43 results on '"Tai Ling Chao"'
Search Results
2. Development of nanoparticles incorporated with quercetin and ACE2-membrane as a novel therapy for COVID-19
- Author
-
Jia-You Fang, Kuo-Yen Huang, Tong-Hong Wang, Zih-Chan Lin, Chin-Chuan Chen, Sui-Yuan Chang, En-Li Chen, Tai-Ling Chao, Shuenn-Chen Yang, Pan-Chyr Yang, and Chi-Yuan Chen
- Subjects
Nanoparticles ,Quercetin ,AXL ,ACE2 ,COVID-19 ,Biotechnology ,TP248.13-248.65 ,Medical technology ,R855-855.5 - Abstract
Abstract Introduction Angiotensin-converting enzyme 2 (ACE2) and AXL tyrosine kinase receptor are known to be involved in the SARS-CoV-2 entry of the host cell. Therefore, targeting ACE2 and AXL should be an effective strategy to inhibit virus entry into cells. However, developing agents that can simultaneously target ACE2 and AXL remains a formidable task. The natural compound quercetin has been shown to inhibit AXL expression. Materials and methods In this study, we employed PLGA nanoparticles to prepare nanoparticles encapsulated with quercetin, coated with ACE2-containing cell membranes, or encapsulated with quercetin and then coated with ACE-2-containing cell membranes. These nanoparticles were tested for their abilities to neutralize or inhibit viral infection. Results Our data showed that nanoparticles encapsulated with quercetin and then coated with ACE2-containing cell membrane inhibited the expression of AXL without causing cytotoxic activity. Nanoparticles incorporated with both quercetin and ACE2-containing cell membrane were found to be able to neutralize pseudo virus infection and were more effective than free quercetin and nanoparticles encapsulated with quercetin at inhibition of pseudo virus and SARS-CoV-2 infection. Conclusions We have shown that the biomimetic nanoparticles incorporated with both ACE-2 membrane and quercetin showed the most antiviral activity and may be further explored for clinical application.
- Published
- 2024
- Full Text
- View/download PDF
3. Evolution of neutralizing antibodies and cross-activity against different variants of SARS-CoV-2 in patients recovering from COVID-19
- Author
-
Wang-Da Liu, Jann-Tay Wang, Tai-Ling Chao, Si-Man Ieong, Ya-Min Tsai, Po-Hsien Kuo, Ming-Jui Tsai, Yi-Jie Chen, Guei-Chi Li, Shu-Yuan Ho, Hui-Hou Chen, Yu-Shan Huang, Chien-Ching Hung, Yee-Chun Chen, Sui-Yuan Chang, and Shan-Chwen Chang
- Subjects
Plaque reduction neutralization test (PRNT) ,Humoral immunity ,Spike protein ,Variant of concern ,Delta variant ,B.1.617.2 ,Medicine (General) ,R5-920 - Abstract
Background: Patients recovering from COVID-19 may need vaccination against SARS-CoV-2 because acquired immunity from primary infection may wane, given the emergence of new SARS-CoV-2 variants. Understanding the trends of anti-spike IgG and neutralizing antibody titers in patients recovering from COVID-19 may inform the decision made on the appropriate interval between recovery and vaccination. Methods: Participants aged 20 years or older and diagnosed with COVID-19 between January and December, 2020 were enrolled. Serum specimens were collected every three months from 10 days to 12 months after the onset of symptom for determinations of anti-spike IgG and neutralizing antibody titers against SARS-CoV-2 Wuhan strain with D614G mutation, alpha, gamma and delta variants. Results: Of 19 participants, we found a decreasing trend of geometric mean titers of anti-spike IgG from 560.9 to 217 and 92 BAU/mL after a 4-month and a 7-month follow-up, respectively. The anti-spike IgG titers declined more quickly in the ten participants with severe or critical disease than the nine participants with only mild to moderate disease between one month and seven months after SARS-CoV-2 infection (−8.49 vs - 2.34-fold, p
- Published
- 2023
- Full Text
- View/download PDF
4. Immune response and safety of heterologous ChAdOx1-nCoV-19/mRNA-1273 vaccination compared with homologous ChAdOx1-nCoV-19 or homologous mRNA-1273 vaccination
- Author
-
Wang-Huei Sheng, Sui-Yuan Chang, Pin-Hung Lin, Ming-Ju Hsieh, Hao-Hsiang Chang, Chien-Yu Cheng, Hung-Chih Yang, Ching-Fu Pan, Si-Man Ieong, Tai-Ling Chao, Jang-Pin Chen, Shu-Hsing Cheng, and Shan-Chwen Chang
- Subjects
Adenovirus-vector vaccine ,Messenger RNA vaccine ,Coronavirus disease 2019 (COVID-19) ,Severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) ,Immune response ,Medicine (General) ,R5-920 - Abstract
Background/Purpose: Efficacy and safety data of heterologous prime-boost vaccination against SARS-CoV-2 remains limited. Methods: We recruited adult volunteers for homologous or heterologous prime-boost vaccinations with adenoviral (ChAdOx1, AstraZeneca) and/or mRNA (mRNA-1273, Moderna) vaccines. Four groups of prime-boost vaccination schedules were designed: Group 1, ChAdOx1/ChAdOx1 8 weeks apart; Group 2, ChAdOx1/mRNA-1273 8 weeks apart; Group 3, ChAdOx1/mRNA-1273 4 weeks apart; and Group 4, mRNA-1273/mRNA-1273 4 weeks apart. The primary outcome was serum anti-SARS-CoV-2 IgG titers and neutralizing antibody titers against B.1.1.7 (alpha) and B.1.617.2 (delta) variants on day 28 after the second dose. Adverse events were recorded up until 84 days after the second dose. Results: We enrolled 399 participants with a median age of 41 years and 75% were female. On day 28 after the second dose, the anti-SARS-CoV-2 IgG titers of both heterologous vaccinations (Group 2 and Group 3) were significantly higher than that of homologous ChAdOx1 vaccination (Group 1), and comparable with homologous mRNA-1273 vaccination (Group 4). The heterologous vaccination group had better neutralizing antibody responses against the alpha and delta variant as compared to the homologous ChAdOx1 group. Most of the adverse events (AEs) were mild and transient. AEs were less frequent when heterologous boosting was done at 8 weeks rather than at 4 weeks. Conclusion: Heterologous ChAdOx1/mRNA-1273 vaccination provided higher immunogenicity than homologous ChAdOx1 vaccination and comparable immunogenicity with the homologous mRNA-1273 vaccination. Our results support the safety and efficacy of heterologous prime-boost vaccination using the ChAdOx1 and mRNA-1273 COVID-19 vaccines. (ClinicalTrials.gov number, NCT05074368).
- Published
- 2022
- Full Text
- View/download PDF
5. A siRNA targets and inhibits a broad range of SARS‐CoV‐2 infections including Delta variant
- Author
-
Yi‐Chung Chang, Chi‐Fan Yang, Yi‐Fen Chen, Chia‐Chun Yang, Yuan‐Lin Chou, Hung‐Wen Chou, Tein‐Yao Chang, Tai‐Ling Chao, Shu‐Chen Hsu, Si‐Man Ieong, Ya‐Min Tsai, Ping‐Cheng Liu, Yuan‐Fan Chin, Jun‐Tung Fang, Han‐Chieh Kao, Hsuan‐Ying Lu, Jia‐Yu Chang, Ren‐Shiuan Weng, Qian‐Wen Tu, Fang‐Yu Chang, Kuo‐Yen Huang, Tong‐Young Lee, Sui‐Yuan Chang, and Pan‐Chyr Yang
- Subjects
COVID‐19 ,inhalation ,K18‐hACE2‐transgenic mice ,SARS‐CoV‐2 ,siRNA ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract The emergence of severe acute respiratory syndrome coronavirus‐2 (SARS‐CoV‐2) variants has altered the trajectory of the COVID‐19 pandemic and raised some uncertainty on the long‐term efficiency of vaccine strategy. The development of new therapeutics against a wide range of SARS‐CoV‐2 variants is imperative. We, here, have designed an inhalable siRNA, C6G25S, which covers 99.8% of current SARS‐CoV‐2 variants and is capable of inhibiting dominant strains, including Alpha, Delta, Gamma, and Epsilon, at picomolar ranges of IC50in vitro. Moreover, C6G25S could completely inhibit the production of infectious virions in lungs by prophylactic treatment, and decrease 96.2% of virions by cotreatment in K18‐hACE2‐transgenic mice, accompanied by a significant prevention of virus‐associated extensive pulmonary alveolar damage, vascular thrombi, and immune cell infiltrations. Our data suggest that C6G25S provides an alternative and effective approach to combating the COVID‐19 pandemic.
- Published
- 2022
- Full Text
- View/download PDF
6. Clinical assessment of SARS-CoV-2 infectivity by rapid antigen test compared with virus isolation
- Author
-
Tai-Ling Chao, Wen-Hau Lee, Hui-Chun Hu, Yi-Chun Lin, Shu-Yuan Ho, Hui-Hou Chen, Cheng-Pin Chen, Ya-Min Tsai, Jun-Tung Fang, Si-Man Leong, Yu-Chen Cheng, Mavis Peng, Shu-Hsing Cheng, Chien-Yu Cheng, and Sui-Yuan Chang
- Subjects
SARS-COV-2 ,Real-time RT-PCR ,Rapid antigen test ,Infectivity ,Infectious and parasitic diseases ,RC109-216 - Abstract
Although real-time reverse transcriptase polymerase chain reaction (real-time RT-PCR) remains as a golden standard for detection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, it can not be easily expanded to large-scaled screening during outbreaks, and the positive results do not necessarily correlate with infectious status of the identified subjects. In this study, the performance of Vstrip® RV2 COVID-19 Antigen Rapid Test (RAT) and its correlation with virus infectivity was examined by virus culture using 163 sequential respiratory specimens collected from 26 SARS-CoV-2 infected patients. When the presence of cytopathic effects (CPE) in cell culture was used as a reference method for virus infectivity, the sensitivity, specificity and accuracy of Vstrip® RV2 COVID-19 Antigen Rapid Test was 96.43%, 89.63%, and 90.8%, respectively. The highest Ct value was 27.7 for RdRp gene and 25.79 for E gene within CPE-positive samples, and the highest Ct value was 31.9 for RdRp gene and 29.1 for E gene within RAT positive samples. When the Ct values of specimens were below 25, the CPE and RAT results had high degree of consistency. We concluded that the RAT could be a great alternative method for determining the infectious potential of individuals with high viral load.
