157 results on '"Cheng‐Feng Qin"'
Search Results
2. Aerosolized Zika virus infection in Guinea pigs
- Author
-
Hong-Ying, Qiu, Na-Na, Zhang, Qing-Qing, Ma, Rui-Ting, Li, Meng-Yue, Guan, Li-Li, Zhang, Jia, Zhou, Rong-Rong, Zhang, Xing-Yao, Huang, Wen-Hui, Yang, Yong-Qiang, Deng, Cheng-Feng, Qin, and Dong-Sheng, Zhou
- Subjects
Zika Virus Infection ,Epidemiology ,Guinea Pigs ,Immunology ,Zika Virus ,General Medicine ,Microbiology ,Infectious Disease Transmission, Vertical ,Immunity, Humoral ,Infectious Diseases ,Virology ,Drug Discovery ,Animals ,Humans ,Parasitology ,Viremia - Abstract
Zika virus (ZIKV) is primarily transmitted through mosquito bites and sexual contact, and vertical transmission of ZIKV has also been observed in humans. In addition, ZIKV infection via unknown transmission routes has been frequently reported in clinical settings. However, whether ZIKV can be transmitted via aerosol routes remains unknown. In this study, we demonstrated that aerosolized ZIKV is fully infectious in vitro and in vivo. Remarkably, intratracheal (i.t.) inoculation with aerosolized ZIKV led to rapid viremia and viral secretion in saliva, as well as robust humoral and innate immune responses in guinea pigs. Transcriptome analysis further revealed that the expression of genes related to viral processes, biological regulation and the immune response was significantly changed. Together, our results confirm that aerosolized ZIKV can result in systemic infection and induce both innate and adaptive immune responses in guinea pigs, highlighting the possibility of ZIKV transmission via aerosols.
- Published
- 2022
- Full Text
- View/download PDF
3. Comparative characterization of SARS‐CoV‐2 variants of concern and mouse‐adapted strains in mice
- Author
-
Qi Chen, Xing‐Yao Huang, Yu Liu, Meng‐Xu Sun, Bin Ji, Chao Zhou, Hang Chi, Rong‐Rong Zhang, Dan Luo, Ying Tian, Xiao‐Feng Li, Hui Zhao, and Cheng‐Feng Qin
- Subjects
Mice ,Infectious Diseases ,SARS-CoV-2 ,Virology ,Spike Glycoprotein, Coronavirus ,Animals ,COVID-19 ,Humans ,Protein Binding - Abstract
SARS-CoV-2 has evolved into a panel of variants of concern (VOCs) and constituted a sustained threat to global health. The wildtype (WT) SARS-CoV-2 isolates fail to infect mice, while the Beta variant, one of the VOCs, has acquired the capability to infect standard laboratory mice, raising a spreading risk of SARS-CoV-2 from humans to mice. However, the infectivity and pathogenicity of other VOCs in mice remain not fully understood. In this study, we systematically investigated the infectivity and pathogenicity of three VOCs, Alpha, Beta, and Delta, in mice in comparison with two well-understood SARS-CoV-2 mouse-adapted strains, MASCp6 and MASCp36, sharing key mutations in the receptor-binding domain (RBD) with Alpha or Beta, respectively. Our results showed that the Beta variant had the strongest infectivity and pathogenicity among the three VOCs, while the Delta variant only caused limited replication and mild pathogenic changes in the mouse lung, which is much weaker than what the Alpha variant did. Meanwhile, Alpha showed comparable infectivity in lungs in comparison with MASCp6, and Beta only showed slightly lower infectivity in lungs when compared with MASCp36. These results indicated that all three VOCs have acquired the capability to infect mice, highlighting the ongoing spillover risk of SARS-CoV-2 from humans to mice during the continued evolution of SARS-CoV-2, and that the key amino acid mutations in the RBD of mouse-adapted strains may be referenced as an early-warning indicator for predicting the spillover risk of newly emerging SARS-CoV-2 variants.
- Published
- 2022
- Full Text
- View/download PDF
4. Lipid nanoparticle-encapsulated mRNA antibody provides long-term protection against SARS-CoV-2 in mice and hamsters
- Author
-
Yong-Qiang Deng, Na-Na Zhang, Yi-Fei Zhang, Xia Zhong, Sue Xu, Hong-Ying Qiu, Tie-Cheng Wang, Hui Zhao, Chao Zhou, Shu-Long Zu, Qi Chen, Tian-Shu Cao, Qing Ye, Hang Chi, Xiang-Hui Duan, Dan-Dan Lin, Xiao-Jing Zhang, Liang-Zhi Xie, Yu-Wei Gao, Bo Ying, and Cheng-Feng Qin
- Subjects
SARS-CoV-2 ,COVID-19 ,Cell Biology ,Antibodies, Viral ,Antibodies, Neutralizing ,Mice ,Cricetinae ,Liposomes ,Spike Glycoprotein, Coronavirus ,Animals ,Humans ,Nanoparticles ,RNA, Messenger ,Pandemics ,Molecular Biology - Abstract
Monoclonal antibodies represent important weapons in our arsenal to against the COVID-19 pandemic. However, this potential is severely limited by the time-consuming process of developing effective antibodies and the relative high cost of manufacturing. Herein, we present a rapid and cost-effective lipid nanoparticle (LNP) encapsulated-mRNA platform for in vivo delivery of SARS-CoV-2 neutralization antibodies. Two mRNAs encoding the light and heavy chains of a potent SARS-CoV-2 neutralizing antibody HB27, which is currently being evaluated in clinical trials, were encapsulated into clinical grade LNP formulations (named as mRNA-HB27-LNP). In vivo characterization demonstrated that intravenous administration of mRNA-HB27-LNP in mice resulted in a longer circulating half-life compared with the original HB27 antibody in protein format. More importantly, a single prophylactic administration of mRNA-HB27-LNP provided protection against SARS-CoV-2 challenge in mice at 1, 7 and even 63 days post administration. In a close contact transmission model, prophylactic administration of mRNA-HB27-LNP prevented SARS-CoV-2 infection between hamsters in a dose-dependent manner. Overall, our results demonstrate a superior long-term protection against SARS-CoV-2 conferred by a single administration of this unique mRNA antibody, highlighting the potential of this universal platform for antibody-based disease prevention and therapy against COVID-19 as well as a variety of other infectious diseases.
- Published
- 2022
- Full Text
- View/download PDF
5. Memory B cell repertoire from triple vaccinees against diverse SARS-CoV-2 variants
- Author
-
Kang Wang, Zijing Jia, Linilin Bao, Lei Wang, Lei Cao, Hang Chi, Yaling Hu, Qianqian Li, Yunjiao Zhou, Yinan Jiang, Qianhui Zhu, Yongqiang Deng, Pan Liu, Nan Wang, Lin Wang, Min Liu, Yurong Li, Boling Zhu, Kaiyue Fan, Wangjun Fu, Peng Yang, Xinran Pei, Zhen Cui, Lili Qin, Pingju Ge, Jiajing Wu, Shuo Liu, Yiding Chen, Weijin Huang, Qiao Wang, Cheng-Feng Qin, Youchun Wang, Chuan Qin, and Xiangxi Wang
- Subjects
COVID-19 Vaccines ,Multidisciplinary ,SARS-CoV-2 ,Antibodies, Monoclonal ,COVID-19 ,Antibodies, Viral ,Antibodies, Neutralizing ,Disease Models, Animal ,Mice ,Memory B Cells ,Neutralization Tests ,Spike Glycoprotein, Coronavirus ,Animals ,Humans - Abstract
Omicron (B.1.1.529), the most heavily mutated SARS-CoV-2 variant so far, is highly resistant to neutralizing antibodies, raising concerns about the effectiveness of antibody therapies and vaccines1,2. Here we examined whether sera from individuals who received two or three doses of inactivated SARS-CoV-2 vaccine could neutralize authentic Omicron. The seroconversion rates of neutralizing antibodies were 3.3% (2 out of 60) and 95% (57 out of 60) for individuals who had received 2 and 3 doses of vaccine, respectively. For recipients of three vaccine doses, the geometric mean neutralization antibody titre for Omicron was 16.5-fold lower than for the ancestral virus (254). We isolated 323 human monoclonal antibodies derived from memory B cells in triple vaccinees, half of which recognized the receptor-binding domain, and showed that a subset (24 out of 163) potently neutralized all SARS-CoV-2 variants of concern, including Omicron. Therapeutic treatments with representative broadly neutralizing monoclonal antibodies were highly protective against infection of mice with SARS-CoV-2 Beta (B.1.351) and Omicron. Atomic structures of the Omicron spike protein in complex with three classes of antibodies that were active against all five variants of concern defined the binding and neutralizing determinants and revealed a key antibody escape site, G446S, that confers greater resistance to a class of antibodies that bind on the right shoulder of the receptor-binding domain by altering local conformation at the binding interface. Our results rationalize the use of three-dose immunization regimens and suggest that the fundamental epitopes revealed by these broadly ultrapotent antibodies are rational targets for a universal sarbecovirus vaccine.
