153 results on '"Kin-Hang Kok"'
Search Results
2. The significance of recurrent de novo amino acid substitutions that emerged during chronic SARS-CoV-2 infection: an observational studyResearch in context
- Author
-
Jonathan Daniel Ip, Wing-Ming Chu, Wan-Mui Chan, Allen Wing-Ho Chu, Rhoda Cheuk-Ying Leung, Qi Peng, Anthony Raymond Tam, Brian Pui-Chun Chan, Jian-Piao Cai, Kwok-Yung Yuen, Kin-Hang Kok, Yi Shi, Ivan Fan-Ngai Hung, and Kelvin Kai-Wang To
- Subjects
COVID-19 ,SARS-CoV-2 ,Chronic infection ,EG.5 ,BA.2.86 ,JN.1 ,Medicine ,Medicine (General) ,R5-920 - Abstract
Summary: Background: De novo amino acid substitutions (DNS) frequently emerge among immunocompromised patients with chronic SARS-CoV-2 infection. While previous studies have reported these DNS, their significance has not been systematically studied. Methods: We performed a review of DNS that emerged during chronic SARS-CoV-2 infection. We searched PubMed until June 2023 using the keywords “(SARS-CoV-2 or COVID-19) and (mutation or sequencing) and ((prolonged infection) or (chronic infection) or (long term))”. We included patients with chronic SARS-CoV-2 infection who had SARS-CoV-2 sequencing performed for at least 3 time points over at least 60 days. We also included 4 additional SARS-CoV-2 patients with chronic infection of our hospital not reported previously. We determined recurrent DNS that has appeared in multiple patients and determined the significance of these mutations among epidemiologically-significant variants. Findings: A total of 34 cases were analyzed, including 30 that were published previously and 4 from our hospital. Twenty two DNS appeared in ≥3 patients, with 14 (64%) belonging to lineage-defining mutations (LDMs) of epidemiologically-significant variants and 10 (45%) emerging among chronically-infected patients before the appearance of the corresponding variant. Notably, nsp9-T35I substitution (Orf1a T4175I) emerged in all three patients with BA.2.2 infection in 2022 before the appearance of Variants of Interest that carry nsp9-T35I as LDM (EG.5 and BA.2.86/JN.1). Structural analysis suggests that nsp9-T35I substitution may affect nsp9-nsp12 interaction, which could be critical for the function of the replication and transcription complex. Interpretation: DNS that emerges recurrently in different chronically-infected patients may be used as a marker for potential epidemiologically-significant variants. Funding: Theme-Based Research Scheme [T11/709/21-N] of the Research Grants Council (See acknowledgements for full list).
- Published
- 2024
- Full Text
- View/download PDF
3. An interferon-integrated mucosal vaccine provides pan-sarbecovirus protection in small animal models
- Author
-
Chun-Kit Yuen, Wan-Man Wong, Long-Fung Mak, Joy-Yan Lam, Lok-Yi Cheung, Derek Tsz-Yin Cheung, Yau-Yee Ng, Andrew Chak-Yiu Lee, Nanshan Zhong, Kwok-Yung Yuen, and Kin-Hang Kok
- Subjects
Science - Abstract
Abstract A pan-sarbecovirus or pan-betacoronavirus vaccine that can prevent current and potential future beta-coronavirus infections is important for fighting possible future pandemics. Here, we report a mucosal vaccine that cross-protects small animal models from sarbecoviruses including SARS-CoV-1, SARS-CoV-2 and its variants. The vaccine comprises a live-but-defective SARS-CoV-2 virus that is envelope deficient and has the orf8 segment replaced by interferon-beta, hence named Interferon Beta Integrated SARS-CoV-2 (IBIS) vaccine. Nasal vaccination with IBIS protected mice from lethal homotypic SARS-CoV-2 infection and hamsters from co-housing-mediated transmission of homotypic virus. Moreover, IBIS provided complete protection against heterotypic sarbecoviruses, including SARS-CoV-2 Delta and Omicron variants, and SARS-CoV-1 in both mice and hamsters. Besides inducing a strong lung CD8 + T cell response, IBIS specifically heightened the activation of mucosal virus-specific CD4 + T cells compared to the interferon-null vaccine. The direct production of interferon by IBIS also suppressed virus co-infection of SARS-CoV-2 in human cells, reducing the risk of genetic recombination when using as live vaccines. Altogether, IBIS is a next-generation pan-sarbecovirus vaccine and warrants further clinical investigations.
- Published
- 2023
- Full Text
- View/download PDF
4. Differential innate immune responses of human macrophages and bronchial epithelial cells against Talaromyces marneffei
- Author
-
Yen-Pei Tan, Chi-Ching Tsang, Ka-Fai Chan, Siu-Leung Fung, Kin-Hang Kok, Susanna K. P. Lau, and Patrick C. Y. Woo
- Subjects
Talaromyces marneffei ,human peripheral blood-derived macrophages ,RNA sequencing ,bronchial epithelial cells ,Microbiology ,QR1-502 - Abstract
ABSTRACT Talaromyces marneffei is a thermally dimorphic fungal pathogen endemic in Southeast Asia. As inhalation of airborne conidia is believed as the major infection route, airway epithelial cells followed by pulmonary macrophages are the first cell types which the fungus encounters inside the host. In this study, we established an in vitro infection model based on human peripheral blood-derived macrophages (hPBDMs) cultured with the supplementation of autologous plasma. Using this model, we determined the transcriptomic changes of hPBDMs in response to T. marneffei infection by quantitative real-time reverse-transcription polymerase chain reaction as well as high-throughput RNA sequencing. Results showed that T. marneffei infection could activate hPBDMs to the M1-like phenotype and trigger a potent induction of chemokine and pro-inflammatory cytokine production as well as the expression of other immunoregulatory genes. In contrast to hPBDMs, there was no detectable innate cytokine response against T. marneffei in human bronchial epithelial cells (hBECs). Using a green fluorescent protein-tagged T. marneffei strain and confocal microscopy, internalization of the fungus by hBECs was confirmed. Live cell imaging further demonstrated that the infected cells exhibited normal cellular physiology, especially that the process of cell division could be observed. Moreover, T. marneffei also survived better inside hBECs than hPBDMs. Our results illustrated a potential role of hBECs to serve as reservoir cells for T. marneffei to evade immunosurveillance by phagocytes, from which the fungus reactivates when the host immunity is weakened and causes infection. Such immunoevasion and reactivation may also help explain the long incubation period observed for talaromycosis, in particular the travel-related cases. IMPORTANCE Talaromyces marneffei is an important fungal pathogen especially in Southeast Asia. To understand the innate immune response to talaromycosis, a suitable infection model is needed. Here, we established an in vitro T. marneffei infection model using human peripheral blood-derived macrophages (hPBDMs). We then examined the transcriptomic changes of hPBDMs in response to T. marneffei infection with this model. We found that contact with T. marneffei could activate hPBDMs to the M1-like phenotype and induced mRNA expressions of five cytokines and eight immunoregulatory genes. Contrary to hPBDMs, such immunoresponse was not elicited in human bronchial epithelial cells (hBECs), despite normal physiology observed in infected cells. We also found that infected hBECs did not eliminate T. marneffei as efficiently as hPBDMs. Our observation suggested that hBECs may potentially serve as reservoir cells for T. marneffei to evade immunosurveillance. When the host immunity deteriorates later, then the fungus reactivates and causes infection.
- Published
- 2023
- Full Text
- View/download PDF
5. SARS-CoV-2 IgG seropositivity after the severe Omicron wave of COVID-19 in Hong Kong
- Author
-
Rosana Wing-Shan Poon, Brian Pui-Chun Chan, Wan-Mui Chan, Carol Ho-Yan Fong, Xiaojuan Zhang, Lu Lu, Lin-Lei Chen, Joy-Yan Lam, Vincent Chi-Chung Cheng, Samson S. Y. Wong, Kin-Hang Kok, Kwok-Yung Yuen, and Kelvin Kai-Wang To
- Subjects
COVID-19 ,SARS-CoV-2 ,serosurveillance ,receptor binding domain ,nucleoprotein ,ORF8 ,Infectious and parasitic diseases ,RC109-216 ,Microbiology ,QR1-502 - Abstract
The SARS-CoV-2 Omicron variant has led to a major wave of COVID-19 in Hong Kong between January and May 2022. Here, we used seroprevalence to estimate the combined incidence of vaccination and SARS-CoV-2 infection, including subclinical infection which were not diagnosed at the acute stage. The overall seropositive rate of IgG against receptor binding domain (anti-RBD IgG) increased from 52.2% in December 2021 to 89.3% in May 2022. The level of anti-RBD IgG was lowest in the 0–9 and ≥80 year-old age groups in May 2022. The seropositive rate of antibody against ORF8, which reflects the rate of prior infection, was 23.4% in May 2022. Our data suggest that although most individuals were either vaccinated or infected after the fifth wave, children and older adults remain most vulnerable. Public health measures should target these age groups in order to ameliorate the healthcare consequences of upcoming waves.
- Published
- 2022
- Full Text
- View/download PDF
6. A nasal omicron vaccine booster elicits potent neutralizing antibody response against emerging SARS-CoV-2 variants
- Author
-
Joy-Yan Lam, Yau-Yee Ng, Chun-Kit Yuen, Wan-Man Wong, Kwok-Yung Yuen, and Kin-Hang Kok
- Subjects
COVID-19 ,SARS-CoV-2 ,nasal protein vaccine booster ,delta variant ,omicron variant ,Infectious and parasitic diseases ,RC109-216 ,Microbiology ,QR1-502 - Abstract
SARS-CoV-2 has caused the COVID-19 pandemic since early 2020. As of January 2022, the worldwide spreading of SARS-CoV-2 leads to approximately 0.35 billion of human infections and five millions of deaths. Current vaccination is one of the effective ways to control SARS-CoV-2 transmission and reduce the disease severity. However, the antibody level against the immunogen significantly drops several months after the standard two-dose vaccination, and hence a third or fourth dose booster (the same immunogen) has been suggested to boost the antibody response. Here, we described an ultra-effective nasal vaccine booster that potently induced the extraordinary high-level of neutralizing antibody in pre-vaccinated mice. The vaccine booster is composed of a recombinant receptor binding domain of SARS-CoV-2 spike (either wild-type or omicron) fused with a domain of SARS-CoV-2 nucleoprotein. In the absence of adjuvants, a single intranasal administration of the booster in pre-vaccinated mice significantly induced systemic and mucosal antibody responses as evidenced by the elevation of the cross-variant neutralizing antibody and induction of IgA in bronchoalveolar lavage respectively. Most importantly, the single dose nasal vaccine booster (omicron version) potently enhanced the neutralizing activity against authentic SARS-CoV-2 omicron virus infection. Taken together, the induction of respiratory mucosal immunity and the enhancement of cross-variant neutralizing activity by the nasal vaccine booster warrants further clinical trials in humans.
- Published
- 2022
- Full Text
- View/download PDF
7. An RNA-Scaffold Protein Subunit Vaccine for Nasal Immunization
- Author
-
Joy-Yan Lam, Wan-Man Wong, Chun-Kit Yuen, Yau-Yee Ng, Chun-Hin San, Kwok-Yung Yuen, and Kin-Hang Kok
- Subjects
protein subunit vaccine ,SARS-CoV-2 ,self-adjuvanted ,nasal vaccine ,RNA scaffold ,mucosal immunity ,Medicine - Abstract
Developing recombinant proteins as nasal vaccines for inducing systemic and mucosal immunity against respiratory viruses is promising. However, additional adjuvants are required to overcome the low immunogenicity of protein antigens. Here, a self-adjuvanted protein-RNA ribonucleoprotein vaccine was developed and found to be an effective nasal vaccine in mice and the SARS-CoV-2 infection model. The vaccine consisted of spike RBD (as an antigen), nucleoprotein (as an adaptor), and ssRNA (as an adjuvant and RNA scaffold). This combination robustly induced mucosal IgA, neutralizing antibodies and activated multifunctional T-cells, while also providing sterilizing immunity against live virus challenge. In addition, high-resolution scRNA-seq analysis highlighted airway-resident immune cells profile during prime-boost immunization. The vaccine also possesses modularity (antigen/adaptor/RNA scaffold) and can be made to target other viruses. This protein-RNA ribonucleoprotein vaccine is a novel and promising approach for developing safe and potent nasal vaccines to combat respiratory virus infections.
- Published
- 2023
- Full Text
- View/download PDF
8. Cocirculation of two SARS-CoV-2 variant strains within imported pet hamsters in Hong Kong
- Author
-
Kin-Hang Kok, Shuk-Ching Wong, Wan-Mui Chan, Wen Lei, Allen Wing-Ho Chu, Jonathan Daniel Ip, Lam-Kwong Lee, Ivan Tak-Fai Wong, Hazel Wing-Hei Lo, Vincent Chi-Chung Cheng, Alex Yat-Man Ho, Bosco Hoi-Shiu Lam, Herman Tse, David Lung, Kenneth Ho-Leung Ng, Albert Ka-Wing Au, Gilman Kit-Hang Siu, and Kwok-Yung Yuen
- Subjects
animal ,coronavirus ,hamster ,interspecies ,transmission ,sars-cov-2 ,Infectious and parasitic diseases ,RC109-216 ,Microbiology ,QR1-502 - Abstract
During the investigation of a pet shop outbreak of severe acute respiratory coronavirus 2 (SARS-CoV-2) with probable hamster-to-human transmission, the environmental and hamster samples in epidemiologically linked pet shops were found positive for SARS-CoV-2 Delta variant AY.127 strains which are phylogenetically closely related to patient and reported European strains. This interspecies spill-over has triggered transmission chains involving 58 patients epidemiologically linked to three pet shops. Incidentally, three dwarf hamsters imported from Netherlands and centralized in a warehouse distributing animals to pet shops were positive for SARS-CoV-2 spike variant phylogenetically related to European B.1.258 strains since March 2020. This B.1.258 strain has almost disappeared since July 2021. While no hamster-to-human transmission of B.1.258-like strain was found in this outbreak, molecular docking showed that its spike receptor binding domain (RBD) has similar binding energy to human ACE2 when compared with that of Delta variant AY.127. Therefore, the potential of this B.1.258-related spike variant for interspecies jumping cannot be ignored. The co-circulation of both B.1.258-related spike variants with Delta AY.127 which originated in Europe and was not previously found in Hong Kong suggested that hamsters in our wholesale warehouse and retail pet shops more likely have acquired these viruses in Netherlands or stopovers during delivery by aviation than locally. The risk of human-to-hamster reverse zoonosis by multiple SARS-CoV-2 variants leading to further adaptive spike mutations with subsequent transmission back to human cannot be underestimated as outbreak source of COVID-19. Testing of imported pet animals susceptible to SARS-CoV-2 is warranted to prevent future outbreaks.