- Published
- 2023
- Full Text
- View/download PDF
7. Evolution of SARS-CoV-2 neutralizing antibody in an HIV-positive patient with COVID-19
- Author
-
Wang-Da Liu, Chien-Ching Hung, Jann-Tay Wang, Ming-Jui Tsai, Po-Hsien Kuo, Tai-Ling Chao, Szu-Min Hsieh, Wang-Huei Sheng, Yee-Chun Chen, Sui-Yuan Chang, and Shan-Chwen Chang
- Subjects
Antiretroviral therapy ,Coronavirus ,Enzyme-linked immunosorbent assay (ELISA) ,Plaque reduction assay ,Severe acute respiratory syndrome ,Medicine (General) ,R5-920 - Abstract
We presented the clinical course and immune responses of a well-controlled HIV-positive patient with COVID-19. The clinical presentation and antibody production to SARS-CoV-2 were similar to other COVID-19 patients without HIV infection. Neutralizing antibody reached a plateau from 26th to 47th day onset but decreased on 157th day after symptoms.
- Published
- 2021
- Full Text
- View/download PDF
8. Nanoparticle composite TPNT1 is effective against SARS-CoV-2 and influenza viruses
- Author
-
Sui-Yuan Chang, Kuo-Yen Huang, Tai-Ling Chao, Han-Chieh Kao, Yu-Hao Pang, Lin Lu, Chun-Lun Chiu, Hsin-Chang Huang, Ting-Jen Rachel Cheng, Jim-Min Fang, and Pan-Chyr Yang
- Subjects
Medicine ,Science - Abstract
Abstract A metal nanoparticle composite, namely TPNT1, which contains Au-NP (1 ppm), Ag-NP (5 ppm), ZnO-NP (60 ppm) and ClO2 (42.5 ppm) in aqueous solution was prepared and characterized by spectroscopy, transmission electron microscopy, dynamic light scattering analysis and potentiometric titration. Based on the in vitro cell-based assay, TPNT1 inhibited six major clades of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) with effective concentration within the range to be used as food additives. TPNT1 was shown to block viral entry by inhibiting the binding of SARS-CoV-2 spike proteins to the angiotensin-converting enzyme 2 (ACE2) receptor and to interfere with the syncytium formation. In addition, TPNT1 also effectively reduced the cytopathic effects induced by human (H1N1) and avian (H5N1) influenza viruses, including the wild-type and oseltamivir-resistant virus isolates. Together with previously demonstrated efficacy as antimicrobials, TPNT1 can block viral entry and inhibit or prevent viral infection to provide prophylactic effects against both SARS-CoV-2 and opportunistic infections.
- Published
- 2021
- Full Text
- View/download PDF
9. Neutralizing Monoclonal Antibodies Inhibit SARS-CoV-2 Infection through Blocking Membrane Fusion
- Author
-
Chia-Jung Li, Tai-Ling Chao, Ting-Yu Chang, Chia-Chun Hsiao, De-Chao Lu, Yi-Wei Chiang, Guan-Chun Lai, Ya-Min Tsai, Jun-Tung Fang, Siman Ieong, Jann-Tay Wang, Sui-Yuan Chang, and Shih-Chung Chang
- Subjects
neutralizing antibodies ,SARS-CoV-2 ,spike (S) protein ,S2 subunit ,membrane fusion ,Microbiology ,QR1-502 - Abstract
ABSTRACT Most of SARS-CoV-2 neutralizing antibodies (nAbs) targeted the receptor binding domain (RBD) of the SARS-CoV-2 spike (S) protein. However, mutations at RBD sequences found in the emerging SARS-CoV-2 variants greatly reduced the effectiveness of nAbs. Here we showed that four nAbs, S2-4D, S2-5D, S2-8D, and S2-4A, which recognized a conserved epitope in the S2 subunit of the S protein, can inhibit SARS-CoV-2 infection through blocking the S protein-mediated membrane fusion. Notably, these four nAbs exhibited broadly neutralizing activity against SARS-CoV-2 Alpha, Gamma, Delta, and Epsilon variants. Antisera collected from mice immunized with the identified epitope peptides of these four nAbs also exhibited potent virus neutralizing activity. Discovery of the S2-specific nAbs and their unique antigenic epitopes paves a new path for development of COVID-19 therapeutics and vaccines. IMPORTANCE The spike (S) protein on the surface of SARS-CoV-2 mediates receptor binding and virus-host cell membrane fusion during virus entry. Many neutralizing antibodies (nAbs), which targeted the receptor binding domain (RBD) of S protein, lost the neutralizing activity against the newly emerging SARS-CoV-2 variants with sequence mutations at the RBD. In contrast, the nAb against the highly conserved S2 subunit, which plays the key role in virus–host cell membrane fusion, was poorly discovered. We showed that four S2-specific nAbs, S2-4D, S2-5D, S2-8D, and S2-4A, inhibited SARS-CoV-2 infection through blocking the S protein-mediated membrane fusion. These nAbs exhibited broadly neutralizing activity against Alpha, Gamma, Delta, and Epsilon variants. Antisera induced by the identified epitope peptides also possessed potent neutralizing activity. This work not only unveiled the S2-specific nAbs but also discovered an immunodominant epitope in the S2 subunit that can be rationally designed as the broad-spectrum vaccine against the SARS-like coronaviruses.
- Published
- 2022
- Full Text
- View/download PDF
10. Humanized COVID‐19 decoy antibody effectively blocks viral entry and prevents SARS‐CoV‐2 infection
- Author
-
Kuo‐Yen Huang, Ming‐Shiu Lin, Ting‐Chun Kuo, Ci‐Ling Chen, Chung‐Chih Lin, Yu‐Chi Chou, Tai‐Ling Chao, Yu‐Hao Pang, Han‐Chieh Kao, Rih‐Sheng Huang, Steven Lin, Sui‐Yuan Chang, and Pan‐Chyr Yang
- Subjects
ACE2‐Fc ,COVID‐19 ,decoy antibody ,SARS‐CoV‐2 ,virus infection ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract To circumvent the devastating pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) infection, a humanized decoy antibody (ACE2‐Fc fusion protein) was designed to target the interaction between viral spike protein and its cellular receptor, angiotensin‐converting enzyme 2 (ACE2). First, we demonstrated that ACE2‐Fc could specifically abrogate virus replication by blocking the entry of SARS‐CoV‐2 spike‐expressing pseudotyped virus into both ACE2‐expressing lung cells and lung organoids. The impairment of viral entry was not affected by virus variants, since efficient inhibition was also observed in six SARS‐CoV‐2 clinical strains, including the D614G variants which have been shown to exhibit increased infectivity. The preservation of peptidase activity also enables ACE2‐Fc to reduce the angiotensin II‐mediated cytokine cascade. Furthermore, this Fc domain of ACE2‐Fc was shown to activate NK cell degranulation after co‐incubation with Spike‐expressing H1975 cells. These promising characteristics potentiate the therapeutic prospects of ACE2‐Fc as an effective treatment for COVID‐19.
- Published
- 2020
- Full Text
- View/download PDF
11. Correction: Lee et al. The Synergistic Inhibition of Coronavirus Replication and Induced Cytokine Production by Ciclesonide and the Tylophorine-Based Compound Dbq33b. Pharmaceutics 2022, 14, 1511
- Author
-
Yue-Zhi Lee, Hsing-Yu Hsu, Cheng-Wei Yang, Yi-Ling Lin, Sui-Yuan Chang, Ruey-Bing Yang, Jian-Jong Liang, Tai-Ling Chao, Chun-Che Liao, Han-Chieh Kao, Jang-Yang Chang, Huey-Kang Sytwu, Chiung-Tong Chen, and Shiow-Ju Lee
- Subjects
n/a ,Pharmacy and materia medica ,RS1-441 - Abstract
In the original publication [...]
- Published
- 2023
- Full Text
- View/download PDF
12. Tag-Free SARS-CoV-2 Receptor Binding Domain (RBD), but Not C-Terminal Tagged SARS-CoV-2 RBD, Induces a Rapid and Potent Neutralizing Antibody Response
- Author
-
Ting-Wei Lin, Ping-Han Huang, Bo-Hung Liao, Tai-Ling Chao, Ya-Min Tsai, Shih-Chung Chang, Sui-Yuan Chang, and Hui-Wen Chen
- Subjects
COVID-19 ,vaccine ,tag-free RBD ,His-tag RBD ,neutralizing antibodies ,Delta variant ,Medicine - Abstract
Recombinant proteins are essential in the development of subunit vaccines. In the design of many recombinant proteins, polyhistidine residues are added to the N- or C-termini of target sequences to facilitate purification. However, whether the addition of tag residues influences the immunogenicity of proteins remains unknown. In this study, the tag-free SARS-CoV-2 RBD and His-tag SARS-CoV-2 RBD proteins were investigated to determine whether there were any differences in their receptor binding affinity and immunogenicity. The results showed that the tag-free RBD protein had a higher affinity for binding with hACE2 receptors than His-tag RBD proteins (EC50: 1.78 µM vs. 7.51 µM). On day 21 after primary immunization with the proteins, the serum ELISA titers of immunized mice were measured and found to be 1:1418 for those immunized with tag-free RBD and only 1:2.4 for His-tag RBD. Two weeks after the booster dose, tag-free-RBD-immunized mice demonstrated a significantly higher neutralizing titer of 1:369 compared with 1:7.9 for His-tag-RBD-immunized mice. Furthermore, neutralizing antibodies induced by tag-free RBD persisted for up to 5 months and demonstrated greater cross-neutralization of the SARS-CoV-2 Delta variant. Evidence from Western blotting showed that the serum of His-tag-RBD-immunized mice recognized irrelevant His-tag proteins. Collectively, we conclude that the addition of a polyhistidine tag on a recombinant protein, when used as a COVID-19 vaccine antigen, may significantly impair protein immunogenicity against SARS-CoV-2. Antibody responses induced were clearly more rapid and robust for the tag-free SARS-CoV-2 RBD than the His-tag SARS-CoV-2 RBD. These findings provide important information for the design of antigens used in the development of COVID-19 subunit vaccines.
