65 results on '"Poon VK"'
Search Results
2. SARS-CoV-2 hijacks neutralizing dimeric IgA for nasal infection and injury in Syrian hamsters 1 .
- Author
-
Zhou B, Zhou R, Chan JF, Zeng J, Zhang Q, Yuan S, Liu L, Robinot R, Shan S, Liu N, Ge J, Kwong HY, Zhou D, Xu H, Chan CC, Poon VK, Chu H, Yue M, Kwan KY, Chan CY, Chan CC, Chik KK, Du Z, Au KK, Huang H, Man HO, Cao J, Li C, Wang Z, Zhou J, Song Y, Yeung ML, To KK, Ho DD, Chakrabarti LA, Wang X, Zhang L, Yuen KY, and Chen Z
- Subjects
- Cricetinae, Animals, Humans, SARS-CoV-2, Mesocricetus, Antibodies, Viral, Antibodies, Neutralizing, Immunoglobulin A, Spike Glycoprotein, Coronavirus, COVID-19, Common Cold
- Abstract
Prevention of robust severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection in nasal turbinate (NT) requires in vivo evaluation of IgA neutralizing antibodies. Here, we report the efficacy of receptor binding domain (RBD)-specific monomeric B8-mIgA1 and B8-mIgA2, and dimeric B8-dIgA1, B8-dIgA2 and TH335-dIgA1 against intranasal SARS-CoV-2 challenge in Syrian hamsters. These antibodies exhibited comparable neutralization potency against authentic virus by competing with human angiotensin converting enzyme-2 (ACE2) receptor for RBD binding. While reducing viral loads in lungs significantly, prophylactic intranasal B8-dIgA unexpectedly led to high amount of infectious viruses and extended damage in NT compared to controls. Mechanistically, B8-dIgA failed to inhibit SARS-CoV-2 cell-to-cell transmission, but was hijacked by the virus through dendritic cell-mediated trans-infection of NT epithelia leading to robust nasal infection. Cryo-EM further revealed B8 as a class II antibody binding trimeric RBDs in 3-up or 2-up/1-down conformation. Neutralizing dIgA, therefore, may engage an unexpected mode of SARS-CoV-2 nasal infection and injury.
- Published
- 2023
- Full Text
- View/download PDF
3. Antibodies targeting a quaternary site on SARS-CoV-2 spike glycoprotein prevent viral receptor engagement by conformational locking.
- Author
-
Liu L, Casner RG, Guo Y, Wang Q, Iketani S, Chan JF, Yu J, Dadonaite B, Nair MS, Mohri H, Reddem ER, Yuan S, Poon VK, Chan CC, Yuen KY, Sheng Z, Huang Y, Bloom JD, Shapiro L, and Ho DD
- Subjects
- Animals, Cricetinae, Humans, COVID-19 Vaccines, SARS-CoV-2, Receptors, Virus, Antibodies, Monoclonal, Antibodies, Viral, Antibodies, Neutralizing, Spike Glycoprotein, Coronavirus, COVID-19
- Abstract
SARS-CoV-2 continues to evolve, with many variants evading clinically authorized antibodies. To isolate monoclonal antibodies (mAbs) with broadly neutralizing capacities against the virus, we screened serum samples from convalescing COVID-19 patients. We isolated two mAbs, 12-16 and 12-19, which neutralized all SARS-CoV-2 variants tested, including the XBB subvariants, and prevented infection in hamsters challenged with Omicron BA.1 intranasally. Structurally, both antibodies targeted a conserved quaternary epitope located at the interface between the N-terminal domain and subdomain 1, uncovering a site of vulnerability on SARS-CoV-2 spike. These antibodies prevented viral receptor engagement by locking the receptor-binding domain (RBD) of spike in the down conformation, revealing a mechanism of virus neutralization for non-RBD antibodies. Deep mutational scanning showed that SARS-CoV-2 could mutate to escape 12-19, but such mutations are rarely found in circulating viruses. Antibodies 12-16 and 12-19 hold promise as prophylactic agents for immunocompromised persons who do not respond robustly to COVID-19 vaccines., Competing Interests: Declaration of interests L.L., S.I., M.S.N., J.Y., Y.H., and D.D.H. are inventors on a provisional patent application for the new antibodies described in this manuscript, titled “Isolation, characterization, and sequences of potent and broadly neutralizing monoclonal antibodies against SARS-CoV-2 and its variants, as well as related coronaviruses” (63/271,627). D.D.H. is a co-founder of TaiMed Biologics and RenBio, a consultant to WuXi Biologics, Brii Biosciences, Apexigen, and Veru Inc., and a board director for Vicarious Surgical. J.D.B. consults for Apriorio Bio, Invivyd, Aerium Therapeutics, and the Vaccine Company. J.D.B. and B.D. are inventors on Fred Hutch licensed patents related to deep mutational scanning of viral proteins., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
4. P21-activated kinase 1 (PAK1)-mediated cytoskeleton rearrangement promotes SARS-CoV-2 entry and ACE2 autophagic degradation.
- Author
-
Liu M, Lu B, Li Y, Yuan S, Zhuang Z, Li G, Wang D, Ma L, Zhu J, Zhao J, Chan CC, Poon VK, Chik KK, Zhao Z, Xian H, Zhao J, Zhao J, Chan JF, and Zhang Y
- Subjects
- Humans, Angiotensin-Converting Enzyme 2 metabolism, Cytoskeleton, Peptidyl-Dipeptidase A metabolism, Spike Glycoprotein, Coronavirus genetics, Spike Glycoprotein, Coronavirus metabolism, COVID-19 Drug Treatment, COVID-19 pathology, COVID-19 virology, p21-Activated Kinases metabolism, SARS-CoV-2 metabolism, Virus Internalization
- Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), has had a significant impact on healthcare systems and economies worldwide. The continuous emergence of new viral strains presents a major challenge in the development of effective antiviral agents. Strategies that possess broad-spectrum antiviral activities are desirable to control SARS-CoV-2 infection. ACE2, an angiotensin-containing enzyme that prevents the overactivation of the renin angiotensin system, is the receptor for SARS-CoV-2. ACE2 interacts with the spike protein and facilitates viral attachment and entry into host cells. Yet, SARS-CoV-2 infection also promotes ACE2 degradation. Whether restoring ACE2 surface expression has an impact on SARS-CoV-2 infection is yet to be determined. Here, we show that the ACE2-spike complex is endocytosed and degraded via autophagy in a manner that depends on clathrin-mediated endocytosis and PAK1-mediated cytoskeleton rearrangement. In contrast, free cellular spike protein is selectively cleaved into S1 and S2 subunits in a lysosomal-dependent manner. Importantly, we show that the pan-PAK inhibitor FRAX-486 restores ACE2 surface expression and suppresses infection by different SARS-CoV-2 strains. FRAX-486-treated Syrian hamsters exhibit significantly decreased lung viral load and alleviated pulmonary inflammation compared with untreated hamsters. In summary, our findings have identified novel pathways regulating viral entry, as well as therapeutic targets and candidate compounds for controlling the emerging strains of SARS-CoV-2 infection., (© 2023. West China Hospital, Sichuan University.)
- Published
- 2023
- Full Text
- View/download PDF
5. Antibodies that neutralize all current SARS-CoV-2 variants of concern by conformational locking.
- Author
-
Liu L, Casner RG, Guo Y, Wang Q, Iketani S, Chan JF, Yu J, Dadonaite B, Nair MS, Mohri H, Reddem ER, Yuan S, Poon VK, Chan CC, Yuen KY, Sheng Z, Huang Y, Bloom JD, Shapiro L, and Ho DD
- Abstract
SARS-CoV-2 continues to evolve and evade most existing neutralizing antibodies, including all clinically authorized antibodies. We have isolated and characterized two human monoclonal antibodies, 12-16 and 12-19, which exhibited neutralizing activities against all SARS-CoV-2 variants tested, including BQ.1.1 and XBB.1.5. They also blocked infection in hamsters challenged with Omicron BA.1 intranasally. Structural analyses revealed both antibodies targeted a conserved quaternary epitope located at the interface between the N-terminal domain and subdomain 1, revealing a previously unrecognized site of vulnerability on SARS-CoV-2 spike. These antibodies prevent viral receptor engagement by locking the receptor-binding domain of spike in the down conformation, revealing a novel mechanism of virus neutralization for non-RBD antibodies. Deep mutational scanning showed that SARS-CoV-2 could mutate to escape 12-19, but the responsible mutations are rarely found in circulating viruses. Antibodies 12-16 and 12-19 hold promise as prophylactic agents for immunocompromised persons who do not respond robustly to COVID-19 vaccines.
- Published
- 2023
- Full Text
- View/download PDF
6. The spike receptor-binding motif G496S substitution determines the replication fitness of SARS-CoV-2 Omicron sublineage.
- Author
-
Liang R, Ye ZW, Ong CP, Qin Z, Xie Y, Fan Y, Tang K, Poon VK, Chan CC, Yang X, Cao H, Wang K, Sun H, Hu B, Cai JP, Luo C, Chik KK, Chu H, Zheng Y, Yuen KY, Chan JF, Jin DY, and Yuan S
- Subjects
- Animals, Antibodies, Monoclonal, Cricetinae, Humans, Mesocricetus, Mutation, Missense, Spike Glycoprotein, Coronavirus genetics, COVID-19, SARS-CoV-2 genetics
- Abstract
The replication and pathogenicity of SARS-CoV-2 Omicron BA.2 are comparable to that of BA.1 in experimental animal models. However, BA.2 has rapidly emerged to overtake BA.1 to become the predominant circulating SARS-CoV-2 variant worldwide. Here, we compared the replication fitness of BA.1 and BA.2 in cell culture and in the Syrian hamster model of COVID-19. Using a reverse genetics approach, we found that the BA.1-specific spike mutation G496S compromises its replication fitness, which may contribute to BA.1 being outcompeted by BA.2 in the real world. Additionally, the BA.1-unique G496S substitution confers differentiated sensitivity to therapeutic monoclonal antibodies, which partially recapitulates the immunoevasive phenotype of BA.1 and BA.2. In summary, our study identified G496S as an important determinant during the evolutionary trajectory of SARS-CoV-2.
- Published
- 2022
- Full Text
- View/download PDF
7. A molecularly engineered, broad-spectrum anti-coronavirus lectin inhibits SARS-CoV-2 and MERS-CoV infection in vivo.
- Author
-
Chan JF, Oh YJ, Yuan S, Chu H, Yeung ML, Canena D, Chan CC, Poon VK, Chan CC, Zhang AJ, Cai JP, Ye ZW, Wen L, Yuen TT, Chik KK, Shuai H, Wang Y, Hou Y, Luo C, Chan WM, Qin Z, Sit KY, Au WK, Legendre M, Zhu R, Hain L, Seferovic H, Tampé R, To KK, Chan KH, Thomas DG, Klausberger M, Xu C, Moon JJ, Stadlmann J, Penninger JM, Oostenbrink C, Hinterdorfer P, Yuen KY, and Markovitz DM
- Subjects
- Humans, SARS-CoV-2, Lectins pharmacology, Mannose pharmacology, Angiotensin-Converting Enzyme 2, Spike Glycoprotein, Coronavirus pharmacology, Antiviral Agents pharmacology, Middle East Respiratory Syndrome Coronavirus, COVID-19
- Abstract
"Pan-coronavirus" antivirals targeting conserved viral components can be designed. Here, we show that the rationally engineered H84T-banana lectin (H84T-BanLec), which specifically recognizes high mannose found on viral proteins but seldom on healthy human cells, potently inhibits Middle East respiratory syndrome coronavirus (MERS-CoV), severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) (including Omicron), and other human-pathogenic coronaviruses at nanomolar concentrations. H84T-BanLec protects against MERS-CoV and SARS-CoV-2 infection in vivo. Importantly, intranasally and intraperitoneally administered H84T-BanLec are comparably effective. Mechanistic assays show that H84T-BanLec targets virus entry. High-speed atomic force microscopy depicts real-time multimolecular associations of H84T-BanLec dimers with the SARS-CoV-2 spike trimer. Single-molecule force spectroscopy demonstrates binding of H84T-BanLec to multiple SARS-CoV-2 spike mannose sites with high affinity and that H84T-BanLec competes with SARS-CoV-2 spike for binding to cellular ACE2. Modeling experiments identify distinct high-mannose glycans in spike recognized by H84T-BanLec. The multiple H84T-BanLec binding sites on spike likely account for the drug compound's broad-spectrum antiviral activity and the lack of resistant mutants., Competing Interests: Declaration of interests J.F.-W.C. has received travel grants from Pfizer Corporation Hong Kong and Astellas Pharma Hong Kong Corporation Limited and was an invited speaker for Gilead Sciences Hong Kong Limited and Luminex Corporation. D.M.M. is an inventor on patents U.S. no. 8,865,867 “Lectins and Uses Thereof”; U.S. no. 9,481,717 “Lectins and Uses Thereof”; European no. 2558488 (DE, GB, FR) “Banana Lectins and Uses Thereof”; Chinese no. 103025756 “Banana Lectins and Uses Thereof”; and U.S. no. 10,450,355 “Lectins and Uses Thereof.”, (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
8. Coronaviruses exploit a host cysteine-aspartic protease for replication.
- Author
-
Chu H, Hou Y, Yang D, Wen L, Shuai H, Yoon C, Shi J, Chai Y, Yuen TT, Hu B, Li C, Zhao X, Wang Y, Huang X, Lee KS, Luo C, Cai JP, Poon VK, Chan CC, Zhang AJ, Yuan S, Sit KY, Foo DC, Au WK, Wong KK, Zhou J, Kok KH, Jin DY, Chan JF, and Yuen KY
- Subjects
- Animals, Apoptosis, Coronavirus Nucleocapsid Proteins immunology, Coronavirus Nucleocapsid Proteins metabolism, Cricetinae, Dipeptidyl Peptidase 4 genetics, Dipeptidyl Peptidase 4 metabolism, Humans, Interferons antagonists & inhibitors, Interferons immunology, Lung pathology, Mesocricetus, Mice, Middle East Respiratory Syndrome Coronavirus, Severe acute respiratory syndrome-related coronavirus, SARS-CoV-2, Survival Rate, Weight Loss, Aspartic Acid metabolism, Caspase 6 metabolism, Coronavirus growth & development, Coronavirus pathogenicity, Coronavirus Infections enzymology, Coronavirus Infections virology, Cysteine metabolism, Host-Pathogen Interactions, Virus Replication
- Abstract
Highly pathogenic coronaviruses, including severe acute respiratory syndrome coronavirus 2 (refs.