- Published
- 2022
- Full Text
- View/download PDF
6. Ångstrom-scale silver particles potently combat SARS-CoV-2 infection by suppressing the ACE2 expression and inflammatory responses
- Author
-
Yong-Qiang Deng, Zhen-Xing Wang, Xin Liu, Yi-Yi Wang, Qi Chen, Zhao-Long Li, Bai-Song Zheng, Qing Ye, Jiang-Shan Gong, Guo-Qiang Zhu, Tian-Shu Cao, Wei-Yi Situ, Cheng-Feng Qin, Hui Xie, and Wen-Yan Zhang
- Subjects
Silver ,SARS-CoV-2 ,Biomedical Engineering ,Humans ,General Materials Science ,Angiotensin-Converting Enzyme 2 ,General Chemistry ,General Medicine ,Peptidyl-Dipeptidase A ,COVID-19 Drug Treatment - Abstract
The SARS-CoV-2 pandemic has become a severe global public health event, and the development of protective and therapeutic strategies is urgently needed. Downregulation of angiotensin converting enzyme 2 (ACE2; one of the important SARS-CoV-2 entry receptors) and aberrant inflammatory responses (cytokine storm) are the main targets to inhibit and control COVID-19 invasion. Silver nanomaterials have well-known pharmaceutical properties, including antiviral, antibacterial, and anticancer properties. Here, based on a self-established metal evaporation-condensation-size graded collection system, smaller silver particles reaching the Ångstrom scale (AgÅPs) were fabricated and coated with fructose to obtain a stabilized AgÅP solution (F-AgÅPs). F-AgÅPs potently inactivated SARS-CoV-2 and prevented viral infection. Considering the application of anti-SARS-CoV-2, a sterilized F-AgÅP solution was produced
- Published
- 2022
- Full Text
- View/download PDF
7. Long-term stability and protection efficacy of the RBD-targeting COVID-19 mRNA vaccine in nonhuman primates
- Author
-
Hui Zhao, Tie-Cheng Wang, Xiao-Feng Li, Na-Na Zhang, Liang Li, Chao Zhou, Yong-Qiang Deng, Tian-Shu Cao, Guan Yang, Rui-Ting Li, Yi-Jiao Huang, Yuan-Guo Li, Yi-Ming Zhang, Fang-Xu Li, Yu-Ren Zhou, Yu-Hang Jiang, Xi-Shan Lu, Shi-Hui Sun, Meng-Li Cheng, Kai-Ping Gu, Mei Zhang, Qing-Qing Ma, Xiao Yang, Bo Ying, Yu-Wei Gao, and Cheng-Feng Qin
- Subjects
Cancer Research ,Vaccines ,COVID-19 Vaccines ,SARS-CoV-2 ,QH301-705.5 ,COVID-19 ,Antibodies, Viral ,Antibodies, Neutralizing ,Article ,Macaca fascicularis ,Immunogenicity, Vaccine ,Chlorocebus aethiops ,Spike Glycoprotein, Coronavirus ,Genetics ,Animals ,Humans ,Medicine ,mRNA Vaccines ,Biology (General) ,Vero Cells - Abstract
Messenger RNA (mRNA) vaccine technology has shown its power in preventing the ongoing COVID-19 pandemic. Two mRNA vaccines targeting the full-length S protein of SARS-CoV-2 have been authorized for emergency use. Recently, we have developed a lipid nanoparticle-encapsulated mRNA (mRNA-LNP) encoding the receptor-binding domain (RBD) of SARS-CoV-2 (termed ARCoV), which confers complete protection in mouse model. Herein, we further characterized the protection efficacy of ARCoV in nonhuman primates and the long-term stability under normal refrigerator temperature. Intramuscular immunization of two doses of ARCoV elicited robust neutralizing antibodies as well as cellular response against SARS-CoV-2 in cynomolgus macaques. More importantly, ARCoV vaccination in macaques significantly protected animals from acute lung lesions caused by SARS-CoV-2, and viral replication in lungs and secretion in nasal swabs were completely cleared in all animals immunized with low or high doses of ARCoV. No evidence of antibody-dependent enhancement of infection was observed throughout the study. Finally, extensive stability assays showed that ARCoV can be stored at 2–8 °C for at least 6 months without decrease of immunogenicity. All these promising results strongly support the ongoing clinical trial.
- Published
- 2021
8. Enhanced protective immunity against SARS-CoV-2 elicited by a VSV vector expressing a chimeric spike protein
- Author
-
Cheng-Feng Qin, Yongchun Li, Fei Yuan, Jiankai Liu, Jianguo Zhao, Shujun Liu, Aihua Zheng, Junfeng Hao, Chaoyue Zhao, Hongyue Li, Yong-Qiang Deng, Dan Wen, Dong Li, Yuhang Zhang, Hongde Xu, Shuguang Duo, Jiandong Liu, and Yong Wu
- Subjects
Cancer Research ,COVID-19 Vaccines ,QH301-705.5 ,Recombinant Fusion Proteins ,viruses ,medicine.disease_cause ,Antibodies, Viral ,Article ,law.invention ,Chimera (genetics) ,Mice ,Immune system ,Antigen ,law ,Neutralization Tests ,Genetics ,medicine ,Animals ,Humans ,Gene Knock-In Techniques ,Biology (General) ,Neutralizing antibody ,skin and connective tissue diseases ,Coronavirus ,Vaccines ,biology ,SARS-CoV-2 ,fungi ,COVID-19 ,Virology ,Antibodies, Neutralizing ,respiratory tract diseases ,Transplantation ,Mice, Inbred C57BL ,body regions ,Titer ,HEK293 Cells ,Spike Glycoprotein, Coronavirus ,Recombinant DNA ,biology.protein ,Medicine - Abstract
SARS-CoV-2 and SARS-CoV are genetically related coronavirus and share the same cellular receptor ACE2. By replacing the VSV glycoprotein with the spikes (S) of SARS-CoV-2 and SARS-CoV, we generated two replication-competent recombinant viruses, rVSV-SARS-CoV-2 and rVSV-SARS-CoV. Using wild-type and human ACE2 (hACE2) knock-in mouse models, we found a single dose of rVSV-SARS-CoV could elicit strong humoral immune response via both intranasal (i.n.) and intramuscular (i.m.) routes. Despite the high genetic similarity between SARS-CoV-2 and SARS-CoV, no obvious cross-neutralizing activity was observed in the immunized mice sera. In macaques, neutralizing antibody (NAb) titers induced by one i.n. dose of rVSV-SARS-CoV-2 were eight-fold higher than those by a single i.m. dose. Thus, our data indicates that rVSV-SARS-CoV-2 might be suitable for i.n. administration instead of the traditional i.m. immunization in human. Because rVSV-SARS-CoV elicited significantly stronger NAb responses than rVSV-SARS-CoV-2 in a route-independent manner, we generated a chimeric antigen by replacing the receptor binding domain (RBD) of SARS-CoV S with that from the SARS-CoV-2. rVSV expressing the chimera (rVSV-SARS-CoV/2-RBD) induced significantly increased NAbs against SARS-CoV-2 in mice and macaques than rVSV-SARS-CoV-2, with a safe Th1-biased response. Serum immunized with rVSV-SARS-CoV/2-RBD showed no cross-reactivity with SARS-CoV. hACE2 mice receiving a single i.m. dose of either rVSV-SARS-CoV-2 or rVSV-SARS-CoV/2-RBD were fully protected against SARS-CoV-2 challenge without obvious lesions in the lungs. Our results suggest that transplantation of SARS-CoV-2 RBD into the S protein of SARS-CoV might be a promising antigen design for COVID-19 vaccines.
- Published
- 2021
9. The Infection and Pathogenicity of SARS-CoV-2 Variant B.1.351 in hACE2 Mice
- Author
-
Xing-Yao Huang, Meng-Xu Sun, Ruiting Li, Guizhen Wu, Changfa Fan, Ying Tian, Chao Zhou, Cheng-Feng Qin, Qi Chen, and Rong-Rong Zhang
- Subjects
Models, Molecular ,2019-20 coronavirus outbreak ,medicine.medical_specialty ,Letter ,Time Factors ,Coronavirus disease 2019 (COVID-19) ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,DNA Mutational Analysis ,Immunology ,Mice, Transgenic ,Biology ,Evolution, Molecular ,South Africa ,Mice ,Medical microbiology ,Virology ,medicine ,Animals ,Humans ,Selection, Genetic ,Phylogeny ,Immune Evasion ,Virulence ,SARS-CoV-2 ,COVID-19 ,Pathogenicity ,Phylogeography ,Mutation ,Spike Glycoprotein, Coronavirus ,Molecular Medicine ,Genetic Fitness - Abstract
Continued uncontrolled transmission of SARS-CoV-2 in many parts of the world is creating conditions for substantial evolutionary changes to the virus
- Published
- 2021
- Full Text
- View/download PDF
10. 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
11. Development of a Bicistronic Yellow Fever Live Attenuated Vaccine with Reduced Neurovirulence and Viscerotropism
- Author
-
Hong-Jiang Wang, Yan Guo, Meng-Jiao He, Zhong-Yu Liu, Qing Ye, Xing-Yao Huang, Yong-Qiang Deng, Xiao-Feng Li, and Cheng-Feng Qin
- Subjects
Microbiology (medical) ,Mammals ,General Immunology and Microbiology ,Ecology ,Physiology ,Yellow Fever Vaccine ,Cell Biology ,Internal Ribosome Entry Sites ,Vaccines, Attenuated ,Mice ,Infectious Diseases ,Yellow Fever ,Interferon Type I ,Genetics ,Humans ,Animals ,Yellow fever virus ,Antigens, Viral - Abstract
The yellow fever (YF) live attenuated vaccine strain 17D (termed 17D) has been widely used for the prevention and control of YF disease. However, 17D retains significant neurovirulence and viscerotropism in mice, which is probably linked to the increased occurrences of serious adverse events following 17D vaccination. Thus, the development of an updated version of the YF vaccine with an improved safety profile is of high priority. Here, we generated a viable bicistronic YF virus (YFV) by incorporating the internal ribosome entry site (IRES) from
- Published
- 2022
12. Immune profiles in mouse brain and testes infected by Zika virus with variable pathogenicity
- Author
-
Jingzhe Shang, Chunfeng Li, Zhujia Jin, Shulong Zu, Songjie Chen, Junlan Chen, Ziyi Chen, Hua Tang, Cheng-Feng Qin, Qing Ye, and Aiping Wu
- Subjects
Male ,Microbiology (medical) ,Virulence ,Zika Virus Infection ,Immunology ,Brain ,Zika Virus ,Microbiology ,Mice ,Infectious Diseases ,Testis ,Animals ,Humans ,Child - Abstract
The Zika virus is responsible for neurological diseases such as microcephaly, Guillain-Barré syndrome, neuropathy, and myelitis in human adults and children. Previous studies have shown that the Zika virus can infect nerve progenitor cells and interfere with neural development. However, it is unclear how the immune system responds to infection with Zika viruses with variable pathogenicity. Here, we used two Zika strains with relatively different pathogenicity, the Asian ancestral strain CAM/2010 and the America pandemic strain GZ01/2016, to infect the brains of mice. We found that both strains elicited a strong immune response. Notably, the strain with relatively high pathogenicity, GZ01/2016, caused more intense immune regulation, with stronger CD8+ T cell and macrophage activation at 14 days post infection (dpi), as well as a greater immune gene disturbance. Notably, several TNF family genes were upregulated at 14 dpi, including Tnfrsf9, Tnfsf13, Tnfrsf8, Cd40, and Tnfsf10. It was notable that GZ01/2016 could maintain the survival of nerve cells at 7dpi but caused neurological disorders at 14dpi. These results indicate that Zika viruses with high pathogenicity may induce sustained activation of the immune system leading to nerve tissue damage.