- Published
- 2022
- Full Text
- View/download PDF
13. Remdesivir and Cyclosporine Synergistically Inhibit the Human Coronaviruses OC43 and SARS-CoV-2
- Author
-
Hsing-Yu Hsu, Cheng-Wei Yang, Yue-Zhi Lee, Yi-Ling Lin, Sui-Yuan Chang, Ruey-Bing Yang, Jian-Jong Liang, Tai-Ling Chao, Chun-Che Liao, Han-Chieh Kao, Szu-Huei Wu, Jang-Yang Chang, Huey-Kang Sytwu, Chiung-Tong Chen, and Shiow-Ju Lee
- Subjects
COVID-19 ,cyclosporine ,IL-6 ,IL-8 ,OC43 ,remdesivir ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Remdesivir, a prodrug targeting RNA-dependent-RNA-polymerase, and cyclosporine, a calcineurin inhibitor, individually exerted inhibitory activity against human coronavirus OC43 (HCoV-OC43) in HCT-8 and MRC-5 cells at EC50 values of 96 ± 34 ∼ 85 ± 23 nM and 2,920 ± 364 ∼ 4,419 ± 490 nM, respectively. When combined, these two drugs synergistically inhibited HCoV-OC43 in both HCT-8 and MRC-5 cells assayed by immunofluorescence assay (IFA). Remdesivir and cyclosporine also separately reduced IL-6 production induced by HCoV-OC43 in human lung fibroblasts MRC-5 cells with EC50 values of 224 ± 53 nM and 1,292 ± 352 nM, respectively; and synergistically reduced it when combined. Similar trends were observed for SARS-CoV-2, which were 1) separately inhibited by remdesivir and cyclosporine with respective EC50 values of 3,962 ± 303 nM and 7,213 ± 143 nM by IFA, and 291 ± 91 nM and 6,767 ± 1,827 nM by a plaque-formation assay; and 2) synergistically inhibited by their combination, again by IFA and plaque-formation assay. Collectively, these results suggest that the combination of remdesivir and cyclosporine merits further study as a possible treatment for COVID-19 complexed with a cytokine storm.
- Published
- 2021
- Full Text
- View/download PDF
14. D614G Substitution of SARS-CoV-2 Spike Protein Increases Syncytium Formation and Virus Titer via Enhanced Furin-Mediated Spike Cleavage
- Author
-
Ya-Wen Cheng, Tai-Ling Chao, Chiao-Ling Li, Sheng-Han Wang, Han-Chieh Kao, Ya-Min Tsai, Hurng-Yi Wang, Chi-Ling Hsieh, You-Yu Lin, Pei-Jer Chen, Sui-Yuan Chang, and Shiou-Hwei Yeh
- Subjects
furin ,SARS-CoV-2 ,spike ,syncytium ,Microbiology ,QR1-502 - Abstract
ABSTRACT Since the D614G substitution in the spike (S) protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged, the variant strain has undergone a rapid expansion to become the most abundant strain worldwide. Therefore, this substitution may provide an advantage for viral spreading. To explore the mechanism, we analyzed 18 viral isolates containing S proteins with either G614 or D614 (S-G614 and S-D614, respectively). The plaque assay showed a significantly higher virus titer in S-G614 than in S-D614 isolates. We further found increased cleavage of the S protein at the furin substrate site, a key event that promotes syncytium formation, in S-G614 isolates. The enhancement of the D614G substitution in the cleavage of the S protein and in syncytium formation has been validated in cells expressing S protein. The effect on the syncytium was abolished by furin inhibitor treatment and mutation of the furin cleavage site, suggesting its dependence on cleavage by furin. Our study pointed to the impact of the D614G substitution on syncytium formation through enhanced furin-mediated S cleavage, which might increase the transmissibility and infectivity of SARS-CoV-2 strains containing S-G614. IMPORTANCE Analysis of viral genomes and monitoring of the evolutionary trajectory of SARS-CoV-2 over time has identified the D614G substitution in spike (S) as the most prevalent expanding variant worldwide, which might confer a selective advantage in transmission. Several studies showed that the D614G variant replicates and transmits more efficiently than the wild-type virus, but the mechanism is unclear. By comparing 18 virus isolates containing S with either D614 or G614, we found significantly higher virus titers in association with higher furin protease-mediated cleavage of S, an event that promotes syncytium formation and virus infectivity, in the S-G614 viruses. The effect of the D614G substitution on furin-mediated S cleavage and the resulting enhancement of the syncytium phenotype has been validated in S-expressing cells. This study suggests a possible effect of the D614G substitution on S of SARS-CoV-2; the antiviral effect through targeting furin protease is worthy of being investigated in proper animal models.
- Published
- 2021
- Full Text
- View/download PDF
15. The Synergistic Inhibition of Coronavirus Replication and Induced Cytokine Production by Ciclesonide and the Tylophorine-Based Compound Dbq33b
- Author
-
Yue-Zhi Lee, Hsing-Yu Hsu, Cheng-Wei Yang, Yi-Ling Lin, Sui-Yuan Chang, Ruey-Bing Yang, Jian-Jong Liang, Tai-Ling Chao, Chun-Che Liao, Han-Chieh Kao, Jang-Yang Chang, Huey-Kang Sytwu, Chiung-Tong Chen, and Shiow-Ju Lee
- Subjects
COVID-19 ,ciclesonide ,cytokine ,IL-6 ,IL-8 ,MCP-1 ,Pharmacy and materia medica ,RS1-441 - Abstract
Ciclesonide is an inhaled corticosteroid used to treat asthma and has been repurposed as a treatment for mildly ill COVID-19 patients, but its precise mechanism of action is unclear. Herein, we report that ciclesonide blocks the coronavirus-induced production of the cytokines IL-6, IL-8, and MCP-1 by increasing IκBα protein levels and significantly decreasing p65 nuclear translocation. Furthermore, we found that the combination of ciclesonide and dbq33b, a potent tylophorine-based coronavirus inhibitor that affects coronavirus-induced NF-κB activation a little, additively and synergistically decreased coronavirus-induced IL-6, IL-8, and MCP-1 cytokine levels, and synergistically inhibited the replication of both HCoV-OC43 and SARS-CoV-2. Collectively, the combination of ciclesonide and dbq33b merits consideration as a treatment for COVID-19 patients who may otherwise be overwhelmed by high viral loads and an NF-κB-mediated cytokine storm.
- Published
- 2022
- Full Text
- View/download PDF
16. Differentiation of Cytopathic Effects (CPE) induced by influenza virus infection using deep Convolutional Neural Networks (CNN).
- Author
-
Ting-En Wang, Tai-Ling Chao, Hsin-Tsuen Tsai, Pi-Han Lin, Yen-Lung Tsai, and Sui-Yuan Chang
- Published
- 2020
- Full Text
- View/download PDF
17. Elicitation of potent neutralizing antibodies in obese mice by ISA 51-adjuvanted SARS-CoV-2 spike RBD-Fc vaccine
- Author
-
Chia-Jung Li, Chung-Lin Jiang, Tai-Ling Chao, Shiau-Yu Lin, Ya-Min Tsai, Chong-Syun Chao, Yu-Ting Su, Chun-Jen Chen, Sui-Yuan Chang, Fu-Jung Lin, and Shih-Chung Chang
- Subjects
General Medicine ,Applied Microbiology and Biotechnology ,Biotechnology - Published
- 2023
- Full Text
- View/download PDF
18. Immunogenicity and safety of third-dose mRNA COVID-19 vaccines in healthy adults previously vaccinated with two doses of the ChAdOx1 vaccine
- Author
-
Wang-Huei Sheng, Si-Man Ieong, Pin-Hung Lin, Ming-Ju Hsieh, Hung-Chih Yang, Ching-Fu Pan, Tai-Ling Chao, Sui-Yuan Chang, and Shan-Chwen Chang
- Subjects
General Medicine - Abstract
The efficacy and safety of coronavirus disease 2019 (COVID-19) booster vaccines remain limited. We investigated the immunogenicity and adverse events of the third dose of mRNA vaccines in healthy adults.Volunteers vaccinated with two doses of the adenoviral vaccine (ChAdOx1) 12 weeks before were administered with an mRNA COVID-19 vaccine. These were divided into three groups, full-dose mRNA-1273 (group 1); half-dose mRNA-1273 (group 2); and full-dose BNT-162b2 (group 3). Primary outcomes included serum anti-SARS-CoV-2 spike immunoglobulin G (IgG) titers and neutralizing antibody titers against B.1.1.7 (alpha), B.1.617.2 (delta), and B.1.1.529 (omicron) variants. Secondary outcomes included the evaluation of humoral and cellular immunity and vaccine-associated adverse events after the boost.Totally 300 participants were recruited, and 298 participants were enrolled. For all three groups, an increase in anti-SARS-CoV-2 spike IgG geometric mean titers (30.12- to 71.80-fold) and neutralizing antibody titers against the alpha variant (69.80- to 173.23-folds), delta variant (132.69- to 324.63-folds), and omicron variant (135.36- to 222.37-folds) were observed on day 28. All groups showed robust T- and B-cell responses after boosting. Adverse events were overall mild and transient but with higher prevalence and severity in group 1 participants than in other groups.Third dose mRNA COVID-19 vaccines markedly enhanced cellular and humoral responses and were safe. Immunological responses and adverse events were higher in individuals receiving the full-dose mRNA-1273 vaccine, followed by a half-dose mRNA-1273 vaccine and BNT-162b2 vaccine.
- Published
- 2023
- Full Text
- View/download PDF
19. Enhancement of the IFN-β-induced host signature informs repurposed drugs for COVID-19
- Author
-
Chen-Tsung Huang, Tai-Ling Chao, Han-Chieh Kao, Yu-Hao Pang, Wen-Hau Lee, Chiao-Hui Hsieh, Sui-Yuan Chang, Hsuan-Cheng Huang, and Hsueh-Fen Juan
- Subjects
Systems biology ,COVID-19 ,Host-directed therapy ,Type I interferon ,Drug repurposing ,Bioinformatics ,Science (General) ,Q1-390 ,Social sciences (General) ,H1-99 - Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a causative agent for the outbreak of coronavirus disease 2019 (COVID-19). This global pandemic is now calling for efforts to develop more effective COVID-19 therapies. Here we use a host-directed approach, which focuses on cellular responses to diverse small-molecule treatments, to identify potentially effective drugs for COVID-19. This framework looks at the ability of compounds to elicit a similar transcriptional response to IFN-β, a type I interferon that fails to be induced at notable levels in response to SARS-CoV-2 infection. By correlating the perturbation profiles of ~3,000 small molecules with a high-quality signature of IFN-β-responsive genes in primary normal human bronchial epithelial cells, our analysis revealed four candidate COVID-19 compounds, namely homoharringtonine, narciclasine, anisomycin, and emetine. We experimentally confirmed that the predicted compounds significantly inhibited SARS-CoV-2 replication in Vero E6 cells at nanomolar, relatively non-toxic concentrations, with half-maximal inhibitory concentrations of 165.7 nM, 16.5 nM, and 31.4 nM for homoharringtonine, narciclasine, and anisomycin, respectively. Together, our results corroborate a host-centric strategy to inform protective antiviral therapies for COVID-19.