- Published
- 2022
- Full Text
- View/download PDF
9. A platform technology for generating subunit vaccines against diverse viral pathogens
- Author
-
Andrew Young, Ariel Isaacs, Connor A. P. Scott, Naphak Modhiran, Christopher L. D. McMillan, Stacey T. M. Cheung, Jennifer Barr, Glenn Marsh, Nazia Thakur, Dalan Bailey, Kenneth S. M. Li, Hayes K. H. Luk, Kin-Hang Kok, Susanna K. P. Lau, Patrick C. Y. Woo, Wakako Furuyama, Andrea Marzi, Paul R. Young, Keith J. Chappell, and Daniel Watterson
- Subjects
subunit ,platform ,viral ,fusion ,clamp ,vaccine ,Immunologic diseases. Allergy ,RC581-607 - Abstract
The COVID-19 pandemic response has shown how vaccine platform technologies can be used to rapidly and effectively counteract a novel emerging infectious disease. The speed of development for mRNA and vector-based vaccines outpaced those of subunit vaccines, however, subunit vaccines can offer advantages in terms of safety and stability. Here we describe a subunit vaccine platform technology, the molecular clamp, in application to four viruses from divergent taxonomic families: Middle Eastern respiratory syndrome coronavirus (MERS-CoV), Ebola virus (EBOV), Lassa virus (LASV) and Nipah virus (NiV). The clamp streamlines subunit antigen production by both stabilising the immunologically important prefusion epitopes of trimeric viral fusion proteins while enabling purification without target-specific reagents by acting as an affinity tag. Conformations for each viral antigen were confirmed by monoclonal antibody binding, size exclusion chromatography and electron microscopy. Notably, all four antigens tested remained stable over four weeks of incubation at 40°C. Of the four vaccines tested, a neutralising immune response was stimulated by clamp stabilised MERS-CoV spike, EBOV glycoprotein and NiV fusion protein. Only the clamp stabilised LASV glycoprotein precursor failed to elicit virus neutralising antibodies. MERS-CoV and EBOV vaccine candidates were both tested in animal models and found to provide protection against viral challenge.
- Published
- 2022
- Full Text
- View/download PDF
10. Host-derived lipids orchestrate pulmonary γδ T cell response to provide early protection against influenza virus infection
- Author
-
Xiaohui Wang, Xiang Lin, Zihan Zheng, Bingtai Lu, Jun Wang, Andy Hee-Meng Tan, Meng Zhao, Jia Tong Loh, Sze Wai Ng, Qian Chen, Fan Xiao, Enyu Huang, King-Hung Ko, Zhong Huang, Jingyi Li, Kin-Hang Kok, Gen Lu, Xiaohui Liu, Kong-Peng Lam, Wanli Liu, Yuxia Zhang, Kwok-Yung Yuen, Tak Wah Mak, and Liwei Lu
- Subjects
Science - Abstract
Influenza A infection results in γδ T cell influx and production of IL-17 in the lungs. Here, the authors show that this effect is primed by CD1-restricted ligands that are released by infected cells and presented by B1a cells in the lungs.
- Published
- 2021
- Full Text
- View/download PDF
11. Intranasal Boosting with Spike Fc-RBD of Wild-Type SARS-CoV-2 Induces Neutralizing Antibodies against Omicron Subvariants and Reduces Viral Load in the Nasal Turbinate of Mice
- Author
-
Jian-Piao Cai, Cuiting Luo, Kun Wang, Hehe Cao, Lin-Lei Chen, Xiaojuan Zhang, Yuting Han, Feifei Yin, Anna Jinxia Zhang, Hin Chu, Shuofeng Yuan, Kin-Hang Kok, Kelvin Kai-Wang To, Honglin Chen, Zhiwei Chen, Dong-Yan Jin, Kwok-Yung Yuen, and Jasper Fuk-Woo Chan
- Subjects
COVID-19 ,SARS-CoV-2 ,Omicron variant ,BA.5.2 ,XBB.1 ,Fc-RBD ,Microbiology ,QR1-502 - Abstract
The emergence of new immune-evasive severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants and subvariants outpaces the development of vaccines specific against the dominant circulating strains. In terms of the only accepted immune correlate of protection, the inactivated whole-virion vaccine using wild-type SARS-CoV-2 spike induces a much lower serum neutralizing antibody titre against the Omicron subvariants. Since the inactivated vaccine given intramuscularly is one of the most commonly used coronavirus disease 2019 (COVID-19) vaccines in developing regions, we tested the hypothesis that intranasal boosting after intramuscular priming would provide a broader level of protection. Here, we showed that one or two intranasal boosts with the Fc-linked trimeric spike receptor-binding domain from wild-type SARS-CoV-2 can induce significantly higher serum neutralizing antibodies against wild-type SARS-CoV-2 and the Omicron subvariants, including BA.5.2 and XBB.1, with a lower titre in the bronchoalveolar lavage of vaccinated Balb/c mice than vaccination with four intramuscular doses of inactivated whole virion vaccine. The intranasally vaccinated K18-hACE2-transgenic mice also had a significantly lower nasal turbinate viral load, suggesting a better protection of the upper airway, which is the predilected site of infection by Omicron subvariants. This intramuscular priming and intranasal boosting approach that achieves broader cross-protection against Omicron variants and subvariants may lengthen the interval required for changing the vaccine immunogen from months to years.
- Published
- 2023
- Full Text
- View/download PDF
12. Isolation of MERS-related coronavirus from lesser bamboo bats that uses DPP4 and infects human-DPP4-transgenic mice
- Author
-
Susanna K. P. Lau, Rachel Y. Y. Fan, Longchao Zhu, Kenneth S. M. Li, Antonio C. P. Wong, Hayes K. H. Luk, Emily Y. M. Wong, Carol S. F. Lam, George C. S. Lo, Joshua Fung, Zirong He, Felix C. H. Fok, Rex K. H. Au-Yeung, Libiao Zhang, Kin-Hang Kok, Kwok-Yung Yuen, and Patrick C. Y. Woo
- Subjects
Science - Abstract
Several human coronaviruses (CoV) have been proposed to emerge from bats but evidence of direct bat-to-human transmission is slim. In this work, the authors isolate a MERS-related CoV strain directly from bats and show that it infects target cells in vitro and engineered mice through the human DDP4 receptor.
- Published
- 2021
- Full Text
- View/download PDF
13. Host and viral determinants for efficient SARS-CoV-2 infection of the human lung
- Author
-
Hin Chu, Bingjie Hu, Xiner Huang, Yue Chai, Dongyan Zhou, Yixin Wang, Huiping Shuai, Dong Yang, Yuxin Hou, Xi Zhang, Terrence Tsz-Tai Yuen, Jian-Piao Cai, Anna Jinxia Zhang, Jie Zhou, Shuofeng Yuan, Kelvin Kai-Wang To, Ivy Hau-Yee Chan, Ko-Yung Sit, Dominic Chi-Chung Foo, Ian Yu-Hong Wong, Ada Tsui-Lin Ng, Tan To Cheung, Simon Ying-Kit Law, Wing-Kuk Au, Melinda A. Brindley, Zhiwei Chen, Kin-Hang Kok, Jasper Fuk-Woo Chan, and Kwok-Yung Yuen
- Subjects
Science - Abstract
Here, using lung epithelial cells and ex vivo tissue explants, the authors show that, in addition to ACE2, host heparan sulfate is directly involved in SARS-CoV-2 attachment and entry and provide data suggesting that host sialic acids may act as viral restriction factor in lung tissues.
- Published
- 2021
- Full Text
- View/download PDF
14. Mining of linear B cell epitopes of SARS-CoV-2 ORF8 protein from COVID-19 patients
- Author
-
Xiaohui Wang, Joy-Yan Lam, Linlei Chen, Shannon Wing-Ngor Au, Kelvin K. W. To, Kwok-Yung Yuen, and Kin-Hang Kok
- Subjects
SARS-CoV-2 ,ORF8 ,COVID-19 ,peptide ,epitope ,antibody ,Infectious and parasitic diseases ,RC109-216 ,Microbiology ,QR1-502 - Abstract
Given the on-going SARS-CoV-2 pandemic, identification of immunogenic targets against the viral protein will provide crucial advances towards the development of sensitive diagnostic tools and vaccination strategies. Our previous study has found that ORF8 protein of SARS-CoV-2 is highly immunogenic and shows high sensitivity in identifying COVID-19 disease. In this study, by employing overlapping linear peptides, we characterized the IgG immunodominant regions on SARS-CoV-2 ORF8 protein that are seropositive in the sera from SARS-CoV-2-infected patients. The major immunogenic epitopes are localized at (1) N-termini alpha helix, (2) the resides spanning beta 2 and 3 sheets, and (3) the loop between beta 4 and 5 sheets. Additionally, hamster model infected by SARS-CoV-2 further validates the seropositivity of the linear epitopes in vivo, demonstrating a potential application of the linear peptide-based immunization strategy. Taken together, identification and validation of these B-cell linear epitopes will provide insights into the design of serological diagnostics and peptide-based vaccination approach against this pandemic virus of high priority.
- Published
- 2021
- Full Text
- View/download PDF
15. Loss of orf3b in the circulating SARS-CoV-2 strains
- Author
-
Joy-Yan Lam, Chun-Kit Yuen, Jonathan Daniel Ip, Wan-Man Wong, Kelvin Kai-Wang To, Kwok-Yung Yuen, and Kin-Hang Kok
- Subjects
SARS-CoV-2 ,orf3b ,orf3a ,Q57H ,D614G ,Infectious and parasitic diseases ,RC109-216 ,Microbiology ,QR1-502 - Abstract
ABSTRACTThe newly emerged betacoronavirus, SARS-CoV-2, causes the COVID-19 pandemic since December 2019 with more than 35 million laboratory confirmed human infections and over one million deaths within nine months. The genome of SARS-CoV-2 continues to evolve during the global transmission with the notable emergence of the spike D614G substitution that enhances infectivity. Some of these viral adaptations may alter not only the infectivity but also viral pathogenesis. Continuous phylogenomic analysis of circulating viral strains and functional investigation of new non-synonymous substitutions may help to understand the evolution of virus, its virulence and transmissibility. Here we describe a loss of an accessory protein orf3b (57 amino acids) in current circulating SARS-CoV-2 strains, contributing around 24% of more than 100,000 complete viral genomes analysed. The loss of 3b is caused by the presence of an early stop codon which is created by an orf3a Q57H substitution. There is an increasing trend in the loss of orf3b which has reached 32% in May 2020. Geographically, loss of 3b is more prevalent in certain countries including Colombia (46%), USA (48%), South Korea (51%), France (66%), Saudi Arabia (72%), Finland (76%) and Egypt (77%). Interestingly, the loss of 3b coincides with the emergence of spike D614G substitution. In addition, we found that truncated orf3b has lost the interferon antagonism compared to the full-length orf3b, suggesting a loss of function by the newly adapted virus. Further investigation of orf3b deletion and spike D614G substitution on virulence and infectivity respectively will provide important insights into SARS-CoV-2 evolution.
- Published
- 2020
- Full Text
- View/download PDF
16. SARS-CoV-2 nsp13, nsp14, nsp15 and orf6 function as potent interferon antagonists
- Author
-
Chun-Kit Yuen, Joy-Yan Lam, Wan-Man Wong, Long-Fung Mak, Xiaohui Wang, Hin Chu, Jian-Piao Cai, Dong-Yan Jin, Kelvin Kai-Wang To, Jasper Fuk-Woo Chan, Kwok-Yung Yuen, and Kin-Hang Kok
- Subjects
COVID-19 ,SARS-CoV-2 ,interferon antagonist ,PLpro ,orf6 ,Infectious and parasitic diseases ,RC109-216 ,Microbiology ,QR1-502 - Abstract
ABSTRACTThe Coronavirus disease 2019 (COVID-19), which is caused by the novel SARS-CoV-2 virus, is now causing a tremendous global health concern. Since its first appearance in December 2019, the outbreak has already caused over 5.8 million infections worldwide (till 29 May 2020), with more than 0.35 million deaths. Early virus-mediated immune suppression is believed to be one of the unique characteristics of SARS-CoV-2 infection and contributes at least partially to the viral pathogenesis. In this study, we identified the key viral interferon antagonists of SARS-CoV-2 and compared them with two well-characterized SARS-CoV interferon antagonists, PLpro and orf6. Here we demonstrated that the SARS-CoV-2 nsp13, nsp14, nsp15 and orf6, but not the unique orf8, could potently suppress primary interferon production and interferon signalling. Although SARS-CoV PLpro has been well-characterized for its potent interferon-antagonizing, deubiquitinase and protease activities, SARS-CoV-2 PLpro, despite sharing high amino acid sequence similarity with SARS-CoV, loses both interferon-antagonising and deubiquitinase activities. Among the 27 viral proteins, SARS-CoV-2 orf6 demonstrated the strongest suppression on both primary interferon production and interferon signalling. Orf6-deleted SARS-CoV-2 may be considered for the development of intranasal live-but-attenuated vaccine against COVID-19.
- Published
- 2020
- Full Text
- View/download PDF
17. Genomic characterization of the 2019 novel human-pathogenic coronavirus isolated from a patient with atypical pneumonia after visiting Wuhan
- Author
-
Jasper Fuk-Woo Chan, Kin-Hang Kok, Zheng Zhu, Hin Chu, Kelvin Kai-Wang To, Shuofeng Yuan, and Kwok-Yung Yuen
- Subjects
Coronavirus ,Wuhan ,SARS ,emerging ,genome ,respiratory ,Infectious and parasitic diseases ,RC109-216 ,Microbiology ,QR1-502 - Abstract
ABSTRACTA mysterious outbreak of atypical pneumonia in late 2019 was traced to a seafood wholesale market in Wuhan of China. Within a few weeks, a novel coronavirus tentatively named as 2019 novel coronavirus (2019-nCoV) was announced by the World Health Organization. We performed bioinformatics analysis on a virus genome from a patient with 2019-nCoV infection and compared it with other related coronavirus genomes. Overall, the genome of 2019-nCoV has 89% nucleotide identity with bat SARS-like-CoVZXC21 and 82% with that of human SARS-CoV. The phylogenetic trees of their orf1a/b, Spike, Envelope, Membrane and Nucleoprotein also clustered closely with those of the bat, civet and human SARS coronaviruses. However, the external subdomain of Spike’s receptor binding domain of 2019-nCoV shares only 40% amino acid identity with other SARS-related coronaviruses. Remarkably, its orf3b encodes a completely novel short protein. Furthermore, its new orf8 likely encodes a secreted protein with an alpha-helix, following with a beta-sheet(s) containing six strands. Learning from the roles of civet in SARS and camel in MERS, hunting for the animal source of 2019-nCoV and its more ancestral virus would be important for understanding the origin and evolution of this novel lineage B betacoronavirus. These findings provide the basis for starting further studies on the pathogenesis, and optimizing the design of diagnostic, antiviral and vaccination strategies for this emerging infection.