- Published
- 2022
- Full Text
- View/download PDF
13. Neutralization of ARCoV-induced sera against SARS-CoV-2 variants
- Author
-
Xiao-Feng Li, Na-Na Zhang, Yu-Hua Li, Shou-Chun Cao, Yi-Fei Zhang, and Cheng-Feng Qin
- Subjects
Pharmacology ,China ,Clinical Trials, Phase III as Topic ,Neutralization Tests ,SARS-CoV-2 ,Spike Glycoprotein, Coronavirus ,Immunology ,Humans ,COVID-19 ,Immunology and Allergy ,Antibodies, Viral ,Antibodies, Neutralizing - Abstract
ARCoV is a candidate mRNA vaccine encoding receptor-binding domain of SARS-CoV-2. Its safety, tolerability, and immunogenicity profile have been confirmed in the phase 1 clinical trial in China. A multi-regional phase 3 clinical trial is currently underway to test the efficacy of ARCoV (NCT04847102). Here, we tested the cross-neutralization against SARS-CoV-2 variants of concern (VOCs) of a panel of serum samples from participants in the phase 1 clinical trial of ARCoV by pesudo- and authentic SARS-CoV-2. Our data suggest the immunity induced by the ARCoV vaccine reduced but still has significant neutralization against the Alpha and Delta variants. Moreover, ARCoV maintained activity against the Beta variant, despite of its obvious reduction in neutralizing titers. Our findings further support the solid protective neutralization activity against VOCs induced by ARCoV vaccine.
- Published
- 2022
- Full Text
- View/download PDF
14. Humoral immune response to circulating SARS-CoV-2 variants elicited by inactivated and RBD-subunit vaccines
- Author
-
Yongzheng Li, Jiajing Wu, Xiaoyu Xu, Ran An, Xiaoliang Sunney Xie, Rui Shi, Na-Na Zhang, Chuanping Gao, Liyang Song, Yingmin Ma, Ayijiang Yisimayi, Xuemei Li, Yali Bai, Y Cao, Youchun Wang, Tianhe Xiao, Shuo Du, Yangyang Wei, Yifei Zhang, Wenping Ma, Xiang Li, Junyu Xiao, Weiliang Song, Cheng-Feng Qin, Bin Su, Shuo Liu, Jing Wang, Zhiying Zhang, Xiaofeng Li, Tianjiao Yuan, Ronghua Jin, Yingmei Feng, Yang Dou, Yonghong Zhang, Bai Lu, Weijin Huang, and Xianghua Guo
- Subjects
Immunology ,Complementarity determining region ,medicine.disease_cause ,Article ,Neutralization ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Antigen ,Cryoelectron microscopy ,Neutralization Tests ,Immunity ,medicine ,Humans ,Molecular Biology ,X-ray crystallography ,030304 developmental biology ,0303 health sciences ,Mutation ,biology ,SARS-CoV-2 ,COVID-19 ,Cell Biology ,Antibodies, Neutralizing ,Virology ,Inactivated vaccine ,biology.protein ,Antibody ,030217 neurology & neurosurgery - Abstract
SARS-CoV-2 variants could induce immune escape by mutations on the receptor-binding domain (RBD) and N-terminal domain (NTD). Here we report the humoral immune response to circulating SARS-CoV-2 variants, such as 501Y.V2 (B.1.351), of the plasma and neutralizing antibodies (NAbs) elicited by CoronaVac (inactivated vaccine), ZF2001 (RBD-subunit vaccine) and natural infection. Among 86 potent NAbs identified by high-throughput single-cell VDJ sequencing of peripheral blood mononuclear cells from vaccinees and convalescents, near half anti-RBD NAbs showed major neutralization reductions against the K417N/E484K/N501Y mutation combination, with E484K being the dominant cause. VH3-53/VH3-66 recurrent antibodies respond differently to RBD variants, and K417N compromises the majority of neutralizing activity through reduced polar contacts with complementarity determining regions. In contrast, the 242–244 deletion (242–244Δ) would abolish most neutralization activity of anti-NTD NAbs by interrupting the conformation of NTD antigenic supersite, indicating a much less diversity of anti-NTD NAbs than anti-RBD NAbs. Plasma of convalescents and CoronaVac vaccinees displayed comparable neutralization reductions against pseudo- and authentic 501Y.V2 variants, mainly caused by E484K/N501Y and 242–244Δ, with the effects being additive. Importantly, RBD-subunit vaccinees exhibit markedly higher tolerance to 501Y.V2 than convalescents, since the elicited anti-RBD NAbs display a high diversity and are unaffected by NTD mutations. Moreover, an extended gap between the third and second doses of ZF2001 leads to better neutralizing activity and tolerance to 501Y.V2 than the standard three-dose administration. Together, these results suggest that the deployment of RBD-vaccines, through a third-dose boost, may be ideal for combating SARS-CoV-2 variants when necessary, especially for those carrying mutations that disrupt the NTD supersite.
- Published
- 2021
- Full Text
- View/download PDF
15. Safety and superior immunogenicity of heterologous boosting with an RBD-based SARS-CoV-2 mRNA vaccine in Chinese adults
- Author
-
Xiaoqiang Liu, Yuhua Li, Zhongfang Wang, Shouchun Cao, Weijin Huang, Lin Yuan, Yi-Jiao Huang, Yan Zheng, Jingjing Chen, Bo Ying, Zuoyun Xiang, Jin Shi, Jincun Zhao, Zhen Huang, and Cheng-Feng Qin
- Subjects
Adult ,China ,Vaccines, Synthetic ,COVID-19 Vaccines ,SARS-CoV-2 ,COVID-19 ,Humans ,mRNA Vaccines ,Cell Biology ,Antibodies, Viral ,Antibodies, Neutralizing ,Molecular Biology - Abstract
Homologous and heterologous booster with COVID-19 mRNA vaccines represent the most effective strategy to prevent the ongoing Omicron pandemic. The additional protection from these prototype SARS-CoV-2 S-targeting vaccine was attributed to the increased RBD-specific memory B cells with expanded potency and breadth. Herein, we show the safety and immunogenicity of heterologous boosting with the RBD-targeting mRNA vaccine AWcorna (also term ARCoV) in Chinese adults who have received two doses inactivated vaccine. The superiority over inactivated vaccine in neutralization antibodies, as well as the safety profile, support the use of AWcorna as heterologous booster in China.
- Published
- 2022
- Full Text
- View/download PDF
16. Longitudinal Dynamics of Cellular Responses in Recovered COVID-19 Patients
- Author
-
Meng-Li, Cheng, Hui-Ying, Liu, Chao, Zhou, Rui-Ting, Li, Jing, Zheng, Yan-Hong, Qin, Ning, Yang, Yue, Zhang, Juan-Juan, Huang, Zhu, Zhu, Qing-Yu, Meng, Guo-Qing, Wang, Hui, Zhao, Yun, Chen, Chang-Qing, Bai, Cheng-Feng, Qin, and Fan, Li
- Subjects
Cohort Studies ,Immunity, Cellular ,COVID-19 Vaccines ,SARS-CoV-2 ,Immunology ,COVID-19 ,Humans ,Interleukin-2 ,Immunology and Allergy ,CD8-Positive T-Lymphocytes ,Pandemics - Abstract
Safe and effective vaccines and therapeutics based on the understanding of antiviral immunity are urgently needed to end the COVID-19 pandemic. However, the understanding of these immune responses, especially cellular immune responses to SARS-CoV-2 infection, is limited. Here, we conducted a cohort study of COVID-19 patients who were followed and had blood collected to characterize the longitudinal dynamics of their cellular immune responses. Compared with healthy controls, the percentage of activation of SARS-CoV-2 S/N-specific T cells in recovered patients was significantly higher. And the activation percentage of S/N-specific CD8+ T cells in recovered patients was significantly higher than that of CD4+ T cells. Notably, SARS-CoV-2 specific T-cell responses were strongly biased toward the expression of Th1 cytokines, included the cytokines IFNγ, TNFα and IL2. Moreover, the secreted IFNγ and IL2 level in severe patients was higher than that in mild patients. Additionally, the number of IFNγ-secreting S-specific T cells in recovered patients were higher than that of N-specific T cells. Overall, the SARS-CoV-2 S/N-specific T-cell responses in recovered patients were strong, and virus-specific immunity was present until 14-16 weeks after symptom onset. Our work provides a basis for understanding the immune responses and pathogenesis of COVID-19. It also has implications for vaccine development and optimization and speeding up the licensing of the next generation of COVID-19 vaccines.