- Published
- 2020
- Full Text
- View/download PDF
20. Inhibition of SARS-CoV-2 by Highly Potent Broad-Spectrum Anti-Coronaviral Tylophorine-Based Derivatives
- Author
-
Cheng-Wei Yang, Yue-Zhi Lee, Hsing-Yu Hsu, Jia-Tsrong Jan, Yi-Ling Lin, Sui-Yuan Chang, Tzu-Ting Peng, Ruey-Bing Yang, Jian-Jong Liang, Chun-Che Liao, Tai-Ling Chao, Yu-Hau Pang, Han-Chieh Kao, Wen-Zheng Huang, Jiunn-Horng Lin, Chun-Ping Chang, Guang-Hao Niu, Szu-Huei Wu, Huey-Kang Sytwu, Chiung-Tong Chen, and Shiow-Ju Lee
- Subjects
HCoV-OC43 ,HCoV-229E ,FIPV ,tylophorine ,SARS-CoV-2 ,ouabain ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Tylophorine-based compounds and natural cardiotonic steroids (cardenolides and bufadienolides) are two classes of transmissible gastroenteritis coronavirus inhibitors, targeting viral RNA and host cell factors, respectively. We tested both types of compounds against two types of coronaviruses, to compare and contrast their antiviral properties, and with view to their further therapeutic development. Examples of both types of compounds potently inhibited the replication of both feline infectious peritonitis virus and human coronavirus OC43 with EC50 values of up to 8 and 16 nM, respectively. Strikingly, the tylophorine-based compounds tested inhibited viral yields of HCoV-OC43 to a much greater extent (7–8 log magnitudes of p.f.u./ml) than the cardiotonic steroids (about 2–3 log magnitudes of p.f.u./ml), as determined by end point assays. Based on these results, three tylophorine-based compounds were further examined for their anti-viral activities on two other human coronaviruses, HCoV-229E and SARS-CoV-2. These three tylophorine-based compounds inhibited HCoV-229E with EC50 values of up to 6.5 nM, inhibited viral yields of HCoV-229E by 6–7 log magnitudes of p.f.u./ml, and were also found to inhibit SARS-CoV-2 with EC50 values of up to 2.5–14 nM. In conclusion, tylophorine-based compounds are potent, broad-spectrum inhibitors of coronaviruses including SARS-CoV-2, and could be used for the treatment of COVID-19.
- Published
- 2020
- Full Text
- View/download PDF
21. Characterization of Influenza A Virus Infection in Mouse Pulmonary Stem/Progenitor Cells
- Author
-
Tai-Ling Chao, Sing-Yi Gu, Pi-Han Lin, Yu-Tien Chou, Thai-Yen Ling, and Sui-Yuan Chang
- Subjects
influenza ,influenza A virus ,mouse pulmonary stem cells ,mouse pulmonary progenitor cells ,pro-inflammatory responses ,Microbiology ,QR1-502 - Abstract
The pulmonary stem/progenitor cells, which could be differentiated into downstream cells to repair tissue damage caused by influenza A virus, have also been shown to be the target cells of influenza virus infection. In this study, mouse pulmonary stem/progenitor cells (mPSCs) with capability to differentiate into type I or type II alveolar cells were used as an in vitro cell model to characterize replication and pathogenic effects of influenza viruses in PSCs. First, mPSCs and its immortalized cell line mPSCsOct4+ were shown to be susceptible to PR8, seasonal H1N1, 2009 pandemic H1N1, and H7N9 influenza viruses and can generate infectious virus particles, although with a lower virus titer, which could be attributed by the reduced vRNA replication and nucleoprotein (NP) aggregation in the cytoplasm. Nevertheless, a significant increase of interleukin (IL)-6 and interferon (IFN)-γ at 12 h and IFN-β at 24 h post infection in mPSCs implicates that mPSCs might function as a sensor to modulate immune responses to influenza virus infection. In summary, our results demonstrated mPSCs, as one of the target cells for influenza A viruses, could modulate early proinflammatory responses to influenza virus infection.
- Published
- 2020
- Full Text
- View/download PDF
22. Drug Repurposing for the Identification of Compounds with Anti-SARS-CoV-2 Capability via Multiple Targets
- Author
-
Pei-Chen Yu, Chen-Hao Huang, Chih-Jung Kuo, Po-Huang Liang, Lily Hui-Ching Wang, Max Yu-Chen Pan, Sui-Yuan Chang, Tai-Ling Chao, Si-Man Ieong, Jun-Tung Fang, Hsuan-Cheng Huang, and Hsueh-Fen Juan
- Subjects
docking simulation ,severe acute respiratory syndrome coronavirus 2 ,transmembrane protease serine 2 (TMPRSS2) ,3C-like protease (3CLpro/Mpro) ,papain-like protease (PLpro) ,tamoxifen ,Pharmacy and materia medica ,RS1-441 - Abstract
Since 2019, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been rapidly spreading worldwide, causing hundreds of millions of infections. Despite the development of vaccines, insufficient protection remains a concern. Therefore, the screening of drugs for the treatment of coronavirus disease 2019 (COVID-19) is reasonable and necessary. This study utilized bioinformatics for the selection of compounds approved by the U.S. Food and Drug Administration with therapeutic potential in this setting. In addition, the inhibitory effect of these compounds on the enzyme activity of transmembrane protease serine 2 (TMPRSS2), papain-like protease (PLpro), and 3C-like protease (3CLpro) was evaluated. Furthermore, the capability of compounds to attach to the spike-receptor-binding domain (RBD) was considered an important factor in the present assessment. Finally, the antiviral potency of compounds was validated using a plaque reduction assay. Our funnel strategy revealed that tamoxifen possesses an anti-SARS-CoV-2 property owing to its inhibitory performance in multiple assays. The proposed time-saving and feasible strategy may accelerate drug screening for COVID-19 and other diseases.
- Published
- 2022
- Full Text
- View/download PDF
23. The Src–ZNRF1 axis controls TLR3 trafficking and interferon responses to limit lung barrier damage
- Author
-
You-Sheng Lin, Yung-Chi Chang, Tai-Ling Chao, Ya-Min Tsai, Shu-Jhen Jhuang, Yu-Hsin Ho, Ting-Yu Lai, Yi-Ling Liu, Chiung-Ya Chen, Ching-Yen Tsai, Yi-Ping Hsueh, Sui-Yuan Chang, Tsung-Hsien Chuang, Chih-Yuan Lee, and Li-Chung Hsu
- Subjects
Immunology ,Immunology and Allergy - Abstract
Type I interferons are important antiviral cytokines, but prolonged interferon production is detrimental to the host. The TLR3-driven immune response is crucial for mammalian antiviral immunity, and its intracellular localization determines induction of type I interferons; however, the mechanism terminating TLR3 signaling remains obscure. Here, we show that the E3 ubiquitin ligase ZNRF1 controls TLR3 sorting into multivesicular bodies/lysosomes to terminate signaling and type I interferon production. Mechanistically, c-Src kinase activated by TLR3 engagement phosphorylates ZNRF1 at tyrosine 103, which mediates K63-linked ubiquitination of TLR3 at lysine 813 and promotes TLR3 lysosomal trafficking and degradation. ZNRF1-deficient mice and cells are resistant to infection by encephalomyocarditis virus and SARS-CoV-2 because of enhanced type I interferon production. However, Znrf1−/− mice have exacerbated lung barrier damage triggered by antiviral immunity, leading to enhanced susceptibility to respiratory bacterial superinfections. Our study highlights the c-Src–ZNRF1 axis as a negative feedback mechanism controlling TLR3 trafficking and the termination of TLR3 signaling.
- Published
- 2023
- Full Text
- View/download PDF
24. Antagonistic pleiotropy plays an important role in governing the evolution and genetic diversity of SARS-CoV-2
- Author
-
Ding-Chin Lee, Jui-Hung Tai, Hsin-Fu Lin, Tai-Ling Chao, Yongsen Ruan, Ya-Wen Cheng, Yu-Chi Chou, You-Yu Lin, Sui-Yuan Chang, Pei-Jer Chen, Shiou-Hwei Yeh, and Hurng-Yi Wang
- Abstract
Analyses of the genomic diversity of SARS-CoV-2 found that some sites across the genome appear to have mutated independently multiple times with frequency significantly higher than four-fold sites, which can be either due to mutational bias, i.e., elevated mutation rate in some sites of the genome, or selection of the variants due to antagonistic pleiotropy, a condition where mutations increase some components of fitness at a cost to others. To examine how different forces shaped evolution of SARS-CoV-2 in 2020–2021, we analyzed a large set of genome sequences (~ 2 million). Here we show that while evolution of SARS-CoV-2 during the pandemic was largely mutation-driven, a group of nonsynonymous changes is probably maintained by antagonistic pleiotropy. To test this hypothesis, we studied the function of one such mutation, spike M1237I. Spike I1237 increases viral assembly and secretion, but decreases efficiency of transmissionin vitro. Therefore, while the frequency of spike M1237I may increase within hosts, viruses carrying this mutation would be outcompeted at the population level. We also discuss how the antagonistic pleiotropy might facilitate positive epistasis to promote virus adaptation and reconcile discordant estimates of SARS-CoV-2 transmission bottleneck sizes in previous studies.
- Published
- 2023
- Full Text
- View/download PDF
25. IFN‐stimulated metabolite transporter ENT3 facilitates viral genome release
- Author
-
Yu‐Ting Hsieh, Tsung‐Lin Tsai, Shen‐Yan Huang, Jian‐Wen Heng, Yu‐Chia Huang, Pei‐Yuan Tsai, Chia‐Chun Tu, Tai‐Ling Chao, Ya‐Min Tsai, Pei‐Ching Chang, Chien‐Kuo Lee, Guann‐Yi Yu, Sui‐Yuan Chang, Ivan L. Dzhagalov, and Chia‐Lin Hsu
- Subjects
Genetics ,Molecular Biology ,Biochemistry - Published
- 2023
- Full Text
- View/download PDF
26. Evolution of SARS-CoV-2 neutralizing antibody in an HIV-positive patient with COVID-19
- Author
-
Sui-Yuan Chang, Szu-Min Hsieh, Yee-Chun Chen, Jann-Tay Wang, Wang-Huei Sheng, Po-Hsien Kuo, Wang-Da Liu, Tai-Ling Chao, Chien-Ching Hung, Shan-Chwen Chang, and Tsai Mj
- Subjects
Medicine (General) ,Coronavirus disease 2019 (COVID-19) ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Human immunodeficiency virus (HIV) ,Case Report ,medicine.disease_cause ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,R5-920 ,medicine ,Neutralizing antibody ,Coronavirus ,biology ,Plaque reduction assay ,business.industry ,Clinical course ,Enzyme-linked immunosorbent assay (ELISA) ,virus diseases ,General Medicine ,Positive patient ,Antiretroviral therapy ,Severe acute respiratory syndrome ,030220 oncology & carcinogenesis ,Immunology ,biology.protein ,030211 gastroenterology & hepatology ,business - Abstract
We presented the clinical course and immune responses of a well-controlled HIV-positive patient with COVID-19. The clinical presentation and antibody production to SARS-CoV-2 were similar to other COVID-19 patients without HIV infection. Neutralizing antibody reached a plateau from 26th to 47th day onset but decreased on 157th day after symptoms.