- Published
- 2020
- Full Text
- View/download PDF
18. SMRT sequencing revealed the diversity and characteristics of defective interfering RNAs in influenza A (H7N9) virus infection
- Author
-
Wing-Yu Lui, Chun-Kit Yuen, Can Li, Wan Man Wong, Pak-Yin Lui, Chi-Ho Lin, Kwok-Hung Chan, Hanjun Zhao, Honglin Chen, Kelvin K. W. To, Anna J. X Zhang, Kwok-Yung Yuen, and Kin-Hang Kok
- Subjects
Avian influenza A/H7N9 virus ,defective interfering viral genome ,Single Molecule Real Time sequencing ,Illumina sequencing ,Infectious and parasitic diseases ,RC109-216 ,Microbiology ,QR1-502 - Abstract
ABSTRACTInfluenza defective interfering (DI) particles are replication-incompetent viruses carrying large internal deletion in the genome. The loss of essential genetic information causes abortive viral replication, which can be rescued by co-infection with a helper virus that possesses an intact genome. Despite reports of DI particles present in seasonal influenza A H1N1 infections, their existence in human infections by the avian influenza A viruses, such as H7N9, has not been studied. Here we report the ubiquitous presence of DI-RNAs in nasopharyngeal aspirates of H7N9-infected patients. Single Molecule Real Time (SMRT) sequencing was first applied and long-read sequencing analysis showed that a variety of H7N9 DI-RNA species were present in the patient samples and human bronchial epithelial cells. In several abundantly expressed DI-RNA species, long overlapping sequences have been identified around at the breakpoint region and the other side of deleted region. Influenza DI-RNA is known as a defective viral RNA with single large internal deletion. Beneficial to the long-read property of SMRT sequencing, double and triple internal deletions were identified in half of the DI-RNA species. In addition, we examined the expression of DI-RNAs in mice infected with sublethal dose of H7N9 virus at different time points. Interestingly, DI-RNAs were abundantly expressed as early as day 2 post-infection. Taken together, we reveal the diversity and characteristics of DI-RNAs found in H7N9-infected patients, cells and animals. Further investigations on this overwhelming generation of DI-RNA may provide important insights into the understanding of H7N9 viral replication and pathogenesis.
- Published
- 2019
- Full Text
- View/download PDF
19. SREBP-dependent lipidomic reprogramming as a broad-spectrum antiviral target
- Author
-
Shuofeng Yuan, Hin Chu, Jasper Fuk-Woo Chan, Zi-Wei Ye, Lei Wen, Bingpeng Yan, Pok-Man Lai, Kah-Meng Tee, Jingjing Huang, Dongdong Chen, Cun Li, Xiaoyu Zhao, Dong Yang, Man Chun Chiu, Cyril Yip, Vincent Kwok-Man Poon, Chris Chung-Sing Chan, Kong-Hung Sze, Jie Zhou, Ivy Hau-Yee Chan, Kin-Hang Kok, Kelvin Kai-Wang To, Richard Yi-Tsun Kao, Johnson Yiu-Nam Lau, Dong-Yan Jin, Stanley Perlman, and Kwok-Yung Yuen
- Subjects
Science - Abstract
Viruses rely on host cell metabolism for replication, making these pathways potential therapeutic targets. Here, the authors show that AM580, a retinoid derivative and RAR-α agonist, affects replication of several RNA viruses by interfering with the activity of SREBP.
- Published
- 2019
- Full Text
- View/download PDF
20. Accurate Diagnosis of COVID-19 by a Novel Immunogenic Secreted SARS-CoV-2 orf8 Protein
- Author
-
Xiaohui Wang, Joy-Yan Lam, Wan-Man Wong, Chun-Kit Yuen, Jian-Piao Cai, Shannon Wing-Ngor Au, Jasper Fuk-Woo Chan, Kelvin K. W. To, Kin-Hang Kok, and Kwok-Yung Yuen
- Subjects
COVID-19 ,diagnosis ,SARS-CoV-2 ,orf8 ,Microbiology ,QR1-502 - Abstract
ABSTRACT An accurate diagnostic test for early severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is the key weapon to control the coronavirus disease 2019 (COVID-19) pandemic. We previously reported that the SARS-CoV-2 genome contains a unique orf8 accessory gene absent from other human-pathogenic coronaviruses. Here, we characterized the SARS-CoV-2 orf8 as a novel immunogenic secreted protein and utilized it for the accurate diagnosis of COVID-19. Extracellular orf8 protein was detected in cell culture supernatant and in sera of COVID-19 patients. In addition, orf8 was found highly immunogenic in COVID-19 patients, who showed early seropositivity for anti-orf8 IgM, IgG, and IgA. We hypothesize that orf8 secretion during SARS-CoV-2 infection facilitates early mounting of B cell response. The serological test detecting anti-orf8 IgG antibody can be used for the early and accurate diagnosis of COVID-19. IMPORTANCE Current commercially available serological tests for COVID-19 patients are detecting antibodies against SARS-CoV-2 nucleoprotein and spike glycoprotein. The antinucleoprotein and antispike antibodies can be accurately detected in patients during the mid or late stage of infection, and therefore, these assays have not been widely used for early diagnosis of COVID-19. In this study, we characterized the secretory property of a SARS-CoV-2 orf8 protein and proposed that orf8 secretion during infection facilitates early mounting of the B cell response. We demonstrated the presence of anti-orf8 antibodies in both symptomatic and asymptomatic patients during the early stage of infection, while the anti-N antibody is not detected. Our serological test detecting anti-orf8 antibodies may facilitate the development of early and accurate diagnosis for COVID-19.
- Published
- 2020
- Full Text
- View/download PDF
21. Seroprevalence of SARS-CoV-2 in Hong Kong and in residents evacuated from Hubei province, China: a multicohort study
- Author
-
Kelvin Kai-Wang To, MD, Vincent Chi-Chung Cheng, MD, Jian-Piao Cai, BSc, Kwok-Hung Chan, PhD, Lin-Lei Chen, MPhil, Lok-Hin Wong, MPhil, Charlotte Yee-Ki Choi, BSc, Carol Ho-Yan Fong, PhD, Anthony Chin-Ki Ng, BSc, Lu Lu, BSc, Cui-Ting Luo, BSc, Jianwen Situ, MPhil, Tom Wai-Hin Chung, MRCP, Shuk-Ching Wong, MNurs, Grace See-Wai Kwan, MMedSc, Siddharth Sridhar, FRCPath, Jasper Fuk-Woo Chan, MD, Cecilia Yuen-Man Fan, FRACGP, Vivien W M Chuang, FRCPath, Kin-Hang Kok, PhD, Ivan Fan-Ngai Hung, ProfMD, and Kwok-Yung Yuen, ProfMD
- Subjects
Medicine (General) ,R5-920 ,Microbiology ,QR1-502 - Abstract
Summary: Background: The role of subclinical severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections in perpetuating the COVID-19 pandemic is unknown because population seroprevalence data are absent. We aimed to establish the sensitivity and specificity of our enzyme immunoassay and microneutralisation assay, and the seroprevalence of SARS-CoV-2 in Hong Kong before and after the pandemic, as well as in Hong Kong residents evacuated from Hubei province, China. Methods: We did a multicohort study in a hospital and university in Hong Kong. We evaluated the sensitivity of our enzyme immunoassay and microneutralisation assay with RT-PCR data from patients positive for SARS-CoV-2 and the specificity of our enzyme immunoassay and microneutralisation assay with archived serum samples collected before 2019. We compared the seropositivity of the general population of Hong Kong before and after the pandemic had begun, and determined the seropositivity of Hong Kong residents evacuated from Hubei province, China, in March, 2020. Findings: Between Feb 26 and March 18, 2020, we assessed RT-PCR samples from 45 patients who had recovered from COVID-19 to establish the sensitivity of our enzyme immunoassay and microneutralisation assay. To establish the specificity of these assays, we retrieved archived serum. The sensitivity was 91·1% (41 of 45 [95% CI 78·8–97·5]) for the microneutralisation assay, 57·8% (26 of 45 [42·2–72·3]) for anti-nucleoprotein IgG, 66·7% (30 of 45 [51·1–80·0]) for anti-spike protein receptor binding domain (RBD) IgG, and 73·3% (33 of 45 [58·1–85·4]) for enzyme immunoassay (either positive for anti-nucleoprotein or anti-RBD IgG). The specificity was 100% (152 of 152 [95% CI 97·6–100·0]) for both the enzyme immunoassay and microneutralisation assay. Among the Hong Kong general population, 53 (2·7%) of 1938 were enzyme immunoassay positive, but of those who were positive, all 53 were microneutralisation negative, and no significant increase was seen in the seroprevalence between April 12, 2018, and Feb 13, 2020. Among asymptomatic Hubei returnees, 17 (4%) of 452 were seropositive with the enzyme immunoassay or the microneutralisation assay, with 15 (88%) of 17 seropositive with the microneutralisation assay, and two familial clusters were identified. Interpretation: Our serological data suggest that SARS-CoV-2 is a new emerging virus. The seropositivity rate in Hubei returnees indicates that RT-PCR-confirmed patients only represent a small proportion of the total number of cases. The low seroprevalence suggests that most of the Hong Kong and Hubei population remain susceptible to COVID-19. Future waves of the outbreak are inevitable without a vaccine or antiviral prophylaxis. The role of age-related cross reactive non-neutralising antibodies in the pathogenesis of COVID-19 warrants further investigation. Funding: Richard and Carol Yu, May Tam Mak Mei Yin, Shaw Foundation (Hong Kong), Michael Tong, Marina Lee, and the Government Consultancy Service (see acknowledgments for full list).
- Published
- 2020
- Full Text
- View/download PDF
22. Comparative tropism, replication kinetics, and cell damage profiling of SARS-CoV-2 and SARS-CoV with implications for clinical manifestations, transmissibility, and laboratory studies of COVID-19: an observational study
- Author
-
Hin Chu, PhD, Jasper Fuk-Woo Chan, MD, Terrence Tsz-Tai Yuen, BA, Huiping Shuai, PhD, Shuofeng Yuan, PhD, Yixin Wang, MPhil, Bingjie Hu, MPhil, Cyril Chik-Yan Yip, PhD, Jessica Oi-Ling Tsang, BSc, Xiner Huang, BSc, Yue Chai, MPhil, Dong Yang, MPhil, Yuxin Hou, MPhil, Kenn Ka-Heng Chik, MMedSc, Xi Zhang, BSc, Agnes Yim-Fong Fung, BSc, Hoi-Wah Tsoi, MPhil, Jian-Piao Cai, BSc, Wan-Mui Chan, PhD, Jonathan Daniel Ip, MSc, Allen Wing-Ho Chu, MSc, Jie Zhou, PhD, David Christopher Lung, FRCPath, Kin-Hang Kok, PhD, Kelvin Kai-Wang To, MD, Owen Tak-Yin Tsang, FRCP, Kwok-Hung Chan, PhD, and Kwok-Yung Yuen, ProfMD
- Subjects
Medicine (General) ,R5-920 ,Microbiology ,QR1-502 - Abstract
Summary: Background: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was reported from China in January, 2020. SARS-CoV-2 is efficiently transmitted from person to person and, in 2 months, has caused more than 82 000 laboratory-confirmed cases of coronavirus disease 2019 (COVID-19) and 2800 deaths in 46 countries. The total number of cases and deaths has surpassed that of the 2003 severe acute respiratory syndrome coronavirus (SARS-CoV). Although both COVID-19 and severe acute respiratory syndrome (SARS) manifest as pneumonia, COVID-19 is associated with apparently more efficient transmission, fewer cases of diarrhoea, increased mental confusion, and a lower crude fatality rate. However, the underlying virus–host interactive characteristics conferring these observations on transmissibility and clinical manifestations of COVID-19 remain unknown. Methods: We systematically investigated the cellular susceptibility, species tropism, replication kinetics, and cell damage of SARS-CoV-2 and compared findings with those for SARS-CoV. We compared SARS-CoV-2 and SARS-CoV replication in different cell lines with one-way ANOVA. For the area under the curve comparison between SARS-CoV-2 and SARS-CoV replication in Calu3 (pulmonary) and Caco2 (intestinal) cells, we used Student's t test. We analysed cell damage induced by SARS-CoV-2 and SARS-CoV with one-way ANOVA. Findings: SARS-CoV-2 infected and replicated to comparable levels in human Caco2 cells and Calu3 cells over a period of 120 h (p=0·52). By contrast, SARS-CoV infected and replicated more efficiently in Caco2 cells than in Calu3 cells under the same multiplicity of infection (p=0·0098). SARS-CoV-2, but not SARS-CoV, replicated modestly in U251 (neuronal) cells (p=0·036). For animal species cell tropism, both SARS-CoV and SARS-CoV-2 replicated in non-human primate, cat, rabbit, and pig cells. SARS-CoV, but not SARS-CoV-2, infected and replicated in Rhinolophus sinicus bat kidney cells. SARS-CoV-2 consistently induced significantly delayed and milder levels of cell damage than did SARS-CoV in non-human primate cells (VeroE6, p=0·016; FRhK4, p=0·0004). Interpretation: As far as we know, our study presents the first quantitative data for tropism, replication kinetics, and cell damage of SARS-CoV-2. These data provide novel insights into the lower incidence of diarrhoea, decreased disease severity, and reduced mortality in patients with COVID-19, with respect to the pathogenesis and high transmissibility of SARS-CoV-2 compared with SARS-CoV. Funding: May Tam Mak Mei Yin, The Shaw Foundation Hong Kong, Richard Yu and Carol Yu, Michael Seak-Kan Tong, Respiratory Viral Research Foundation, Hui Ming, Hui Hoy and Chow Sin Lan Charity Fund, Chan Yin Chuen Memorial Charitable Foundation, Marina Man-Wai Lee, The Hong Kong Hainan Commercial Association South China Microbiology Research Fund, The Jessie & George Ho Charitable Foundation, Perfect Shape Medical, The Consultancy Service for Enhancing Laboratory Surveillance of Emerging Infectious Diseases and Research Capability on Antimicrobial Resistance for the Department of Health of the Hong Kong Special Administrative Region Government, The Theme-Based Research Scheme of the Research Grants Council, Sanming Project of Medicine in Shenzhen, and The High Level-Hospital Program, Health Commission of Guangdong Province, China.