- Published
- 2022
- Full Text
- View/download PDF
17. Characterization of m
- Author
-
Yu, Liu, Kai, Li, Yan-Peng, Xu, Zhu, Zhu, Hui, Zhao, Xiao-Feng, Li, Qing, Ye, Chengqi, Yi, and Cheng-Feng, Qin
- Subjects
Zika Virus Infection ,Humans ,RNA, Viral ,Genome, Viral ,Zika Virus ,Virus Replication ,3' Untranslated Regions - Abstract
Zika virus (ZIKV) suddenly evolved from a neglected arthropod-borne flavivirus into a pandemic pathogen during 2015-2016. A panel of amino acid mutations has been shown to be responsible for the enhanced neurovirulence and transmissibility of ZIKV. Recent studies have demonstrated that ZIKV genomic RNA is modified by host N6-methyladenosine (m
- Published
- 2022
18. 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
19. 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
20. Development of an automatic integrated gene detection system for novel severe acute respiratory syndrome-related coronavirus (SARS-CoV2)
- Author
-
Ying Zhang, Lizhong Dai, Tao Jiang, Hu Yi, Juan Liu, Sen Zhang, Li Jing, Kang Xiaoping, Lin Li, Cheng-Feng Qin, Wu Xiaoyan, and Yu-Chang Li
- Subjects
0301 basic medicine ,China ,automatic integrated gene detection system ,Virus Cultivation ,Coronavirus disease 2019 (COVID-19) ,Epidemiology ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Pneumonia, Viral ,030106 microbiology ,Immunology ,Real-Time Polymerase Chain Reaction ,medicine.disease_cause ,Sensitivity and Specificity ,Microbiology ,Rapid detection ,Betacoronavirus ,03 medical and health sciences ,Limit of Detection ,Virology ,Drug Discovery ,Humans ,Medicine ,Respiratory system ,Pandemics ,Gene ,DNA Primers ,Coronavirus ,Automation, Laboratory ,Detection limit ,SARS-CoV-2 ,business.industry ,COVID-19 ,qRT-PCR ,Articles ,General Medicine ,rapid detection ,030104 developmental biology ,Infectious Diseases ,Real-time polymerase chain reaction ,SARS-CoV2 ,RNA, Viral ,Parasitology ,Coronavirus Infections ,business ,Research Article - Abstract
In December 2019, Wuhan, China suffered a serious outbreak of a novel coronavirus infectious disease (COVID) caused by novel severe acute respiratory syndrome-related coronavirus (SARS-CoV 2). To quickly identify the pathogen, we designed and screened primer sets, and established a sensitive and specific qRT-PCR assay for SARS-CoV 2; the lower limit of detection (LOD) was 15 (95% CI: 9.8–21) copies per reaction. We combined this qRT-PCR assay with an automatic integration system for nucleic acid extraction and amplification, thereby establishing an automatic integrated gene detection system (AIGS) for SARS-CoV 2. Cross reactive analysis performed in 20 other respiratory viruses and 37 nasopharyngeal swabs confirmed a 100% specificity of the assay. Using two fold diluted SARS-CoV 2 culture, the LOD of AIGS was confirmed to be 365 copies/ml (95% CI: 350–375), which was Comparable to that of conventional qRT-PCR (740 copies/ml, 95% CI: 690–750). Clinical performances of AIGS assay were assessed in 266 suspected COVID-19 clinical respiratory tract samples tested in parallel with a commercial kit. The clinical sensitivity of the AIGS test was 97.62% (95% CI: 0.9320–0.9951) based on the commercial kit test result, and concordance analysis showed a high agreement in SARS-CoV-2 detection between the two assays, Pearson R was 0.9623 (95% CI: 0.9523–0.9703). The results indicated that this AIGS could be used for rapid detection of SARS-CoV 2. With the advantage of simple operation and less time consuming, AIGS could be suitable for SARS-CoV2 detection in primary medical institutions, thus would do a great help to improve detection efficiency and control the spread of COVID-19.
- Published
- 2020
- Full Text
- View/download PDF
21. Safety and immunogenicity of the SARS-CoV-2 ARCoV mRNA vaccine in Chinese adults: a randomised, double-blind, placebo-controlled, phase 1 trial
- Author
-
Gui-Ling Chen, Xiao-Feng Li, Xia-Hong Dai, Nan Li, Meng-Li Cheng, Zhen Huang, Jian Shen, Yu-Hua Ge, Zhen-Wei Shen, Yong-Qiang Deng, Shu-Yuan Yang, Hui Zhao, Na-Na Zhang, Yi-Fei Zhang, Ling Wei, Kai-Qi Wu, Meng-Fei Zhu, Cong-Gao Peng, Qi Jiang, Shou-Chun Cao, Yu-Hua Li, Dan-Hua Zhao, Xiao-Hong Wu, Ling Ni, Hua-Hao Shen, Chen Dong, Bo Ying, Guo-Ping Sheng, Cheng-Feng Qin, Hai-Nv Gao, and Lan-Juan Li
- Subjects
Microbiology (medical) ,Adult ,China ,Vaccines, Synthetic ,COVID-19 Vaccines ,SARS-CoV-2 ,COVID-19 ,Articles ,Antibodies, Viral ,Microbiology ,Antibodies, Neutralizing ,Infectious Diseases ,Immunogenicity, Vaccine ,Virology ,Immunoglobulin G ,Spike Glycoprotein, Coronavirus ,Humans ,mRNA Vaccines ,Pandemics - Abstract
Background Safe and effective vaccines are urgently needed to end the COVID-19 pandemic caused by SARS-CoV-2 infection. We aimed to assess the preliminary safety, tolerability, and immunogenicity of an mRNA vaccine ARCoV, which encodes the SARS-CoV-2 spike protein receptor-binding domain (RBD). Methods This single centre, double-blind, randomised, placebo-controlled, dose-escalation, phase 1 trial of ARCoV was conducted at Shulan (Hangzhou) hospital in Hangzhou, Zhejiang province, China. Healthy adults aged 18–59 years negative for SARS-CoV-2 infection were enrolled and randomly assigned using block randomisation to receive an intramuscular injection of vaccine or placebo. Vaccine doses were 5 μg, 10 μg, 15 μg, 20 μg, and 25 μg. The first six participants in each block were sentinels and along with the remaining 18 participants, were randomly assigned to groups (5:1). In block 1 sentinels were given the lowest vaccine dose and after a 4-day observation with confirmed safety analyses, the remaining 18 participants in the same dose group proceeded and sentinels in block 2 were given their first administration on a two-dose schedule, 28 days apart. All participants, investigators, and staff doing laboratory analyses were masked to treatment allocation. Humoral responses were assessed by measuring anti-SARS-CoV-2 RBD IgG using a standardised ELISA and neutralising antibodies using pseudovirus-based and live SARS-CoV-2 neutralisation assays. SARS-CoV-2 RBD-specific T-cell responses, including IFN-γ and IL-2 production, were assessed using an enzyme-linked immunospot (ELISpot) assay. The primary outcome for safety was incidence of adverse events or adverse reactions within 60 min, and at days 7, 14, and 28 after each vaccine dose. The secondary safety outcome was abnormal changes detected by laboratory tests at days 1, 4, 7, and 28 after each vaccine dose. For immunogenicity, the secondary outcome was humoral immune responses: titres of neutralising antibodies to live SARS-CoV-2, neutralising antibodies to pseudovirus, and RBD-specific IgG at baseline and 28 days after first vaccination and at days 7, 15, and 28 after second vaccination. The exploratory outcome was SARS-CoV-2-specific T-cell responses at 7 days after the first vaccination and at days 7 and 15 after the second vaccination. This trial is registered with www.chictr.org.cn (ChiCTR2000039212). Findings Between Oct 30 and Dec 2, 2020, 230 individuals were screened and 120 eligible participants were randomly assigned to receive five-dose levels of ARCoV or a placebo (20 per group). All participants received the first vaccination and 118 received the second dose. No serious adverse events were reported within 56 days after vaccination and the majority of adverse events were mild or moderate. Fever was the most common systemic adverse reaction (one [5%] of 20 in the 5 μg group, 13 [65%] of 20 in the 10 μg group, 17 [85%] of 20 in the 15 μg group, 19 [95%] of 20 in the 20 μg group, 16 [100%] of 16 in the 25 μg group; p
- Published
- 2022
22. Rapid development of an updated mRNA vaccine against the SARS-CoV-2 Omicron variant
- Author
-
Na-Na Zhang, Rong-Rong Zhang, Yi-Fei Zhang, Kai Ji, Xiao-Chuan Xiong, Qian-Shan Qin, Peng Gao, Xi-Shan Lu, Hang-Yu Zhou, Hai-Feng Song, Bo Ying, and Cheng-Feng Qin
- Subjects
Vaccines, Synthetic ,SARS-CoV-2 ,COVID-19 ,Humans ,Cell Biology ,mRNA Vaccines ,Molecular Biology - Published
- 2022
23. Construction and characterization of UAA-controlled recombinant Zika virus by genetic code expansion
- Author
-
Yan-Peng Xu, Xing-Yao Huang, Xiaofeng Li, Rong-Rong Zhang, Yong-Qiang Deng, Qing Xia, Cheng-Feng Qin, and Qing Ye
- Subjects
DNA, Recombinant ,Computational biology ,Biology ,Virus Replication ,General Biochemistry, Genetics and Molecular Biology ,law.invention ,Zika virus ,Viral Proteins ,law ,Chlorocebus aethiops ,Animals ,Humans ,Amino Acids ,Vero Cells ,General Environmental Science ,Recombination, Genetic ,Base Sequence ,Zika Virus Infection ,Zika Virus ,Genetic code ,biology.organism_classification ,HEK293 Cells ,Genetic Code ,Protein Biosynthesis ,Codon, Terminator ,Recombinant DNA ,General Agricultural and Biological Sciences - Published
- 2020
- Full Text
- View/download PDF
24. Update on the Animal Models and Underlying Mechanisms for ZIKV-Induced Microcephaly
- Author
-
Dan Xu, Cui Li, Zhiheng Xu, and Cheng-Feng Qin
- Subjects
medicine.medical_specialty ,Microcephaly ,Biology ,World health ,Zika virus ,03 medical and health sciences ,0302 clinical medicine ,Virology ,ZikV Infection ,medicine ,Animals ,Humans ,030304 developmental biology ,0303 health sciences ,Zika Virus Infection ,Mechanism (biology) ,Public health ,Brain ,Zika Virus ,Mammalian brain ,biology.organism_classification ,medicine.disease ,Disease Models, Animal ,Public Health ,Neuroscience ,030217 neurology & neurosurgery - Abstract
The circulation of Zika virus (ZIKV) in nearly 80 countries and territories poses a significant global threat to public health. ZIKV is causally linked to severe developmental defects in the brain, recognized as congenital Zika syndrome (CZS), which includes microcephaly and other serious congenital neurological complications. Since the World Health Organization declared the ZIKV outbreak a public health emergency of international concern, remarkable progress has been made in the generation of different ZIKV infection animal models to gain insight into cellular targets and pathogenesis and to explore the associated underlying mechanisms. Here we focus on summarizing our current understanding of the effects of ZIKV on mammalian brain development in different developmental stages and discuss the potential underlying mechanisms of ZIKV-induced CZS, as well as future perspectives.