- Published
- 2021
27. Evolution of neutralizing antibodies and cross-activity against different variants of SARS-CoV-2 in patients recovering from COVID-19
- Author
-
Wang-Da Liu, Jann-Tay Wang, Tai-Ling Chao, Si-Man Ieong, Ya-Min Tsai, Po-Hsien Kuo, Ming-Jui Tsai, Yi-Jie Chen, Guei-Chi Li, Shu-Yuan Ho, Hui-Hou Chen, Yu-Shan Huang, Chien-Ching Hung, Yee-Chun Chen, Sui-Yuan Chang, and Shan-Chwen Chang
- Subjects
General Medicine - Abstract
Patients recovering from COVID-19 may need vaccination against SARS-CoV-2 because acquired immunity from primary infection may wane, given the emergence of new SARS-CoV-2 variants. Understanding the trends of anti-spike IgG and neutralizing antibody titers in patients recovering from COVID-19 may inform the decision made on the appropriate interval between recovery and vaccination.Participants aged 20 years or older and diagnosed with COVID-19 between January and December, 2020 were enrolled. Serum specimens were collected every three months from 10 days to 12 months after the onset of symptom for determinations of anti-spike IgG and neutralizing antibody titers against SARS-CoV-2 Wuhan strain with D614G mutation, alpha, gamma and delta variants.Of 19 participants, we found a decreasing trend of geometric mean titers of anti-spike IgG from 560.9 to 217 and 92 BAU/mL after a 4-month and a 7-month follow-up, respectively. The anti-spike IgG titers declined more quickly in the ten participants with severe or critical disease than the nine participants with only mild to moderate disease between one month and seven months after SARS-CoV-2 infection (-8.49 vs - 2.34-fold, p 0.001). The neutralizing activity of the convalescent serum specimens collected from participants recovering from wild-type SARS-CoV-2 infection against different variants was lower, especially against the delta variants (p 0.01 for each variant with Wuhan strain as reference).Acquired immunity from primary infection with SARS-CoV-2 waned within 4-7 months in COVID-19 patients, and neutralizing cross-activities against different SARS-CoV-2 variants were lower compared with those against wild-type strain.
- Published
- 2022
28. Equilibrative Nucleoside Transporter 3 is an IFN-stimulated Gene that Facilitates Viral Genome Release
- Author
-
Yu-Ting Hsieh, Tsung-Lin Tsai, Chia-Chun Tu, Shen-Yan Huang, Jian-Wen Heng, Pei-Yuan Tsai, Tai-Ling Chao, Ya-Min Tsai, Pei-Ching Chang, Chien-Kuo Lee, Guann-Yi Yu, Sui-Yuan Chang, Ivan L. Dzhagalov, and Chia-Lin Hsu
- Abstract
An increasing body of evidence emphasizes the role of metabolic reprogramming in immune cells to fight off infections. However, little is known about the regulation of metabolite transporters that facilitate and support metabolic demands. In this study, we found that equilibrative nucleoside transporter 3 (ENT3) expression is part of the innate immune response, and is rapidly upregulated upon bacterial and viral infection. The transcription of ENT3 is directly under the regulation of IFN-induced signaling, positioning this metabolite transporter as an Interferon-stimulated gene (ISG). Moreover, we unveil that several viruses, including SARS-CoV2, require ENT3 to facilitate their entry into the cytoplasm. The removal or suppression of ENT3 expression is sufficient to significantly decrease viral replication in vitro and in vivo.
- Published
- 2022
- Full Text
- View/download PDF
29. Vaccination with SARS-CoV-2 spike protein lacking glycan shields elicits enhanced protective responses in animal models
- Author
-
Han-Yi Huang, Hsin-Yu Liao, Xiaorui Chen, Szu-Wen Wang, Cheng-Wei Cheng, Md. Shahed-Al-Mahmud, Yo-Min Liu, Arpita Mohapatra, Ting-Hua Chen, Jennifer M. Lo, Yi-Min Wu, Hsiu-Hua Ma, Yi-Hsuan Chang, Ho-Yang Tsai, Yu-Chi Chou, Yi-Ping Hsueh, Ching-Yen Tsai, Pau-Yi Huang, Sui-Yuan Chang, Tai-Ling Chao, Han-Chieh Kao, Ya-Min Tsai, Yen-Hui Chen, Chung-Yi Wu, Jia-Tsrong Jan, Ting-Jen Rachel Cheng, Kuo-I Lin, Che Ma, and Chi-Huey Wong
- Subjects
Mice ,COVID-19 Vaccines ,Polysaccharides ,SARS-CoV-2 ,Models, Animal ,Spike Glycoprotein, Coronavirus ,Vaccination ,Animals ,COVID-19 ,Humans ,General Medicine ,Antibodies, Viral ,Antibodies, Neutralizing - Abstract
A major challenge to end the pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is to develop a broadly protective vaccine that elicits long-term immunity. As the key immunogen, the viral surface spike (S) protein is frequently mutated, and conserved epitopes are shielded by glycans. Here, we revealed that S protein glycosylation has site-differential effects on viral infectivity. We found that S protein generated by lung epithelial cells has glycoforms associated with increased infectivity. Compared to the fully glycosylated S protein, immunization of S protein with N-glycans trimmed to the mono-GlcNAc–decorated state (S MG ) elicited stronger immune responses and better protection for human angiotensin-converting enzyme 2 (hACE2) transgenic mice against variants of concern (VOCs). In addition, a broadly neutralizing monoclonal antibody was identified from S MG -immunized mice that could neutralize wild-type SARS-CoV-2 and VOCs with subpicomolar potency. Together, these results demonstrate that removal of glycan shields to better expose the conserved sequences has the potential to be an effective and simple approach for developing a broadly protective SARS-CoV-2 vaccine.
- Published
- 2022
30. Neutralization or enhancement of SARS-CoV-2 infection by a monoclonal antibody targeting a specific epitope in the spike receptor-binding domain
- Author
-
Guan-Chun Lai, Tai-Ling Chao, Shiau-Yu Lin, Han-Chieh Kao, Ya-Min Tsai, De-Chao Lu, Yi-Wei Chiang, Sui-Yuan Chang, and Shih-Chung Chang
- Subjects
Pharmacology ,Epitopes ,Mice ,HEK293 Cells ,SARS-CoV-2 ,Virology ,Spike Glycoprotein, Coronavirus ,Animals ,Antibodies, Monoclonal ,COVID-19 ,Humans - Abstract
Neutralizing antibodies (NAbs) are believed to be promising prophylactic and therapeutic treatment against the coronavirus disease 2019 (COVID-19), which is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Here, we reported two mouse monoclonal antibodies 7 Eb-4G and 1Ba-3H that specifically recognized the receptor-binding domain (RBD) of SARS-CoV-2 spike (S) protein without exhibiting cross-reactivity with the S proteins of SARS-CoV and MERS-CoV. The binding epitopes of 7 Eb-4G and 1Ba-3H were respectively located in the regions of residues 457-476 and 477-496 in the S protein. Only 1Ba-3H exhibited the neutralizing activity for preventing the pseudotyped lentivirus from binding to the angiotensin-converting enzyme 2 (ACE2)-transfected HEK293T cells. The competitive ELISA further showed that 1Ba-3H interfered with the binding between RBD and ACE2. Epitope mapping experiments demonstrated that a single alanine replacement at residues 480, 482, 484, 485, and 488-491 in the RBD abrogated 1Ba-3H binding. 1Ba-3H exhibited the neutralizing activity against the wild-type, Alpha, Delta, and Epsilon variants of SARS-CoV-2, but lost the neutralizing activity against Gamma variant in the plaque reduction assay. On the contrary, 1Ba-3H enhanced the cellular infection of Gamma variant in a dose-dependent manner. Our findings suggest that the antibody-dependent enhancement of infection mediated by the RBD-specific antibody for different SARS-CoV-2 variants must be considered while developing the NAb.
- Published
- 2021
31. Remdesivir and Cyclosporine Synergistically Inhibit the Human Coronaviruses OC43 and SARS-CoV-2
- Author
-
Sui-Yuan Chang, Cheng Wei Yang, Yue Zhi Lee, Tai Ling Chao, Chiung-Tong Chen, Jian Jong Liang, Ruey-Bing Yang, Szu Huei Wu, Han Chieh Kao, Chun Che Liao, Huey-Kang Sytwu, Jang Yang Chang, Hsing Yu Hsu, Shiow Ju Lee, and Yi-Ling Lin
- Subjects
synergistic ,remdesivir ,RM1-950 ,Pharmacology ,Immunofluorescence ,medicine ,Pharmacology (medical) ,Human coronavirus OC43 ,cyclosporine ,Interleukin 8 ,Interleukin 6 ,Original Research ,EC50 ,IL-6 ,IL-8 ,biology ,medicine.diagnostic_test ,SARS-CoV-2 ,Chemistry ,COVID-19 ,virus diseases ,OC43 ,Prodrug ,biology.organism_classification ,medicine.disease ,Calcineurin ,biology.protein ,Therapeutics. Pharmacology ,Cytokine storm - Abstract
Remdesivir, a prodrug targeting RNA-dependent-RNA-polymerase, and cyclosporine, a calcineurin inhibitor, individually exerted inhibitory activity against human coronavirus OC43 (HCoV-OC43) in HCT-8 and MRC-5 cells at EC50 values of 96 ± 34 ∼ 85 ± 23 nM and 2,920 ± 364 ∼ 4,419 ± 490 nM, respectively. When combined, these two drugs synergistically inhibited HCoV-OC43 in both HCT-8 and MRC-5 cells assayed by immunofluorescence assay (IFA). Remdesivir and cyclosporine also separately reduced IL-6 production induced by HCoV-OC43 in human lung fibroblasts MRC-5 cells with EC50 values of 224 ± 53 nM and 1,292 ± 352 nM, respectively; and synergistically reduced it when combined. Similar trends were observed for SARS-CoV-2, which were 1) separately inhibited by remdesivir and cyclosporine with respective EC50 values of 3,962 ± 303 nM and 7,213 ± 143 nM by IFA, and 291 ± 91 nM and 6,767 ± 1,827 nM by a plaque-formation assay; and 2) synergistically inhibited by their combination, again by IFA and plaque-formation assay. Collectively, these results suggest that the combination of remdesivir and cyclosporine merits further study as a possible treatment for COVID-19 complexed with a cytokine storm.