- Published
- 2020
- Full Text
- View/download PDF
23. Dual-functional peptide with defective interfering genes effectively protects mice against avian and seasonal influenza
- Author
-
Hanjun Zhao, Kelvin K. W. To, Hin Chu, Qiulu Ding, Xiaoyu Zhao, Cun Li, Huiping Shuai, Shuofeng Yuan, Jie Zhou, Kin-Hang Kok, Shibo Jiang, and Kwok-Yung Yuen
- Subjects
Science - Abstract
A limited number of therapeutics is available to treat influenza A virus (IAV) infections. Here, the authors show that defective interfering genes, delivered with a dual-functional peptide that enables intracellular accumulation and prevents endosomal acidification, inhibit IAV replication in vitro and in vivo.
- Published
- 2018
- Full Text
- View/download PDF
24. Repurposing of Miltefosine as an Adjuvant for Influenza Vaccine
- Author
-
Lu Lu, Carol Ho-Yan Fong, Anna Jinxia Zhang, Wai-Lan Wu, Iris Can Li, Andrew Chak-Yiu Lee, Thrimendra Kaushika Dissanayake, Linlei Chen, Ivan Fan-Ngai Hung, Kwok-Hung Chan, Hin Chu, Kin-Hang Kok, Kwok-Yung Yuen, and Kelvin Kai-Wang To
- Subjects
influenza vaccine ,vaccine ,adjuvant ,TFH ,miltefosine ,Medicine - Abstract
We previously reported that topical imiquimod can improve the immunogenicity of the influenza vaccine. This study investigated another FDA-approved drug, miltefosine (MTF), as a vaccine adjuvant. Mice immunized with an influenza vaccine with or without MTF adjuvant were challenged by a lethal dose of influenza virus 3 or 7 days after vaccination. Survival, body weight, antibody response, histopathological changes, viral loads, cytokine levels, and T cell frequencies were compared. The MTF-adjuvanted vaccine (MTF-VAC) group had a significantly better survival rate than the vaccine-only (VAC) group, when administered 3 days (80% vs. 26.7%, p = 0.0063) or 7 days (96% vs. 65%, p = 0.0041) before influenza virus challenge. Lung damage was significantly ameliorated in the MTF-VAC group. Antibody response was significantly augmented in the MTF-VAC group against both homologous and heterologous influenza strains. There was a greater T follicular helper cell (TFH) response and an enhanced germinal center (GC) reaction in the MTF-VAC group. MTF-VAC also induced both TH1 and TH2 antigen-specific cytokine responses. MTF improved the efficacy of the influenza vaccine against homologous and heterologous viruses by improving the TFH and antibody responses. Miltefosine may also be used for other vaccines, including the upcoming vaccines for COVID-19.
- Published
- 2020
- Full Text
- View/download PDF
25. Differential cell line susceptibility to the emerging Zika virus: implications for disease pathogenesis, non-vector-borne human transmission and animal reservoirs
- Author
-
Jasper Fuk-Woo Chan, Cyril Chik-Yan Yip, Jessica Oi-Ling Tsang, Kah-Meng Tee, Jian-Piao Cai, Kenn Ka-Heng Chik, Zheng Zhu, Chris Chung-Sing Chan, Garnet Kwan-Yue Choi, Siddharth Sridhar, Anna Jinxia Zhang, Gang Lu, Kin Chiu, Amy Cheuk-Yin Lo, Sai-Wah Tsao, Kin-Hang Kok, Dong-Yan Jin, Kwok-Hung Chan, and Kwok-Yung Yuen
- Subjects
animal ,cell line ,flavivirus ,placenta ,transmission ,tropism ,Infectious and parasitic diseases ,RC109-216 ,Microbiology ,QR1-502 - Abstract
Zika virus (ZIKV) is unique among human-pathogenic flaviviruses by its association with congenital anomalies and trans-placental and sexual human-to-human transmission. Although the pathogenesis of ZIKV-associated neurological complications has been reported in recent studies, key questions on the pathogenesis of the other clinical manifestations, non-vector-borne transmission and potential animal reservoirs of ZIKV remain unanswered. We systematically characterized the differential cell line susceptibility of 18 human and 15 nonhuman cell lines to two ZIKV isolates (human and primate) and dengue virus type 2 (DENV-2). Productive ZIKV replication (⩾2 log increase in viral load, ZIKV nonstructural protein-1 (NS1) protein expression and cytopathic effects (CPE)) was found in the placental (JEG-3), neuronal (SF268), muscle (RD), retinal (ARPE19), pulmonary (Hep-2 and HFL), colonic (Caco-2),and hepatic (Huh-7) cell lines. These findings helped to explain the trans-placental transmission and other clinical manifestations of ZIKV. Notably, the prostatic (LNCaP), testicular (833KE) and renal (HEK) cell lines showed increased ZIKV load and/or NS1 protein expression without inducing CPE, suggesting their potential roles in sexual transmission with persistent viral replication at these anatomical sites. Comparatively, none of the placental and genital tract cell lines allowed efficient DENV-2 replication. Among the nonhuman cell lines, nonhuman primate (Vero and LLC-MK2), pig (PK-15), rabbit (RK-13), hamster (BHK21) and chicken (DF-1) cell lines supported productive ZIKV replication. These animal species may be important reservoirs and/or potential animal models for ZIKV. The findings in our study help to explain the viral shedding pattern, transmission and pathogenesis of the rapidly disseminating ZIKV, and are useful for optimizing laboratory diagnostics and studies on the pathogenesis and counter-measures of ZIKV.Emerging Microbes & Infections (2016) 5, e93; doi:10.1038/emi.2016.99; published online 24 August 2016
- Published
- 2016
- Full Text
- View/download PDF
26. A novel bunyavirus causing fever and thrombocytopenia: More questions than answers
- Author
-
Kin-Hang Kok and Dong-Yan Jin
- Subjects
Medicine (General) ,R5-920 - Published
- 2011
- Full Text
- View/download PDF
27. Retraction for Siu et al., 'Middle East Respiratory Syndrome Coronavirus 4a Protein Is a Double-Stranded RNA-Binding Protein That Suppresses PACT-Induced Activation of RIG-I and MDA5 in the Innate Antiviral Response'
- Author
-
Kam-Leung Siu, Man Lung Yeung, Kin-Hang Kok, Kit-San Yuen, Chun Kew, Pak-Yin Lui, Chi-Ping Chan, Herman Tse, Patrick C. Y. Woo, Kwok-Yung Yuen, and Dong-Yan Jin
- Subjects
Interferon-Induced Helicase, IFIH1 ,Immunology ,RNA-Binding Proteins ,Microbiology ,Retraction ,Cell Line ,Coronavirus ,DEAD-box RNA Helicases ,Viral Proteins ,Virology ,Insect Science ,Host-Pathogen Interactions ,Protein Interaction Mapping ,DEAD Box Protein 58 ,Humans ,Interferons ,Receptors, Immunologic ,Immune Evasion ,Protein Binding - Abstract
Middle East respiratory syndrome coronavirus (MERS-CoV) is an emerging pathogen that causes severe disease in human. MERS-CoV is closely related to bat coronaviruses HKU4 and HKU5. Evasion of the innate antiviral response might contribute significantly to MERS-CoV pathogenesis, but the mechanism is poorly understood. In this study, we characterized MERS-CoV 4a protein as a novel immunosuppressive factor that antagonizes type I interferon production. MERS-CoV 4a protein contains a double-stranded RNA-binding domain capable of interacting with poly(I · C). Expression of MERS-CoV 4a protein suppressed the interferon production induced by poly(I · C) or Sendai virus. RNA binding of MERS-CoV 4a protein was required for IFN antagonism, a property shared by 4a protein of bat coronavirus HKU5 but not by the counterpart in bat coronavirus HKU4. MERS-CoV 4a protein interacted with PACT in an RNA-dependent manner but not with RIG-I or MDA5. It inhibited PACT-induced activation of RIG-I and MDA5 but did not affect the activity of downstream effectors such as RIG-I, MDA5, MAVS, TBK1, and IRF3. Taken together, our findings suggest a new mechanism through which MERS-CoV employs a viral double-stranded RNA-binding protein to circumvent the innate antiviral response by perturbing the function of cellular double-stranded RNA-binding protein PACT. PACT targeting might be a common strategy used by different viruses, including Ebola virus and herpes simplex virus 1, to counteract innate immunity.Middle East respiratory syndrome coronavirus (MERS-CoV) is an emerging and highly lethal human pathogen. Why MERS-CoV causes severe disease in human is unclear, and one possibility is that MERS-CoV is particularly efficient in counteracting host immunity, including the sensing of virus invasion. It will therefore be critical to clarify how MERS-CoV cripples the host proteins that sense viruses and to compare MERS-CoV with its ancestral viruses in bats in the counteraction of virus sensing. This work not only provides a new understanding of the abilities of MERS-CoV and closely related bat viruses to subvert virus sensing but also might prove useful in revealing new strategies for the development of vaccines and antivirals.
- Published
- 2022
28. Intravenous Injection of Coronavirus Disease 2019 (COVID-19) mRNA Vaccine Can Induce Acute Myopericarditis in Mouse Model
- Author
-
Yan Zhao, Jasper Fuk-Woo Chan, Kelvin K. W. To, Can Li, Yanxia Chen, Wan-Man Wong, Ivan Hung, Fei-Fei Liu, David Christopher Lung, Kin-Hang Kok, Wenchen Song, Dong-Yan Jin, Kwok-Yung Yuen, Zhanhong Ye, Hin Chu, Anna Jinxia Zhang, Jian-Piao Cai, Siddharth Sridhar, and Cyril C. Y. Yip
- Subjects
Microbiology (medical) ,Chemokine ,Pathology ,medicine.medical_specialty ,COVID-19 Vaccines ,Necrosis ,mouse model ,medicine.medical_treatment ,Antibodies, Viral ,Intracardiac injection ,Mice ,Ballooning degeneration ,Major Article ,medicine ,Animals ,Humans ,RNA, Messenger ,intramuscular ,Vaccines, Synthetic ,biology ,SARS-CoV-2 ,business.industry ,COVID-19 ,Endothelial Cells ,medicine.disease ,Troponin ,AcademicSubjects/MED00290 ,mRNA vaccine ,Infectious Diseases ,medicine.anatomical_structure ,Cytokine ,intravenous ,Injections, Intravenous ,biology.protein ,Cytokines ,mRNA Vaccines ,Chemokines ,medicine.symptom ,business ,Immunostaining ,Artery ,Myopericarditis - Abstract
Background Post-vaccination myopericarditis is reported after immunization with coronavirus disease 2019 (COVID-19) messenger RNA (mRNA) vaccines. The effect of inadvertent intravenous injection of this vaccine on the heart is unknown. Methods We compared the clinical manifestations, histopathological changes, tissue mRNA expression, and serum levels of cytokine/chemokine and troponin in Balb/c mice at different time points after intravenous (IV) or intramuscular (IM) vaccine injection with normal saline (NS) control. Results Although significant weight loss and higher serum cytokine/chemokine levels were found in IM group at 1–2 days post-injection (dpi), only IV group developed histopathological changes of myopericarditis as evidenced by cardiomyocyte degeneration, apoptosis, and necrosis with adjacent inflammatory cell infiltration and calcific deposits on visceral pericardium, although evidence of coronary artery or other cardiac pathologies was absent. Serum troponin level was significantly higher in IV group. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike antigen expression by immunostaining was occasionally found in infiltrating immune cells of the heart or injection site, in cardiomyocytes and intracardiac vascular endothelial cells, but not skeletal myocytes. The histological changes of myopericarditis after the first IV-priming dose persisted for 2 weeks and were markedly aggravated by a second IM- or IV-booster dose. Cardiac tissue mRNA expression of interleukin (IL)-1β, interferon (IFN)-β, IL-6, and tumor necrosis factor (TNF)-α increased significantly from 1 dpi to 2 dpi in the IV group but not the IM group, compatible with presence of myopericarditis in the IV group. Ballooning degeneration of hepatocytes was consistently found in the IV group. All other organs appeared normal. Conclusions This study provided in vivo evidence that inadvertent intravenous injection of COVID-19 mRNA vaccines may induce myopericarditis. Brief withdrawal of syringe plunger to exclude blood aspiration may be one possible way to reduce such risk.
- Published
- 2021
29. Clofazimine broadly inhibits coronaviruses including SARS-CoV-2
- Author
-
Jessica Pihl, Jasper Fuk-Woo Chan, Pok Man Lai, Jeffrey D. Esko, Li Sheng, Ronald A. Li, Yushen Du, Ren Sun, Lars Pache, Ronghui Liang, Thomas Mandel Clausen, Kwok-Yung Yuen, Hongzhe Sun, Chun-Kit Yuen, Yuan Pu, Zi-Wei Ye, Chris Chung-Sing Chan, Jianli Cao, Xue-Hui Cai, Anna Jinxia Zhang, Dong Wang, Sumit K. Chanda, Yan-Dong Tang, Ivan Hung, Andrew Chak-Yiu Lee, Wing-Kuk Au, Ko-Yung Sit, Kong-Hung Sze, Vincent Kwok-Man Poon, Dong-Yan Jin, Honglin Chen, Kin-Hang Kok, Runming Wang, Naoko Matsunaga, Wan Xu, Hin Chu, Kaiming Tang, Chit-Ying Lau, Shuofeng Yuan, Juntaek Oh, Chris Chun-Yiu Chan, Laura Riva, Yu-Yuan Zhang, Xiangzhi Meng, and Xin Yin
- Subjects
0301 basic medicine ,Drug ,Multidisciplinary ,business.industry ,viruses ,media_common.quotation_subject ,medicine.disease ,medicine.disease_cause ,Virology ,Clofazimine ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,In vivo ,medicine ,Middle East respiratory syndrome ,030212 general & internal medicine ,Viral shedding ,business ,Viral load ,media_common ,Respiratory tract ,medicine.drug ,Coronavirus - Abstract
The COVID-19 pandemic is the third outbreak this century of a zoonotic disease caused by a coronavirus, following the emergence of severe acute respiratory syndrome (SARS) in 20031 and Middle East respiratory syndrome (MERS) in 20122. Treatment options for coronaviruses are limited. Here we show that clofazimine-an anti-leprosy drug with a favourable safety profile3-possesses inhibitory activity against several coronaviruses, and can antagonize the replication of SARS-CoV-2 and MERS-CoV in a range of in vitro systems. We found that this molecule, which has been approved by the US Food and Drug Administration, inhibits cell fusion mediated by the viral spike glycoprotein, as well as activity of the viral helicase. Prophylactic or therapeutic administration of clofazimine in a hamster model of SARS-CoV-2 pathogenesis led to reduced viral loads in the lung and viral shedding in faeces, and also alleviated the inflammation associated with viral infection. Combinations of clofazimine and remdesivir exhibited antiviral synergy in vitro and in vivo, and restricted viral shedding from the upper respiratory tract. Clofazimine, which is orally bioavailable and comparatively cheap to manufacture, is an attractive clinical candidate for the treatment of outpatients and-when combined with remdesivir-in therapy for hospitalized patients with COVID-19, particularly in contexts in which costs are an important factor or specialized medical facilities are limited. Our data provide evidence that clofazimine may have a role in the control of the current pandemic of COVID-19 and-possibly more importantly-in dealing with coronavirus diseases that may emerge in the future.