1,2 ) (SARS-CoV-2), Middle East respiratory syndrome coronavirus3 (MERS-CoV) and SARS-CoV-1 (ref.4 ), vary in their transmissibility and pathogenicity. However, infection by all three viruses results in substantial apoptosis in cell culture5-7 and in patient tissues8-10 , suggesting a potential link between apoptosis and pathogenesis of coronaviruses. Here we show that caspase-6, a cysteine-aspartic protease of the apoptosis cascade, serves as an important host factor for efficient coronavirus replication. We demonstrate that caspase-6 cleaves coronavirus nucleocapsid proteins, generating fragments that serve as interferon antagonists, thus facilitating virus replication. Inhibition of caspase-6 substantially attenuates lung pathology and body weight loss in golden Syrian hamsters infected with SARS-CoV-2 and improves the survival of mice expressing human DPP4 that are infected with mouse-adapted MERS-CoV. Our study reveals how coronaviruses exploit a component of the host apoptosis cascade to facilitate virus replication., (© 2022. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2022
- Full Text
- View/download PDF
9. A small animal model of chronic hepatitis E infection using immunocompromised rats.
- Author
-
Sridhar S, Wu S, Situ J, Shun EH, Li Z, Zhang AJ, Hui K, Fong CH, Poon VK, Chew NF, Yip CC, Chan WM, Cai JP, and Yuen KY
- Abstract
Background & Aims: HEV variants such as swine genotypes within Paslahepevirus species balayani (HEV-A) and rat HEV ( Rocahepevirus ratti ; HEV-C1) cause chronic hepatitis E in immunocompromised individuals. There are few reliable and accessible small animal models that accurately reflect chronic HEV infection. We aimed to develop an immunocompromised rat model of chronic hepatitis E infection., Methods: In this animal model infection study, rats were immunosuppressed with a drug combination (prednisolone, tacrolimus, and mycophenolate mofetil) commonly taken by transplant recipients. Rats were challenged with human- and rat-derived HEV-C1 strains or a human-derived HEV-A strain. Viral load, liver function, liver histology, humoural, and cellular immune responses were monitored., Results: A high-dose (HD) immunosuppressive regimen consistently prolonged human- and rat-derived HEV-C1 infection in rats (up to 12 weeks post infection) compared with transient infections in low-dose (LD) immunosuppressant-treated and immunocompetent (IC) rats. Mean HEV-C1 viral loads in stool, serum, and liver tissue were higher in HD regimen-treated rats than in LD or IC rats ( p <0.05). Alanine aminotransferase elevation was observed in chronically infected rats, which was consistent with histological hepatitis and HEV-C1 antigen expression in liver tissue. None (0/6) of the HD regimen-treated, 5/6 LD regimen-treated, and 6/6 IC rats developed antibodies to HEV-C1 in species-specific immunoblots. Reversal of immunosuppression was associated with clearance of viraemia and restoration of HEV-C1-specific humoural and cellular immune responses in HD regimen-treated rats, mimicking patterns in treated patients with chronic hepatitis E. Viral load suppression was observed with i.p. ribavirin treatment. HD regimen-treated rats remained unsusceptible to HEV-A infection., Conclusions: We developed a scalable immunosuppressed rat model of chronic hepatitis E that closely mimics this infection phenotype in transplant recipients., Lay Summary: Convenient small animal models are required for the study of chronic hepatitis E in humans. We developed an animal model of chronic hepatitis E by suppressing immune responses of rats with drugs commonly taken by humans as organ transplant rejection prophylaxis. This model closely mimicked features of chronic hepatitis E in humans., Competing Interests: SS has received speaker’s honoraria from Abbott Medical Laboratories. The other authors report that there are no competing interests to declare. Please refer to the accompanying ICMJE disclosure forms for further details., (© 2022 The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
10. Pathogenicity, transmissibility, and fitness of SARS-CoV-2 Omicron in Syrian hamsters.
- Author
-
Yuan S, Ye ZW, Liang R, Tang K, Zhang AJ, Lu G, Ong CP, Man Poon VK, Chan CC, Mok BW, Qin Z, Xie Y, Chu AW, Chan WM, Ip JD, Sun H, Tsang JO, Yuen TT, Chik KK, Chan CC, Cai JP, Luo C, Lu L, Yip CC, Chu H, To KK, Chen H, Jin DY, Yuen KY, and Chan JF
- Subjects
- Animals, Disease Models, Animal, Mesocricetus, Virulence, COVID-19 transmission, SARS-CoV-2 pathogenicity
- Abstract
The in vivo pathogenicity, transmissibility, and fitness of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron (B.1.1.529) variant are not well understood. We compared these virological attributes of this new variant of concern (VOC) with those of the Delta (B.1.617.2) variant in a Syrian hamster model of COVID-19. Omicron-infected hamsters lost significantly less body weight and exhibited reduced clinical scores, respiratory tract viral burdens, cytokine and chemokine dysregulation, and lung damage than Delta-infected hamsters. Both variants were highly transmissible through contact transmission. In noncontact transmission studies Omicron demonstrated similar or higher transmissibility than Delta. Delta outcompeted Omicron without selection pressure, but this scenario changed once immune selection pressure with neutralizing antibodies-active against Delta but poorly active against Omicron-was introduced. Next-generation vaccines and antivirals effective against this new VOC are therefore urgently needed.
- Published
- 2022
- Full Text
- View/download PDF
11. A broadly neutralizing antibody protects Syrian hamsters against SARS-CoV-2 Omicron challenge.
- Author
-
Zhou B, Zhou R, Tang B, Chan JF, Luo M, Peng Q, Yuan S, Liu H, Mok BW, Chen B, Wang P, Poon VK, Chu H, Chan CC, Tsang JO, Chan CC, Au KK, Man HO, Lu L, To KK, Chen H, Yuen KY, Dang S, and Chen Z
- Subjects
- Animals, BNT162 Vaccine, Cricetinae, Humans, Mesocricetus, SARS-CoV-2 genetics, Spike Glycoprotein, Coronavirus genetics, Antibodies, Viral immunology, Broadly Neutralizing Antibodies immunology, COVID-19 prevention & control
- Abstract
The strikingly high transmissibility and antibody evasion of SARS-CoV-2 Omicron variants have posed great challenges to the efficacy of current vaccines and antibody immunotherapy. Here, we screen 34 BNT162b2-vaccinees and isolate a public broadly neutralizing antibody ZCB11 derived from the IGHV1-58 family. ZCB11 targets viral receptor-binding domain specifically and neutralizes all SARS-CoV-2 variants of concern, especially with great potency against authentic Omicron and Delta variants. Pseudovirus-based mapping of 57 naturally occurred spike mutations or deletions reveals that S371L results in 11-fold neutralization resistance, but it is rescued by compensating mutations in Omicron variants. Cryo-EM analysis demonstrates that ZCB11 heavy chain predominantly interacts with Omicron spike trimer with receptor-binding domain in up conformation blocking ACE2 binding. In addition, prophylactic or therapeutic ZCB11 administration protects lung infection against Omicron viral challenge in golden Syrian hamsters. These results suggest that vaccine-induced ZCB11 is a promising broadly neutralizing antibody for biomedical interventions against pandemic SARS-CoV-2., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
12. Author Correction: SARS-CoV-2 infection induces inflammatory bone loss in golden Syrian hamsters.
- Author
-
Qiao W, Lau HE, Xie H, Poon VK, Chan CC, Chu H, Yuan S, Yuen TT, Chik KK, Tsang JO, Chan CC, Cai JP, Luo C, Yuen KY, Cheung KM, Chan JF, and Yeung KW
- Published
- 2022
- Full Text
- View/download PDF
13. An antibody class with a common CDRH3 motif broadly neutralizes sarbecoviruses.
- Author
-
Liu L, Iketani S, Guo Y, Reddem ER, Casner RG, Nair MS, Yu J, Chan JF, Wang M, Cerutti G, Li Z, Morano NC, Castagna CD, Corredor L, Chu H, Yuan S, Poon VK, Chan CC, Chen Z, Luo Y, Cunningham M, Chavez A, Yin MT, Perlin DS, Tsuji M, Yuen KY, Kwong PD, Sheng Z, Huang Y, Shapiro L, and Ho DD
- Subjects
- Antibodies, Viral, Humans, Immunoglobulin Isotypes, Spike Glycoprotein, Coronavirus, COVID-19, SARS-CoV-2
- Abstract
The devastation caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has made clear the importance of pandemic preparedness. To address future zoonotic outbreaks due to related viruses in the sarbecovirus subgenus, we identified a human monoclonal antibody, 10-40, that neutralized or bound all sarbecoviruses tested in vitro and protected against SARS-CoV-2 and SARS-CoV in vivo. Comparative studies with other receptor-binding domain (RBD)-directed antibodies showed 10-40 to have the greatest breadth against sarbecoviruses, suggesting that 10-40 is a promising agent for pandemic preparedness. Moreover, structural analyses on 10-40 and similar antibodies not only defined an epitope cluster in the inner face of the RBD that is well conserved among sarbecoviruses but also uncovered a distinct antibody class with a common CDRH3 motif. Our analyses also suggested that elicitation of this class of antibodies may not be overly difficult, an observation that bodes well for the development of a pan-sarbecovirus vaccine.
- Published
- 2022
- Full Text
- View/download PDF
14. SARS-CoV-2 infection induces inflammatory bone loss in golden Syrian hamsters.
- Author
-
Qiao W, Lau HE, Xie H, Poon VK, Chan CC, Chu H, Yuan S, Yuen TT, Chik KK, Tsang JO, Chan CC, Cai JP, Luo C, Yuen KY, Cheung KM, Chan JF, and Yeung KW
- Subjects
- Animals, Cricetinae, Disease Models, Animal, Humans, Mesocricetus, SARS-CoV-2, COVID-19 complications
- Abstract
Extrapulmonary complications of different organ systems have been increasingly recognized in patients with severe or chronic Coronavirus Disease 2019 (COVID-19). However, limited information on the skeletal complications of COVID-19 is known, even though inflammatory diseases of the respiratory tract have been known to perturb bone metabolism and cause pathological bone loss. In this study, we characterize the effects of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection on bone metabolism in an established golden Syrian hamster model for COVID-19. SARS-CoV-2 causes significant multifocal loss of bone trabeculae in the long bones and lumbar vertebrae of all infected hamsters. Moreover, we show that the bone loss is associated with SARS-CoV-2-induced cytokine dysregulation, as the circulating pro-inflammatory cytokines not only upregulate osteoclastic differentiation in bone tissues, but also trigger an amplified pro-inflammatory cascade in the skeletal tissues to augment their pro-osteoclastogenesis effect. Our findings suggest that pathological bone loss may be a neglected complication which warrants more extensive investigations during the long-term follow-up of COVID-19 patients. The benefits of potential prophylactic and therapeutic interventions against pathological bone loss should be further evaluated., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
15. A novel linker-immunodominant site (LIS) vaccine targeting the SARS-CoV-2 spike protein protects against severe COVID-19 in Syrian hamsters.
- Author
-
Zhang BZ, Wang X, Yuan S, Li W, Dou Y, Poon VK, Chan CC, Cai JP, Chik KK, Tang K, Chan CC, Hu YF, Hu JC, Badea SR, Gong HR, Lin X, Chu H, Li X, To KK, Liu L, Chen Z, Hung IF, Yuen KY, Chan JF, and Huang JD
- Subjects
- Animals, Cricetinae, Female, HEK293 Cells, Humans, Male, Mesocricetus, Mice, Mice, Inbred BALB C, Vaccination, COVID-19 prevention & control, COVID-19 Vaccines immunology, Immunodominant Epitopes immunology, SARS-CoV-2 immunology, Spike Glycoprotein, Coronavirus immunology
- Abstract
The Coronavirus Disease 2019 (COVID-19) pandemic is unlikely to abate until sufficient herd immunity is built up by either natural infection or vaccination. We previously identified ten linear immunodominant sites on the SARS-CoV-2 spike protein of which four are located within the RBD. Therefore, we designed two linkerimmunodominant site (LIS) vaccine candidates which are composed of four immunodominant sites within the RBD (RBD-ID) or all the 10 immunodominant sites within the whole spike (S-ID). They were administered by subcutaneous injection and were tested for immunogenicity and in vivo protective efficacy in a hamster model for COVID-19. We showed that the S-ID vaccine induced significantly better neutralizing antibody response than RBD-ID and alum control. As expected, hamsters vaccinated by S-ID had significantly less body weight loss, lung viral load, and histopathological changes of pneumonia. The S-ID has the potential to be an effective vaccine for protection against COVID-19.
- Published
- 2021
- Full Text
- View/download PDF
16. Beneficial effect of combinational methylprednisolone and remdesivir in hamster model of SARS-CoV-2 infection.
- Author
-
Ye ZW, Yuan S, Chan JF, Zhang AJ, Yu CY, Ong CP, Yang D, Chan CC, Tang K, Cao J, Poon VK, Chan CC, Cai JP, Chu H, Yuen KY, and Jin DY
- Subjects
- Adenosine Monophosphate pharmacology, Adenosine Monophosphate therapeutic use, Alanine pharmacology, Alanine therapeutic use, Animals, Antibodies, Viral blood, Antiviral Agents pharmacology, COVID-19 pathology, COVID-19 virology, Cytokines biosynthesis, Cytokines immunology, Disease Models, Animal, Drug Therapy, Combination, Female, Humans, Macrophages immunology, Macrophages virology, Male, Mesocricetus, Methylprednisolone pharmacology, RNA, Viral, Respiratory System pathology, Respiratory System virology, SARS-CoV-2 physiology, Spike Glycoprotein, Coronavirus immunology, Viral Load drug effects, Virus Replication drug effects, Adenosine Monophosphate analogs & derivatives, Alanine analogs & derivatives, Antiviral Agents therapeutic use, Methylprednisolone therapeutic use, SARS-CoV-2 drug effects, COVID-19 Drug Treatment
- Abstract
Effective treatments for coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are urgently needed. Dexamethasone has been shown to confer survival benefits to certain groups of hospitalized patients, but whether glucocorticoids such as dexamethasone and methylprednisolone should be used together with antivirals to prevent a boost of SARS-CoV-2 replication remains to be determined. Here, we show the beneficial effect of methylprednisolone alone and in combination with remdesivir in the hamster model of SARS-CoV-2 infection. Treatment with methylprednisolone boosted RNA replication of SARS-CoV-2 but suppressed viral induction of proinflammatory cytokines in human monocyte-derived macrophages. Although methylprednisolone monotherapy alleviated body weight loss as well as nasal and pulmonary inflammation, viral loads increased and antibody response against the receptor-binding domain of spike protein attenuated. In contrast, a combination of methylprednisolone with remdesivir not only prevented body weight loss and inflammation, but also dampened viral protein expression and viral loads. In addition, the suppressive effect of methylprednisolone on antibody response was alleviated in the presence of remdesivir. Thus, combinational anti-inflammatory and antiviral therapy might be an effective, safer and more versatile treatment option for COVID-19. These data support testing of the efficacy of a combination of methylprednisolone and remdesivir for the treatment of COVID-19 in randomized controlled clinical trials.