- Published
- 2019
- Full Text
- View/download PDF
25. Zika virus infection induces RNAi-mediated antiviral immunity in human neural progenitors and brain organoids
- Author
-
Jiuyue Xu, Zhiheng Xu, Da Zhang, Xi Zhou, Na-Na Zhang, Boya Zhang, Guilai Chen, Yang Qiu, Wenliang Zhu, Baoyang Hu, Jie Cui, Menghua Wu, Rong-Rong Zhang, Bao Lyu, Xiaofeng Li, Fan Mo, Qi Chen, Miao Wang, Yan-Peng Xu, Jin Wu, Jianghong Man, Cheng-Feng Qin, Meng-Li Cheng, and Qing Ye
- Subjects
Antiviral Agents ,Article ,Zika virus ,03 medical and health sciences ,0302 clinical medicine ,Immunity ,RNA interference ,Humans ,Progenitor cell ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,biology ,Zika Virus Infection ,Brain ,Zika Virus ,Cell Biology ,biology.organism_classification ,Virology ,Phenotype ,Neural stem cell ,Organoids ,Viral replication ,Cell culture ,RNA Interference ,030217 neurology & neurosurgery - Abstract
The re-emergence of Zika virus (ZIKV) in the Western Hemisphere has resulted in global public health crisis since 2015. ZIKV preferentially infects and targets human neural progenitor cells (hNPCs) and causes fetal microcephaly upon maternal infection. hNPCs not only play critical roles during fetal brain development, but also persist in adult brain throughout life. Yet the mechanism of innate antiviral immunity in hNPCs remains largely unknown. Here, we show that ZIKV infection triggers the abundant production of virus-derived small interfering RNAs in hNPCs, but not in the more differentiated progenies or somatic cells. Ablation of key RNAi machinery components significantly enhances ZIKV replication in hNPCs. Furthermore, enoxacin, a broad-spectrum antibiotic that is known as an RNAi enhancer, exerts potent anti-ZIKV activity in hNPCs and other RNAi-competent cells. Strikingly, enoxacin treatment completely prevents ZIKV infection and circumvents ZIKV-induced microcephalic phenotypes in brain organoid models that recapitulate human fetal brain development. Our findings highlight the physiological importance of RNAi-mediated antiviral immunity during the early stage of human brain development, uncovering a novel strategy to combat human congenital viral infections through enhancing RNAi.
- Published
- 2019
- Full Text
- View/download PDF
26. Convergent evolution of SARS-CoV-2 in human and animals
- Author
-
Cheng-Feng Qin, Xiaofeng Li, Aiping Wu, Hang Fan, Na Han, Hangyu Zhou, and Chengyang Ji
- Subjects
2019-20 coronavirus outbreak ,Coronavirus disease 2019 (COVID-19) ,SARS-CoV-2 ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,COVID-19 ,Cell Biology ,Biology ,Biochemistry ,Virology ,Polymorphism, Single Nucleotide ,Human genetics ,Evolution, Molecular ,Disease Models, Animal ,Convergent evolution ,Drug Discovery ,Spike Glycoprotein, Coronavirus ,Commentary ,Animals ,Humans ,Stem cell ,Developmental biology ,Biotechnology - Published
- 2021
27. 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
28. 25-Hydroxycholesterol is a potent SARS-CoV-2 inhibitor
- Author
-
Lili Li, Heng Yang, Chao Zhou, Sarah Gold, Cheng-Feng Qin, Jie Li, Genhong Cheng, Changqing Zhang, Jun He, Xiaofeng Li, Xiang Li, Shulong Zu, Qi Chen, Hui Zhao, and Yong-Qiang Deng
- Subjects
2019-20 coronavirus outbreak ,Fatal outcome ,Coronavirus disease 2019 (COVID-19) ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Clinical Sciences ,Biology ,Peptidyl-Dipeptidase A ,Virus Replication ,Transgenic ,Mice ,Betacoronavirus ,Fatal Outcome ,Chlorocebus aethiops ,Animals ,Humans ,Viral ,Letter to the Editor ,Molecular Biology ,Vero Cells ,Pandemics ,Inbred BALB C ,Aged ,Innate immunity ,Extramural ,Animal ,Reverse Transcriptase Polymerase Chain Reaction ,SARS-CoV-2 ,COVID-19 ,Cell Biology ,Pneumonia ,Molecular biology ,Hydroxycholesterols ,Mice transgenic ,Mechanisms of disease ,Disease Models ,Female ,Angiotensin-Converting Enzyme 2 ,Biochemistry and Cell Biology ,Coronavirus Infections ,Developmental Biology - Abstract
Author(s): Zu, Shulong; Deng, Yong-Qiang; Zhou, Chao; Li, Jie; Li, Lili; Chen, Qi; Li, Xiao-Feng; Zhao, Hui; Gold, Sarah; He, Jun; Li, Xiang; Zhang, Changqing; Yang, Heng; Cheng, Genhong; Qin, Cheng-Feng
- Published
- 2020
29. Type-IInterferon-Inducible SERTAD3 Inhibits Influenza A Virus Replication by Blocking the Assembly of Viral RNA Polymerase Complex
- Author
-
Cheng-Feng Qin, Xiaofeng Li, Na-Na Zhang, Ning Lu, Sarah Gold, Ping Liu, Nina N. Sun, Peishuang Du, Lili Li, Shulong Zu, Rong-Rong Zhang, Chunfeng Li, Yong-Qiang Deng, Jingfeng Wang, Genhong Cheng, Xiang Chen, and Tao Jiang
- Subjects
0301 basic medicine ,RNA-dependent RNA polymerase ,Peptide ,Biology ,IFN-stimulated genes ,Virus Replication ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,Madin Darby Canine Kidney Cells ,03 medical and health sciences ,chemistry.chemical_compound ,Dogs ,0302 clinical medicine ,SERTAD3 ,Interferon ,RNA polymerase ,Influenza A virus ,medicine ,Animals ,Humans ,Viral Replicase Complex Proteins ,Viral rna ,Gene ,lcsh:QH301-705.5 ,Polymerase ,Mice, Knockout ,chemistry.chemical_classification ,virus diseases ,Virology ,Mice, Inbred C57BL ,Protein Subunits ,IAV ,HEK293 Cells ,030104 developmental biology ,chemistry ,lcsh:Biology (General) ,A549 Cells ,Interferon Type I ,Trans-Activators ,biology.protein ,type I interferon ,Peptides ,030217 neurology & neurosurgery ,Protein Binding ,medicine.drug - Abstract
Summary Influenza A virus (IAV) infection stimulates a type I interferon (IFN-I) response in host cells that exerts antiviral effects by inducing the expression of hundreds of IFN-stimulated genes (ISGs). However, most ISGs are poorly studied for their roles in the infection of IAV. Herein, we demonstrate that SERTA domain containing 3 (SERTAD3) has a significant inhibitory effect on IAV replication in vitro. More importantly, Sertad3−/− mice develop more severe symptoms upon IAV infection. Mechanistically, we find SERTAD3 reduces IAV replication through interacting with viral polymerase basic protein 2 (PB2), polymerase basic protein 1 (PB1), and polymerase acidic protein (PA) to disrupt the formation of the RNA-dependent RNA polymerase (RdRp) complex. We further identify an 8-amino-acid peptide of SERTAD3 as a minimum interacting motif that can disrupt RdRp complex formation and inhibit IAV replication. Thus, our studies not only identify SERTAD3 as an antiviral ISG, but also provide the mechanism of potential application of SERTAD3-derived peptide in suppressing influenza replication.