- Published
- 2021
30. Emergence of a Severe Acute Respiratory Syndrome Coronavirus 2 Virus Variant With Novel Genomic Architecture in Hong Kong
- Author
-
Sally Cheuk-Ying Wong, Kin-Hang Kok, David Christopher Lung, Kelvin K. W. To, Herman Tse, Kwok-Yung Yuen, Garnet K. Y. Choi, Ka-Fai Ip, and Tak-Chiu Wu
- Subjects
0301 basic medicine ,Microbiology (medical) ,Untranslated region ,viruses ,030106 microbiology ,Genome ,Virus ,Transposition (music) ,03 medical and health sciences ,Pandemic ,Gene duplication ,Humans ,Medicine ,Whole genome sequencing ,SARS-CoV-2 ,business.industry ,Brief Report ,COVID-19 ,virus diseases ,Genomics ,Gene rearrangement ,Virology ,AcademicSubjects/MED00290 ,030104 developmental biology ,Infectious Diseases ,genomic rearrangement ,whole-genome sequencing ,Hong Kong ,business - Abstract
Throughout the coronavirus disease 2019 (COVID-19) pandemic, divergent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) lineages have emerged continuously, mostly through the genomic accumulation of substitutions. We report the discovery of a SARS-CoV-2 variant with a novel genomic architecture characterized by absent ORF7a, ORF7b, and ORF8, and a C-terminally modified ORF6 product resulting from partial 5′-untranslated region (UTR) duplication and transposition.
- Published
- 2021
31. Host-derived lipids orchestrate pulmonary γδ T cell response to provide early protection against influenza virus infection
- Author
-
Jingyi Li, Yuxia Zhang, Gen Lu, Tak Wah Mak, Sze Wai Ng, Kong-Peng Lam, Meng Zhao, Zhong Huang, Andy Hee-Meng Tan, Xiang Lin, Xiaohui Liu, Zihan Zheng, Kin-Hang Kok, Liwei Lu, Kwok-Yung Yuen, Qian Chen, Jia Tong Loh, Bingtai Lu, Jun Wang, Wanli Liu, King-Hung Ko, Fan Xiao, Xiaohui Wang, and Enyu Huang
- Subjects
0301 basic medicine ,T cell ,Science ,General Physics and Astronomy ,Mice, Transgenic ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,0302 clinical medicine ,Influenza A Virus, H1N1 Subtype ,Antigen ,Orthomyxoviridae Infections ,Immunity ,Influenza, Human ,medicine ,Influenza A virus ,Animals ,Humans ,Lung ,Mice, Knockout ,Multidisciplinary ,Innate immune system ,biology ,Interleukins ,Interleukin-17 ,Interleukin ,Receptors, Antigen, T-Cell, gamma-delta ,General Chemistry ,respiratory system ,Acquired immune system ,Lipids ,Immunity, Innate ,respiratory tract diseases ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,CD1D ,Immunology ,Host-Pathogen Interactions ,Interferon Regulatory Factors ,biology.protein ,Female ,Antigens, CD1d ,Influenza virus ,Gammadelta T cells ,030215 immunology - Abstract
Innate immunity is important for host defense by eliciting rapid anti-viral responses and bridging adaptive immunity. Here, we show that endogenous lipids released from virus-infected host cells activate lung γδ T cells to produce interleukin 17 A (IL-17A) for early protection against H1N1 influenza infection. During infection, the lung γδ T cell pool is constantly supplemented by thymic output, with recent emigrants infiltrating into the lung parenchyma and airway to acquire tissue-resident feature. Single-cell studies identify IL-17A-producing γδ T (Tγδ17) cells with a phenotype of TCRγδhiCD3hiAQP3hiCXCR6hi in both infected mice and patients with pneumonia. Mechanistically, host cell-released lipids during viral infection are presented by lung infiltrating CD1d+ B-1a cells to activate IL-17A production in γδ T cells via γδTCR-mediated IRF4-dependent transcription. Reduced IL-17A production in γδ T cells is detected in mice either lacking B-1a cells or with ablated CD1d in B cells. Our findings identify a local host-immune crosstalk and define important cellular and molecular mediators for early innate defense against lung viral infection., Influenza A infection results in γδ T cell influx and production of IL-17 in the lungs. Here, the authors show that this effect is primed by CD1-restricted ligands that are released by infected cells and presented by B1a cells in the lungs.
- Published
- 2021
32. Mining of linear B cell epitopes of SARS-CoV-2 ORF8 protein from COVID-19 patients
- Author
-
Lin-Lei Chen, Kwok-Yung Yuen, Shannon Wing-Ngor Au, Kin-Hang Kok, Kelvin K. W. To, Joy-Yan Lam, and Xiaohui Wang
- Subjects
Models, Molecular ,0301 basic medicine ,Coronavirus disease 2019 (COVID-19) ,Protein Conformation ,Epidemiology ,Viral protein ,viruses ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,030106 microbiology ,Immunology ,Biology ,Antibodies, Viral ,medicine.disease_cause ,Microbiology ,Epitope ,Viral Proteins ,03 medical and health sciences ,Protein structure ,antibody ,Cricetinae ,Virology ,Drug Discovery ,Pandemic ,medicine ,Animals ,Humans ,B-Cell Epitopes ,epitope ,Mesocricetus ,SARS-CoV-2 ,Immunodominant Epitopes ,COVID-19 ,ORF8 ,General Medicine ,peptide ,030104 developmental biology ,Infectious Diseases ,biology.protein ,Epitopes, B-Lymphocyte ,Parasitology ,Antibody ,Research Article - Abstract
Given the on-going SARS-CoV-2 pandemic, identification of immunogenic targets against the viral protein will provide crucial advances towards the development of sensitive diagnostic tools and vaccination strategies. Our previous study has found that ORF8 protein of SARS-CoV-2 is highly immunogenic and shows high sensitivity in identifying COVID-19 disease. In this study, by employing overlapping linear peptides, we characterized the IgG immunodominant regions on SARS-CoV-2 ORF8 protein that are seropositive in the sera from SARS-CoV-2-infected patients. The major immunogenic epitopes are localized at (1) N-termini alpha helix, (2) the resides spanning beta 2 and 3 sheets, and (3) the loop between beta 4 and 5 sheets. Additionally, hamster model infected by SARS-CoV-2 further validates the seropositivity of the linear epitopes in vivo, demonstrating a potential application of the linear peptide-based immunization strategy. Taken together, identification and validation of these B-cell linear epitopes will provide insights into the design of serological diagnostics and peptide-based vaccination approach against this pandemic virus of high priority.
- Published
- 2021
33. Coronavirus Disease 2019 (COVID-19) Re-infection by a Phylogenetically Distinct Severe Acute Respiratory Syndrome Coronavirus 2 Strain Confirmed by Whole Genome Sequencing
- Author
-
Ivan Hung, Polk Wan, Carol Ho-Yan Fong, Allen Wing-Ho Chu, Eugene Yuk-Keung Tso, Dominic N.C. Tsang, Shuofeng Yuan, Kelvin K. W. To, Kwok-Hung Chan, Hoi-Wah Tsoi, Kwok-Yung Yuen, Jian-Dong Huang, Vincent C.C. Cheng, Anthony Raymond Tam, Wing-Kin To, Larry Lap-Yip Lee, Wan-Mui Chan, Anthony Chin-Ki Ng, Jonathan Daniel Ip, and Kin-Hang Kok
- Subjects
0301 basic medicine ,Microbiology (medical) ,Health Personnel ,India ,Genome, Viral ,Antibodies, Viral ,medicine.disease_cause ,Genome ,Virus ,Serology ,03 medical and health sciences ,0302 clinical medicine ,Major Article ,medicine ,Humans ,030212 general & internal medicine ,Viral shedding ,Seroconversion ,Coronavirus ,re-infection ,First episode ,Whole genome sequencing ,Whole Genome Sequencing ,SARS-CoV-2 ,business.industry ,COVID-19 ,D614G ,Virology ,AcademicSubjects/MED00290 ,030104 developmental biology ,Infectious Diseases ,Reinfection ,business - Abstract
Background Waning immunity occurs in patients who have recovered from Coronavirus Disease 2019 (COVID-19). However, it remains unclear whether true re-infection occurs. Methods Whole genome sequencing was performed directly on respiratory specimens collected during 2 episodes of COVID-19 in a patient. Comparative genome analysis was conducted to differentiate re-infection from persistent viral shedding. Laboratory results, including RT-PCR Ct values and serum Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) IgG, were analyzed. Results The second episode of asymptomatic infection occurred 142 days after the first symptomatic episode in an apparently immunocompetent patient. During the second episode, there was evidence of acute infection including elevated C-reactive protein and SARS-CoV-2 IgG seroconversion. Viral genomes from first and second episodes belong to different clades/lineages. The virus genome from the first episode contained a a stop codon at position 64 of ORF8, leading to a truncation of 58 amino acids. Another 23 nucleotide and 13 amino acid differences located in 9 different proteins, including positions of B and T cell epitopes, were found between viruses from the first and second episodes. Compared to viral genomes in GISAID, the first virus genome was phylogenetically closely related to strains collected in March/April 2020, while the second virus genome was closely related to strains collected in July/August 2020. Conclusions Epidemiological, clinical, serological, and genomic analyses confirmed that the patient had re-infection instead of persistent viral shedding from first infection. Our results suggest SARS-CoV-2 may continue to circulate among humans despite herd immunity due to natural infection. Further studies of patients with re-infection will shed light on protective immunological correlates for guiding vaccine design.
- Published
- 2020
34. Seroprevalence of SARS-CoV-2 in Hong Kong and in residents evacuated from Hubei province, China: a multicohort study
- Author
-
Grace See-Wai Kwan, Kwok-Hung Chan, Cuiting Luo, Shuk-Ching Wong, Anthony Chin-Ki Ng, Lin-Lei Chen, Vincent C.C. Cheng, Lu Lu, Lok-Hin Wong, Kwok-Yung Yuen, Siddharth Sridhar, Charlotte Yee Ki Choi, Ivan Hung, Cecilia Yuen-Man Fan, Jianwen Situ, Kelvin K. W. To, Jasper Fuk-Woo Chan, Tom Wai-Hin Chung, Jian-Piao Cai, Kin-Hang Kok, Vivien W M Chuang, and Carol Ho-Yan Fong
- Subjects
Microbiology (medical) ,lcsh:R5-920 ,education.field_of_study ,medicine.diagnostic_test ,business.industry ,Population ,lcsh:QR1-502 ,Outbreak ,Microbiology ,Virology ,Asymptomatic ,lcsh:Microbiology ,Serology ,Infectious Diseases ,Immunoassay ,Pandemic ,Medicine ,Seroprevalence ,medicine.symptom ,lcsh:Medicine (General) ,business ,education ,Subclinical infection - Abstract
Summary Background The role of subclinical severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections in perpetuating the COVID-19 pandemic is unknown because population seroprevalence data are absent. We aimed to establish the sensitivity and specificity of our enzyme immunoassay and microneutralisation assay, and the seroprevalence of SARS-CoV-2 in Hong Kong before and after the pandemic, as well as in Hong Kong residents evacuated from Hubei province, China. Methods We did a multicohort study in a hospital and university in Hong Kong. We evaluated the sensitivity of our enzyme immunoassay and microneutralisation assay with RT-PCR data from patients positive for SARS-CoV-2 and the specificity of our enzyme immunoassay and microneutralisation assay with archived serum samples collected before 2019. We compared the seropositivity of the general population of Hong Kong before and after the pandemic had begun, and determined the seropositivity of Hong Kong residents evacuated from Hubei province, China, in March, 2020. Findings Between Feb 26 and March 18, 2020, we assessed RT-PCR samples from 45 patients who had recovered from COVID-19 to establish the sensitivity of our enzyme immunoassay and microneutralisation assay. To establish the specificity of these assays, we retrieved archived serum. The sensitivity was 91·1% (41 of 45 [95% CI 78·8–97·5]) for the microneutralisation assay, 57·8% (26 of 45 [42·2–72·3]) for anti-nucleoprotein IgG, 66·7% (30 of 45 [51·1–80·0]) for anti-spike protein receptor binding domain (RBD) IgG, and 73·3% (33 of 45 [58·1–85·4]) for enzyme immunoassay (either positive for anti-nucleoprotein or anti-RBD IgG). The specificity was 100% (152 of 152 [95% CI 97·6–100·0]) for both the enzyme immunoassay and microneutralisation assay. Among the Hong Kong general population, 53 (2·7%) of 1938 were enzyme immunoassay positive, but of those who were positive, all 53 were microneutralisation negative, and no significant increase was seen in the seroprevalence between April 12, 2018, and Feb 13, 2020. Among asymptomatic Hubei returnees, 17 (4%) of 452 were seropositive with the enzyme immunoassay or the microneutralisation assay, with 15 (88%) of 17 seropositive with the microneutralisation assay, and two familial clusters were identified. Interpretation Our serological data suggest that SARS-CoV-2 is a new emerging virus. The seropositivity rate in Hubei returnees indicates that RT-PCR-confirmed patients only represent a small proportion of the total number of cases. The low seroprevalence suggests that most of the Hong Kong and Hubei population remain susceptible to COVID-19. Future waves of the outbreak are inevitable without a vaccine or antiviral prophylaxis. The role of age-related cross reactive non-neutralising antibodies in the pathogenesis of COVID-19 warrants further investigation. Funding Richard and Carol Yu, May Tam Mak Mei Yin, Shaw Foundation (Hong Kong), Michael Tong, Marina Lee, and the Government Consultancy Service (see acknowledgments for full list).