- Published
- 2021
- Full Text
- View/download PDF
17. ACE2-like carboxypeptidase B38-CAP protects from SARS-CoV-2-induced lung injury.
- Author
-
Yamaguchi T, Hoshizaki M, Minato T, Nirasawa S, Asaka MN, Niiyama M, Imai M, Uda A, Chan JF, Takahashi S, An J, Saku A, Nukiwa R, Utsumi D, Kiso M, Yasuhara A, Poon VK, Chan CC, Fujino Y, Motoyama S, Nagata S, Penninger JM, Kamada H, Yuen KY, Kamitani W, Maeda K, Kawaoka Y, Yasutomi Y, Imai Y, and Kuba K
- Subjects
- Acute Lung Injury, Angiotensin II, Animals, COVID-19 pathology, Carboxypeptidases, Chlorocebus aethiops, Cricetinae, Disease Models, Animal, Female, Humans, Lung pathology, Male, Mice, Mice, Transgenic, Pulmonary Edema pathology, Pulmonary Edema prevention & control, Spike Glycoprotein, Coronavirus drug effects, Vero Cells, Angiotensin-Converting Enzyme 2 metabolism, COVID-19 prevention & control, Lung Injury prevention & control, SARS-CoV-2 drug effects, Virus Internalization drug effects, COVID-19 Drug Treatment
- Abstract
Angiotensin-converting enzyme 2 (ACE2) is a receptor for cell entry of SARS-CoV-2, and recombinant soluble ACE2 protein inhibits SARS-CoV-2 infection as a decoy. ACE2 is a carboxypeptidase that degrades angiotensin II, thereby improving the pathologies of cardiovascular disease or acute lung injury. Here we show that B38-CAP, an ACE2-like enzyme, is protective against SARS-CoV-2-induced lung injury. Endogenous ACE2 expression is downregulated in the lungs of SARS-CoV-2-infected hamsters, leading to elevation of angiotensin II levels. Recombinant Spike also downregulates ACE2 expression and worsens the symptoms of acid-induced lung injury. B38-CAP does not neutralize cell entry of SARS-CoV-2. However, B38-CAP treatment improves the pathologies of Spike-augmented acid-induced lung injury. In SARS-CoV-2-infected hamsters or human ACE2 transgenic mice, B38-CAP significantly improves lung edema and pathologies of lung injury. These results provide the first in vivo evidence that increasing ACE2-like enzymatic activity is a potential therapeutic strategy to alleviate lung pathologies in COVID-19 patients., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
18. Emerging SARS-CoV-2 variants expand species tropism to murines.
- Author
-
Shuai H, Chan JF, Yuen TT, Yoon C, Hu JC, Wen L, Hu B, Yang D, Wang Y, Hou Y, Huang X, Chai Y, Chan CC, Poon VK, Lu L, Zhang RQ, Chan WM, Ip JD, Chu AW, Hu YF, Cai JP, Chan KH, Zhou J, Sridhar S, Zhang BZ, Yuan S, Zhang AJ, Huang JD, To KK, Yuen KY, and Chu H
- Subjects
- Angiotensin-Converting Enzyme 2 genetics, Angiotensin-Converting Enzyme 2 metabolism, Animals, Antibodies, Viral blood, Antibodies, Viral immunology, COVID-19 virology, Female, Humans, Lung pathology, Lung virology, Mice, Mice, Inbred C57BL, Mice, Transgenic, Neutralization Tests, Nucleocapsid Proteins immunology, Nucleocapsid Proteins metabolism, RNA, Viral analysis, RNA, Viral metabolism, Rats, Rats, Sprague-Dawley, SARS-CoV-2 immunology, SARS-CoV-2 isolation & purification, Turbinates pathology, Turbinates virology, Virus Internalization, COVID-19 pathology, SARS-CoV-2 physiology, Viral Tropism
- Abstract
Background: Wildtype mice are not susceptible to SARS-CoV-2 infection. Emerging SARS-CoV-2 variants, including B.1.1.7, B.1.351, P.1, and P.3, contain mutations in spike that has been suggested to associate with an increased recognition of mouse ACE2, raising the postulation that these SARS-CoV-2 variants may have evolved to expand species tropism to wildtype mouse and potentially other murines. Our study evaluated this possibility with substantial public health importance., Methods: We investigated the capacity of wildtype (WT) SARS-CoV-2 and SARS-CoV-2 variants in infecting mice (Mus musculus) and rats (Rattus norvegicus) under in vitro and in vivo settings. Susceptibility to infection was evaluated with RT-qPCR, plaque assays, immunohistological stainings, and neutralization assays., Findings: Our results reveal that B.1.1.7 and other N501Y-carrying variants but not WT SARS-CoV-2 can infect wildtype mice. High viral genome copies and high infectious virus particle titres are recovered from the nasal turbinate and lung of B.1.1.7-inocluated mice for 4-to-7 days post infection. In agreement with these observations, robust expression of viral nucleocapsid protein and histopathological changes are detected from the nasal turbinate and lung of B.1.1.7-inocluated mice but not that of the WT SARS-CoV-2-inoculated mice. Similarly, B.1.1.7 readily infects wildtype rats with production of infectious virus particles., Interpretation: Our study provides direct evidence that the SARS-CoV-2 variant, B.1.1.7, as well as other N501Y-carrying variants including B.1.351 and P.3, has gained the capability to expand species tropism to murines and public health measures including stringent murine control should be implemented to facilitate the control of the ongoing pandemic., Funding: A full list of funding bodies that contributed to this study can be found in the Acknowledgements section., Competing Interests: Declaration of Competing Interest Authors declare that they have no competing interests., (Copyright © 2021 The Author(s). Published by Elsevier B.V. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
19. Severe Acute Respiratory Syndrome Coronavirus 2 Infects and Damages the Mature and Immature Olfactory Sensory Neurons of Hamsters.
- Author
-
Zhang AJ, Lee AC, Chu H, Chan JF, Fan Z, Li C, Liu F, Chen Y, Yuan S, Poon VK, Chan CC, Cai JP, Wu KL, Sridhar S, Chan YS, and Yuen KY
- Subjects
- Animals, Cricetinae, Humans, Mesocricetus, Olfactory Mucosa, SARS-CoV-2, COVID-19, Olfactory Receptor Neurons
- Abstract
Background: Coronavirus disease 2019 (COVID-19) is primarily an acute respiratory tract infection. Distinctively, a substantial proportion of COVID-19 patients develop olfactory dysfunction. Especially in young patients, loss of smell can be the first or only symptom. The roles of inflammatory obstruction of the olfactory clefts, inflammatory cytokines affecting olfactory neuronal function, destruction of olfactory neurons or their supporting cells, and direct invasion of olfactory bulbs in causing olfactory dysfunction are uncertain., Methods: We investigated the location for the pathogenesis of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) from the olfactory epithelium (OE) to the olfactory bulb in golden Syrian hamsters., Results: After intranasal inoculation with SARS-CoV-2, inflammatory cell infiltration and proinflammatory cytokine/chemokine responses were detected in the nasal turbinate tissues. The responses peaked between 2 and 4 days postinfection, with the highest viral load detected at day 2 postinfection. In addition to the pseudo-columnar ciliated respiratory epithelial cells, SARS-CoV-2 viral antigens were also detected in the mature olfactory sensory neurons labeled by olfactory marker protein, in the less mature olfactory neurons labeled by neuron-specific class III β-tubulin at the more basal position, and in the sustentacular cells, resulting in apoptosis and severe destruction of the OE. During the entire course of infection, SARS-CoV-2 viral antigens were not detected in the olfactory bulb., Conclusions: In addition to acute inflammation at the OE, infection of mature and immature olfactory neurons and the supporting sustentacular cells by SARS-CoV-2 may contribute to the unique olfactory dysfunction related to COVID-19, which is not reported with SARS-CoV-2., (© The Author(s) 2020. Published by Oxford University Press for the Infectious Diseases Society of America. All rights reserved. For permissions, e-mail: journals.permissions@oup.com.)
- Published
- 2021
- Full Text
- View/download PDF
20. Targeting highly pathogenic coronavirus-induced apoptosis reduces viral pathogenesis and disease severity.
- Author
-
Chu H, Shuai H, Hou Y, Zhang X, Wen L, Huang X, Hu B, Yang D, Wang Y, Yoon C, Wong BH, Li C, Zhao X, Poon VK, Cai JP, Wong KK, Yeung ML, Zhou J, Au-Yeung RK, Yuan S, Jin DY, Kok KH, Perlman S, Chan JF, and Yuen KY
- Subjects
- Adenine analogs & derivatives, Adenine pharmacology, Angiotensin-Converting Enzyme 2 genetics, Animals, Apoptosis physiology, COVID-19 etiology, COVID-19 pathology, Cell Line, Coronavirus Infections etiology, Coronavirus Infections pathology, Dipeptidyl Peptidase 4 genetics, Epithelial Cells virology, Female, Humans, Indoles pharmacology, Lung virology, Male, Mice, Transgenic, eIF-2 Kinase antagonists & inhibitors, eIF-2 Kinase genetics, Antiviral Agents pharmacology, Apoptosis drug effects, Coronavirus Infections drug therapy, eIF-2 Kinase metabolism, COVID-19 Drug Treatment
- Abstract
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., (Copyright © 2021 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works. Distributed under a Creative Commons Attribution NonCommercial License 4.0 (CC BY-NC).)
- Published
- 2021
- Full Text
- View/download PDF
21. Coinfection by Severe Acute Respiratory Syndrome Coronavirus 2 and Influenza A(H1N1)pdm09 Virus Enhances the Severity of Pneumonia in Golden Syrian Hamsters.
- Author
-
Zhang AJ, Lee AC, Chan JF, Liu F, Li C, Chen Y, Chu H, Lau SY, Wang P, Chan CC, Poon VK, Yuan S, To KK, Chen H, and Yuen KY
- Subjects
- Animals, Cricetinae, Disease Models, Animal, Humans, Mesocricetus, Mice, SARS-CoV-2, COVID-19, Coinfection, Influenza A Virus, H1N1 Subtype, Influenza, Human
- Abstract
Background: Clinical outcomes of the interaction between the co-circulating pandemic severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and seasonal influenza viruses are unknown., Methods: We established a golden Syrian hamster model coinfected by SARS-CoV-2 and mouse-adapted A(H1N1)pdm09 simultaneously or sequentially. The weight loss, clinical scores, histopathological changes, viral load and titer, and serum neutralizing antibody titer were compared with hamsters challenged by either virus., Results: Coinfected hamsters had more weight loss, more severe lung inflammatory damage, and tissue cytokine/chemokine expression. Lung viral load, infectious virus titers, and virus antigen expression suggested that hamsters were generally more susceptible to SARS-CoV-2 than to A(H1N1)pdm09. Sequential coinfection with A(H1N1)pdm09 one day prior to SARS-CoV-2 exposure resulted in a lower lung SARS-CoV-2 titer and viral load than with SARS-CoV-2 monoinfection, but a higher lung A(H1N1)pdm09 viral load. Coinfection also increased intestinal inflammation with more SARS-CoV-2 nucleoprotein expression in enterocytes. Simultaneous coinfection was associated with delay in resolution of lung damage, lower serum SARS-CoV-2 neutralizing antibody, and longer SARS-CoV-2 shedding in oral swabs compared to that of SARS-CoV-2 monoinfection., Conclusions: Simultaneous or sequential coinfection by SARS-CoV-2 and A(H1N1)pdm09 caused more severe disease than monoinfection by either virus in hamsters. Prior A(H1N1)pdm09 infection lowered SARS-CoV-2 pulmonary viral loads but enhanced lung damage. Whole-population influenza vaccination for prevention of coinfection, and multiplex molecular diagnostics for both viruses to achieve early initiation of antiviral treatment for improvement of clinical outcome should be considered., (© The Author(s) 2020. Published by Oxford University Press for the Infectious Diseases Society of America.)