- Published
- 2021
- Full Text
- View/download PDF
32. D614G Substitution of SARS-CoV-2 Spike Protein Increases Syncytium Formation and Virus Titer via Enhanced Furin-Mediated Spike Cleavage
- Author
-
Sui-Yuan Chang, Hurng-Yi Wang, Han Chieh Kao, Sheng Han Wang, Shiou-Hwei Yeh, You Yu Lin, Tai Ling Chao, Chiao Ling Li, Chi Ling Hsieh, Ya Min Tsai, Pei-Jer Chen, and Ya Wen Cheng
- Subjects
0301 basic medicine ,viruses ,Genome, Viral ,Virus Replication ,Cleavage (embryo) ,medicine.disease_cause ,Giant Cells ,Microbiology ,Virus ,Cell Line ,03 medical and health sciences ,Virology ,Chlorocebus aethiops ,medicine ,Animals ,Humans ,Vero Cells ,Furin ,Infectivity ,Virus quantification ,Syncytium ,Mutation ,030102 biochemistry & molecular biology ,biology ,Chemistry ,SARS-CoV-2 ,COVID-19 ,spike ,Viral Load ,QR1-502 ,HEK293 Cells ,030104 developmental biology ,Amino Acid Substitution ,Spike Glycoprotein, Coronavirus ,Vero cell ,biology.protein ,Genetic Fitness ,furin ,syncytium ,Research Article - Abstract
Since the D614G substitution in the spike (S) protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged, the variant strain has undergone a rapid expansion to become the most abundant strain worldwide. Therefore, this substitution may provide an advantage for viral spreading. To explore the mechanism, we analyzed 18 viral isolates containing S proteins with either G614 or D614 (S-G614 and S-D614, respectively). The plaque assay showed a significantly higher virus titer in S-G614 than in S-D614 isolates. We further found increased cleavage of the S protein at the furin substrate site, a key event that promotes syncytium formation, in S-G614 isolates. The enhancement of the D614G substitution in the cleavage of the S protein and in syncytium formation has been validated in cells expressing S protein. The effect on the syncytium was abolished by furin inhibitor treatment and mutation of the furin cleavage site, suggesting its dependence on cleavage by furin. Our study pointed to the impact of the D614G substitution on syncytium formation through enhanced furin-mediated S cleavage, which might increase the transmissibility and infectivity of SARS-CoV-2 strains containing S-G614. IMPORTANCE Analysis of viral genomes and monitoring of the evolutionary trajectory of SARS-CoV-2 over time has identified the D614G substitution in spike (S) as the most prevalent expanding variant worldwide, which might confer a selective advantage in transmission. Several studies showed that the D614G variant replicates and transmits more efficiently than the wild-type virus, but the mechanism is unclear. By comparing 18 virus isolates containing S with either D614 or G614, we found significantly higher virus titers in association with higher furin protease-mediated cleavage of S, an event that promotes syncytium formation and virus infectivity, in the S-G614 viruses. The effect of the D614G substitution on furin-mediated S cleavage and the resulting enhancement of the syncytium phenotype has been validated in S-expressing cells. This study suggests a possible effect of the D614G substitution on S of SARS-CoV-2; the antiviral effect through targeting furin protease is worthy of being investigated in proper animal models.
- Published
- 2021
- Full Text
- View/download PDF
33. Impact of glycosylation on a broad-spectrum vaccine against SARS-CoV-2
- Author
-
Sui-Yuan Chang, Kuo-I Lin, Ya-Min Tsai, Hsin-Yu Liao, Yi-Ping Hsueh, Che Ma, Cheng-Wei Cheng, Yu Chi Chou, Chung-Yi Wu, Ting-Jen R. Cheng, Yi-Min Wu, Md. Shahed-Al-Mahmud, Jia-Tsrong Jan, Ting-Hua Chen, Jennifer M. Lo, Chi-Huey Wong, Han-Chieh Kao, Yi-Hsuan Chang, Szu-Wen Wang, Ching-Yen Tsai, Xiaorui Chen, Yen-Hui Chen, Tai-Ling Chao, Han-Yi Huang, Hsiu-Hua Ma, Yo-Min Liu, Ho-Yang Tsai, and Pau-Yi Huang
- Subjects
Infectivity ,Glycan ,Immunogen ,Glycosylation ,biology ,medicine.drug_class ,Monoclonal antibody ,Virology ,Epitope ,Conserved sequence ,chemistry.chemical_compound ,Immune system ,chemistry ,medicine ,biology.protein - Abstract
A major challenge to end the pandemic caused by SARS-CoV-2 is to develop a broadly protective vaccine. As the key immunogen, the spike protein is frequently mutated with conserved epitopes shielded by glycans. Here, we reveal that spike glycosylation has site-differential effects on viral infectivity and lung epithelial cells generate spike with more infective glycoforms. Compared to the fully glycosylated spike, immunization of spike protein with N-glycans trimmed to the monoglycosylated state (Smg) elicits stronger immune responses and better protection for hACE2 transgenic mice against variants of concern. In addition, a broadly neutralizing monoclonal antibody was identified from the Smg immunized mice, demonstrating that removal of glycan shields to better expose the conserved sequences is an effective and simple approach to broad-spectrum vaccine development.One-Sentence SummaryRemoving glycan shields to expose conserved epitopes is an effective approach to develop a broad-spectrum SARS-CoV-2 vaccine.
- Published
- 2021
- Full Text
- View/download PDF
34. Kinetic Characterization and Inhibitor Screening for the Proteases Leading to Identification of Drugs against SARS-CoV-2
- Author
-
Sui-Yuan Chang, Chih-Jung Kuo, Yi-Kai Liu, Lily Hui-Ching Wang, Po-Huang Liang, Han-Chieh Kao, Ya-Min Tsai, Tai-Ling Chao, and Ming-Chang Hsieh
- Subjects
Drug ,Proteases ,Polyproteins ,medicine.medical_treatment ,media_common.quotation_subject ,viruses ,medicine.disease_cause ,Antiviral Agents ,Cell Line ,Substrate Specificity ,03 medical and health sciences ,0302 clinical medicine ,antivirals ,inhibitors ,Chlorocebus aethiops ,Medicine ,Animals ,Humans ,Pharmacology (medical) ,Protease Inhibitors ,Vero Cells ,030304 developmental biology ,EC50 ,Coronavirus ,media_common ,Pharmacology ,0303 health sciences ,Protease ,drug repurposing ,business.industry ,SARS-CoV-2 ,virus diseases ,COVID-19 ,3CLpro ,biochemical phenomena, metabolism, and nutrition ,Virology ,respiratory tract diseases ,Drug repositioning ,PLpro ,Kinetics ,Infectious Diseases ,030220 oncology & carcinogenesis ,Vero cell ,business ,Peptide Hydrolases - Abstract
Coronavirus (CoV) disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has claimed many lives worldwide and is still spreading since December 2019. The 3C-like protease (3CLpro) and papain-like protease (PLpro) are essential for maturation of viral polyproteins in SARS-CoV-2 life cycle and thus regarded as key drug targets for the disease., Coronavirus (CoV) disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has claimed many lives worldwide and is still spreading since December 2019. The 3C-like protease (3CLpro) and papain-like protease (PLpro) are essential for maturation of viral polyproteins in SARS-CoV-2 life cycle and thus regarded as key drug targets for the disease. In this study, 3CLpro and PLpro assay platforms were established, and their substrate specificities were characterized. The assays were used to screen collections of 1,068 and 2,701 FDA-approved drugs. After excluding the externally used drugs which are too toxic, we totally identified 12 drugs as 3CLpro inhibitors and 36 drugs as PLpro inhibitors active at 10 μM. Among these inhibitors, six drugs were found to suppress SARS-CoV-2 with the half-maximal effective concentration (EC50) below or close to 10 μM. This study enhances our understanding on the proteases and provides FDA-approved drugs for prevention and/or treatment of COVID-19.
- Published
- 2021
35. Design, synthesis and biological evaluations of niclosamide analogues against SARS-CoV-2
- Author
-
Yu-Pu Juang, Yu-Ting Chou, Ru-Xian Lin, Hsiu-Hua Ma, Tai-Ling Chao, Jia-Tsrong Jan, Sui-Yuan Chang, and Pi-Hui Liang
- Subjects
Molecular Docking Simulation ,Pharmacology ,SARS-CoV-2 ,Organic Chemistry ,Drug Discovery ,Humans ,Niclosamide ,General Medicine ,Antiviral Agents ,COVID-19 Drug Treatment - Abstract
Niclosamide, a widely-used anthelmintic drug, inhibits SARS-CoV-2 virus entry through TMEM16F inhibition and replication through autophagy induction, but the relatively high cytotoxicity and poor oral bioavailability limited its application. We synthesized 22 niclosamide analogues of which compound 5 was found to exhibit the best anti-SARS-CoV-2 efficacy (IC
- Published
- 2022
- Full Text
- View/download PDF
36. Identification of Entry Inhibitors against Delta and Omicron Variants of SARS-CoV-2
- Author
-
Richard Kuan-Lin Lee, Tian-Neng Li, Sui-Yuan Chang, Tai-Ling Chao, Chun-Hsien Kuo, Max Yu-Chen Pan, Yu-Ting Chiou, Kuan-Ju Liao, Yi Yang, Yi-Hsuan Wu, Chen-Hao Huang, Hsueh-Fen Juan, Hsing-Pang Hsieh, and Lily Hui-Ching Wang
- Subjects
SARS-CoV-2 ,viruses ,Organic Chemistry ,General Medicine ,Catalysis ,COVID-19 Drug Treatment ,Computer Science Applications ,Molecular Docking Simulation ,Inorganic Chemistry ,Spike Glycoprotein, Coronavirus ,Humans ,RNA, Viral ,COVID-19 ,viral entry ,receptor-binding domain ,entry inhibitor ,ACE2 ,Angiotensin-Converting Enzyme 2 ,Physical and Theoretical Chemistry ,Molecular Biology ,Spectroscopy - Abstract
Entry inhibitors against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are urgently needed to control the outbreak of coronavirus disease 2019 (COVID-19). This study developed a robust and straightforward assay that detected the molecular interaction between the receptor-binding domain (RBD) of viral spike protein and the angiotensin-converting enzyme 2 (ACE2) receptor in just 10 min. A drug library of 1068 approved compounds was used to screen for SARS-CoV2 entry inhibition, and 9 active drugs were identified as specific pseudovirus entry inhibitors. A plaque reduction neutralization test using authentic SARS-CoV-2 virus in Vero E6 cells confirmed that 2 of these drugs (Etravirine and Dolutegravir) significantly inhibited the infection of SARS-CoV-2. With molecular docking, we showed that both Etravirine and Dolutegravir are preferentially bound to primary ACE2-interacting residues on the RBD domain, implying that these two drug blocks may prohibit the viral attachment of SARS-CoV-2. We compared the neutralizing activities of these entry inhibitors against different pseudoviruses carrying spike proteins from alpha, beta, gamma, and delta variants. Both Etravirine and Dolutegravir showed similar neutralizing activities against different variants, with EC50 values between 4.5 to 5.8 nM for Etravirine and 10.2 to 22.9 nM for Dolutegravir. These data implied that Etravirine and Dolutegravir may serve as general spike inhibitors against dominant viral variants of SARS-CoV-2.