- Published
- 2020
35. Identification of nsp1 gene as the target of SARS‐CoV‐2 real‐time RT‐PCR using nanopore whole‐genome sequencing
- Author
-
Kin-Hang Kok, Anthony Chin-Ki Ng, Owen Tak Yin Tsang, Wing Kin To, Mike Yat Wah Kwan, Kwok-Yung Yuen, Kwok-Hung Chan, Kelvin K. W. To, Wan Mui Chan, Ivan Hung, Wai Shing Leung, Jonathan Daniel Ip, Jasper Fuk-Woo Chan, Gilbert T. Chua, Allen Wing-Ho Chu, Tom Wai-Hin Chung, Rosana W.S. Poon, Cyril C. Y. Yip, Lap Sum Lo, and Vincent C.C. Cheng
- Subjects
Male ,RT‐PCR ,diagnosis ,viruses ,nsp1 ,Viral Nonstructural Proteins ,Biology ,medicine.disease_cause ,Sensitivity and Specificity ,Genome ,SARS‐CoV‐2 ,Open Reading Frames ,03 medical and health sciences ,0302 clinical medicine ,COVID‐19 ,Nasopharynx ,Virology ,medicine ,Humans ,030212 general & internal medicine ,Saliva ,Gene ,Research Articles ,Whole genome sequencing ,NSP1 ,Mutation ,Whole Genome Sequencing ,SARS-CoV-2 ,COVID-19 ,virus diseases ,Middle Aged ,RNA-Dependent RNA Polymerase ,Nanopore Sequencing ,Infectious Diseases ,Real-time polymerase chain reaction ,COVID-19 Nucleic Acid Testing ,RNA, Viral ,Female ,030211 gastroenterology & hepatology ,Nanopore sequencing ,Primer (molecular biology) ,Research Article - Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused the coronavirus disease 2019 (COVID-19) pandemic. Accurate detection of SARS-CoV-2 using molecular assays is critical for patient management and the control of the COVID-19 pandemic. However, there is an increasing number of SARS-CoV-2 viruses with mutations at the primer or probe binding sites, and these mutations may affect the sensitivity of currently available real-time reverse transcription-polymerase chain reaction (RT-PCR) assays targeting the nucleocapsid (N), envelope (E), and open reading frame 1a or 1b genes. Using sequence-independent single-primer amplification and nanopore whole-genome sequencing, we have found that the nonstructural protein 1 (nsp1) gene, located at the 5' end of the SARS-CoV-2 genome, was highly expressed in the nasopharyngeal or saliva specimens of 9 COVID-19 patients of different clinical severity. Based on this finding, we have developed a novel nsp1 real-time RT-PCR assay. The primers and probes are highly specific for SARS-CoV-2. Validation with 101 clinical specimens showed that our nsp1 RT-PCR assay has a sensitivity of 93.1% (95% confidence interval [CI]: 86.2%-97.2%), which was similar to those of N and E gene RT-PCR assays. The diagnostic specificity was 100% (95% CI: 92.9%-100%). The addition of nsp1 for multitarget detection of SARS-CoV-2 can avoid false-negative results due to mutations at the primers/probes binding sites of currently available RT-PCR assays.
- Published
- 2020
36. Targeting the Inositol-Requiring Enzyme-1 Pathway Efficiently Reverts Zika Virus-Induced Neurogenesis and Spermatogenesis Marker Perturbations
- Author
-
Kin-Hang Kok, Terrence T. T. Yuen, Hin Chu, Zheng Zhu, Jie Zhou, Shuofeng Yuan, Jasper Fuk-Woo Chan, Dong Yang, Huiping Shuai, Kwok-Yung Yuen, Yixin Wang, Chris Chung-Sing Chan, Kenn K. H. Chik, Zijiao Zou, Vincent Kwok-Man Poon, and Feifei Yin
- Subjects
0301 basic medicine ,Neurogenesis ,030106 microbiology ,Naphthalenes ,Protein Serine-Threonine Kinases ,03 medical and health sciences ,Downregulation and upregulation ,Interferon ,medicine ,Humans ,Progenitor cell ,Spermatogenesis ,Induced pluripotent stem cell ,biology ,Zika Virus Infection ,Imidazoles ,Zika Virus ,biology.organism_classification ,Flavivirus ,030104 developmental biology ,Infectious Diseases ,Pyrazines ,Unfolded protein response ,Cancer research ,Signal transduction ,Inositol ,medicine.drug - Abstract
Zika virus (ZIKV) is an emerging flavivirus that may be associated with congenital anomalies in infected fetuses and severe neurological and genital tract complications in infected adults. Currently, antiviral treatments to revert these ZIKV-induced complications are lacking. ZIKV infection has recently been suggested to upregulate the host unfolded protein response, which may contribute to the congenital neurological anomalies. As an extension from these findings, we thoroughly investigated the ZIKV-induced unfolded protein response using a combination of the neuronal cell line, induced pluripotent stem cell-derived human neuronal stem and progenitor cells, and an interferon receptor-deficient A129 mouse model. Our results revealed a critical contribution of the inositol-requiring enzyme-1 (IRE1) arm of the unfolded protein response to ZIKV-induced neurological and testicular complications. Importantly, the inhibition of the IRE1 signaling pathway activation with KIRA6 (kinase-inhibiting RNAse attenuator 6), a selective small molecule IRE1 inhibitor that promotes cell survival, potently reverted the ZIKV-induced perturbations of the key gene expressions associated with neurogenesis and spermatogenesis in vitro and in vivo, highlighting the potential of IRE1 inhibition as a novel host-targeting antiviral strategy in combating against ZIKV-induced neurological and testicular pathologies.
- Published
- 2020
37. Comparative Replication and Immune Activation Profiles of SARS-CoV-2 and SARS-CoV in Human Lungs: An Ex Vivo Study With Implications for the Pathogenesis of COVID-19
- Author
-
Shuofeng Yuan, Kwok-Yung Yuen, Kelvin K. W. To, Ivy Hau-Yee Chan, Wing-Kuk Au, Bingjie Hu, Terrence Tsz-Tai Yuen, Ko-Yung Sit, Yue Chai, Anna Jinxia Zhang, Yuxin Hou, Jian-Piao Cai, Dong Yang, Yixin Wang, Jie Zhou, Kin-Hang Kok, Huiping Shuai, Jasper Fuk-Woo Chan, Xi Zhang, Hin Chu, and Xiner Huang
- Subjects
0301 basic medicine ,Microbiology (medical) ,Chemokine ,viruses ,Pneumonia, Viral ,Virus Replication ,medicine.disease_cause ,Proinflammatory cytokine ,Pathogenesis ,Betacoronavirus ,03 medical and health sciences ,0302 clinical medicine ,Interferon ,medicine ,Humans ,skin and connective tissue diseases ,Lung ,Pandemics ,Tropism ,Coronavirus ,biology ,SARS-CoV-2 ,business.industry ,fungi ,COVID-19 ,virus diseases ,Immunity, Innate ,respiratory tract diseases ,030104 developmental biology ,Infectious Diseases ,medicine.anatomical_structure ,Severe acute respiratory syndrome-related coronavirus ,Viral replication ,030220 oncology & carcinogenesis ,Immunology ,biology.protein ,Cytokines ,Interferons ,Chemokines ,Coronavirus Infections ,business ,medicine.drug - Abstract
BackgroundSevere acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is an emerging coronavirus that has resulted in more than 2 000 000 laboratory-confirmed cases including over 145 000 deaths. Although SARS-CoV-2 and SARS-CoV share a number of common clinical manifestations, SARS-CoV-2 appears to be highly efficient in person-to-person transmission and frequently causes asymptomatic or presymptomatic infections. However, the underlying mechanisms that confer these viral characteristics of high transmissibility and asymptomatic infection remain incompletely understood.MethodsWe comprehensively investigated the replication, cell tropism, and immune activation profile of SARS-CoV-2 infection in human lung tissues with SARS-CoV included as a comparison.ResultsSARS-CoV-2 infected and replicated in human lung tissues more efficiently than SARS-CoV. Within the 48-hour interval, SARS-CoV-2 generated 3.20-fold more infectious virus particles than did SARS-CoV from the infected lung tissues (P < .024). SARS-CoV-2 and SARS-CoV were similar in cell tropism, with both targeting types I and II pneumocytes and alveolar macrophages. Importantly, despite the more efficient virus replication, SARS-CoV-2 did not significantly induce types I, II, or III interferons in the infected human lung tissues. In addition, while SARS-CoV infection upregulated the expression of 11 out of 13 (84.62%) representative proinflammatory cytokines/chemokines, SARS-CoV-2 infection only upregulated 5 of these 13 (38.46%) key inflammatory mediators despite replicating more efficiently.ConclusionsOur study provides the first quantitative data on the comparative replication capacity and immune activation profile of SARS-CoV-2 and SARS-CoV infection in human lung tissues. Our results provide important insights into the pathogenesis, high transmissibility, and asymptomatic infection of SARS-CoV-2.
- Published
- 2020
38. SARS-CoV-2 nsp13, nsp14, nsp15 and orf6 function as potent interferon antagonists
- Author
-
Xiaohui Wang, Kelvin K. W. To, Wan Man Wong, Jian Piao Cai, Chun Kit Yuen, Long Fung Mak, Jasper Fuk-Woo Chan, Kwok-Yung Yuen, Joy Lam, Kin-Hang Kok, Hin Chu, and Dong-Yan Jin
- Subjects
0301 basic medicine ,Epidemiology ,viruses ,Viral pathogenesis ,medicine.medical_treatment ,Viral Nonstructural Proteins ,Deubiquitinating enzyme ,Interferon ,Drug Discovery ,skin and connective tissue diseases ,Peptide sequence ,virus diseases ,Articles ,interferon antagonist ,General Medicine ,PLpro ,Infectious Diseases ,Host-Pathogen Interactions ,orf6 ,Coronavirus Infections ,RNA Helicases ,Research Article ,medicine.drug ,Pneumonia, Viral ,030106 microbiology ,Immunology ,Biology ,Microbiology ,Virus ,Cell Line ,Betacoronavirus ,Viral Proteins ,03 medical and health sciences ,Immune system ,Virology ,Endoribonucleases ,medicine ,Humans ,Pandemics ,Protease ,SARS-CoV-2 ,fungi ,COVID-19 ,Methyltransferases ,body regions ,030104 developmental biology ,Cell culture ,Exoribonucleases ,biology.protein ,Parasitology ,Interferons - Abstract
The Coronavirus disease 2019 (COVID-19), which is caused by the novel SARS-CoV-2 virus, is now causing a tremendous global health concern. Since its first appearance in December 2019, the outbreak has already caused over 5.8 million infections worldwide (till 29 May 2020), with more than 0.35 million deaths. Early virus-mediated immune suppression is believed to be one of the unique characteristics of SARS-CoV-2 infection and contributes at least partially to the viral pathogenesis. In this study, we identified the key viral interferon antagonists of SARS-CoV-2 and compared them with two well-characterized SARS-CoV interferon antagonists, PLpro and orf6. Here we demonstrated that the SARS-CoV-2 nsp13, nsp14, nsp15 and orf6, but not the unique orf8, could potently suppress primary interferon production and interferon signalling. Although SARS-CoV PLpro has been well-characterized for its potent interferon-antagonizing, deubiquitinase and protease activities, SARS-CoV-2 PLpro, despite sharing high amino acid sequence similarity with SARS-CoV, loses both interferon-antagonising and deubiquitinase activities. Among the 27 viral proteins, SARS-CoV-2 orf6 demonstrated the strongest suppression on both primary interferon production and interferon signalling. Orf6-deleted SARS-CoV-2 may be considered for the development of intranasal live-but-attenuated vaccine against COVID-19.
- Published
- 2020
39. Genomic characterization of the 2019 novel human-pathogenic coronavirus isolated from a patient with atypical pneumonia after visiting Wuhan
- Author
-
Zheng Zhu, Kin-Hang Kok, Hin Chu, Kelvin K. W. To, Kwok-Yung Yuen, Shuofeng Yuan, and Jasper Fuk-Woo Chan
- Subjects
0301 basic medicine ,Epidemiology ,medicine.disease_cause ,Genome ,Sequence Analysis, Protein ,Drug Discovery ,Phylogeny ,Coronavirus ,Travel ,biology ,emerging ,bioinformatics ,General Medicine ,Infectious Diseases ,Civet ,Coronavirus Infections ,Wuhan ,China ,Sequence analysis ,Pneumonia, Viral ,030106 microbiology ,Immunology ,virus ,Genome, Viral ,Microbiology ,Virus ,Betacoronavirus ,Viral Proteins ,03 medical and health sciences ,Phylogenetics ,Virology ,medicine ,Humans ,Amino Acid Sequence ,genome ,SARS ,COVID-19 ,respiratory ,SARS-CoV ,SARS-CoV-2 ,Correction ,Original Articles ,medicine.disease ,biology.organism_classification ,030104 developmental biology ,Atypical pneumonia ,Parasitology - Abstract
A mysterious outbreak of atypical pneumonia in late 2019 was traced to a seafood wholesale market in Wuhan of China. Within a few weeks, a novel coronavirus tentatively named as 2019 novel coronavirus (2019-nCoV) was announced by the World Health Organization. We performed bioinformatics analysis on a virus genome from a patient with 2019-nCoV infection and compared it with other related coronavirus genomes. Overall, the genome of 2019-nCoV has 89% nucleotide identity with bat SARS-like-CoVZXC21 and 82% with that of human SARS-CoV. The phylogenetic trees of their orf1a/b, Spike, Envelope, Membrane and Nucleoprotein also clustered closely with those of the bat, civet and human SARS coronaviruses. However, the external subdomain of Spike’s receptor binding domain of 2019-nCoV shares only 40% amino acid identity with other SARS-related coronaviruses. Remarkably, its orf3b encodes a completely novel short protein. Furthermore, its new orf8 likely encodes a secreted protein with an alpha-helix, following with a beta-sheet(s) containing six strands. Learning from the roles of civet in SARS and camel in MERS, hunting for the animal source of 2019-nCoV and its more ancestral virus would be important for understanding the origin and evolution of this novel lineage B betacoronavirus. These findings provide the basis for starting further studies on the pathogenesis, and optimizing the design of diagnostic, antiviral and vaccination strategies for this emerging infection.