- Published
- 2021
- Full Text
- View/download PDF
22. Clofazimine broadly inhibits coronaviruses including SARS-CoV-2.
- Author
-
Yuan S, Yin X, Meng X, Chan JF, Ye ZW, Riva L, Pache L, Chan CC, Lai PM, Chan CC, Poon VK, Lee AC, Matsunaga N, Pu Y, Yuen CK, Cao J, Liang R, Tang K, Sheng L, Du Y, Xu W, Lau CY, Sit KY, Au WK, Wang R, Zhang YY, Tang YD, Clausen TM, Pihl J, Oh J, Sze KH, Zhang AJ, Chu H, Kok KH, Wang D, Cai XH, Esko JD, Hung IF, Li RA, Chen H, Sun H, Jin DY, Sun R, Chanda SK, and Yuen KY
- Subjects
- Adenosine Monophosphate analogs & derivatives, Adenosine Monophosphate pharmacology, Adenosine Monophosphate therapeutic use, Alanine analogs & derivatives, Alanine pharmacology, Alanine therapeutic use, Animals, Anti-Inflammatory Agents pharmacokinetics, Anti-Inflammatory Agents pharmacology, Anti-Inflammatory Agents therapeutic use, Antiviral Agents pharmacokinetics, Antiviral Agents therapeutic use, Biological Availability, Cell Fusion, Cell Line, Clofazimine pharmacokinetics, Clofazimine therapeutic use, Coronavirus growth & development, Coronavirus pathogenicity, Cricetinae, DNA Helicases antagonists & inhibitors, Drug Synergism, Female, Humans, Life Cycle Stages drug effects, Male, Mesocricetus, Pre-Exposure Prophylaxis, SARS-CoV-2 growth & development, Species Specificity, Spike Glycoprotein, Coronavirus antagonists & inhibitors, Transcription, Genetic drug effects, Transcription, Genetic genetics, Antiviral Agents pharmacology, Clofazimine pharmacology, Coronavirus classification, Coronavirus drug effects, SARS-CoV-2 drug effects
- 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 2003
1 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
- Full Text
- View/download PDF
23. Robust SARS-CoV-2 infection in nasal turbinates after treatment with systemic neutralizing antibodies.
- Author
-
Zhou D, Chan JF, Zhou B, Zhou R, Li S, Shan S, Liu L, Zhang AJ, Chen SJ, Chan CC, Xu H, Poon VK, Yuan S, Li C, Chik KK, Chan CC, Cao J, Chan CY, Kwan KY, Du Z, Lau TT, Zhang Q, Zhou J, To KK, Zhang L, Ho DD, Yuen KY, and Chen Z
- Subjects
- Angiotensin-Converting Enzyme 2 physiology, Animals, Antibodies, Neutralizing immunology, Antibodies, Viral immunology, COVID-19 immunology, COVID-19 virology, Cricetinae, Female, HEK293 Cells, Humans, Male, Mesocricetus, Viral Load, Antibodies, Neutralizing therapeutic use, Antibodies, Viral therapeutic use, COVID-19 therapy, SARS-CoV-2 immunology, Turbinates virology
- Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is characterized by a burst in the upper respiratory portal for high transmissibility. To determine human neutralizing antibodies (HuNAbs) for entry protection, we tested three potent HuNAbs (IC
50 range, 0.0007-0.35 μg/mL) against live SARS-CoV-2 infection in the golden Syrian hamster model. These HuNAbs inhibit SARS-CoV-2 infection by competing with human angiotensin converting enzyme-2 for binding to the viral receptor binding domain (RBD). Prophylactic intraperitoneal or intranasal injection of individual HuNAb or DNA vaccination significantly reduces infection in the lungs but not in the nasal turbinates of hamsters intranasally challenged with SARS-CoV-2. Although postchallenge HuNAb therapy suppresses viral loads and lung damage, robust infection is observed in nasal turbinates treated within 1-3 days. Our findings demonstrate that systemic HuNAb suppresses SARS-CoV-2 replication and injury in lungs; however, robust viral infection in nasal turbinate may outcompete the antibody with significant implications to subprotection, reinfection, and vaccine., Competing Interests: Declaration of interests J.F.-W.C. has received travel grants from Pfizer Corporation Hong Kong and Astellas Pharma Hong Kong Corporation Limited and was an invited speaker for Gilead Sciences Hong Kong Limited and Luminex Corporation. The other authors declare no conflicts of interest except for a provisional patent application filed for human monoclonal antibodies generated in our laboratory by the University of Hong Kong. The funding sources had no role in study design, data collection, analysis, interpretation, or writing of the report., (Copyright © 2021 Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
24. Simulation of the Clinical and Pathological Manifestations of Coronavirus Disease 2019 (COVID-19) in a Golden Syrian Hamster Model: Implications for Disease Pathogenesis and Transmissibility.
- Author
-
Chan JF, Zhang AJ, Yuan S, Poon VK, Chan CC, Lee AC, Chan WM, Fan Z, Tsoi HW, Wen L, Liang R, Cao J, Chen Y, Tang K, Luo C, Cai JP, Kok KH, Chu H, Chan KH, Sridhar S, Chen Z, Chen H, To KK, and Yuen KY
- Subjects
- Animals, Antibodies, Neutralizing blood, Antibodies, Viral blood, COVID-19 blood, COVID-19 immunology, Cricetinae, Disease Models, Animal, Lung virology, Molecular Docking Simulation, Viral Load, COVID-19 pathology, SARS-CoV-2
- Abstract
Background: A physiological small-animal model that resembles COVID-19 with low mortality is lacking., Methods: Molecular docking on the binding between angiotensin-converting enzyme 2 (ACE2) of common laboratory mammals and the receptor-binding domain of the surface spike protein of SARS-CoV-2 suggested that the golden Syrian hamster is an option. Virus challenge, contact transmission, and passive immunoprophylaxis studies were performed. Serial organ tissues and blood were harvested for histopathology, viral load and titer, chemokine/cytokine level, and neutralizing antibody titer., Results: The Syrian hamster could be consistently infected by SARS-CoV-2. Maximal clinical signs of rapid breathing, weight loss, histopathological changes from the initial exudative phase of diffuse alveolar damage with extensive apoptosis to the later proliferative phase of tissue repair, airway and intestinal involvement with viral nucleocapsid protein expression, high lung viral load, and spleen and lymphoid atrophy associated with marked chemokine/cytokine activation were observed within the first week of virus challenge. The mean lung virus titer was between 105 and 107 TCID50/g. Challenged index hamsters consistently infected naive contact hamsters housed within the same cages, resulting in similar pathology but not weight loss. All infected hamsters recovered and developed mean serum neutralizing antibody titers ≥1:427 14 days postchallenge. Immunoprophylaxis with early convalescent serum achieved significant decrease in lung viral load but not in lung pathology. No consistent nonsynonymous adaptive mutation of the spike was found in viruses isolated from the infected hamsters., Conclusions: Besides satisfying Koch's postulates, this readily available hamster model is an important tool for studying transmission, pathogenesis, treatment, and vaccination against SARS-CoV-2., (© The Author(s) 2020. Published by Oxford University Press for the Infectious Diseases Society of America. All rights reserved. For permissions, e-mail: journals.permissions@oup.com.)
- Published
- 2020
- Full Text
- View/download PDF
25. Surgical Mask Partition Reduces the Risk of Noncontact Transmission in a Golden Syrian Hamster Model for Coronavirus Disease 2019 (COVID-19).
- Author
-
Chan JF, Yuan S, Zhang AJ, Poon VK, Chan CC, Lee AC, Fan Z, Li C, Liang R, Cao J, Tang K, Luo C, Cheng VC, Cai JP, Chu H, Chan KH, To KK, Sridhar S, and Yuen KY
- Subjects
- Animals, Coronavirus pathogenicity, Cricetinae, Female, Male, Pandemics, Viral Load, COVID-19 transmission, Masks, SARS-CoV-2 pathogenicity
- Abstract
Background: Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is believed to be mostly transmitted by medium- to large-sized respiratory droplets, although airborne transmission may be possible in healthcare settings involving aerosol-generating procedures. Exposure to respiratory droplets can theoretically be reduced by surgical mask usage. However, there is a lack of experimental evidence supporting surgical mask usage for prevention of COVID-19., Methods: We used a well-established golden Syrian hamster SARS-CoV-2 model. We placed SARS-CoV-2-challenged index hamsters and naive hamsters into closed system units each comprising 2 different cages separated by a polyvinyl chloride air porous partition with unidirectional airflow within the isolator. The effect of a surgical mask partition placed between the cages was investigated. Besides clinical scoring, hamster specimens were tested for viral load, histopathology, and viral nucleocapsid antigen expression., Results: Noncontact transmission was found in 66.7% (10/15) of exposed naive hamsters. Surgical mask partition for challenged index or naive hamsters significantly reduced transmission to 25% (6/24, P = .018). Surgical mask partition for challenged index hamsters significantly reduced transmission to only 16.7% (2/12, P = .019) of exposed naive hamsters. Unlike the severe manifestations of challenged hamsters, infected naive hamsters had lower clinical scores, milder histopathological changes, and lower viral nucleocapsid antigen expression in respiratory tract tissues., Conclusions: SARS-CoV-2 could be transmitted by respiratory droplets or airborne droplet nuclei which could be reduced by surgical mask partition in the hamster model. This is the first in vivo experimental evidence to support the possible benefit of surgical mask in prevention of COVID-19 transmission, especially when masks were worn by infected individuals., (© The Author(s) 2020. Published by Oxford University Press for the Infectious Diseases Society of America. All rights reserved. For permissions, e-mail: journals.permissions@oup.com.)
- Published
- 2020
- Full Text
- View/download PDF
26. Oral SARS-CoV-2 Inoculation Establishes Subclinical Respiratory Infection with Virus Shedding in Golden Syrian Hamsters.
- Author
-
Lee AC, Zhang AJ, Chan JF, Li C, Fan Z, Liu F, Chen Y, Liang R, Sridhar S, Cai JP, Poon VK, Chan CC, To KK, Yuan S, Zhou J, Chu H, and Yuen KY
- Subjects
- Animals, COVID-19 immunology, COVID-19 pathology, Cricetinae, Cytokines metabolism, Gastrointestinal Tract immunology, Gastrointestinal Tract pathology, Gastrointestinal Tract virology, Humans, Inflammation, Lung pathology, Lung virology, Mesocricetus, Severity of Illness Index, Viral Load, Asymptomatic Infections, COVID-19 virology, Disease Models, Animal, SARS-CoV-2 physiology, Virus Shedding
- Abstract
Severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) is transmitted largely by respiratory droplets or airborne aerosols. Despite being frequently found in the immediate environment and feces of patients, evidence supporting the oral acquisition of SARS-CoV-2 is unavailable. Using the Syrian hamster model, we demonstrate that the severity of pneumonia induced by the intranasal inhalation of SARS-CoV-2 increases with virus inoculum. SARS-CoV-2 retains its infectivity in vitro in simulated human-fed-gastric and fasted-intestinal fluid after 2 h. Oral inoculation with the highest intranasal inoculum (10
5 PFUs) causes mild pneumonia in 67% (4/6) of the animals, with no weight loss. The lung histopathology score and viral load are significantly lower than those infected by the lowest intranasal inoculum (100 PFUs). However, 83% of the oral infections (10/12 hamsters) have a level of detectable viral shedding from oral swabs and feces similar to that of intranasally infected hamsters. Our findings indicate that the oral acquisition of SARS-CoV-2 can establish subclinical respiratory infection with less efficiency., Competing Interests: K.-Y.Y. serves as a member of the Advisory Committee on Centre for Health Protection in Hong Kong and is the chair of the Scientific Committee on Emerging and Zoonotic Diseases of the Centre for Health Protection in Hong Kong. J.F.-W.C. is an invited member of the WHO ad hoc expert group on COVID-19 animal models., (© 2020 The Authors.)- Published
- 2020
- Full Text
- View/download PDF
27. Discovery of SARS-CoV-2 antiviral drugs through large-scale compound repurposing.
- Author
-
Riva L, Yuan S, Yin X, Martin-Sancho L, Matsunaga N, Pache L, Burgstaller-Muehlbacher S, De Jesus PD, Teriete P, Hull MV, Chang MW, Chan JF, Cao J, Poon VK, Herbert KM, Cheng K, Nguyen TH, Rubanov A, Pu Y, Nguyen C, Choi A, Rathnasinghe R, Schotsaert M, Miorin L, Dejosez M, Zwaka TP, Sit KY, Martinez-Sobrido L, Liu WC, White KM, Chapman ME, Lendy EK, Glynne RJ, Albrecht R, Ruppin E, Mesecar AD, Johnson JR, Benner C, Sun R, Schultz PG, Su AI, García-Sastre A, Chatterjee AK, Yuen KY, and Chanda SK
- Subjects
- Adenosine Monophosphate analogs & derivatives, Adenosine Monophosphate pharmacology, Alanine analogs & derivatives, Alanine pharmacology, Alveolar Epithelial Cells cytology, Alveolar Epithelial Cells drug effects, Betacoronavirus growth & development, COVID-19, Cell Line, Cysteine Proteinase Inhibitors analysis, Cysteine Proteinase Inhibitors pharmacology, Dose-Response Relationship, Drug, Drug Synergism, Gene Expression Regulation drug effects, Humans, Hydrazones, Induced Pluripotent Stem Cells cytology, Models, Biological, Morpholines analysis, Morpholines pharmacology, Pandemics, Pyrimidines, Reproducibility of Results, SARS-CoV-2, Small Molecule Libraries analysis, Small Molecule Libraries pharmacology, Triazines analysis, Triazines pharmacology, Virus Internalization drug effects, Virus Replication drug effects, COVID-19 Drug Treatment, Antiviral Agents analysis, Antiviral Agents pharmacology, Betacoronavirus drug effects, Coronavirus Infections drug therapy, Coronavirus Infections virology, Drug Evaluation, Preclinical, Drug Repositioning, Pneumonia, Viral drug therapy, Pneumonia, Viral virology
- Abstract
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in 2019 has triggered an ongoing global pandemic of the severe pneumonia-like disease coronavirus disease 2019 (COVID-19)
1 . The development of a vaccine is likely to take at least 12-18 months, and the typical timeline for approval of a new antiviral therapeutic agent can exceed 10 years. Thus, repurposing of known drugs could substantially accelerate the deployment of new therapies for COVID-19. Here we profiled a library of drugs encompassing approximately 12,000 clinical-stage or Food and Drug Administration (FDA)-approved small molecules to identify candidate therapeutic drugs for COVID-19. We report the identification of 100 molecules that inhibit viral replication of SARS-CoV-2, including 21 drugs that exhibit dose-response relationships. Of these, thirteen were found to harbour effective concentrations commensurate with probable achievable therapeutic doses in patients, including the PIKfyve kinase inhibitor apilimod2-4 and the cysteine protease inhibitors MDL-28170, Z LVG CHN2, VBY-825 and ONO 5334. Notably, MDL-28170, ONO 5334 and apilimod were found to antagonize viral replication in human pneumocyte-like cells derived from induced pluripotent stem cells, and apilimod also demonstrated antiviral efficacy in a primary human lung explant model. Since most of the molecules identified in this study have already advanced into the clinic, their known pharmacological and human safety profiles will enable accelerated preclinical and clinical evaluation of these drugs for the treatment of COVID-19.- Published
- 2020
- Full Text
- View/download PDF
28. Viruses harness YxxØ motif to interact with host AP2M1 for replication: A vulnerable broad-spectrum antiviral target.