- Published
- 2022
- Full Text
- View/download PDF
37. Inhibition of SARS-CoV-2 by Highly Potent Broad-Spectrum Anti-Coronaviral Tylophorine-Based Derivatives
- Author
-
Chiung-Tong Chen, Jiunn Horng Lin, Guang Hao Niu, Shiow Ju Lee, Sui-Yuan Chang, Wen Zheng Huang, Jia Tsrong Jan, Tzu Ting Peng, Hsing Yu Hsu, Yi-Ling Lin, Szu Huei Wu, Yu Hau Pang, Han Chieh Kao, Chun-Ping Chang, Jian Jong Liang, Ruey-Bing Yang, Chun Che Liao, Yue Zhi Lee, Tai Ling Chao, Huey-Kang Sytwu, and Cheng Wei Yang
- Subjects
0301 basic medicine ,HCoV-OC43 ,Coronavirus disease 2019 (COVID-19) ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,viruses ,Transmissible gastroenteritis coronavirus ,coronavirus ,medicine.disease_cause ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Pharmacology (medical) ,Human coronavirus OC43 ,Original Research ,Coronavirus ,EC50 ,Cytopathic effect ,Pharmacology ,Virus quantification ,FIPV ,biology ,Chemistry ,SARS-CoV-2 ,tylophorine ,lcsh:RM1-950 ,ouabain ,COVID-19 ,virus diseases ,HCoV-229E ,biology.organism_classification ,Molecular biology ,030104 developmental biology ,lcsh:Therapeutics. Pharmacology ,030220 oncology & carcinogenesis - Abstract
Tylophorine-based compounds and natural cardiotonic steroids (cardenolides and bufadienolides) are two classes of transmissible gastroenteritis coronavirus inhibitors, targeting viral RNA and host cell factors, respectively We tested both types of compounds against two types of coronaviruses, to compare and contrast their antiviral properties, and with view to their further therapeutic development Examples of both types of compounds potently inhibited the replication of both feline infectious peritonitis virus and human coronavirus OC43 with EC50 values of up to 8 and 16 nM, respectively Strikingly, the tylophorine-based compounds tested inhibited viral yields of HCoV-OC43 to a much greater extent (7–8 log magnitudes of p f u /ml) than the cardiotonic steroids (about 2–3 log magnitudes of p f u /ml), as determined by end point assays Based on these results, three tylophorine-based compounds were further examined for their anti-viral activities on two other human coronaviruses, HCoV-229E and SARS-CoV-2 These three tylophorine-based compounds inhibited HCoV-229E with EC50 values of up to 6 5 nM, inhibited viral yields of HCoV-229E by 6–7 log magnitudes of p f u /ml, and were also found to inhibit SARS-CoV-2 with EC50 values of up to 2 5–14 nM In conclusion, tylophorine-based compounds are potent, broad-spectrum inhibitors of coronaviruses including SARS-CoV-2, and could be used for the treatment of COVID-19 © Copyright © 2020 Yang, Lee, Hsu, Jan, Lin, Chang, Peng, Yang, Liang, Liao, Chao, Pang, Kao, Huang, Lin, Chang, Niu, Wu, Sytwu, Chen and Lee
- Published
- 2020
- Full Text
- View/download PDF
38. Furin Inhibitors Block SARS-CoV-2 Spike Protein Cleavage to Suppress Virus Production and Cytopathic Effects
- Author
-
Chih Hui Lin, Han Chieh Kao, Tung Ching Ho, Shiou-Hwei Yeh, Sui-Yuan Chang, Wen Hau Lee, Mi-Hua Tao, Yu Hao Pang, Chiao Ling Li, Ya-Wen Cheng, Sheng-Han Wang, Li Ting Jang, Tai Ling Chao, Ping-Yi Wu, Pei-Jer Chen, Mu Fan Chiu, and Ya Min Tsai
- Subjects
0301 basic medicine ,Camostat ,Proteases ,viruses ,Biology ,Cleavage (embryo) ,Virus Replication ,Antiviral Agents ,Virus ,General Biochemistry, Genetics and Molecular Biology ,Article ,cytopathic effect ,Amino Acid Chloromethyl Ketones ,03 medical and health sciences ,chemistry.chemical_compound ,Betacoronavirus ,0302 clinical medicine ,Viral entry ,Chlorocebus aethiops ,Animals ,Humans ,Protease Inhibitors ,Furin ,Vero Cells ,Cytopathic effect ,Syncytium ,SARS-CoV-2 ,virus diseases ,spike ,Fluoresceins ,Transmembrane Protease Serine 2 ,Virology ,Cell biology ,030104 developmental biology ,chemistry ,Proteolysis ,Spike Glycoprotein, Coronavirus ,biology.protein ,furin ,030217 neurology & neurosurgery ,syncytium - Abstract
Development of specific antivirals is an urgent unmet need for SARS-coronavirus 2 (SARS-CoV-2) infections. This study focuses on host proteases that proteolytically activate the SARS-CoV-2 spike protein, critical for its fusion after binding to angiotensin-converting enzyme 2 (ACE2), as antiviral targets. We first validated cleavage at a putative furin substrate motif at SARS-CoV-2 spike by expressing it in VeroE6 cells and found prominent syncytium formation. Both cleavage and syncytium were abolished by treatment with furin inhibitors decanoyl-RVKR-chloromethylketone (CMK) and naphthofluorescein but not by transmembrane protease serine 2 (TMPRSS2) inhibitor camostat. CMK and naphthofluorescein showed antiviral effects in SARS-CoV-2-infected cells by decreasing viral production and cytopathic effects. Further analysis revealed that, similar to camostat, CMK blocks virus entry, but it further suppresses the cleavage of spike and syncytium. Naphthofluorescein instead acts primarily by suppressing viral RNA transcription. Therefore, furin inhibitors may become promising antivirals for prevention and treatment of SARS-CoV-2 infections., Graphical Abstract, Highlights ● The furin cleavage site in the SARS-CoV-2 spike protein mediates syncytium formation. ● The SARS-CoV-2 spike-mediated syncytium is suppressed by specific furin inhibitors. ● Furin inhibitors block SARS-CoV-2 viral entry and viral replication. ● Furin inhibitors are potential antivirals for SARS-CoV-2 infection and pathogenesis., Development of effective antivirals is an urgent unmet need for SARS-CoV-2 infections. Cheng et al. find that the cleavage of the furin substrate site in the viral spike protein is critical for viral production and cytopathic effects. Two inhibitors targeting furin are potential antivirals to control SARS-CoV-2 infection and pathogenesis.
- Published
- 2020
39. Nanoparticle composite TPNT1 is effective against SARS-CoV-2 and influenza viruses
- Author
-
Chun Lun Chiu, Jim-Min Fang, Lu Lin, Tai Ling Chao, Han Chieh Kao, Kuo Yen Huang, Ting-Jen R. Cheng, Pan-Chyr Yang, Hsin Chang Huang, Sui-Yuan Chang, and Yu Hao Pang
- Subjects
0301 basic medicine ,Oseltamivir ,Silver ,Science ,viruses ,Metal Nanoparticles ,02 engineering and technology ,medicine.disease_cause ,Antiviral Agents ,Virus ,Article ,Nanocomposites ,03 medical and health sciences ,chemistry.chemical_compound ,Influenza A Virus, H1N1 Subtype ,Viral entry ,Drug Resistance, Viral ,medicine ,Influenza A virus ,Humans ,Particle Size ,Syncytium ,Multidisciplinary ,Influenza A Virus, H5N1 Subtype ,Chemistry ,SARS-CoV-2 ,virus diseases ,Virus Internalization ,021001 nanoscience & nanotechnology ,Antimicrobial ,Virology ,Influenza A virus subtype H5N1 ,In vitro ,030104 developmental biology ,HEK293 Cells ,Spike Glycoprotein, Coronavirus ,Medicine ,Nanoparticles ,Food Additives ,Angiotensin-Converting Enzyme 2 ,Gold ,Zinc Oxide ,0210 nano-technology ,Protein Binding - Abstract
A metal nanoparticle composite, namely TPNT1, which contains Au-NP (1 ppm), Ag-NP (5 ppm), ZnO-NP (60 ppm) and ClO2 (42.5 ppm) in aqueous solution was prepared and characterized by spectroscopy, transmission electron microscopy, dynamic light scattering analysis and potentiometric titration. Based on the in vitro cell-based assay, TPNT1 inhibited six major clades of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) with effective concentration within the range to be used as food additives. TPNT1 was shown to block viral entry by inhibiting the binding of SARS-CoV-2 spike proteins to the angiotensin-converting enzyme 2 (ACE2) receptor and to interfere with the syncytium formation. In addition, TPNT1 also effectively reduced the cytopathic effects induced by human (H1N1) and avian (H5N1) influenza viruses, including the wild-type and oseltamivir-resistant virus isolates. Together with previously demonstrated efficacy as antimicrobials, TPNT1 can block viral entry and inhibit or prevent viral infection to provide prophylactic effects against both SARS-CoV-2 and opportunistic infections.