- Published
- 2020
30. 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
31. Impaired Cellular Immunity to SARS-CoV-2 in Severe COVID-19 Patients
- Author
-
Ling Ni, Meng-Li Cheng, Yu Feng, Hui Zhao, Jingyuan Liu, Fang Ye, Qing Ye, Gengzhen Zhu, Xiaoli Li, Pengzhi Wang, Jing Shao, Yong-Qiang Deng, Peng Wei, Fang Chen, Cheng-Feng Qin, Guoqing Wang, Fan Li, Hui Zeng, and Chen Dong
- Subjects
lcsh:Immunologic diseases. Allergy ,CD4-Positive T-Lymphocytes ,Male ,Cellular immunity ,ARDS ,neutralization antibody ,Immunology ,T cells ,Cell Count ,CD8-Positive T-Lymphocytes ,Antibodies, Viral ,Virus ,Pathogenesis ,Interferon-gamma ,Immune system ,medicine ,Immunology and Allergy ,Humans ,Interferon gamma ,Cells, Cultured ,Original Research ,Immunity, Cellular ,Respiratory Distress Syndrome ,biology ,business.industry ,SARS-CoV-2 ,COVID-19 ,adaptive immunity ,biochemical phenomena, metabolism, and nutrition ,Middle Aged ,acute respiratory distress syndrome ,Acquired immune system ,medicine.disease ,Antibodies, Neutralizing ,Killer Cells, Natural ,interferon gamma ,Humoral immunity ,biology.protein ,Disease Progression ,Female ,Antibody ,business ,lcsh:RC581-607 ,CD8 ,medicine.drug - Abstract
SUMMARYThe World Health Organization has declared SARS-CoV-2 virus outbreak a world-wide pandemic. Individuals infected by the virus exhibited different degrees of symptoms, the basis of which remains largely unclear. Currently, though convalescent individuals have been shown with both cellular and humoral immune responses, there is very limited understanding on the immune responses, especially adaptive immune responses, in patients with severe COVID-19. Here, we examined 10 blood samples from COVID-19 patients with acute respiratory distress syndrome (ARDS). The majority of them (70%) mounted SARS-CoV-2-specific humoral immunity with production of neutralizing antibodies. However, compared to healthy controls, the percentages and absolute numbers of both NK cells and CD8+ T cells were significantly reduced, accompanied with decreased IFNγ expression in CD4+ T cells in peripheral blood from severe patients. Most notably, we failed in detecting SARS-CoV-2-specific IFNγ production by peripheral blood lymphocytes from these patients. Our work thus indicates that COVID-19 patients with severe symptoms are associated with defective cellular immunity, which not only provides insights on understanding the pathogenesis of COVID-19, but also has implications in developing an effective vaccine to SARS-CoV-2.
- Published
- 2020
32. 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
33. The m
- Author
-
Jun'e, Liu, Yan-Peng, Xu, Kai, Li, Qing, Ye, Hang-Yu, Zhou, Hanxiao, Sun, Xiaoyu, Li, Liu, Yu, Yong-Qiang, Deng, Rui-Ting, Li, Meng-Li, Cheng, Bo, He, Jia, Zhou, Xiao-Feng, Li, Aiping, Wu, Chengqi, Yi, and Cheng-Feng, Qin
- Subjects
Adenosine ,SARS-CoV-2 ,COVID-19 ,Genome, Viral ,DNA Methylation ,Virus Replication ,RNA modification ,Article ,Cell Line ,Mechanisms of disease ,Gene Expression Regulation ,Chlorocebus aethiops ,Host-Pathogen Interactions ,Mutagenesis, Site-Directed ,Animals ,Humans ,RNA, Messenger ,Vero Cells ,Phylogeny - Abstract
The newly identified Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has resulted in a global health emergency because of its rapid spread and high mortality. The molecular mechanism of interaction between host and viral genomic RNA is yet unclear. We demonstrate herein that SARS-CoV-2 genomic RNA, as well as the negative-sense RNA, is dynamically N6-methyladenosine (m6A)-modified in human and monkey cells. Combined RIP-seq and miCLIP analyses identified a total of 8 m6A sites at single-base resolution in the genome. Especially, epidemic strains with mutations at these identified m6A sites have emerged worldwide, and formed a unique cluster in the US as indicated by phylogenetic analysis. Further functional experiments showed that m6A methylation negatively regulates SARS-CoV-2 infection. SARS-CoV-2 infection also triggered a global increase in host m6A methylome, exhibiting altered localization and motifs of m6A methylation in mRNAs. Altogether, our results identify m6A as a dynamic epitranscriptomic mark mediating the virus–host interaction.
- Published
- 2020
34. 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
35. Structure-based development of human antibody cocktails against SARS-CoV-2
- Author
-
Zhen Cui, Yong-Qiang Deng, Yan Guo, Li Zhang, Yuxi Wang, Nan Wang, Yanjun Zhang, Xiangxi Wang, Lei Cao, Fengcai Zhu, Weimin Li, Rui Feng, Lei Wang, Cheng-Feng Qin, and Yao Sun
- Subjects
2019-20 coronavirus outbreak ,Coronavirus disease 2019 (COVID-19) ,SARS-CoV-2 ,Molecular biology ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,COVID-19 ,Cell Biology ,Biology ,Antibodies, Viral ,Virology ,Antibodies, Neutralizing ,COVID-19 Drug Treatment ,Cryoelectron microscopy ,Spike Glycoprotein, Coronavirus ,biology.protein ,Structure based ,Humans ,Antibody ,Viral immunology ,Letter to the Editor - Published
- 2020
36. 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
37. A Thermostable mRNA Vaccine against COVID-19
- Author
-
Hua Yuan Yan, Cheng-Feng Qin, Xing Yao Huang, Shu Long Zu, Qing Ye, Tian Shu Cao, Xiaofeng Li, Hang Fan, Chao Zhou, Yi Jiao Huang, Peng Gao, Yan Guo, Xia Zhong, Yuhang Jiang, Qi He, Zhen Yu Zha, Na Na Zhang, Dongsheng Zhou, Rong Rong Zhang, Weijin Huang, Shi Hui Sun, Yong Qiang Deng, You Chun Wang, Xiao Yang, Guan Yang, Qi Chen, Hui Zhao, Hong Ying Qiu, Bo Ying, Xia Lin Xue, and Jian Hui Nie
- Subjects
Male ,Mice ,0302 clinical medicine ,Immunogenicity, Vaccine ,Chlorocebus aethiops ,0303 health sciences ,Mice, Inbred ICR ,Vaccines, Synthetic ,Protection ,biology ,Viral Vaccine ,Spike Glycoprotein, Coronavirus ,RNA, Viral ,Female ,Antibody ,Coronavirus Infections ,COVID-19 Vaccines ,Lipid nanoparticle ,Injections, Intramuscular ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Animals ,Humans ,RNA, Messenger ,Binding site ,Gene ,Vaccine Potency ,Vero Cells ,030304 developmental biology ,Messenger RNA ,Binding Sites ,SARS-CoV-2 ,COVID-19 ,Viral Vaccines ,Th1 Cells ,Non-human primate ,Virology ,Antibodies, Neutralizing ,Macaca fascicularis ,mRNA vaccine ,HEK293 Cells ,Immunization ,Mouse adapted strain ,Vero cell ,biology.protein ,Nanoparticles ,030217 neurology & neurosurgery ,HeLa Cells - Abstract
Summary There has been an urgent need of vaccines against coronavirus disease 2019 (COVID-19) due to the ongoing SARS-CoV-2 pandemic. Among all approaches, messenger RNA (mRNA) -based vaccine has emerged as a rapid and versatile platform to quickly respond to such a challenge. Here, we developed a lipid-nanoparticle-encapsulated mRNA (mRNA-LNP) encoding the receptor binding domain (RBD) of SARS-CoV-2 as a vaccine candidate (termed ARCoV). Intramuscular immunization of ARCoV mRNA-LNPs elicited robust neutralizing antibodies against SARS-CoV-2 as well as Th1-biased cellular response in mice and non-human primates. Two doses of ARCoV immunization in mice conferred complete protection against the challenge of a SARS-CoV-2 mouse adapted strain. Additionally, ARCoV was manufactured in liquid formulation and can be stored at room temperature for at least one week. This novel COVID-19 mRNA vaccine, ARCoV, is currently being evaluated in phase 1 clinical trials., Highlights Development of LNP-encapsulated mRNA vaccine (ARCoV) targeting the RBD of SARS-CoV-2 ARCoV induces neutralizing antibodies and T-cell immunity in mice and NHPs. ARCoV vaccination confers full protection against SARS-CoV-2 challenge in mice. ARCoV is a thermostable vaccine candidate for phase I studies, ARCoV is an LNP-encapsulated mRNA vaccine platform that is highly immunogenic and safe in mice and non-human primates, conferring protection against the challenge a SARS-CoV-2 mouse adapted strain.