- Published
- 2020
40. Low Environmental Temperature Exacerbates Severe Acute Respiratory Syndrome Coronavirus 2 Infection in Golden Syrian Hamsters
- Author
-
Andrew Chak-Yiu Lee, Kwok-Hung Chan, Cuiting Luo, Kenn Ka-Heng Chik, Ronghui Liang, Feifei Yin, Siddharth Sridhar, Chris Chung-Sing Chan, Kin-Hang Kok, Bingjie Hu, Hin Chu, Vincent Kwok-Man Poon, Terrence Tsz-Tai Yuen, Zijiao Zou, Chris Chun-Yiu Chan, Jasper Fuk-Woo Chan, Huiping Shuai, Kwok-Yung Yuen, Xiner Huang, Kaiming Tang, Jianli Cao, Shuofeng Yuan, Anna Jinxia Zhang, Jessica Oi-Ling Tsang, Jian-Piao Cai, Can Li, and Yue Chai
- Subjects
Microbiology (medical) ,Inflammation ,medicine.disease_cause ,Proinflammatory cytokine ,Alveolar cells ,Cricetinae ,Medicine ,Animals ,Humans ,Viral shedding ,Neutralizing antibody ,Lung ,Coronavirus ,biology ,Mesocricetus ,business.industry ,SARS-CoV-2 ,Temperature ,COVID-19 ,Antibodies, Neutralizing ,Actins ,Disease Models, Animal ,Infectious Diseases ,medicine.anatomical_structure ,Immunology ,biology.protein ,medicine.symptom ,business ,Viral load - Abstract
Background The effect of low environmental temperature on viral shedding and disease severity of Coronavirus Disease 2019 (COVID-19) is uncertain. Methods We investigated the virological, clinical, pathological, and immunological changes in hamsters housed at room (21°C), low (12–15°C), and high (30–33°C) temperature after challenge by 105 plaque-forming units of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Results The nasal turbinate, trachea, and lung viral load and live virus titer were significantly higher (~0.5-log10 gene copies/β-actin, P Conclusions This study provided in vivo evidence that low environmental temperature exacerbated the degree of virus shedding, disease severity, and tissue proinflammatory cytokines/chemokines expression, and suppressed the neutralizing antibody response of SARS-CoV-2-infected hamsters. Keeping warm in winter may reduce the severity of COVID-19.
- Published
- 2021
41. SMRT sequencing revealed the diversity and characteristics of defective interfering RNAs in influenza A (H7N9) virus infection
- Author
-
Kin-Hang Kok, Pak-Yin Lui, Kelvin K. W. To, Can Li, Anna J. X. Zhang, Hanjun Zhao, Chi-Ho Lin, Wing-Yu Lui, Kwok-Hung Chan, Kwok-Yung Yuen, Chun-Kit Yuen, Honglin Chen, and Wan Man Wong
- Subjects
0301 basic medicine ,Epidemiology ,viruses ,030106 microbiology ,Immunology ,Avian influenza A/H7N9 virus ,Bronchi ,Genome, Viral ,Biology ,Influenza A Virus, H7N9 Subtype ,Microbiology ,Genome ,Virus ,03 medical and health sciences ,Mice ,Orthomyxoviridae Infections ,Virology ,Nasopharynx ,Drug Discovery ,Influenza, Human ,Animals ,Humans ,Illumina dye sequencing ,Sequence Deletion ,Genetics ,Illumina sequencing ,Defective Viruses ,Influenza a ,Epithelial Cells ,General Medicine ,Original Articles ,Sequence Analysis, DNA ,Single Molecule Real Time sequencing ,defective interfering viral genome ,Disease Models, Animal ,030104 developmental biology ,Infectious Diseases ,RNA, Viral ,Parasitology ,Single molecule real time sequencing - Abstract
Influenza defective interfering (DI) particles are replication-incompetent viruses carrying large internal deletion in the genome. The loss of essential genetic information causes abortive viral replication, which can be rescued by co-infection with a helper virus that possesses an intact genome. Despite reports of DI particles present in seasonal influenza A H1N1 infections, their existence in human infections by the avian influenza A viruses, such as H7N9, has not been studied. Here we report the ubiquitous presence of DI-RNAs in nasopharyngeal aspirates of H7N9-infected patients. Single Molecule Real Time (SMRT) sequencing was first applied and long-read sequencing analysis showed that a variety of H7N9 DI-RNA species were present in the patient samples and human bronchial epithelial cells. In several abundantly expressed DI-RNA species, long overlapping sequences have been identified around at the breakpoint region and the other side of deleted region. Influenza DI-RNA is known as a defective viral RNA with single large internal deletion. Beneficial to the long-read property of SMRT sequencing, double and triple internal deletions were identified in half of the DI-RNA species. In addition, we examined the expression of DI-RNAs in mice infected with sublethal dose of H7N9 virus at different time points. Interestingly, DI-RNAs were abundantly expressed as early as day 2 post-infection. Taken together, we reveal the diversity and characteristics of DI-RNAs found in H7N9-infected patients, cells and animals. Further investigations on this overwhelming generation of DI-RNA may provide important insights into the understanding of H7N9 viral replication and pathogenesis.
- Published
- 2019
42. Coronaviruses exploit a host cysteine-aspartic protease for efficient replication
- Author
-
Yue Chai, Yuxin Hou, Yixin Wang, Jinxia Zhang, Lei Wen, Vincent Kwok-Man Poon, Kenneth K. Y. Wong, Jian-Piao Cai, Terrence Tsz-Tai Yuen, Dong Yang, Shuofeng Yuan, Wing-Kuk Au, Ko-Yung Sit, Chaemin Yoon, Kin-Hang Kok, Cun Li, Hin Chu, Xiner Huang, Kwok-Yung Yuen, Bingjie Hu, Dong-Yan Jin, Xiaoyu Zhao, Jasper Fuk-Woo Chan, Jie Zhou, Chris Chung-Sing Chan, Huiping Shuai, and Dominic Chi-Chung Foo
- Subjects
Host (biology) ,viruses ,virus diseases ,Cysteine-aspartic Protease ,Computational biology ,Biology ,Replication (computing) - Abstract
Highly pathogenic coronaviruses including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)1,2, Middle East respiratory syndrome coronavirus (MERS-CoV)3,4, and SARS-CoV-15 vary in their transmissibility and pathogenicity. However, infection by all three viruses result in substantial apoptosis in cell culture6-8 and in patient samples9-11, suggesting a potential link between apoptosis and the pathogenesis of coronaviruses. To date, the underlying mechanism of how apoptosis modulates coronavirus pathogenesis is unknown. Here we show that a cysteine-aspartic protease of the apoptosis cascade, caspase-6, serves as an essential host factor for efficient coronavirus replication. We demonstrate that caspase-6 cleaves coronavirus nucleocapsid (N) proteins, generating N fragments that serve as interferon (IFN) antagonists, thus facilitating virus replication. Inhibition of caspase-6 substantially attenuates the lung pathology and body weight loss of SARS-CoV-2-infected golden Syrian hamsters and improves the survival of mouse-adapted MERS-CoV (MERS-CoVMA)-infected human DPP4 knock-in (hDPP4 KI) mice. Overall, our study reveals how coronaviruses exploit a component of the host apoptosis cascade to facilitate their replication. These results further suggest caspase-6 as a potential target of intervention for the treatment of highly pathogenic coronavirus infections including COVID-19 and MERS.
- Published
- 2021
43. Coronaviruses exploit a host cysteine-aspartic protease for replication
- Author
-
Hin Chu, Yuxin Hou, Dong Yang, Lei Wen, Huiping Shuai, Chaemin Yoon, Jialu Shi, Yue Chai, Terrence Tsz-Tai Yuen, Bingjie Hu, Cun Li, Xiaoyu Zhao, Yixin Wang, Xiner Huang, Kin Shing Lee, Cuiting Luo, Jian-Piao Cai, Vincent Kwok-Man Poon, Chris Chung-Sing Chan, Anna Jinxia Zhang, Shuofeng Yuan, Ko-Yung Sit, Dominic Chi-Chung Foo, Wing-Kuk Au, Kenneth Kak-Yuen Wong, Jie Zhou, Kin-Hang Kok, Dong-Yan Jin, Jasper Fuk-Woo Chan, and Kwok-Yung Yuen
- Subjects
Aspartic Acid ,Multidisciplinary ,Caspase 6 ,Mesocricetus ,SARS-CoV-2 ,Dipeptidyl Peptidase 4 ,Apoptosis ,Virus Replication ,Coronavirus ,Survival Rate ,Mice ,Severe acute respiratory syndrome-related coronavirus ,Cricetinae ,Host-Pathogen Interactions ,Weight Loss ,Middle East Respiratory Syndrome Coronavirus ,Animals ,Coronavirus Nucleocapsid Proteins ,Humans ,Cysteine ,Interferons ,Coronavirus Infections ,Lung - Abstract
Highly pathogenic coronaviruses, including severe acute respiratory syndrome coronavirus 2 (refs.
- Published
- 2021
44. Corrigendum to: Intravenous Injection of Coronavirus Disease 2019 (COVID-19) mRNA Vaccine Can Induce Acute Myopericarditis in Mouse Model
- Author
-
Wenchen Song, Kwok-Yung Yuen, Jasper Fuk-Woo Chan, Kin-Hang Kok, Ivan Hung, Hin Chu, David Christopher Lung, Anna JinxiaZhang, Jian-Piao Cai, Yanxia Chen, Wan-Man Wong, Yan Zhao, Zhanhong Ye, Kelvin K. W. To, Dong-Yan Jin, Cyril Chik-YanYip, Siddharth Sridhar, Can Li, and Fei-Fei Liu
- Subjects
Microbiology (medical) ,COVID-19 Vaccines ,Coronavirus disease 2019 (COVID-19) ,Antibodies, Viral ,Mice ,Medicine ,Animals ,Humans ,RNA, Messenger ,Messenger RNA ,Vaccines, Synthetic ,Acute myopericarditis ,business.industry ,SARS-CoV-2 ,COVID-19 ,Endothelial Cells ,Virology ,Corrigenda ,Troponin ,Infectious Diseases ,AcademicSubjects/MED00290 ,Injections, Intravenous ,Cytokines ,mRNA Vaccines ,Chemokines ,business - Abstract
Post-vaccination myopericarditis is reported after immunization with coronavirus disease 2019 (COVID-19) messenger RNA (mRNA) vaccines. The effect of inadvertent intravenous injection of this vaccine on the heart is unknown.We compared the clinical manifestations, histopathological changes, tissue mRNA expression, and serum levels of cytokine/chemokine and troponin in Balb/c mice at different time points after intravenous (IV) or intramuscular (IM) vaccine injection with normal saline (NS) control.Although significant weight loss and higher serum cytokine/chemokine levels were found in IM group at 1-2 days post-injection (dpi), only IV group developed histopathological changes of myopericarditis as evidenced by cardiomyocyte degeneration, apoptosis, and necrosis with adjacent inflammatory cell infiltration and calcific deposits on visceral pericardium, although evidence of coronary artery or other cardiac pathologies was absent. Serum troponin level was significantly higher in IV group. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike antigen expression by immunostaining was occasionally found in infiltrating immune cells of the heart or injection site, in cardiomyocytes and intracardiac vascular endothelial cells, but not skeletal myocytes. The histological changes of myopericarditis after the first IV-priming dose persisted for 2 weeks and were markedly aggravated by a second IM- or IV-booster dose. Cardiac tissue mRNA expression of interleukin (IL)-1β, interferon (IFN)-β, IL-6, and tumor necrosis factor (TNF)-α increased significantly from 1 dpi to 2 dpi in the IV group but not the IM group, compatible with presence of myopericarditis in the IV group. Ballooning degeneration of hepatocytes was consistently found in the IV group. All other organs appeared normal.This study provided in vivo evidence that inadvertent intravenous injection of COVID-19 mRNA vaccines may induce myopericarditis. Brief withdrawal of syringe plunger to exclude blood aspiration may be one possible way to reduce such risk.
- Published
- 2021
45. Isolation of MERS-related coronavirus from lesser bamboo bats that uses DPP4 and infects human-DPP4-transgenic mice
- Author
-
Antonio C.P. Wong, Libiao Zhang, Kin-Hang Kok, Zirong He, George C.S. Lo, Kwok-Yung Yuen, Kenneth S. M. Li, Hayes K.H. Luk, Rachel Y.Y. Fan, Longchao Zhu, Felix C.H. Fok, Patrick C. Y. Woo, Joshua Fung, Emily Y.M. Wong, Carol S. F. Lam, Susanna K. P. Lau, and Rex Au-Yeung
- Subjects
0301 basic medicine ,Genetically modified mouse ,Chemokine ,endocrine system ,animal structures ,Middle East respiratory syndrome coronavirus ,Science ,viruses ,Dipeptidyl Peptidase 4 ,030106 microbiology ,General Physics and Astronomy ,Mice, Transgenic ,medicine.disease_cause ,Virus ,Host Specificity ,Article ,General Biochemistry, Genetics and Molecular Biology ,Proinflammatory cytokine ,03 medical and health sciences ,Mice ,Viral reservoirs ,Chiroptera ,medicine ,Animals ,Humans ,Receptor ,Lung ,Coronavirus ,Multidisciplinary ,biology ,virus diseases ,Brain ,General Chemistry ,Tylonycteris ,biology.organism_classification ,Virology ,respiratory tract diseases ,Mice, Inbred C57BL ,030104 developmental biology ,HEK293 Cells ,biology.protein ,Middle East Respiratory Syndrome Coronavirus ,Cytokines ,Caco-2 Cells ,Coronavirus Infections ,Viral pathogenesis - Abstract
While a number of human coronaviruses are believed to be originated from ancestral viruses in bats, it remains unclear if bat coronaviruses are ready to cause direct bat-to-human transmission. Here, we report the isolation of a MERS-related coronavirus, Tylonycteris-bat-CoV-HKU4, from lesser bamboo bats. Tylonycteris-bat-CoV-HKU4 replicates efficiently in human colorectal adenocarcinoma and hepatocarcinoma cells with cytopathic effects, and can utilize human-dipeptidyl-peptidase-4 and dromedary camel-dipeptidyl-peptidase-4 as the receptors for cell entry. Flow cytometry, co-immunoprecipitation and surface plasmon resonance assays show that Tylonycteris-bat-CoV-HKU4-receptor-binding-domain can bind human-dipeptidyl-peptidase-4, dromedary camel-dipeptidyl-peptidase-4, and Tylonycteris pachypus-dipeptidyl-peptidase-4. Tylonycteris-bat-CoV-HKU4 can infect human-dipeptidyl-peptidase-4-transgenic mice by intranasal inoculation with self-limiting disease. Positive virus and inflammatory changes were detected in lungs and brains of infected mice, associated with suppression of antiviral cytokines and activation of proinflammatory cytokines and chemokines. The results suggest that MERS-related bat coronaviruses may overcome species barrier by utilizing dipeptidyl-peptidase-4 and potentially emerge in humans by direct bat-to-human transmission., Several human coronaviruses (CoV) have been proposed to emerge from bats but evidence of direct bat-to-human transmission is slim. In this work, the authors isolate a MERS-related CoV strain directly from bats and show that it infects target cells in vitro and engineered mice through the human DDP4 receptor.