- Author
-
Yuan S, Chu H, Huang J, Zhao X, Ye ZW, Lai PM, Wen L, Cai JP, Mo Y, Cao J, Liang R, Poon VK, Sze KH, Zhou J, To KK, Chen Z, Chen H, Jin DY, Chan JF, and Yuen KY
- Subjects
- A549 Cells, Animals, Betacoronavirus drug effects, Binding Sites genetics, COVID-19, Cell Line, Tumor, Chlorocebus aethiops, Coronavirus Infections pathology, Dogs, HEK293 Cells, HIV Infections pathology, HIV-1 drug effects, Host-Pathogen Interactions drug effects, Humans, Influenza A virus drug effects, Influenza, Human pathology, Madin Darby Canine Kidney Cells, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Knockout, Middle East Respiratory Syndrome Coronavirus drug effects, Pandemics, Pneumonia, Viral pathology, Protein Binding genetics, Protein Transport drug effects, RNA, Viral genetics, Receptor, Interferon alpha-beta genetics, SARS-CoV-2, Transforming Growth Factor beta1 metabolism, Vero Cells, Virus Replication drug effects, Zika Virus drug effects, Zika Virus Infection pathology, Adaptor Proteins, Vesicular Transport metabolism, Antiviral Agents pharmacology, Cinnamates pharmacology, Coronavirus Infections drug therapy, HIV Infections drug therapy, Influenza, Human drug therapy, Pneumonia, Viral drug therapy, ortho-Aminobenzoates pharmacology
- Abstract
Targeting a universal host protein exploited by most viruses would be a game-changing strategy that offers broad-spectrum solution and rapid pandemic control including the current COVID-19. Here, we found a common YxxØ-motif of multiple viruses that exploits host AP2M1 for intracellular trafficking. A library chemical, N -(p-amylcinnamoyl)anthranilic acid (ACA), was identified to interrupt AP2M1-virus interaction and exhibit potent antiviral efficacy against a number of viruses in vitro and in vivo, including the influenza A viruses (IAVs), Zika virus (ZIKV), human immunodeficiency virus, and coronaviruses including MERS-CoV and SARS-CoV-2. YxxØ mutation, AP2M1 depletion, or disruption by ACA causes incorrect localization of viral proteins, which is exemplified by the failure of nuclear import of IAV nucleoprotein and diminished endoplasmic reticulum localization of ZIKV-NS3 and enterovirus-A71-2C proteins, thereby suppressing viral replication. Our study reveals an evolutionarily conserved mechanism of protein-protein interaction between host and virus that can serve as a broad-spectrum antiviral target., (Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works. Distributed under a Creative Commons Attribution NonCommercial License 4.0 (CC BY-NC).)
- Published
- 2020
- Full Text
- View/download PDF
29. A Large-scale Drug Repositioning Survey for SARS-CoV-2 Antivirals.
- Author
-
Riva L, Yuan S, Yin X, Martin-Sancho L, Matsunaga N, Burgstaller-Muehlbacher S, Pache L, De Jesus PP, Hull MV, Chang M, Chan JF, Cao J, Poon VK, Herbert K, Nguyen TT, Pu Y, Nguyen C, Rubanov A, Martinez-Sobrido L, Liu WC, Miorin L, White KM, Johnson JR, Benner C, Sun R, Schultz PG, Su A, Garcia-Sastre A, Chatterjee AK, Yuen KY, and Chanda SK
- Abstract
The emergence of novel SARS coronavirus 2 (SARS-CoV-2) in 2019 has triggered an ongoing global pandemic of severe pneumonia-like disease designated as coronavirus disease 2019 (COVID-19). To date, more than 2.1 million confirmed cases and 139,500 deaths have been reported worldwide, and there are currently no medical countermeasures available to prevent or treat the disease. As the development of a vaccine could require at least 12-18 months, and the typical timeline from hit finding to drug registration of an antiviral is >10 years, repositioning of known drugs can significantly accelerate the development and deployment of therapies for COVID-19. To identify therapeutics that can be repurposed as SARS-CoV-2 antivirals, we profiled a library of known drugs encompassing approximately 12,000 clinical-stage or FDA-approved small molecules. Here, we report the identification of 30 known drugs that inhibit viral replication. Of these, six were characterized for cellular dose-activity relationships, and showed effective concentrations likely to be commensurate with therapeutic doses in patients. These include the PIKfyve kinase inhibitor Apilimod, cysteine protease inhibitors MDL-28170, Z LVG CHN2, VBY-825, and ONO 5334, and the CCR1 antagonist MLN-3897. Since many of these molecules have advanced into the clinic, the known pharmacological and human safety profiles of these compounds will accelerate their preclinical and clinical evaluation for COVID-19 treatment.
- Published
- 2020
- Full Text
- View/download PDF
30. A familial cluster of pneumonia associated with the 2019 novel coronavirus indicating person-to-person transmission: a study of a family cluster.
- Author
-
Chan JF, Yuan S, Kok KH, To KK, Chu H, Yang J, Xing F, Liu J, Yip CC, Poon RW, Tsoi HW, Lo SK, Chan KH, Poon VK, Chan WM, Ip JD, Cai JP, Cheng VC, Chen H, Hui CK, and Yuen KY
- Subjects
- Adult, Aged, Betacoronavirus classification, Betacoronavirus genetics, COVID-19, COVID-19 Testing, China epidemiology, Clinical Laboratory Techniques, Coronavirus Infections diagnosis, Coronavirus Infections epidemiology, Family Health, Genome, Viral, Humans, Middle Aged, Phylogeny, Pneumonia, Viral diagnosis, Pneumonia, Viral epidemiology, Radiography, Thoracic, SARS-CoV-2, Tomography, X-Ray Computed, Whole Genome Sequencing methods, Coronavirus Infections transmission, Pneumonia, Viral transmission
- Abstract
Background: An ongoing outbreak of pneumonia associated with a novel coronavirus was reported in Wuhan city, Hubei province, China. Affected patients were geographically linked with a local wet market as a potential source. No data on person-to-person or nosocomial transmission have been published to date., Methods: In this study, we report the epidemiological, clinical, laboratory, radiological, and microbiological findings of five patients in a family cluster who presented with unexplained pneumonia after returning to Shenzhen, Guangdong province, China, after a visit to Wuhan, and an additional family member who did not travel to Wuhan. Phylogenetic analysis of genetic sequences from these patients were done., Findings: From Jan 10, 2020, we enrolled a family of six patients who travelled to Wuhan from Shenzhen between Dec 29, 2019 and Jan 4, 2020. Of six family members who travelled to Wuhan, five were identified as infected with the novel coronavirus. Additionally, one family member, who did not travel to Wuhan, became infected with the virus after several days of contact with four of the family members. None of the family members had contacts with Wuhan markets or animals, although two had visited a Wuhan hospital. Five family members (aged 36-66 years) presented with fever, upper or lower respiratory tract symptoms, or diarrhoea, or a combination of these 3-6 days after exposure. They presented to our hospital (The University of Hong Kong-Shenzhen Hospital, Shenzhen) 6-10 days after symptom onset. They and one asymptomatic child (aged 10 years) had radiological ground-glass lung opacities. Older patients (aged >60 years) had more systemic symptoms, extensive radiological ground-glass lung changes, lymphopenia, thrombocytopenia, and increased C-reactive protein and lactate dehydrogenase levels. The nasopharyngeal or throat swabs of these six patients were negative for known respiratory microbes by point-of-care multiplex RT-PCR, but five patients (four adults and the child) were RT-PCR positive for genes encoding the internal RNA-dependent RNA polymerase and surface Spike protein of this novel coronavirus, which were confirmed by Sanger sequencing. Phylogenetic analysis of these five patients' RT-PCR amplicons and two full genomes by next-generation sequencing showed that this is a novel coronavirus, which is closest to the bat severe acute respiatory syndrome (SARS)-related coronaviruses found in Chinese horseshoe bats., Interpretation: Our findings are consistent with person-to-person transmission of this novel coronavirus in hospital and family settings, and the reports of infected travellers in other geographical regions., Funding: The Shaw Foundation Hong Kong, Michael Seak-Kan Tong, Respiratory Viral Research Foundation Limited, Hui Ming, Hui Hoy and Chow Sin Lan Charity Fund Limited, Marina Man-Wai Lee, the Hong Kong Hainan Commercial Association South China Microbiology Research Fund, Sanming Project of Medicine (Shenzhen), and High Level-Hospital Program (Guangdong Health Commission)., (Copyright © 2020 Elsevier Ltd. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
31. Activation of C-Type Lectin Receptor and (RIG)-I-Like Receptors Contributes to Proinflammatory Response in Middle East Respiratory Syndrome Coronavirus-Infected Macrophages.
- Author
-
Zhao X, Chu H, Wong BH, Chiu MC, Wang D, Li C, Liu X, Yang D, Poon VK, Cai J, Chan JF, To KK, Zhou J, and Yuen KY
- Subjects
- Animals, CARD Signaling Adaptor Proteins, Chemokines metabolism, Chlorocebus aethiops, Coronavirus Infections metabolism, Coronavirus Infections virology, Cytokines immunology, Cytokines metabolism, DEAD Box Protein 58 genetics, Gene Knockdown Techniques, Humans, Lectins, C-Type genetics, Lung immunology, Middle East Respiratory Syndrome Coronavirus genetics, Middle East Respiratory Syndrome Coronavirus pathogenicity, RNA, Small Interfering metabolism, Receptors, Immunologic genetics, Receptors, Immunologic metabolism, Signal Transduction, Transcriptome, Tretinoin pharmacology, Vero Cells, Virus Replication, Coronavirus Infections immunology, DEAD Box Protein 58 metabolism, Lectins, C-Type metabolism, Macrophages immunology, Macrophages virology, Middle East Respiratory Syndrome Coronavirus metabolism
- Abstract
Background: Human infection with Middle East respiratory syndrome coronavirus (MERS-CoV) poses an ongoing threat to public health worldwide. The studies of MERS patients with severe disease and experimentally infected animals showed that robust viral replication and intensive proinflammatory response in lung tissues contribute to high pathogenicity of MERS-CoV. We sought to identify pattern recognition receptor (PRR) signaling pathway(s) that mediates the inflammatory cascade in human macrophages upon MERS-CoV infection., Methods: The potential signaling pathways were manipulated individually by pharmacological inhibition, small interfering ribonucleic acid (siRNA) depletion, and antibody blocking. The MERS-CoV-induced proinflammatory response was evaluated by measuring the expression levels of key cytokines and/or chemokines. Reverse transcription-quantitative polymerase chain reaction assay, flow cytometry analysis, and Western blotting were applied to evaluate the activation of related PRRs and engagement of adaptors., Results: MERS-CoV replication significantly upregulated C-type lectin receptor (CLR) macrophage-inducible Ca2+-dependent lectin receptor (Mincle). The role of Mincle for MERS-CoV-triggered cytokine/chemokine induction was established based on the results of antibody blockage, siRNA depletion of Mincle and its adaptor spleen tyrosine kinase (Syk), and Syk pharmacological inhibition. The cytokine and/or chemokine induction was significantly attenuated by siRNA depletion of retinoic acid-inducible-I-like receptors (RLR) or adaptor, indicating that RLR signaling also contributed to MERS-CoV-induced proinflammatory response., Conclusions: The CLR and RLR pathways are activated and contribute to the proinflammatory response in MERS-CoV-infected macrophages., (© The Author(s) 2019. Published by Oxford University Press for the Infectious Diseases Society of America. All rights reserved. For permissions, e-mail: journals.permissions@oup.com.)
- Published
- 2020
- Full Text
- View/download PDF
32. SREBP-dependent lipidomic reprogramming as a broad-spectrum antiviral target.
- Author
-
Yuan S, Chu H, Chan JF, Ye ZW, Wen L, Yan B, Lai PM, Tee KM, Huang J, Chen D, Li C, Zhao X, Yang D, Chiu MC, Yip C, Poon VK, Chan CC, Sze KH, Zhou J, Chan IH, Kok KH, To KK, Kao RY, Lau JY, Jin DY, Perlman S, and Yuen KY
- Subjects
- Antiviral Agents pharmacology, Benzoates chemistry, Benzoates metabolism, Biosynthetic Pathways drug effects, Influenza A virus drug effects, Influenza A virus physiology, Lipids biosynthesis, Middle East Respiratory Syndrome Coronavirus drug effects, Middle East Respiratory Syndrome Coronavirus physiology, Protein Binding, Retinoids chemistry, Retinoids metabolism, Retinoids pharmacology, Tetrahydronaphthalenes chemistry, Tetrahydronaphthalenes metabolism, Virus Diseases prevention & control, Virus Diseases virology, Benzoates pharmacology, Lipid Metabolism drug effects, Sterol Regulatory Element Binding Proteins metabolism, Tetrahydronaphthalenes pharmacology, Virus Replication drug effects
- Abstract
Viruses are obligate intracellular microbes that exploit the host metabolic machineries to meet their biosynthetic demands, making these host pathways potential therapeutic targets. Here, by exploring a lipid library, we show that AM580, a retinoid derivative and RAR-α agonist, is highly potent in interrupting the life cycle of diverse viruses including Middle East respiratory syndrome coronavirus and influenza A virus. Using click chemistry, the overexpressed sterol regulatory element binding protein (SREBP) is shown to interact with AM580, which accounts for its broad-spectrum antiviral activity. Mechanistic studies pinpoint multiple SREBP proteolytic processes and SREBP-regulated lipid biosynthesis pathways, including the downstream viral protein palmitoylation and double-membrane vesicles formation, that are indispensable for virus replication. Collectively, our study identifies a basic lipogenic transactivation event with broad relevance to human viral infections and represents SREBP as a potential target for the development of broad-spectrum antiviral strategies.
