015. Zika virus (ZIKV) is generally considered as an Aedes-transmitted pathogen. The transmission of ZIKV by mosquito bite has been recognized as the major transmission route. However, it is possible that some important potential vectors may not yet have been examined. Armigeres subalbatus is one of primary mosquito species in China, which could develop in human waste lagoons. In the study, through oral infection experiments, it was found that Ar. subalbatus could be infected with ZIKV and transmit ZIKV through mosquito bites. Therefore, in many rural areas in China and in undeveloped areas of other Asian countries, it is valid to consider that this mosquito species should be given enough attention in the prevention and control of Zika epidemic.
- Published
- 2020
- Full Text
- View/download PDF
47. Differential antiviral immunity to Japanese encephalitis virus in developing cortical organoids
- Author
-
Yali Li, Cheng-Feng Qin, Qing Sunny Shen, Fan Mo, Chunfeng Li, Yan-Peng Xu, Yihui Wu, Yangzhige He, Wenliang Zhu, Jing Liu, Guilai Chen, Ting-Wei Mi, Guomin Zhou, Boya Zhang, and Baoyang Hu
- Subjects
Telencephalon ,0301 basic medicine ,Cancer Research ,Neurogenesis ,viruses ,Immunology ,Population ,Adaptive Immunity ,Virus ,Zika virus ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Interferon ,Cricetinae ,medicine ,Organoid ,Animals ,Humans ,lcsh:QH573-671 ,Progenitor cell ,Encephalitis, Japanese ,education ,Cells, Cultured ,Encephalitis Virus, Japanese ,education.field_of_study ,biology ,lcsh:Cytology ,Brain ,Cell Biology ,Japanese encephalitis ,medicine.disease ,biology.organism_classification ,Virology ,Organoids ,030104 developmental biology ,Encephalitis ,medicine.drug - Abstract
Japanese encephalitis (JE) caused by Japanese encephalitis virus (JEV) poses a serious threat to the world’s public health yet without a cure. Certain JEV-infected neural cells express a subset of previously identified intrinsic antiviral interferon stimulated genes (ISGs), indicating brain cells retain autonomous antiviral immunity. However, whether this happens in composited brain remains unclear. Human pluripotent stem cell (hPSC)-derived organoids can model disorders caused by human endemic pathogens such as Zika virus, which may potentially address this question and facilitate the discovery of a cure for JE. We thus generated telencephalon organoid and infected them with JEV. We found JEV infection caused significant decline of cell proliferation and increase of cell death in brain organoid, resulting in smaller organoid spheres. JEV tended to infect astrocytes and neural progenitors, especially the population representing outer radial glial cells (oRGCs) of developing human brain. In addition, we revealed variable antiviral immunity in brain organoids of different stages of culture. In organoids of longer culture (older than 8 weeks), but not of early ones (less than 4 weeks), JEV infection caused typical activation of interferon signaling pathway. Preferential infection of oRGCs and differential antiviral response at various stages might explain the much more severe outcomes of JEV infection in the younger, which also provide clues to develop effective therapeutics of such diseases.
- Published
- 2018
- Full Text
- View/download PDF
48. PARP12 suppresses Zika virus infection through PARP-dependent degradation of NS1 and NS3 viral proteins
- Author
-
Lili Li, Natalie Quanquin, Ning Lu, Genhong Cheng, Nina N. Sun, Peishuang Du, Chunfeng Li, Cheng-Feng Qin, Xue Ji, Ping Liu, and Hui Zhao
- Subjects
0301 basic medicine ,viruses ,Viral Nonstructural Proteins ,Virus Replication ,Biochemistry ,Zika virus ,Madin Darby Canine Kidney Cells ,Mice ,Interferon ,2.1 Biological and endogenous factors ,Aetiology ,Lung ,biology ,Zika Virus Infection ,Serine Endopeptidases ,virus diseases ,Infectious Diseases ,Interferon Type I ,Interferon Regulatory Factors ,Poly(ADP-ribose) Polymerases ,Infection ,RNA Helicases ,medicine.drug ,Proteasome Endopeptidase Complex ,Strong inhibitor ,Poly ADP ribose polymerase ,Article ,Vaccine Related ,03 medical and health sciences ,Dogs ,ADP-Ribosylation ,Biodefense ,medicine ,Genetics ,Animals ,Humans ,Molecular Biology ,Gene ,NS3 ,Host (biology) ,Ubiquitin ,Prevention ,Cell Biology ,Zika Virus ,biology.organism_classification ,Virology ,030104 developmental biology ,Emerging Infectious Diseases ,Good Health and Well Being ,Viral replication ,A549 Cells ,Proteolysis ,Biochemistry and Cell Biology - Abstract
Zika virus infection stimulates a type I interferon (IFN) response in host cells, which suppresses viral replication. Type I IFNs exert antiviral effects by inducing the expression of hundreds of IFN-stimulated genes (ISGs). To screen for antiviral ISGs that restricted Zika virus replication, we individually knocked out 21 ISGs in A549 lung cancer cells and identified PARP12 as a strong inhibitor of Zika virus replication. Our findings suggest that PARP12 mediated the ADP-ribosylation of NS1 and NS3, nonstructural viral proteins that are involved in viral replication and modulating host defense responses. This modification of NS1 and NS3 triggered their proteasome-mediated degradation. These data increase our understanding of the antiviral activity of PARP12 and suggest a molecular basis for the potential development of therapeutics against Zika virus.