- Published
- 2021
- Full Text
- View/download PDF
46. A platform technology for generating subunit vaccines against diverse viral pathogens.
- Author
-
Young, Andrew, Isaacs, Ariel, Scott, Connor A. P., Modhiran, Naphak, McMillan, Christopher L. D., Cheung, Stacey T. M., Barr, Jennifer, Marsh, Glenn, Thakur, Nazia, Bailey, Dalan, Li, Kenneth S. M., Luk, Hayes K. H., Kin-Hang Kok, Lau, Susanna K. P., Woo, Patrick C. Y., Wakako Furuyama, Marzi, Andrea, Young, Paul R., Chappell, Keith J., and Watterson, Daniel
- Subjects
EMERGING infectious diseases ,GEL permeation chromatography ,VIRAL antigens ,VIRAL antibodies ,EBOLA virus - Abstract
The COVID-19 pandemic response has shown how vaccine platform technologies can be used to rapidly and effectively counteract a novel emerging infectious disease. The speed of development for mRNA and vector-based vaccines outpaced those of subunit vaccines, however, subunit vaccines can offer advantages in terms of safety and stability. Here we describe a subunit vaccine platform technology, the molecular clamp, in application to four viruses from divergent taxonomic families: Middle Eastern respiratory syndrome coronavirus (MERS-CoV), Ebola virus (EBOV), Lassa virus (LASV) and Nipah virus (NiV). The clamp streamlines subunit antigen production by both stabilising the immunologically important prefusion epitopes of trimeric viral fusion proteins while enabling purification without target-specific reagents by acting as an affinity tag. Conformations for each viral antigen were confirmed by monoclonal antibody binding, size exclusion chromatography and electronmicroscopy.Notably, all four antigens tested remained stable over four weeks of incubation at 40°C.Of the four vaccines tested, a neutralising immune response was stimulated by clamp stabilised MERS-CoV spike, EBOV glycoprotein and NiV fusion protein. Only the clamp stabilised LASV glycoprotein precursor failed to elicit virus neutralising antibodies. MERS-CoV and EBOV vaccine candidates were both tested in animal models and found to provide protection against viral challenge. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
47. Targeting highly pathogenic coronavirus-induced apoptosis reduces viral pathogenesis and disease severity
- Author
-
Huiping Shuai, Bingjie Hu, Shuofeng Yuan, Dong Yang, Vincent Kwok-Man Poon, Stanley Perlman, Bosco Ho-Yin Wong, Kwok-Yung Yuen, Chaemin Yoon, Cun Li, Kin-Hang Kok, Dong-Yan Jin, Hin Chu, Jie Zhou, Lei Wen, Kenneth K. Y. Wong, Yixin Wang, Man Lung Yeung, Jasper Fuk-Woo Chan, Xi Zhang, Rex Au-Yeung, Xiner Huang, Xiaoyu Zhao, Yuxin Hou, and Jian-Piao Cai
- Subjects
Male ,Indoles ,Middle East respiratory syndrome coronavirus ,Viral pathogenesis ,viruses ,Dipeptidyl Peptidase 4 ,Apoptosis ,Mice, Transgenic ,macromolecular substances ,medicine.disease_cause ,Antiviral Agents ,Microbiology ,Cell Line ,Pathogenesis ,03 medical and health sciences ,eIF-2 Kinase ,0302 clinical medicine ,Virology ,medicine ,Animals ,Humans ,Lung ,Dipeptidyl peptidase-4 ,Research Articles ,030304 developmental biology ,Coronavirus ,0303 health sciences ,EIF-2 kinase ,Multidisciplinary ,biology ,Adenine ,Intrinsic apoptosis ,fungi ,virus diseases ,food and beverages ,COVID-19 ,SciAdv r-articles ,Epithelial Cells ,respiratory tract diseases ,COVID-19 Drug Treatment ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Female ,Angiotensin-Converting Enzyme 2 ,Coronavirus Infections ,Research Article - Abstract
Infection of highly pathogenic coronaviruses triggers apoptosis that can be targeted to reduce disease severity., Infection by highly pathogenic coronaviruses results in substantial apoptosis. However, the physiological relevance of apoptosis in the pathogenesis of coronavirus infections is unknown. Here, with a combination of in vitro, ex vivo, and in vivo models, we demonstrated that protein kinase R–like endoplasmic reticulum kinase (PERK) signaling mediated the proapoptotic signals in Middle East respiratory syndrome coronavirus (MERS-CoV) infection, which converged in the intrinsic apoptosis pathway. Inhibiting PERK signaling or intrinsic apoptosis both alleviated MERS pathogenesis in vivo. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and SARS-CoV induced apoptosis through distinct mechanisms but inhibition of intrinsic apoptosis similarly limited SARS-CoV-2– and SARS-CoV–induced apoptosis in vitro and markedly ameliorated the lung damage of SARS-CoV-2–inoculated human angiotensin-converting enzyme 2 (hACE2) mice. Collectively, our study provides the first evidence that virus-induced apoptosis is an important disease determinant of highly pathogenic coronaviruses and demonstrates that this process can be targeted to attenuate disease severity.
- Published
- 2020
48. Loss of orf3b in the circulating SARS-CoV-2 strains
- Author
-
Kwok-Yung Yuen, Wan-Man Wong, Joy-Yan Lam, Kelvin K. W. To, Kin-Hang Kok, Jonathan Daniel Ip, and Chun-Kit Yuen
- Subjects
0301 basic medicine ,Epidemiology ,Viral pathogenesis ,030106 microbiology ,Immunology ,Virulence ,Q57H ,Biology ,Microbiology ,Virus ,03 medical and health sciences ,Viral Proteins ,Interferon ,Virology ,Drug Discovery ,medicine ,Humans ,Amino Acid Sequence ,Loss function ,Cells, Cultured ,Infectivity ,SARS-CoV-2 ,General Medicine ,biology.organism_classification ,D614G ,Stop codon ,030104 developmental biology ,Infectious Diseases ,orf3b ,orf3a ,Parasitology ,Interferons ,Betacoronavirus ,Gene Deletion ,medicine.drug ,Research Article - Abstract
The newly emerged betacoronavirus, SARS-CoV-2, causes the COVID-19 pandemic since December 2019 with more than 35 million laboratory confirmed human infections and over one million deaths within nine months. The genome of SARS-CoV-2 continues to evolve during the global transmission with the notable emergence of the spike D614G substitution that enhances infectivity. Some of these viral adaptations may alter not only the infectivity but also viral pathogenesis. Continuous phylogenomic analysis of circulating viral strains and functional investigation of new non-synonymous substitutions may help to understand the evolution of virus, its virulence and transmissibility. Here we describe a loss of an accessory protein orf3b (57 amino acids) in current circulating SARS-CoV-2 strains, contributing around 24% of more than 100,000 complete viral genomes analysed. The loss of 3b is caused by the presence of an early stop codon which is created by an orf3a Q57H substitution. There is an increasing trend in the loss of orf3b which has reached 32% in May 2020. Geographically, loss of 3b is more prevalent in certain countries including Colombia (46%), USA (48%), South Korea (51%), France (66%), Saudi Arabia (72%), Finland (76%) and Egypt (77%). Interestingly, the loss of 3b coincides with the emergence of spike D614G substitution. In addition, we found that truncated orf3b has lost the interferon antagonism compared to the full-length orf3b, suggesting a loss of function by the newly adapted virus. Further investigation of orf3b deletion and spike D614G substitution on virulence and infectivity respectively will provide important insights into SARS-CoV-2 evolution.
- Published
- 2020
49. Accurate Diagnosis of COVID-19 by a Novel Immunogenic Secreted SARS-CoV-2 orf8 Protein
- Author
-
Kelvin K. W. To, Jasper Fuk-Woo Chan, Chun Kit Yuen, Kwok-Yung Yuen, Wan Man Wong, Kin-Hang Kok, Xiaohui Wang, Joy Lam, Shannon Wing-Ngor Au, and Jian Piao Cai
- Subjects
diagnosis ,Pneumonia, Viral ,Enzyme-Linked Immunosorbent Assay ,Antibodies, Viral ,Asymptomatic ,Microbiology ,Serology ,Cell Line ,03 medical and health sciences ,Betacoronavirus ,Viral Proteins ,Virology ,Medicine ,Humans ,Secretion ,Antigens, Viral ,Pandemics ,B cell ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,biology ,030306 microbiology ,business.industry ,SARS-CoV-2 ,COVID-19 ,Therapeutics and Prevention ,QR1-502 ,Nucleoprotein ,medicine.anatomical_structure ,Early Diagnosis ,chemistry ,Cell culture ,Immunoglobulin G ,biology.protein ,orf8 ,Antibody ,medicine.symptom ,Glycoprotein ,business ,Coronavirus Infections ,Research Article - Abstract
Current commercially available serological tests for COVID-19 patients are detecting antibodies against SARS-CoV-2 nucleoprotein and spike glycoprotein. The antinucleoprotein and antispike antibodies can be accurately detected in patients during the mid or late stage of infection, and therefore, these assays have not been widely used for early diagnosis of COVID-19. In this study, we characterized the secretory property of a SARS-CoV-2 orf8 protein and proposed that orf8 secretion during infection facilitates early mounting of the B cell response. We demonstrated the presence of anti-orf8 antibodies in both symptomatic and asymptomatic patients during the early stage of infection, while the anti-N antibody is not detected. Our serological test detecting anti-orf8 antibodies may facilitate the development of early and accurate diagnosis for COVID-19., An accurate diagnostic test for early severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is the key weapon to control the coronavirus disease 2019 (COVID-19) pandemic. We previously reported that the SARS-CoV-2 genome contains a unique orf8 accessory gene absent from other human-pathogenic coronaviruses. Here, we characterized the SARS-CoV-2 orf8 as a novel immunogenic secreted protein and utilized it for the accurate diagnosis of COVID-19. Extracellular orf8 protein was detected in cell culture supernatant and in sera of COVID-19 patients. In addition, orf8 was found highly immunogenic in COVID-19 patients, who showed early seropositivity for anti-orf8 IgM, IgG, and IgA. We hypothesize that orf8 secretion during SARS-CoV-2 infection facilitates early mounting of B cell response. The serological test detecting anti-orf8 IgG antibody can be used for the early and accurate diagnosis of COVID-19.
- Published
- 2020
50. Clofazimine is a broad-spectrum coronavirus inhibitor that antagonizes SARS-CoV-2 replication in primary human cell culture and hamsters
- Author
-
Kaiming Tang, Kwok-Yung Yuen, Pok Man Lai, Jinxia Zhang, Laura Riva, Kin-Hang Kok, Hin Chu, Sumit K. Chanda, Shuofeng Yuan, Dong-Yan Jin, Kelvin K. W. To, Ronghui Liang, Lars Pache, Jasper Fuk-Woo Chan, Naoko Matsunaga, Kong-Hung Sze, Yushen Du, Chun-Kit Yuen, Yuan Pu, Chris Chung-Sing Chan, Xiangzhi Meng, Vincent Kwok-Man Poon, Zi-Wei Ye, Wan Xu, Xin Yin, Chris Chun-Yiu Chan, Ren Sun, Li Sheng, and Jianli Cao
- Subjects
Male ,Transcription, Genetic ,viruses ,Anti-Inflammatory Agents ,Biological Availability ,medicine.disease_cause ,Clofazimine ,Antiviral Agents ,Article ,Cell Line ,Cell Fusion ,Species Specificity ,Cricetinae ,medicine ,Animals ,Humans ,Viral shedding ,Coronavirus ,Life Cycle Stages ,Alanine ,Mesocricetus ,SARS-CoV-2 ,business.industry ,DNA Helicases ,COVID-19 ,Drug Synergism ,coronavirus inhibitor ,medicine.disease ,Virology ,Adenosine Monophosphate ,Viral replication ,pan-coronaviral inhibitory activity ,Spike Glycoprotein, Coronavirus ,Middle East respiratory syndrome ,coronavirus (CoV) outbreak ,Female ,Pre-Exposure Prophylaxis ,Cytokine storm ,business ,Viral load ,Ex vivo ,medicine.drug - Abstract
COVID-19 pandemic is the third zoonotic coronavirus (CoV) outbreak of the century after severe acute respiratory syndrome (SARS) in 2003 and Middle East respiratory syndrome (MERS) since 2012. Treatment options for CoVs are largely lacking. Here, we show that clofazimine, an anti-leprosy drug with a favorable safety and pharmacokinetics profile, possesses pan-coronaviral inhibitory activity, and can antagonize SARS-CoV-2 replication in multiple in vitro systems, including the human embryonic stem cell-derived cardiomyocytes and ex vivo lung cultures. The FDA-approved molecule was found to inhibit multiple steps of viral replication, suggesting multiple underlying antiviral mechanisms. In a hamster model of SARS-CoV-2 pathogenesis, prophylactic or therapeutic administration of clofazimine significantly reduced viral load in the lung and fecal viral shedding, and also prevented cytokine storm associated with viral infection. Additionally, clofazimine exhibited synergy when administered with remdesivir. Since clofazimine is orally bioavailable and has a comparatively low manufacturing cost, it is an attractive clinical candidate for outpatient treatment and remdesivir-based combinatorial therapy for hospitalized COVID-19 patients, particularly in developing countries. Taken together, our data provide evidence that clofazimine may have a role in the control of the current pandemic SARS-CoV-2, endemic MERS-CoV in the Middle East, and, possibly most importantly, emerging CoVs of the future.
- Published
- 2020
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.