- Published
- 2019
- Full Text
- View/download PDF
33. Identification and characterization of GLDC as host susceptibility gene to severe influenza.
- Author
-
Zhou J, Wang D, Wong BH, Li C, Poon VK, Wen L, Zhao X, Chiu MC, Liu X, Ye Z, Yuan S, Sze KH, Chan JF, Chu H, To KK, and Yuen KY
- Subjects
- Animals, Disease Models, Animal, Enzyme Inhibitors administration & dosage, Humans, Influenza A Virus, H1N1 Subtype growth & development, Influenza A Virus, H1N1 Subtype immunology, Influenza A Virus, H7N9 Subtype growth & development, Influenza A Virus, H7N9 Subtype immunology, Influenza, Human pathology, Mice, Inbred BALB C, Orthomyxoviridae Infections drug therapy, Orthomyxoviridae Infections pathology, Tacrolimus administration & dosage, Tacrolimus analogs & derivatives, Treatment Outcome, Virus Replication, Genetic Predisposition to Disease, Glycine Dehydrogenase (Decarboxylating) genetics, Immunity, Innate, Influenza, Human genetics
- Abstract
Glycine decarboxylase (GLDC) was prioritized as a candidate susceptibility gene to severe influenza in humans. The higher expression of GLDC derived from genetic variations may confer a higher risk to H7N9 and severe H1N1 infection. We sought to characterize GLDC as functional susceptibility gene that GLDC may intrinsically regulate antiviral response, thereby impacting viral replication and disease outcome. We demonstrated that GLDC inhibitor AOAA and siRNA depletion boosted IFNβ- and IFN-stimulated genes (ISGs) in combination with PolyI:C stimulation. GLDC inhibition and depletion significantly amplified antiviral response of type I IFNs and ISGs upon viral infection and suppressed the replication of H1N1 and H7N9 viruses. Consistently, GLDC overexpression significantly promoted viral replication due to the attenuated antiviral responses. Moreover, GLDC inhibition in H1N1-infected BALB/c mice recapitulated the amplified antiviral response and suppressed viral growth. AOAA provided potent protection to the infected mice from lethal infection, comparable to a standard antiviral against influenza viruses. Collectively, GLDC regulates cellular antiviral response and orchestrates viral growth. GLDC is a functional susceptibility gene to severe influenza in humans., (© 2018 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2019
- Full Text
- View/download PDF
34. The celecoxib derivative kinase inhibitor AR-12 (OSU-03012) inhibits Zika virus via down-regulation of the PI3K/Akt pathway and protects Zika virus-infected A129 mice: A host-targeting treatment strategy.
- Author
-
Chan JF, Zhu Z, Chu H, Yuan S, Chik KK, Chan CC, Poon VK, Yip CC, Zhang X, Tsang JO, Zou Z, Tee KM, Shuai H, Lu G, and Yuen KY
- Subjects
- Administration, Oral, Animals, Antiviral Agents therapeutic use, Cell Line, Disease Models, Animal, Humans, Injections, Intraperitoneal, Mice, Microbial Sensitivity Tests, Protein Kinase Inhibitors therapeutic use, Pyrazoles therapeutic use, Sulfonamides therapeutic use, Survival Analysis, Treatment Outcome, Viral Load, Zika Virus growth & development, Zika Virus Infection pathology, Zika Virus Infection virology, Antiviral Agents pharmacology, Protein Kinase Inhibitors pharmacology, Pyrazoles pharmacology, Sulfonamides pharmacology, Zika Virus drug effects, Zika Virus Infection prevention & control
- Abstract
Zika virus (ZIKV) is a human-pathogenic flavivirus that has recently emerged as a global public health threat. ZIKV infection may be associated with congenital malformations in infected fetuses and severe neurological and systemic complications in infected adults. There are currently limited treatment options for ZIKV infection. AR-12 (OSU-03012) is a celecoxib derivative cellular kinase inhibitor that has broad-spectrum antiviral activities. In this study, we investigated the antiviral activity and mechanism of AR-12 against ZIKV. We evaluated the in vitro anti-ZIKV activity of AR-12, using cell protection and virus yield reduction assays, in multiple clinically relevant cell lines, and the in vivo treatment effects of AR-12 in a lethal mouse model using type I interferon receptor-deficient A129 mice. AR-12 inhibited ZIKV strains belonging to both the African and Asian/American lineages in Huh-7 and/or neuronal cells. AR12's IC
50 against ZIKV was consistently <2 μM in these cells. ZIKV-infected A129 mice treated with intraperitoneally or orally administered AR-12 had significantly higher survival rate (50.0%-83.3% vs 0%, P < 0.05), less body weight loss, and lower blood and tissue ZIKV RNA loads than untreated control A129 mice. These anti-ZIKV effects were likely the results of down-regulation of the PI3K/Akt pathway by AR-12. Clinical trials using the clinically available and broad-spectrum AR-12 as an empirical treatment should be considered especially for patients residing in or returning from areas endemic of ZIKV and other arboviral infections who present with an acute undifferentiated febrile illness., (Copyright © 2018 Elsevier B.V. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
35. Middle East respiratory syndrome coronavirus and bat coronavirus HKU9 both can utilize GRP78 for attachment onto host cells.
- Author
-
Chu H, Chan CM, Zhang X, Wang Y, Yuan S, Zhou J, Au-Yeung RK, Sze KH, Yang D, Shuai H, Hou Y, Li C, Zhao X, Poon VK, Leung SP, Yeung ML, Yan J, Lu G, Jin DY, Gao GF, Chan JF, and Yuen KY
- Subjects
- Animals, Cell Line, Chlorocebus aethiops, Dipeptidyl Peptidase 4 metabolism, Endoplasmic Reticulum Chaperone BiP, Host-Pathogen Interactions, Humans, Protein Interaction Maps, Receptors, Virus metabolism, Spike Glycoprotein, Coronavirus metabolism, Vero Cells, Betacoronavirus physiology, Coronavirus physiology, Coronavirus Infections metabolism, Heat-Shock Proteins metabolism, Middle East Respiratory Syndrome Coronavirus physiology, Virus Attachment
- Abstract
Coronavirus tropism is predominantly determined by the interaction between coronavirus spikes and the host receptors. In this regard, coronaviruses have evolved a complicated receptor-recognition system through their spike proteins. Spikes from highly related coronaviruses can recognize distinct receptors, whereas spikes of distant coronaviruses can employ the same cell-surface molecule for entry. Moreover, coronavirus spikes can recognize a broad range of cell-surface molecules in addition to the receptors and thereby can augment coronavirus attachment or entry. The receptor of Middle East respiratory syndrome coronavirus (MERS-CoV) is dipeptidyl peptidase 4 (DPP4). In this study, we identified membrane-associated 78-kDa glucose-regulated protein (GRP78) as an additional binding target of the MERS-CoV spike. Further analyses indicated that GRP78 could not independently render nonpermissive cells susceptible to MERS-CoV infection but could facilitate MERS-CoV entry into permissive cells by augmenting virus attachment. More importantly, by exploring potential interactions between GRP78 and spikes of other coronaviruses, we discovered that the highly conserved human GRP78 could interact with the spike protein of bat coronavirus HKU9 (bCoV-HKU9) and facilitate its attachment to the host cell surface. Taken together, our study has identified GRP78 as a host factor that can interact with the spike proteins of two Betacoronaviruses , the lineage C MERS-CoV and the lineage D bCoV-HKU9. The capacity of GRP78 to facilitate surface attachment of both a human coronavirus and a phylogenetically related bat coronavirus exemplifies the need for continuous surveillance of the evolution of animal coronaviruses to monitor their potential for human adaptations., (© 2018 Chu et al.)
- Published
- 2018
- Full Text
- View/download PDF
36. Immunization With a Novel Human Type 5 Adenovirus-Vectored Vaccine Expressing the Premembrane and Envelope Proteins of Zika Virus Provides Consistent and Sterilizing Protection in Multiple Immunocompetent and Immunocompromised Animal Models.
- Author
-
Guo Q, Chan JF, Poon VK, Wu S, Chan CC, Hou L, Yip CC, Ren C, Cai JP, Zhao M, Zhang AJ, Song X, Chan KH, Wang B, Kok KH, Wen Y, Yuen KY, and Chen W
- Subjects
- Animal Structures virology, Animals, Antibodies, Neutralizing blood, Antibodies, Viral blood, Blood virology, Disease Models, Animal, Drug Carriers, Female, Immunity, Cellular, Immunity, Humoral, Immunocompromised Host, Mice, Inbred BALB C, Treatment Outcome, Vaccines, Synthetic administration & dosage, Vaccines, Synthetic genetics, Vaccines, Synthetic immunology, Viral Envelope Proteins genetics, Viral Load, Viral Vaccines administration & dosage, Viral Vaccines genetics, Zika Virus genetics, Adenoviruses, Human genetics, Genetic Vectors, Viral Envelope Proteins immunology, Viral Vaccines immunology, Zika Virus immunology, Zika Virus Infection prevention & control
- Abstract
Background: Zika virus (ZIKV) infection may be associated with severe complications and disseminated via both vector-borne and nonvector-borne routes. Adenovirus-vectored vaccines represent a favorable controlling measure for the ZIKV epidemic because they have been shown to be safe, immunogenic, and rapidly generable for other emerging viral infections. Evaluations of 2 previously reported adenovirus-vectored ZIKV vaccines were performed using nonlethal animal models and/or nonepidemic ZIKV strain., Methods: We constructed 2 novel human adenovirus 5 (Ad5)-vectored vaccines containing the ZIKV premembrane-envelope (Ad5-Sig-prM-Env) and envelope (Ad5-Env) proteins, respectively, and evaluated them in multiple nonlethal and lethal animal models using epidemic ZIKV strains., Results: Both vaccines elicited robust humoral and cellular immune responses in immunocompetent BALB/c mice. Dexamethasone-immunosuppressed mice vaccinated with either vaccine demonstrated robust and durable antibody responses and significantly lower blood and tissue viral loads than controls (P < .05). Similar findings were also observed in interferon-α/β receptor-deficient A129 mice. In both of these immunocompromised animal models, Ad5-Sig-prM-Env-vaccinated mice had significantly (P < .05) higher titers of anti-ZIKV-specific neutralizing antibody titers and lower (undetectable) viral loads than Ad5-Env-vaccinated mice. The close correlation between the neutralizing antibody titer and viral load helped to explain the better protective effect of Ad5-Sig-prM-Env than Ad5-Env. Anamnestic response was absent in Ad5-Sig-prM-Env-vaccinated A129 mice., Conclusions: Ad5-Sig-prM-Env provided sterilizing protection against ZIKV infection in mice.
- Published
- 2018
- Full Text
- View/download PDF
37. Differentiated human airway organoids to assess infectivity of emerging influenza virus.
- Author
-
Zhou J, Li C, Sachs N, Chiu MC, Wong BH, Chu H, Poon VK, Wang D, Zhao X, Wen L, Song W, Yuan S, Wong KK, Chan JF, To KK, Chen H, Clevers H, and Yuen KY
- Subjects
- Humans, Influenza A Virus, H1N1 Subtype growth & development, Influenza A Virus, H7N2 Subtype growth & development, Organoids pathology, Respiratory System pathology, Influenza A Virus, H1N1 Subtype pathogenicity, Influenza A Virus, H7N2 Subtype pathogenicity, Influenza, Human, Organoids virology, Respiratory System virology
- Abstract
Novel reassortant avian influenza H7N9 virus and pandemic 2009 H1N1 (H1N1pdm) virus cause human infections, while avian H7N2 and swine H1N1 virus mainly infect birds and pigs, respectively. There is no robust in vitro model for assessing the infectivity of emerging viruses in humans. Based on a recently established method, we generated long-term expanding 3D human airway organoids which accommodate four types of airway epithelial cells: ciliated, goblet, club, and basal cells. We report differentiation conditions which increase ciliated cell numbers to a nearly physiological level with synchronously beating cilia readily discernible in every organoid. In addition, the differentiation conditions induce elevated levels of serine proteases, which are essential for productive infection of human influenza viruses and low-pathogenic avian influenza viruses. We also established improved 2D monolayer culture conditions for the differentiated airway organoids. To demonstrate the ability of differentiated airway organoids to identify human-infective virus, 3D and 2D differentiated airway organoids are applied to evaluate two pairs of viruses with known distinct infectivity in humans, H7N9/Ah versus H7N2 and H1N1pdm versus an H1N1 strain isolated from swine (H1N1sw). The human-infective H7N9/Ah virus replicated more robustly than the poorly human-infective H7N2 virus; the highly human-infective H1N1pdm virus replicated to a higher titer than the counterpart H1N1sw. Collectively, we developed differentiated human airway organoids which can morphologically and functionally simulate human airway epithelium. These differentiated airway organoids can be applied for rapid assessment of the infectivity of emerging respiratory viruses to human., Competing Interests: The authors declare no conflict of interest., (Copyright © 2018 the Author(s). Published by PNAS.)
- Published
- 2018
- Full Text
- View/download PDF
38. Human intestinal tract serves as an alternative infection route for Middle East respiratory syndrome coronavirus.
- Author
-
Zhou J, Li C, Zhao G, Chu H, Wang D, Yan HH, Poon VK, Wen L, Wong BH, Zhao X, Chiu MC, Yang D, Wang Y, Au-Yeung RKH, Chan IH, Sun S, Chan JF, To KK, Memish ZA, Corman VM, Drosten C, Hung IF, Zhou Y, Leung SY, and Yuen KY
- Subjects
- Animals, Caco-2 Cells, Coronavirus Infections drug therapy, Coronavirus Infections virology, Dipeptidyl Peptidase 4 genetics, Dipeptidyl Peptidase 4 metabolism, Epithelial Cells cytology, Epithelial Cells virology, Feces virology, Female, Humans, Intestine, Small cytology, Intestine, Small pathology, Intestine, Small virology, Lung pathology, Lung virology, Mice, Mice, Transgenic, Middle East Respiratory Syndrome Coronavirus genetics, Middle East Respiratory Syndrome Coronavirus pathogenicity, Pantoprazole therapeutic use, RNA, Viral genetics, RNA, Viral metabolism, Coronavirus Infections pathology, Middle East Respiratory Syndrome Coronavirus physiology
- Abstract
Middle East respiratory syndrome coronavirus (MERS-CoV) has caused human respiratory infections with a high case fatality rate since 2012. However, the mode of virus transmission is not well understood. The findings of epidemiological and virological studies prompted us to hypothesize that the human gastrointestinal tract could serve as an alternative route to acquire MERS-CoV infection. We demonstrated that human primary intestinal epithelial cells, small intestine explants, and intestinal organoids were highly susceptible to MERS-CoV and can sustain robust viral replication. We also identified the evidence of enteric MERS-CoV infection in the stool specimen of a clinical patient. MERS-CoV was considerably resistant to fed-state gastrointestinal fluids but less tolerant to highly acidic fasted-state gastric fluid. In polarized Caco-2 cells cultured in Transwell inserts, apical MERS-CoV inoculation was more effective in establishing infection than basolateral inoculation. Notably, direct intragastric inoculation of MERS-CoV caused a lethal infection in human DPP4 transgenic mice. Histological examination revealed MERS-CoV enteric infection in all inoculated mice, as shown by the presence of virus-positive cells, progressive inflammation, and epithelial degeneration in small intestines, which were exaggerated in the mice pretreated with the proton pump inhibitor pantoprazole. With the progression of the enteric infection, inflammation, virus-positive cells, and live viruses emerged in the lung tissues, indicating the development of sequential respiratory infection. Taken together, these data suggest that the human intestinal tract may serve as an alternative infection route for MERS-CoV.