- Published
- 2020
40. Differentiation of Cytopathic Effects (CPE) induced by influenza virus infection using deep Convolutional Neural Networks (CNN)
- Author
-
Hsin-Tsuen Tsai, Sui-Yuan Chang, Tai-Ling Chao, Yen-Lung Tsai, Pi-Han Lin, and Ting-En Wang
- Subjects
0301 basic medicine ,RNA viruses ,Influenza Viruses ,Viral Diseases ,Adenoviruses ,Pulmonology ,viruses ,Pathology and Laboratory Medicine ,Convolutional neural network ,Diagnostic Radiology ,Madin Darby Canine Kidney Cells ,0302 clinical medicine ,Cytopathogenic Effect, Viral ,Medicine and Health Sciences ,Biology (General) ,Ecology ,Artificial neural network ,Radiology and Imaging ,Infection Imaging ,Antibodies, Monoclonal ,Cell Differentiation ,Orthomyxoviridae ,Infectious Diseases ,Computational Theory and Mathematics ,Current practice ,Medical Microbiology ,Modeling and Simulation ,Viral Pathogens ,Viruses ,Antibody ,Pathogens ,Research Article ,Computer and Information Sciences ,Neural Networks ,medicine.drug_class ,Imaging Techniques ,QH301-705.5 ,Biology ,Monoclonal antibody ,Research and Analysis Methods ,Microbiology ,Virus ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Dogs ,Parvoviruses ,Adeno-Associated Viruses ,Orthomyxoviridae Infections ,Diagnostic Medicine ,Genetics ,medicine ,Animals ,Molecular Biology ,Microbial Pathogens ,Ecology, Evolution, Behavior and Systematics ,Biology and life sciences ,Organisms ,Assay sensitivity ,Virology ,Influenza ,030104 developmental biology ,Cell culture ,Respiratory Infections ,biology.protein ,Neural Networks, Computer ,DNA viruses ,030217 neurology & neurosurgery ,Orthomyxoviruses ,Neuroscience ,Developmental Biology - Abstract
Cell culture remains as the golden standard for primary isolation of viruses in clinical specimens. In the current practice, researchers have to recognize the cytopathic effects (CPE) induced by virus infection and subsequently use virus-specific monoclonal antibody to confirm the presence of virus. Considering the broad applications of neural network in various fields, we aimed to utilize convolutional neural networks (CNN) to shorten the timing required for CPE identification and to improve the assay sensitivity. Based on the characteristics of influenza-induced CPE, a CNN model with larger sizes of filters and max-pooling kernels was constructed in the absence of transfer learning. A total of 601 images from mock-infected and influenza-infected MDCK cells were used to train the model. The performance of the model was tested by using extra 400 images and the percentage of correct recognition was 99.75%. To further examine the limit of our model in evaluating the changes of CPE overtime, additional 1190 images from a new experiment were used and the recognition rates at 16 hour (hr), 28 hr, and 40 hr post virus infection were 71.80%, 98.25%, and 87.46%, respectively. The specificity of our model, examined by images of MDCK cells infected by six other non-influenza viruses, was 100%. Hence, a simple CNN model was established to enhance the identification of influenza virus in clinical practice., Author summary Observation of cytopathic effects (CPE) induced by virus infection is a practical method to determine the prsence of viruses in the clinical specimens. However, CPE observation is labor-intensive and time-consuming because it requires medical examiner to inspect cell morphology changes for a period of time. Here, Convolutional Neural Networks (CNN) was applied to improve the disadvantage of CPE observation by using influenza virus as an example. To reduce the requirement for large image input of every clinical test, small amount of data was used to train our CNNs model without transfer learning and the trained model was examined with testing image data taken at 25hr post virus infection. The recognition of testing data shows that the model can identify CPE at 25hr and the high specificity of the model can differentiate the CPE induced by influenza viruses from those by other non-influenza viruses. The limit of our model was further examined by more experimental data of influenza-induced and mock-infected images, and the result shows our model can detect the slight changes at the initial stage of CPE development. Hence, our deep CNN model can significantly shorten the timing required to identify virus-induced cytopathic effects.
- Published
- 2020
41. Distinct Inductions of and Responses to Type I and Type III Interferons Promote Infections in Two SARS-CoV-2 Isolates
- Author
-
Sui-Yuan Chang, Chih-Hui Lin, Jing-Yi Lin, Han-Chieh Kao, Fu Hsin, Helene Minyi Liu, Jann-Tay Wang, Wang-Da Liu, Yu-Hao Pang, Yun-Rui Chan, Ya-Min Tsai, and Tai-Ling Chao
- Subjects
Innate immune system ,biology ,viruses ,virus diseases ,Outbreak ,biochemical phenomena, metabolism, and nutrition ,medicine.disease_cause ,medicine.disease ,Phenotype ,Virology ,Neutralization ,respiratory tract diseases ,Incubation period ,Viral pneumonia ,biology.protein ,medicine ,Antibody ,skin and connective tissue diseases ,Coronavirus - Abstract
The recent emerging coronavirus, SARS-CoV-2, has been rapidly and widely spread and causing an ongoing viral pneumonia outbreak worldwide. It has been observed that SARS-CoV-2 patients show a rather long and asymptomatic incubation time. We characterized the abilities to induce and to response to IFNβ/IFNλ1 of two or our clinical isolates, SARS-CoV-2/NTU01/TWN/human/2020 and SARS-CoV-2/NTU02/TWN/human/2020, which exhibit only two amino acid differences over the ∼30kb viral genome. We found that both isolates may infect Huh7, A549 and Calu-3 cells, yet the RIG-I-like receptor-dependent antiviral signaling was poorly induced in these cells in the early infections. Unexpectedly, we found that the intracellular vRNA levels of these isolates were sustained upon to type I/III IFN treatments, and this phenotype was more pronounced in the Taiwan/NTU01/2020 isolate. The type I/III IFN responses are antiviral but partially proviral in the case of SARS-CoV-2 infections. Poor induction and response to innate immunity may contribute to destitute neutralization index of the antibody produced, and indeed we found that the patient serum could not efficiently neutralize SARS-CoV-2 virions. With better understandings of the interplay between SARS-CoV-2 and the host antiviral innate immunity, our report may provide new insights for the regimen of therapies for SARS-CoV-2 infected patients.
- Published
- 2020
- Full Text
- View/download PDF
42. Enhancement of the IFN-β-induced host signature informs repurposed drugs for COVID-19
- Author
-
Chiao Hui Hsieh, Sui-Yuan Chang, Hsuan Cheng Huang, Wen Hau Lee, Chen Tsung Huang, Yu Hao Pang, Tai Ling Chao, Hsueh Fen Juan, and Han Chieh Kao
- Subjects
0301 basic medicine ,Bioinformatics ,Narciclasine ,Drug repurposing ,Biology ,Pharmacology ,Host-directed therapy ,Transcriptome ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Interferon ,medicine ,lcsh:Social sciences (General) ,Pharmaceutical sciences ,Transcriptomics ,Type I interferon ,lcsh:Science (General) ,Anisomycin ,Infectious disease ,Multidisciplinary ,COVID-19 ,Pharmaceutical science ,Drug repositioning ,030104 developmental biology ,chemistry ,Homoharringtonine ,Vero cell ,lcsh:H1-99 ,Systems biology ,030217 neurology & neurosurgery ,Research Article ,lcsh:Q1-390 ,medicine.drug - Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a causative agent for the outbreak of coronavirus disease 2019 (COVID-19). This global pandemic is now calling for efforts to develop more effective COVID-19 therapies. Here we use a host-directed approach, which focuses on cellular responses to diverse small-molecule treatments, to identify potentially effective drugs for COVID-19. This framework looks at the ability of compounds to elicit a similar transcriptional response to IFN-β, a type I interferon that fails to be induced at notable levels in response to SARS-CoV-2 infection. By correlating the perturbation profiles of ~3,000 small molecules with a high-quality signature of IFN-β-responsive genes in primary normal human bronchial epithelial cells, our analysis revealed four candidate COVID-19 compounds, namely homoharringtonine, narciclasine, anisomycin, and emetine. We experimentally confirmed that the predicted compounds significantly inhibited SARS-CoV-2 replication in Vero E6 cells at nanomolar, relatively non-toxic concentrations, with half-maximal inhibitory concentrations of 165.7 nM, 16.5 nM, and 31.4 nM for homoharringtonine, narciclasine, and anisomycin, respectively. Together, our results corroborate a host-centric strategy to inform protective antiviral therapies for COVID-19., Systems biology; COVID-19; Host-directed therapy; Type I interferon; Drug repurposing; Bioinformatics; Infectious disease; Transcriptomics; Pharmaceutical science
- Published
- 2020
- Full Text
- View/download PDF
43. Virological, serological, and antiviral studies in an imported human case of avian influenza A(H7N9) virus in Taiwan
- Author
-
Yee-Chun Chen, Chien-Ching Hung, Zong-Yu Yang, Pi-Han Lin, Hong-Shiee Lai, Hui-Hou Chen, C. C. Chang, Pan-Chyr Yang, Sui-Yuan Chang, Shu-Yuan Ho, Jann-Tay Wang, Shuenn-Wen Kuo, Tai-Ling Chao, Huan-Chun Lin, Shan-Chwen Chang, and Ming-Siang Huang
- Subjects
Microbiology (medical) ,Oseltamivir ,viruses ,Molecular Sequence Data ,Respiratory System ,Mutation, Missense ,Taiwan ,medicine.disease_cause ,Antibodies, Viral ,Influenza A Virus, H7N9 Subtype ,H5N1 genetic structure ,Antiviral Agents ,Virus ,Serology ,chemistry.chemical_compound ,Drug Resistance, Viral ,Influenza, Human ,Medicine ,Humans ,biology ,business.industry ,Sequence Analysis, DNA ,Viral Load ,Virology ,Influenza A virus subtype H5N1 ,Infectious Diseases ,chemistry ,biology.protein ,RNA, Viral ,Antibody ,business ,Viral load ,Transmission and infection of H5N1 - Abstract
We describe the clinical course and virological characteristics of the first H7N9 influenza case in a Taiwanese patient; this patient had detectable viruses in the airway for 2 weeks, during which time an oseltamivir resistance-associated R292K mutation rapidly emerged. Anti-H7N9 antibody was detected 21 days after onset of symptoms, when H7N9 viral load declined significantly.
- Published
- 2013
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.