- Published
- 2018
49. Development of a chimeric Zika vaccine using a licensed live-attenuated flavivirus vaccine as backbone
- Author
-
Yang Liu, Andrew D. Davidson, Chunfeng Li, Ye Feng Qiu, Yong Qiang Deng, Ya Jun Song, Xing Yao Huang, Fuchun Zhang, Cheng-Feng Qin, Pei Yong Shi, Tao Jiang, Qing Ye, Hong Jiang Wang, Gong Cheng, Hao Long Dong, Xiaofeng Li, and Xue Ji
- Subjects
0301 basic medicine ,Male ,Live attenuated vaccines ,viruses ,General Physics and Astronomy ,Antibodies, Viral ,Zika virus ,law.invention ,Mice ,0302 clinical medicine ,law ,Aedes ,Pregnancy ,030212 general & internal medicine ,lcsh:Science ,Encephalitis Virus, Japanese ,Mice, Inbred BALB C ,Vaccines, Synthetic ,Multidisciplinary ,Zika Virus Infection ,Immunogenicity ,Flavivirus ,Recombinant DNA ,Female ,Antibody ,Infection ,Genetic Engineering ,Mice, 129 Strain ,Science ,Mosquito Vectors ,Biology ,Vaccines, Attenuated ,complex mixtures ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Immune system ,Virology ,medicine ,Animals ,Humans ,Viremia ,Viral Vaccines ,General Chemistry ,Zika Virus ,Japanese encephalitis ,biology.organism_classification ,medicine.disease ,Antibodies, Neutralizing ,Macaca mulatta ,Infectious Disease Transmission, Vertical ,Disease Models, Animal ,030104 developmental biology ,Immunization ,biology.protein ,lcsh:Q - Abstract
The global spread of Zika virus (ZIKV) and its unexpected association with congenital defects necessitates the rapid development of a safe and effective vaccine. Here we report the development and characterization of a recombinant chimeric ZIKV vaccine candidate (termed ChinZIKV) that expresses the prM-E proteins of ZIKV using the licensed Japanese encephalitis live-attenuated vaccine SA14-14-2 as the genetic backbone. ChinZIKV retains its replication activity and genetic stability in vitro, while exhibiting an attenuation phenotype in multiple animal models. Remarkably, immunization of mice and rhesus macaques with a single dose of ChinZIKV elicits robust and long-lasting immune responses, and confers complete protection against ZIKV challenge. Significantly, female mice immunized with ChinZIKV are protected against placental and fetal damage upon ZIKV challenge during pregnancy. Overall, our study provides an alternative vaccine platform in response to the ZIKV emergency, and the safety, immunogenicity, and protection profiles of ChinZIKV warrant further clinical development., Given the recent Zika virus (ZIKV) epidemic, development of an effective vaccine is of high importance. Here, the authors use a licensed live-attenuated flavivirus vaccine backbone to develop a ZIKV vaccine and determine immunogenicity, safety and protection profiles in different animal models.
- Published
- 2018
- Full Text
- View/download PDF
50. Crystal Structures of Enterovirus 71 (EV71) Recombinant Virus Particles Provide Insights into Vaccine Design
- Author
-
Ke Lyu, Ya-Ling He, Guangchuan Wang, Jian-Feng Han, Rong Chen, Cheng-Feng Qin, and Qing Ye
- Subjects
viruses ,Molecular Sequence Data ,Virus Uncoating ,Cell Biology ,Biology ,Crystallography, X-Ray ,Recombinant virus ,biology.organism_classification ,Microbiology ,Biochemistry ,Virology ,Virus ,Enterovirus A, Human ,law.invention ,Capsid ,stomatognathic system ,Viral replication ,law ,Recombinant DNA ,Enterovirus 71 ,Amino Acid Sequence ,Vaccines, Virus-Like Particle ,Molecular Biology ,Peptide sequence - Abstract
Hand-foot-and-mouth disease (HFMD) remains a major health concern in the Asia-Pacific regions, and its major causative agents include human enterovirus 71 (EV71) and coxsackievirus A16. A desirable vaccine against HFMD would be multivalent and able to elicit protective responses against multiple HFMD causative agents. Previously, we have demonstrated that a thermostable recombinant EV71 vaccine candidate can be produced by the insertion of a foreign peptide into the BC loop of VP1 without affecting viral replication. Here we present crystal structures of two different naturally occurring empty particles, one from a clinical C4 strain EV71 and the other from its recombinant virus containing an insertion in the VP1 BC loop. Crystal structure analysis demonstrated that the inserted foreign peptide is well exposed on the particle surface without significant structural changes in the capsid. Importantly, such insertions do not seem to affect the virus uncoating process as illustrated by the conformational similarity between an uncoating intermediate of another recombinant virus and that of EV71. Especially, at least 18 residues from the N terminus of VP1 are transiently externalized. Altogether, our study provides insights into vaccine development against HFMD.
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.