- Published
- 2017
- Full Text
- View/download PDF
39. Structure-based discovery of clinically approved drugs as Zika virus NS2B-NS3 protease inhibitors that potently inhibit Zika virus infection in vitro and in vivo.
- Author
-
Yuan S, Chan JF, den-Haan H, Chik KK, Zhang AJ, Chan CC, Poon VK, Yip CC, Mak WW, Zhu Z, Zou Z, Tee KM, Cai JP, Chan KH, de la Peña J, Pérez-Sánchez H, Cerón-Carrasco JP, and Yuen KY
- Subjects
- Animals, Antiviral Agents administration & dosage, Antiviral Agents chemistry, Computer Simulation, Drug Discovery, Humans, Mice, Models, Molecular, Molecular Docking Simulation, Novobiocin administration & dosage, Novobiocin therapeutic use, Protease Inhibitors administration & dosage, Protease Inhibitors chemistry, Protein Binding, Protein Conformation, Small Molecule Libraries, Viral Load drug effects, Virus Replication drug effects, Zika Virus enzymology, Zika Virus Infection virology, Antiviral Agents pharmacology, Antiviral Agents therapeutic use, Protease Inhibitors pharmacology, Protease Inhibitors therapeutic use, Zika Virus drug effects, Zika Virus Infection drug therapy
- Abstract
Zika virus (ZIKV) infection may be associated with severe complications in fetuses and adults, but treatment options are limited. We performed an in silico structure-based screening of a large chemical library to identify potential ZIKV NS2B-NS3 protease inhibitors. Clinically approved drugs belonging to different drug classes were selected among the 100 primary hit compounds with the highest predicted binding affinities to ZIKV NS2B-NS3-protease for validation studies. ZIKV NS2B-NS3 protease inhibitory activity was validated in most of the selected drugs and in vitro anti-ZIKV activity was identified in two of them (novobiocin and lopinavir-ritonavir). Molecular docking and molecular dynamics simulations predicted that novobiocin bound to ZIKV NS2B-NS3-protease with high stability. Dexamethasone-immunosuppressed mice with disseminated ZIKV infection and novobiocin treatment had significantly (P < 0.05) higher survival rate (100% vs 0%), lower mean blood and tissue viral loads, and less severe histopathological changes than untreated controls. This structure-based drug discovery platform should facilitate the identification of additional enzyme inhibitors of ZIKV., (Copyright © 2017 Elsevier B.V. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
40. Novel antiviral activity and mechanism of bromocriptine as a Zika virus NS2B-NS3 protease inhibitor.
- Author
-
Chan JF, Chik KK, Yuan S, Yip CC, Zhu Z, Tee KM, Tsang JO, Chan CC, Poon VK, Lu G, Zhang AJ, Lai KK, Chan KH, Kao RY, and Yuen KY
- Subjects
- Bromocriptine therapeutic use, Enzyme Assays, Fluorescence, Humans, Interferon alpha-2, Interferon-alpha pharmacology, Peptide Hydrolases drug effects, Recombinant Proteins pharmacology, Serine Proteases metabolism, Serine Proteinase Inhibitors chemistry, Virus Replication drug effects, Zika Virus enzymology, Zika Virus physiology, Antiviral Agents pharmacology, Bromocriptine pharmacology, Serine Proteinase Inhibitors pharmacology, Viral Nonstructural Proteins antagonists & inhibitors, Zika Virus drug effects
- Abstract
Zika virus (ZIKV) infection is associated with congenital malformations in infected fetuses and severe neurological and other systemic complications in adults. There are currently limited anti-ZIKV treatment options that are readily available and safe for use in pregnancy. In this drug repurposing study, bromocriptine was found to have inhibitory effects on ZIKV replication in cytopathic effect inhibition, virus yield reduction, and plaque reduction assays. Time-of-drug-addition assay showed that bromocriptine exerted anti-ZIKV activity between 0 and 12 h post-ZIKV inoculation, corroborating with post-entry events in the virus replication cycle prior to budding. Our docking model showed that bromocriptine interacted with several active site residues of the proteolytic cavity involving H51 and S135 in the ZIKV-NS2B-NS3 protease protein, and might occupy the active site and inhibit the protease activity of the ZIKV-NS2B-NS3 protein. A fluorescence-based protease inhibition assay confirmed that bromocriptine inhibited ZIKV protease activity. Moreover, bromocriptine exhibited synergistic effect with interferon-α2b against ZIKV replication in cytopathic effect inhibition assay. The availability of per vagina administration of bromocriptine as suppositories or vaginoadhesive discs and the synergistic anti-ZIKV activity between bromocriptine and type I interferon may make bromocriptine a potentially useful and readily available treatment option for ZIKV infection. The anti-ZIKV effects of bromocriptine should be evaluated in a suitable animal model., (Copyright © 2017 Elsevier B.V. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
41. Improved detection of Zika virus RNA in human and animal specimens by a novel, highly sensitive and specific real-time RT-PCR assay targeting the 5'-untranslated region of Zika virus.
- Author
-
Chan JF, Yip CC, Tee KM, Zhu Z, Tsang JO, Chik KK, Tsang TG, Chan CC, Poon VK, Sridhar S, Yin F, Hung IF, Chau SK, Zhang AJ, Chan KH, and Yuen KY
- Subjects
- Animals, Cross Reactions, Humans, Mice, Real-Time Polymerase Chain Reaction methods, Sensitivity and Specificity, Zika Virus Infection virology, 5' Untranslated Regions, RNA, Viral analysis, Reverse Transcriptase Polymerase Chain Reaction methods, Zika Virus genetics, Zika Virus Infection diagnosis
- Abstract
Objective and Method: We developed and evaluated five novel real-time RT-PCR assays targeting conserved regions in the 5'-untranslated region (5'-UTR), envelope (E'), non-structural protein 2A (NS2A), NS5 and 3'-UTR of the ZIKV genome., Results: The ZIKV-5'-UTR assay exhibited the lowest in vitro limit of detection (5-10 RNA copies/reaction and 3.0 × 10
-1 plaque-forming units/ml). Compared to the modified version of a widely adopted RT-PCR assay targeting the ZIKV-E gene, the ZIKV-5'-UTR assay showed better sensitivity in human clinical specimens, and representative mouse specimens, including many organs which are known to be involved in human ZIKV infection but difficult to obtain in clinical settings. The ZIKV-5'-UTR assay detected ZIKV RNA in 84/84 (100.0%) ZIKV-E'-positive and an additional 30/296 (10.1%, P < 0.01) ZIKV-E'-negative mouse specimens. The higher sensitivity of the ZIKV-5'-UTR assay was most significant in kidney and testis/epididymis specimens (P < 0.01). No in vitro or in vivo cross-reactivity was found between the ZIKV-5'-UTR assay and dengue virus, yellow fever virus, Japanese encephalitis virus, West Nile virus, hepatitis C virus and Chikungunya virus., Conclusions: The highly sensitive and specific ZIKV-5'-UTR assay may help to improve the laboratory diagnosis of ZIKV infection., (© 2017 John Wiley & Sons Ltd.)- Published
- 2017
- Full Text
- View/download PDF
42. Zika Virus Infection in Dexamethasone-immunosuppressed Mice Demonstrating Disseminated Infection with Multi-organ Involvement Including Orchitis Effectively Treated by Recombinant Type I Interferons.
- Author
-
Chan JF, Zhang AJ, Chan CC, Yip CC, Mak WW, Zhu H, Poon VK, Tee KM, Zhu Z, Cai JP, Tsang JO, Chik KK, Yin F, Chan KH, Kok KH, Jin DY, Au-Yeung RK, and Yuen KY
- Subjects
- Animals, Brain immunology, Brain metabolism, Brain pathology, Dexamethasone adverse effects, Disease Models, Animal, Female, Kidney immunology, Kidney metabolism, Kidney pathology, Male, Mice, Orchitis drug therapy, Viral Load, Zika Virus Infection drug therapy, Zika Virus Infection pathology, Antiviral Agents pharmacology, Immunocompromised Host, Interferon Type I pharmacology, Orchitis virology, Zika Virus drug effects, Zika Virus immunology, Zika Virus Infection immunology, Zika Virus Infection virology
- Abstract
Background: Disseminated or fatal Zika virus (ZIKV) infections were reported in immunosuppressed patients. Existing interferon-signaling/receptor-deficient mouse models may not be suitable for evaluating treatment effects of recombinant interferons., Methods: We developed a novel mouse model for ZIKV infection by immunosuppressing BALB/c mice with dexamethasone., Results: Dexamethasone-immunosuppressed male mice (6-8weeks) developed disseminated infection as evidenced by the detection of ZIKV-NS1 protein expression and high viral loads in multiple organs. They had ≥10% weight loss and high clinical scores soon after dexamethasone withdrawal (10dpi), which warranted euthanasia at 12dpi. Viral loads in blood and most tissues at 5dpi were significantly higher than those at 12dpi (P<0.05). Histological examination revealed prominent inflammatory infiltrates in multiple organs, and CD45+ and CD8+ inflammatory cells were seen in the testis. These findings suggested that clinical deterioration occurred during viral clearance by host immune response. Type I interferon treatments improved clinical outcome of mice (100% vs 0% survival)., Conclusions: Besides virus dissemination, inflammation of various tissues, especially orchitis, may be potential complications of ZIKV infection with significant implications on disease transmission and male fertility. Interferon treatment should be considered in patients at high risks for ZIKV-associated complications when the potential benefits outweigh the side effects of treatment., (Copyright © 2016 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
43. A novel peptide with potent and broad-spectrum antiviral activities against multiple respiratory viruses.
- Author
-
Zhao H, Zhou J, Zhang K, Chu H, Liu D, Poon VK, Chan CC, Leung HC, Fai N, Lin YP, Zhang AJ, Jin DY, Yuen KY, and Zheng BJ
- Subjects
- Animals, Endosomes drug effects, Endosomes metabolism, Endosomes virology, Influenza A virus drug effects, Influenza A virus physiology, Inhibitory Concentration 50, Mice, Microbial Sensitivity Tests, Middle East Respiratory Syndrome Coronavirus drug effects, Orthomyxoviridae Infections drug therapy, Orthomyxoviridae Infections mortality, Orthomyxoviridae Infections pathology, Orthomyxoviridae Infections virology, Peptide Fragments pharmacology, Peptides chemistry, Peptides metabolism, Protein Binding, Severe acute respiratory syndrome-related coronavirus drug effects, Viral Envelope Proteins metabolism, Viral Load, beta-Defensins chemistry, Antiviral Agents pharmacology, Peptides pharmacology
- Abstract
A safe, potent and broad-spectrum antiviral is urgently needed to combat emerging respiratory viruses. In light of the broad antiviral activity of β-defensins, we tested the antiviral activity of 11 peptides derived from mouse β-defensin-4 and found that a short peptide, P9, exhibited potent and broad-spectrum antiviral effects against multiple respiratory viruses in vitro and in vivo, including influenza A virus H1N1, H3N2, H5N1, H7N7, H7N9, SARS-CoV and MERS-CoV. The antiviral activity of P9 was attributed to its high-affinity binding to viral glycoproteins, as well as the abundance of basic amino acids in its composition. After binding viral particles through viral surface glycoproteins, P9 entered into cells together with the viruses via endocytosis and prevented endosomal acidification, which blocked membrane fusion and subsequent viral RNA release. This study has paved the avenue for developing new prophylactic and therapeutic agents with broad-spectrum antiviral activities.
- Published
- 2016
- Full Text
- View/download PDF
44. MERS coronavirus induces apoptosis in kidney and lung by upregulating Smad7 and FGF2.
- Author
-
Yeung ML, Yao Y, Jia L, Chan JF, Chan KH, Cheung KF, Chen H, Poon VK, Tsang AK, To KK, Yiu MK, Teng JL, Chu H, Zhou J, Zhang Q, Deng W, Lau SK, Lau JY, Woo PC, Chan TM, Yung S, Zheng BJ, Jin DY, Mathieson PW, Qin C, and Yuen KY
- Subjects
- Animals, Callithrix, Cytopathogenic Effect, Viral, Disease Models, Animal, Host-Pathogen Interactions, Humans, Organ Culture Techniques, Apoptosis, Coronavirus Infections pathology, Fibroblast Growth Factor 2 metabolism, Kidney pathology, Lung pathology, Middle East Respiratory Syndrome Coronavirus pathogenicity, Smad7 Protein metabolism
- Abstract
Middle East respiratory syndrome coronavirus (MERS-CoV) causes sporadic zoonotic disease and healthcare-associated outbreaks in human. MERS is often complicated by acute respiratory distress syndrome (ARDS) and multi-organ failure(1,2). The high incidence of renal failure in MERS is a unique clinical feature not often found in other human coronavirus infections(3,4). Whether MERS-CoV infects the kidney and how it triggers renal failure are not understood(5,6). Here, we demonstrated renal infection and apoptotic induction by MERS-CoV in human ex vivo organ culture and a nonhuman primate model. High-throughput analysis revealed that the cellular genes most significantly perturbed by MERS-CoV have previously been implicated in renal diseases. Furthermore, MERS-CoV induced apoptosis through upregulation of Smad7 and fibroblast growth factor 2 (FGF2) expression in both kidney and lung cells. Conversely, knockdown of Smad7 effectively inhibited MERS-CoV replication and protected cells from virus-induced cytopathic effects. We further demonstrated that hyperexpression of Smad7 or FGF2 induced a strong apoptotic response in kidney cells. Common marmosets infected by MERS-CoV developed ARDS and disseminated infection in kidneys and other organs. Smad7 and FGF2 expression were elevated in the lungs and kidneys of the infected animals. Our results provide insights into the pathogenesis of MERS-CoV and host targets for treatment.