- Published
- 2020
38. 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
39. 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
40. 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
41. Short Direct Repeats in the 3′ Untranslated Region Are Involved in Subgenomic Flaviviral RNA Production
- Author
-
Zhiming Yuan, Xiao-Dan Li, Han-Qing Ye, Xing Yao Huang, Cheng-Feng Qin, Xiaolin Niu, Na Li, Yan Peng Xu, Hao Long Dong, Bo Zhang, Qiu Yan Zhang, Xiaofeng Li, Hong Jiang Wang, Xianyang Fang, Peng Gong, Pei Yong Shi, Cheng Lin Deng, Qi Chen, and Hui Zhao
- Subjects
Untranslated region ,Immunology ,Genome, Viral ,Biology ,Microbiology ,Genome ,Conserved sequence ,Cricetinae ,Virology ,Exoribonuclease ,Chlorocebus aethiops ,Animals ,Humans ,Direct repeat ,3' Untranslated Regions ,Vero Cells ,Subgenomic mRNA ,Genetics ,Three prime untranslated region ,Flavivirus ,RNA ,Genome Replication and Regulation of Viral Gene Expression ,Culicidae ,A549 Cells ,Tandem Repeat Sequences ,Insect Science ,Nucleic Acid Conformation ,RNA, Viral - Abstract
Mosquito-borne flaviviruses consist of a positive-sense genome RNA flanked by the untranslated regions (UTRs). There is a panel of highly complex RNA structures in the UTRs with critical functions. For instance, Xrn1-resistant RNAs (xrRNAs) halt Xrn1 digestion, leading to the production of subgenomic flaviviral RNA (sfRNA). Conserved short direct repeats (DRs), also known as conserved sequences (CS) and repeated conserved sequences (RCS), have been identified as being among the RNA elements locating downstream of xrRNAs, but their biological function remains unknown. In this study, we revealed that the specific DRs are involved in the production of specific sfRNAs in both mammalian and mosquito cells. Biochemical assays and structural remodeling demonstrate that the base pairings in the stem of these DRs control sfRNA formation by maintaining the binding affinity of the corresponding xrRNAs to Xrn1. On the basis of these findings, we propose that DRs functions like a bracket holding the Xrn1-xrRNA complex for sfRNA formation. IMPORTANCE Flaviviruses include many important human pathogens. The production of subgenomic flaviviral RNAs (sfRNAs) is important for viral pathogenicity as a common feature of flaviviruses. sfRNAs are formed through the incomplete degradation of viral genomic RNA by the cytoplasmic 5ʹ–3ʹ exoribonuclease Xrn1 halted at the Xrn1-resistant RNA (xrRNA) structures within the 3ʹ-UTR. The 3ʹ-UTRs of the flavivirus genome also contain distinct short direct repeats (DRs), such as RCS3, CS3, RCS2, and CS2. However, the biological functions of these ancient primary DR sequences remain largely unknown. Here, we found that DR sequences are involved in sfRNA formation and viral virulence and provide novel targets for the rational design of live attenuated flavivirus vaccine.
- Published
- 2020
- Full Text
- View/download PDF
42. Salivary factor LTRIN from Aedes aegypti facilitates the transmission of Zika virus by interfering with the lymphotoxin-β receptor
- Author
-
Xue Hao, Xiaomin Guo, Hongning Zhou, Cheng-Feng Qin, Tao Xu, Lin Jin, Pengpeng Li, Mingdong Yang, Peng Sun, Xiaopeng Qi, Mingzhao Zhu, Hua Peng, Chuanbin Shen, Chunmiao Hu, Ombati Rose, Ren Lai, Gong Cheng, and Jingya Guo
- Subjects
0301 basic medicine ,030106 microbiology ,Immunology ,Mosquito Vectors ,Aedes aegypti ,Proinflammatory cytokine ,Zika virus ,Mice ,03 medical and health sciences ,Immune system ,Aedes ,Lymphotoxin beta Receptor ,Animals ,Humans ,Immunology and Allergy ,Saliva ,Infectivity ,biology ,Zika Virus Infection ,Zika Virus ,biology.organism_classification ,Virology ,030104 developmental biology ,Lymphotoxin ,Insect Proteins ,Lymphotoxin beta receptor - Abstract
Pathogens have co-evolved with mosquitoes to optimize transmission to hosts. Mosquito salivary-gland extract is known to modulate host immune responses and facilitate pathogen transmission, but the underlying molecular mechanisms of this have remained unknown. In this study, we identified and characterized a prominent 15-kilodalton protein, LTRIN, obtained from the salivary glands of the mosquito Aedes aegypti. LTRIN expression was upregulated in blood-fed mosquitoes, and LTRIN facilitated the transmission of Zika virus (ZIKV) and exacerbated its pathogenicity by interfering with signaling through the lymphotoxin-β receptor (LTβR). Mechanically, LTRIN bound to LTβR and 'preferentially' inhibited signaling via the transcription factor NF-κB and the production of inflammatory cytokines by interfering with the dimerization of LTβR during infection with ZIKV. Furthermore, treatment with antibody to LTRIN inhibited mosquito-mediated infection with ZIKV, and abolishing LTβR potentiated the infectivity of ZIKV both in vitro and in vivo. This study provides deeper insight into the transmission of mosquito-borne diseases in nature and supports the therapeutic potential of inhibiting the action of LTRIN to disrupt ZIKV transmission.
- Published
- 2018
- Full Text
- View/download PDF
43. American Strain of Zika Virus Causes More Severe Microcephaly Than an Old Asian Strain in Neonatal Mice
- Author
-
Guanghui Li, Yong Qiang Deng, Hong Jiang Wang, Feng Zhang, Xing Yao Huang, Haozhen Yang, Ling Yuan, Zhiheng Xu, Zhong-Yu Liu, Qin Wang, Lei Shi, Cheng-Feng Qin, and Qing Ye
- Subjects
0301 basic medicine ,Microcephaly ,lcsh:Medicine ,Brain damage ,Severity of Illness Index ,General Biochemistry, Genetics and Molecular Biology ,American strain ,Zika virus ,Asian strain ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,medicine ,Animals ,Humans ,Progenitor cell ,Neurons ,lcsh:R5-920 ,biology ,Zika Virus Infection ,Strain (biology) ,lcsh:R ,Brain ,General Medicine ,Zika Virus ,biology.organism_classification ,medicine.disease ,Venezuela ,Virology ,Oligodendrocyte ,United States ,Flavivirus ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Immunology ,medicine.symptom ,lcsh:Medicine (General) ,030217 neurology & neurosurgery ,Research Paper - Abstract
Zika virus (ZIKV) has evolved from an overlooked mosquito-borne flavivirus into a global health threat due to its astonishing causal link to microcephaly and other disorders. ZIKV has been shown to infect neuronal progenitor cells of the fetal mouse brain, which is comparable to the first-trimester human fetal brain, and result in microcephaly. However, whether there are different effects between the contemporary ZIKV strain and its ancestral strain in the neonatal mouse brain, which is comparable with the second-trimester human fetal brain, is unclear. Here we adopted a mouse model which enables us to study the postnatal effect of ZIKV infection. We show that even 100 pfu of ZIKV can replicate and infect neurons and oligodendrocytes in most parts of the brain. Compared with the ancestral strain from Cambodia (CAM/2010), infection of the ZIKV strain from Venezuela (VEN/2016) leads to much more severe microcephaly, accompanied by more neuronal cell death, abolishment of oligodendrocyte development, and a more dramatic immune response. The serious brain damage caused by VEN/2016 infection would be helpful to elucidate why the American strain resulted in severe neurovirulence in infants and will provide clinical guidance for the diagnosis and treatment of infection by different ZIKV strains., Highlights • The infection of an American strain of ZIKV leads to more severe microcephaly than the ancestral Asian strain. • American strain infects more cells, and induces more dramatic immune response and cell death than ancestral Asian strain. World attention has been drawn to a global Zika virus (ZIKV) outbreak due to its unexpected causal link to congenital brain abnormalities, especially microcephaly. Infection of pregnant women with the American Zika strain, but not the ancestral Asian strain, can result in microcephaly in infants. However, the phenotypic difference between the contemporary American strain and ancestral Asian strain of ZIKV is still unclear. We employed the ZIKV infection model of a neonatal mouse brain to compare the difference between these two strains. We find that infection by the American strain leads to more severe microcephaly than the ancestral Asian strain.
- Published
- 2017
44. A peptide-based viral inactivator inhibits Zika virus infection in pregnant mice and fetuses
- Author
-
Kwok-Yung Yuen, Shibo Jiang, Xiaohui Zhou, Na-Na Zhang, Yufeng Yu, Yanyan Dai, Yu Meng, Lanying Du, Yuan Li, Lu Lu, Jia-Nan Hao, Qian Wang, Peng Zou, Min Tian, Cheng-Feng Qin, Fei Yu, Yong-Qiang Deng, and Jasper Fuk-Woo Chan
- Subjects
0301 basic medicine ,General Physics and Astronomy ,Antibodies, Viral ,Virus Replication ,Zika virus ,Mice ,Viral Envelope Proteins ,Pregnancy ,Interferon ,Cricetinae ,Chlorocebus aethiops ,reproductive and urinary physiology ,Mice, Inbred ICR ,Multidisciplinary ,Zika Virus Infection ,Transmission (medicine) ,Flow Cytometry ,Flavivirus ,Female ,Antibody ,medicine.drug ,Science ,030106 microbiology ,Biology ,Antiviral Agents ,Article ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,03 medical and health sciences ,medicine ,Animals ,Humans ,Vero Cells ,Fetus ,Virion ,Viral Vaccines ,Zika Virus ,General Chemistry ,Viral membrane ,biology.organism_classification ,Antibodies, Neutralizing ,Virology ,Mice, Inbred C57BL ,Disease Models, Animal ,HEK293 Cells ,030104 developmental biology ,Microscopy, Fluorescence ,Drug Design ,Vero cell ,biology.protein ,Pregnancy, Animal ,Peptides - Abstract
Zika virus (ZIKV), a re-emerging flavivirus associated with neurological disorders, has spread rapidly to more than 70 countries and territories. However, no specific vaccines or antiviral drugs are currently available to prevent or treat ZIKV infection. Here we report that a synthetic peptide derived from the stem region of ZIKV envelope protein, designated Z2, potently inhibits infection of ZIKV and other flaviviruses in vitro. We show that Z2 interacts with ZIKV surface protein and disrupts the integrity of the viral membrane. Z2 can penetrate the placental barrier to enter fetal tissues and is safe for use in pregnant mice. Intraperitoneal administration of Z2 inhibits vertical transmission of ZIKV in pregnant C57BL/6 mice and protects type I or type I/II interferon receptor-deficient mice against lethal ZIKV challenge. Thus, Z2 has potential to be further developed as an antiviral treatment against ZIKV infection in high-risk populations, particularly pregnant women., Zika virus (ZIKV) has spread rapidly in recent years and there is a need for antiviral treatments. Here, the authors develop an antiviral peptide, based on the stem region of ZIKV envelope protein, and show that it is safe in pregnant mice and inhibits ZIKV infection in pregnant mice and fetuses.