- Published
- 2016
- Full Text
- View/download PDF
45. An H5N1-based matrix protein 2 ectodomain tetrameric peptide vaccine provides cross-protection against lethal infection with H7N9 influenza virus.
- Author
-
Leung HC, Chan CC, Poon VK, Zhao HJ, Cheung CY, Ng F, Huang JD, and Zheng BJ
- Subjects
- Allantois, Animals, Antibodies, Viral biosynthesis, Antibodies, Viral immunology, Chickens, Cross Reactions, Dogs, Female, Humans, Influenza A Virus, H5N1 Subtype chemistry, Lung pathology, Lung virology, Madin Darby Canine Kidney Cells, Mice, Mice, Inbred BALB C, Specific Pathogen-Free Organisms, Vaccines, Subunit immunology, Viral Matrix Proteins immunology, Cross Protection, Influenza A Virus, H5N1 Subtype immunology, Influenza A Virus, H7N9 Subtype immunology, Influenza Vaccines immunology, Influenza, Human prevention & control
- Abstract
In March 2013, a patient infected with a novel avian influenza A H7N9 virus was reported in China. Since then, there have been 458 confirmed infection cases and 177 deaths. The virus contains several human-adapted markers, indicating that H7N9 has pandemic potential. The outbreak of this new influenza virus highlighted the need for the development of universal influenza vaccines. Previously, we demonstrated that a tetrameric peptide vaccine based on the matrix protein 2 ectodomain (M2e) of the H5N1 virus (H5N1-M2e) could protect mice from lethal infection with different clades of H5N1 and 2009 pandemic H1N1 influenza viruses. In this study, we investigated the cross-protection of H5N1-M2e against lethal infection with the new H7N9 virus. Although five amino acid differences existed at positions 13, 14, 18, 20, and 21 between M2e of H5N1 and H7N9, H5N1-M2e vaccination with either Freund's adjuvant or the Sigma adjuvant system (SAS) induced a high level of anti-M2e antibody, which cross-reacted with H7N9-M2e peptide. A mouse-adapted H7N9 strain, A/Anhui/01/2013m, was used for lethal challenge in animal experiments. H5N1-M2e vaccination provided potent cross-protection against lethal challenge of the H7N9 virus. Reduced viral replication and histopathological damage of mouse lungs were also observed in the vaccinated mice. Our results suggest that the tetrameric H5N1-M2e peptide vaccine could protect against different subtypes of influenza virus infections. Therefore, this vaccine may be an ideal candidate for developing a universal vaccine to prevent the reemergence of avian influenza A H7N9 virus and the emergence of potential novel reassortants of influenza virus.
- Published
- 2015
- Full Text
- View/download PDF
46. Active replication of Middle East respiratory syndrome coronavirus and aberrant induction of inflammatory cytokines and chemokines in human macrophages: implications for pathogenesis.
- Author
-
Zhou J, Chu H, Li C, Wong BH, Cheng ZS, Poon VK, Sun T, Lau CC, Wong KK, Chan JY, Chan JF, To KK, Chan KH, Zheng BJ, and Yuen KY
- Subjects
- Animals, Antigen Presentation, Cell Survival immunology, Cells, Cultured, Chlorocebus aethiops, Coronavirus immunology, Coronavirus pathogenicity, Coronavirus Infections immunology, Cytokines biosynthesis, Humans, Lung virology, Macrophages immunology, Pneumonia, Viral immunology, Vero Cells, Coronavirus physiology, Coronavirus Infections virology, Cytokines immunology, Macrophages virology, Pneumonia, Viral virology, Virus Replication physiology
- Abstract
Middle East respiratory syndrome coronavirus (MERS-CoV) infection caused severe pneumonia and multiorgan dysfunction and had a higher crude fatality rate (around 50% vs. 10%) than SARS coronavirus (SARS-CoV) infection. To understand the pathogenesis, we studied viral replication, cytokine/chemokine response, and antigen presentation in MERS-CoV-infected human monocyte-derived macrophages (MDMs) versus SARS-CoV-infected MDMs. Only MERS-CoV can replicate in MDMs. Both viruses were unable to significantly stimulate the expression of antiviral cytokines (interferon α [IFN-α] and IFN-β) but induced comparable levels of tumor necrosis factor α and interleukin 6. Notably, MERS-CoV induced significantly higher expression levels of interleukin 12, IFN-γ, and chemokines (IP-10/CXCL-10, MCP-1/CCL-2, MIP-1α/CCL-3, RANTES/CCL-5, and interleukin 8) than SARS-CoV. The expression of major histocompatibility complex class I and costimulatory molecules were significantly higher in MERS-CoV-infected MDMs than in SARS-CoV-infected cells. MERS-CoV replication was validated by immunostaining of infected MDMs and ex vivo lung tissue. We conclusively showed that MERS-CoV can establish a productive infection in human macrophages. The aberrant induction of inflammatory cytokines/chemokines could be important in the disease pathogenesis.
- Published
- 2014
- Full Text
- View/download PDF
47. Productive replication of Middle East respiratory syndrome coronavirus in monocyte-derived dendritic cells modulates innate immune response.
- Author
-
Chu H, Zhou J, Wong BH, Li C, Cheng ZS, Lin X, Poon VK, Sun T, Lau CC, Chan JF, To KK, Chan KH, Lu L, Zheng BJ, and Yuen KY
- Subjects
- B7-2 Antigen metabolism, Cells, Cultured, Cytokines metabolism, Healthy Volunteers, Histocompatibility Antigens Class II metabolism, Humans, Coronavirus growth & development, Coronavirus immunology, Dendritic Cells immunology, Dendritic Cells virology, Immunity, Innate
- Abstract
The Middle East respiratory syndrome coronavirus (MERS-CoV) closely resembled severe acute respiratory syndrome coronavirus (SARS-CoV) in disease manifestation as rapidly progressive acute pneumonia with multi-organ dysfunction. Using monocyte-derived-dendritic cells (Mo-DCs), we discovered fundamental discrepancies in the outcome of MERS-CoV- and SARS-CoV-infection. First, MERS-CoV productively infected Mo-DCs while SARS-CoV-infection was abortive. Second, MERS-CoV induced significantly higher levels of IFN-γ, IP-10, IL-12, and RANTES expression than SARS-CoV. Third, MERS-CoV-infection induced higher surface expression of MHC class II (HLA-DR) and the co-stimulatory molecule CD86 than SARS-CoV-infection. Overall, our data suggests that the dendritic cell can serve as an important target of viral replication and a vehicle for dissemination. MERS-CoV-infection in DCs results in the production of a rich combination of cytokines and chemokines, and modulates innate immune response differently from that of SARS-CoV-infection. Our findings may help to explain the apparent discrepancy in the pathogenicity between MERS-CoV and SARS-CoV., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
48. Identification of a receptor-binding domain in the S protein of the novel human coronavirus Middle East respiratory syndrome coronavirus as an essential target for vaccine development.
- Author
-
Du L, Zhao G, Kou Z, Ma C, Sun S, Poon VK, Lu L, Wang L, Debnath AK, Zheng BJ, Zhou Y, and Jiang S
- Subjects
- Animals, Antibodies, Neutralizing blood, Antibodies, Viral blood, Binding Sites, Cell Line, Coronavirus genetics, Coronavirus immunology, Coronavirus Infections virology, Dipeptidyl Peptidase 4 metabolism, Humans, Mice, Mice, Inbred BALB C, Protein Structure, Tertiary, Receptors, Virus metabolism, Respiratory Tract Infections virology, Spike Glycoprotein, Coronavirus genetics, Spike Glycoprotein, Coronavirus immunology, Viral Vaccines genetics, Viral Vaccines immunology, Coronavirus metabolism, Spike Glycoprotein, Coronavirus metabolism
- Abstract
A novel human Middle East respiratory syndrome coronavirus (MERS-CoV) caused outbreaks of severe acute respiratory syndrome (SARS)-like illness with a high mortality rate, raising concerns of its pandemic potential. Dipeptidyl peptidase-4 (DPP4) was recently identified as its receptor. Here we showed that residues 377 to 662 in the S protein of MERS-CoV specifically bound to DPP4-expressing cells and soluble DPP4 protein and induced significant neutralizing antibody responses, suggesting that this region contains the receptor-binding domain (RBD), which has a potential to be developed as a MERS-CoV vaccine.
- Published
- 2013
- Full Text
- View/download PDF
49. A safe and convenient pseudovirus-based inhibition assay to detect neutralizing antibodies and screen for viral entry inhibitors against the novel human coronavirus MERS-CoV.
- Author
-
Zhao G, Du L, Ma C, Li Y, Li L, Poon VK, Wang L, Yu F, Zheng BJ, Jiang S, and Zhou Y
- Subjects
- Animals, Cell Line, HIV-1 genetics, Humans, Luciferases analysis, Luciferases genetics, Mice, Spike Glycoprotein, Coronavirus genetics, Spike Glycoprotein, Coronavirus metabolism, Staining and Labeling methods, Antibodies, Neutralizing isolation & purification, Antibodies, Viral isolation & purification, Antiviral Agents isolation & purification, Coronavirus drug effects, Coronavirus immunology, Drug Evaluation, Preclinical methods, Neutralization Tests methods
- Abstract
Background: Evidence points to the emergence of a novel human coronavirus, Middle East respiratory syndrome coronavirus (MERS-CoV), which causes a severe acute respiratory syndrome (SARS)-like disease. In response, the development of effective vaccines and therapeutics remains a clinical priority. To accomplish this, it is necessary to evaluate neutralizing antibodies and screen for MERS-CoV entry inhibitors., Methods: In this study, we produced a pseudovirus bearing the full-length spike (S) protein of MERS-CoV in the Env-defective, luciferase-expressing HIV-1 backbone. We then established a pseudovirus-based inhibition assay to detect neutralizing antibodies and anti-MERS-CoV entry inhibitors., Results: Our results demonstrated that the generated MERS-CoV pseudovirus allows for single-cycle infection of a variety of cells expressing dipeptidyl peptidase-4 (DPP4), the confirmed receptor for MERS-CoV. Consistent with the results from a live MERS-CoV-based inhibition assay, the antisera of mice vaccinated with a recombinant protein containing receptor-binding domain (RBD, residues 377-662) of MERS-CoV S fused with Fc of human IgG exhibited neutralizing antibody response against infection of MERS-CoV pseudovirus. Furthermore, one small molecule HIV entry inhibitor targeting gp41 (ADS-J1) and the 3-hydroxyphthalic anhydride-modified human serum albumin (HP-HSA) could significantly inhibit MERS-CoV pseudovirus infection., Conclusion: Taken together, the established MERS-CoV inhibition assay is a safe and convenient pseudovirus-based alternative to BSL-3 live-virus restrictions and can be used to rapidly screen MERS-CoV entry inhibitors, as well as evaluate vaccine-induced neutralizing antibodies against the highly pathogenic MERS-CoV.
- Published
- 2013
- Full Text
- View/download PDF
50. Leptin mediates the pathogenesis of severe 2009 pandemic influenza A(H1N1) infection associated with cytokine dysregulation in mice with diet-induced obesity.
- Author
-
Zhang AJ, To KK, Li C, Lau CC, Poon VK, Chan CC, Zheng BJ, Hung IF, Lam KS, Xu A, and Yuen KY
- Subjects
- Animals, Antibodies administration & dosage, Antibodies immunology, Diet, High-Fat adverse effects, Disease Models, Animal, Epithelium immunology, Epithelium pathology, Female, Interleukin-1beta immunology, Interleukin-6 genetics, Kaplan-Meier Estimate, Leptin metabolism, Leptin pharmacology, Lung immunology, Lung pathology, Lung virology, Macrophages drug effects, Macrophages immunology, Macrophages virology, Mice, Mice, Inbred C57BL, Mice, Obese, Obesity virology, Orthomyxoviridae Infections immunology, Orthomyxoviridae Infections virology, Pandemics, Pneumonia immunology, Pneumonia pathology, Pneumonia virology, RNA, Messenger analysis, RNA, Messenger genetics, Recombinant Proteins immunology, Recombinant Proteins pharmacology, Viral Load, Influenza A Virus, H1N1 Subtype pathogenicity, Interleukin-6 immunology, Leptin immunology, Obesity immunology, Orthomyxoviridae Infections pathology
- Abstract
Background: Obesity is associated with a high circulating leptin level and severe 2009 pandemic influenza A virus subtype H1N1 (A[H1N1]pdm09) infection. The mechanism for severe lung injury in obese patients and the specific treatment strategy remain elusive., Method: We studied the pathogenesis of A(H1N1)pdm09 infection in a mouse model of diet-induced obesity., Results: Obese mice had significantly higher initial pulmonary viral titer and mortality after challenge with A(H1N1)pdm09, compared with age-matched lean mice. Compared with lean mice, obese mice had heightened proinflammatory cytokine and chemokine levels and more severe pulmonary inflammatory damage. Furthermore, obese mice had a higher preexisting serum leptin level but a lower preexisting adiponectin level. Recombinant mouse leptin increased the interleukin 6 (IL-6) messenger RNA expression in mouse single-lung-cell preparations, mouse macrophages, and mouse lung epithelial cell lines infected with A(H1N1)pdm09. Administration of anti-leptin antibody improved the survival of infected obese mice, with associated reductions in pulmonary levels of the proinflammatory cytokines IL-6 and interleukin 1β but not the pulmonary viral titer., Conclusions: Our findings suggest that preexisting high levels of circulating leptin contribute to the development of severe lung injury by A(H1N1)pdm09 in mice with diet-induced obesity. The therapeutic strategy of leptin neutralization for the reduction of proinflammatory responses and pulmonary damage in obese patients warrants further investigations.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.