- Published
- 2017
45. Zika virus directly infects peripheral neurons and induces cell death
- Author
-
Ronghua Li, Feiran Zhang, Zhiheng Xu, In Young Choi, Luoxiu Huang, Tianlei Xu, Hotae Lim, Cheng-Feng Qin, Qing-Feng Wu, Fahimeh Mirakhori, Yohan Oh, Peng Jin, Hengli Tang, Yaqing Wang, Hongjun Song, Hao Wu, Zhexing Wen, Gabsang Lee, Cui Li, Emily M. Lee, and Guo Li Ming
- Subjects
0301 basic medicine ,Programmed cell death ,Mice, 129 Strain ,Global Health ,Article ,Zika virus ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Neural Stem Cells ,In vivo ,Chlorocebus aethiops ,Animals ,Humans ,Vero Cells ,Cells, Cultured ,Neurons ,Mice, Inbred ICR ,biology ,Cell Death ,Molecular pathology ,Zika Virus Infection ,General Neuroscience ,Peripheral Nervous System Diseases ,Neural crest ,Zika Virus ,biology.organism_classification ,Virology ,Neural stem cell ,In vitro ,3. Good health ,030104 developmental biology ,nervous system ,Immunology ,Vero cell ,030217 neurology & neurosurgery - Abstract
Zika virus (ZIKV)-infection is associated with neurological disorders of both the central and peripheral nervous systems (PNS), yet few studies have directly examined PNS-infection. Here we show that intraperitoneally or intraventricularly-injected ZIKV in the mouse could infect and impact peripheral neurons in vivo. Moreover, ZIKV productively infects stem cell-derived human neural crest cells and peripheral neurons in vitro, leading to increased cell death, transcriptional dysregulation and cell-type specific molecular pathology.
- Published
- 2017
46. 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
47. 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
48. Machine Learning Methods for Predicting Human-Adaptive Influenza A Viruses Based on Viral Nucleotide Compositions
- Author
-
Zhongpeng Zhao, Wu Xiaoyan, Bo Li, Li Yuchang, Meng-Ting Huang, Xiaoping Kang, Tao Jiang, Cheng-Feng Qin, Yi Hu, Jing Li, and Sen Zhang
- Subjects
Adaptation, Biological ,Hemagglutinin (influenza) ,Biology ,Machine learning ,computer.software_genre ,Nucleotide composition ,Machine Learning ,03 medical and health sciences ,Viral Proteins ,human adaptation ,Genetics ,Methods ,Humans ,Nucleotide ,genomic nucleotide composition ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,Polymerase ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,030306 microbiology ,business.industry ,influenza A viruses (IAVs) ,machine learning (ML) ,dinucleotide ,Influenza a ,Nucleoprotein ,H1n1 pandemic ,chemistry ,Influenza A virus ,Host-Pathogen Interactions ,biology.protein ,Artificial intelligence ,business ,Neuraminidase ,computer - Abstract
Each influenza pandemic was caused at least partly by avian- and/or swine-origin influenza A viruses (IAVs). The timing of and the potential IAVs involved in the next pandemic are currently unpredictable. We aim to build machine learning (ML) models to predict human-adaptive IAV nucleotide composition. A total of 217,549 IAV full-length coding sequences of the PB2 (polymerase basic protein-2), PB1, PA (polymerase acidic protein), HA (hemagglutinin), NP (nucleoprotein), and NA (neuraminidase) segments were decomposed for their codon position-based mononucleotides (12 nts) and dinucleotides (48 dnts). A total of 68,742 human sequences and 68,739 avian sequences (1:1) were resampled to characterize the human adaptation-associated (d)nts with principal component analysis (PCA) and other ML models. Then, the human adaptation of IAV sequences was predicted based on the characterized (d)nts. Respectively, 9, 12, 11, 13, 10 and 9 human-adaptive (d)nts were optimized for the six segments. PCA and hierarchical clustering analysis revealed the linear separability of the optimized (d)nts between the human-adaptive and avian-adaptive sets. The results of the confusion matrix and the area under the receiver operating characteristic curve indicated a high performance of the ML models to predict human adaptation of IAVs. Our model performed well in predicting the human adaptation of the swine/avian IAVs before and after the 2009 H1N1 pandemic. In conclusion, we identified the human adaptation-associated genomic composition of IAV segments. ML models for IAV human adaptation prediction using large IAV genomic data sets can facilitate the identification of key viral factors that affect virus transmission/pathogenicity. Most importantly, it allows the prediction of pandemic influenza.
- Published
- 2019
49. Zika virus degrades the ω-3 fatty acid transporter Mfsd2a in brain microvascular endothelial cells and impairs lipid homeostasis
- Author
-
Xingyao Huang, Xiaojing Chi, Tianli Lin, Lei Shi, Xiuying Liu, Cheng-Feng Qin, Jia Zhou, Min Cheng, Wei Yang, Hua Xu, and Jingjing Fan
- Subjects
Microcephaly ,Docosahexaenoic Acids ,Biology ,Microbiology ,Pathogenesis ,03 medical and health sciences ,0302 clinical medicine ,Virology ,Fatty Acids, Omega-3 ,Human Umbilical Vein Endothelial Cells ,medicine ,Animals ,Homeostasis ,Humans ,Research Articles ,030304 developmental biology ,Mice, Knockout ,0303 health sciences ,Multidisciplinary ,Symporters ,Zika Virus Infection ,HEK 293 cells ,SciAdv r-articles ,Brain ,Endothelial Cells ,Lipid metabolism ,Zika Virus ,Human brain ,Lipid Metabolism ,medicine.disease ,Cell biology ,HEK293 Cells ,Phenotype ,medicine.anatomical_structure ,Docosahexaenoic acid ,Microvessels ,Proteolysis ,Ectopic expression ,030217 neurology & neurosurgery ,Research Article - Abstract
Both the endothelial transporter Mfsd2a and docosahexaenoic acid uptake are disrupted in Zika virus–induced microcephaly models., Zika virus (ZIKV) infection during pregnancy increases the risk of postnatal microcephaly. Neurovascular function provides a homeostatic environment for proper brain development. The major facilitator superfamily domain-containing protein 2 (Mfsd2a) is selectively expressed in human brain microvascular endothelial cells (hBMECs) and is the major transporter mediating the brain uptake of docosahexaenoic acid (DHA). We have discovered a pivotal role for Mfsd2a in the pathogenesis of ZIKV. ZIKV disrupted Mfsd2a both in cultured primary hBMECs and in the neonatal mouse brain. ZIKV envelope (E) protein specifically interacted with Mfsd2a and promoted Mfsd2a polyubiquitination for proteasome-dependent degradation. Infection with ZIKV or ectopic expression of ZIKV E impaired Mfsd2a-mediated DHA uptake. Lipidomic analysis revealed obvious differences in DHA-containing lipids after ZIKV infection. Supplementation with DHA rescued ZIKV-caused growth restriction and microcephaly. Our findings suggest endothelial Mfsd2a as an important pathogenic mediator and supplementation with DHA as a potential therapeutic option for ZIKV infection.
- Published
- 2019
- Full Text
- View/download PDF
50. Long non‐coding subgenomic flavivirus RNAs have extended 3D structures and are flexible in solution
- Author
-
Yupeng Zhang, Zhong-Yu Liu, Xianyang Fang, Yikan Zhang, Yan Wang, Cheng-Feng Qin, Meng-Li Cheng, and Junfeng Ma
- Subjects
computational modeling ,Models, Molecular ,small angle X‐ray scattering ,West Nile virus ,Genome, Viral ,Computational biology ,Dengue virus ,3D structure ,subgenomic flavivirus RNA ,medicine.disease_cause ,Biochemistry ,Article ,Zika virus ,03 medical and health sciences ,0302 clinical medicine ,X-Ray Diffraction ,Structural Biology ,Genetics ,medicine ,Animals ,Humans ,Molecular Biology ,030304 developmental biology ,Subgenomic mRNA ,0303 health sciences ,Base Sequence ,biology ,Flavivirus ,Articles ,Zika Virus ,biology.organism_classification ,Pathogenicity ,Microbiology, Virology & Host Pathogen Interaction ,Long non-coding RNA ,Solutions ,long non‐coding RNA ,Nucleic Acid Conformation ,RNA, Viral ,RNA, Long Noncoding ,030217 neurology & neurosurgery - Abstract
Most mosquito‐borne flaviviruses, including Zika virus (ZIKV), Dengue virus (DENV), and West Nile virus (WNV), produce long non‐coding subgenomic RNAs (sfRNAs) in infected cells that link to pathogenicity and immune evasion. Until now, the structural characterization of these lncRNAs remains limited. Here, we studied the 3D structures of individual and combined subdomains of sfRNAs, and visualized the accessible 3D conformational spaces of complete sfRNAs from DENV2, ZIKV, and WNV by small angle X‐ray scattering (SAXS) and computational modeling. The individual xrRNA1s and xrRNA2s adopt similar structures in solution as the crystal structure of ZIKV xrRNA1, and all xrRNA1‐2s form compact structures with reduced flexibility. While the DB12 of DENV2 is extended, the DB12s of ZIKV and WNV are compact due to the formation of intertwined double pseudoknots. All 3′ stem‐loops (3′SLs) share similar rod‐like structures. Complete sfRNAs are extended and sample a large conformational space in solution. Our work not only provides structural insight into the function of flavivirus sfRNAs, but also highlights strategies of visualizing other lncRNAs in solution by SAXS and computational methods.
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.