13 results on '"Kim, Seungtaek"'
Search Results
2. No correlation of neutralizing antibody titers against the Omicron variant after a booster dose of COVID-19 vaccines with subsequent breakthrough Omicron infections among healthcare workers.
- Author
-
Lee, Jeongjae, Park, Soonju, Kim, Ji Yeun, Lim, So Yun, Chang, Euijin, Bae, Seongman, Jung, Jiwon, Kim, Min Jae, Chong, Yong Pil, Lee, Sang-Oh, Choi, Sang-Ho, Kim, Yang Soo, Lee, Nakyung, Shum, David, Kim, Seungtaek, Jee, Youngmee, and Kim, Sung-Han
- Published
- 2022
- Full Text
- View/download PDF
3. Potential of 6′‑hydroxy justicidin B from Justicia procumbens as a therapeutic agent against coronavirus disease 2019.
- Author
-
Yoo, Min Heui, Eom, Han Young, Im, Wan-Jung, Lee, Byoung-Seok, Han, Kang-Hyun, Seo, Joung-Wook, Hwang, Yunha, Youm, Jihyun, Lee, Sangho, Kim, Seungtaek, Ko, Kyong-Cheol, and Kim, Yong-Bum
- Abstract
Since the onset of the coronavirus disease 2019 (COVID-19) pandemic, remarkable advances have been made in vaccine development to reduce mortality. However, therapeutic interventions for COVID-19 are comparatively limited despite these intensive efforts. Furthermore, the rapid mutation capability of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a characteristic of its RNA structure, has led to the emergence of multiple variants, necessitating a shift from a predominantly vaccine-centric approach to one that encompasses therapeutic strategies. 6′-Hydroxy justicidin B (6′-HJB), an arylnaphthalene lignan isolated from Justicia procumbens , a traditional Chinese medicine, is known for its antiviral properties. The aim of the present study was to assess the effectiveness and safety of 6′-HJB against SARS-CoV-2 in order to determine its potential as a therapeutic agent against COVID-19. The efficacy of 6′-HJB was evaluated both in vitro using Vero and Calu-3 cell lines and in vivo using ferrets. The safety assessment included toxicokinetics, safety pharmacology, and Good Laboratory Practice (GLP)-compliant toxicity evaluations following single- and repeated-dose toxicity studies in dogs. The anti-SARS-CoV-2 efficacy of 6′-HJB was evaluated through dose-response curve (DRC) analysis using immunofluorescence; 6′-HJB demonstrated superior inhibition of SARS-CoV-2 growth and lower cytotoxicity than remdesivir. In SARS-CoV-2-infected ferret, 6′-HJB showed efficacy comparable to that of the positive control, Truvada. Further GLP toxicity studies corroborated the safety profile of 6′-HJB. Single-dose and 4-week repeated oral toxicity studies in Beagle dogs demonstrated minimal harmful effects at the highest dosages. The lethal dose of 6′-HJB exceeded 2,000 mg kg
-1 in Beagle dogs. Toxicokinetic and GLP safety pharmacology studies demonstrated no adverse effects of 6′-HJB on metabolic processes, respiratory or central nervous systems, or cardiac functions. This research highlights both the antiviral efficacy and safety profile of 6′-HJB, underscoring its potential as a novel COVID-19 treatment option. The potential of 6′-HJB was demonstrated using modern scientific methodologies and standards. [Display omitted] [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
4. Immune responses and reactogenicity after ChAdOx1 in individuals with past SARS-CoV-2 infection and those without.
- Author
-
Lim, So Yun, Kim, Ji Yeun, Lee, Jin Ah, Kwon, Ji-Soo, Park, Ji Young, Cha, Hye Hee, Suh, Mi Hyun, Lee, Hyun Jung, Kim, Hyeonju, Kim, Seungtaek, Bae, Seongman, Jung, Jiwon, Kim, Eui Ho, Jee, Youngmee, and Kim, Sung-Han
- Published
- 2022
- Full Text
- View/download PDF
5. DeepGT: Deep learning-based quantification of nanosized bioparticles in bright-field micrographs of Gires-Tournois biosensor.
- Author
-
Kang, Jiwon, Yoo, Young Jin, Park, Jin-Hwi, Ko, Joo Hwan, Kim, Seungtaek, Stanciu, Stefan G., Stenmark, Harald A., Lee, JinAh, Mahmud, Abdullah Al, Jeon, Hae-Gon, and Song, Young Min
- Subjects
DEEP learning ,CONVOLUTIONAL neural networks ,MACHINE learning ,BIOSENSORS ,REFRACTIVE index ,VISIBLE spectra ,CONTRAST-enhanced magnetic resonance imaging - Abstract
Rapid and decentralized quantification of viral load profiles in infected patients is vital for assessing clinical severity and tailoring appropriate therapeutic strategies. Although microscopic imaging offers potential for label-free and amplification-free quantitative diagnostics, the small size (∼100 nm in diameter) and low refractive index (n ∼1.5) of bioparticles present challenges in achieving accurate estimations, consequently increasing the limit of detection (LoD). In this study, we present a novel synergistic biosensing approach, DeepGT, combining Gires-Tournois (GT) sensing platforms with deep learning algorithms to enhance nanoscale bioparticle counting accuracy. The GT sensing platform serves as a photonic resonator, increasing bioparticle visibility in bright-field microscopy and maximizing chromatic contrast. By employing a back-end with a dilated convolutional neural network architecture, DeepGT effectively refines artifacts and color deviations, significantly improving particle estimation accuracy (MAE ∼2.37 across 1596 images) compared to rule-based algorithms (MAE ∼ 13.47). Notably, the enhanced accuracy in detecting invisible particles (e.g., two- or three-particles) enables an LoD of 138 pg ml
−1 , facilitating a dynamic linear correlation at low viral concentration ranges within the clinical spectrum of infection, from asymptomatic to severe cases. Leveraging transfer learning, DeepGT, which relies on a chromatometry-based strategy instead of a spatial resolution approach, exhibits exceptional precision when analyzing particles of diverse dimensions smaller than the microscopy system's minimum diffraction limit in visible light (< 258 nm). The DeepGT approach holds promise for early screening and triage of emerging viruses, reducing costs and time requirements in diagnostics. [Display omitted] • Quantitative bright-field imaging of nanosized bioparticles using deep learning without any sample preparation. • DeepGT refines artifacts and color deviations, substantially improving particle estimation accuracy. • Exceptionally accurate quantification at low viral concentration ranges within the clinical spectrum of infection. • DeepGT analyzes bioparticles of various sizes smaller than the microscopy system's minimum diffraction limit. • Candidate for early screening and triage of future emerging viruses, reducing costs and time requirements in diagnostics. [ABSTRACT FROM AUTHOR]- Published
- 2023
- Full Text
- View/download PDF
6. Lycorine, a non-nucleoside RNA dependent RNA polymerase inhibitor, as potential treatment for emerging coronavirus infections.
- Author
-
Jin, Young-Hee, Min, Jung Sun, Jeon, Sangeun, Lee, Jihye, Kim, Seungtaek, Park, Tamina, Park, Daeui, Jang, Min Seong, Park, Chul Min, Song, Jong Hwan, Kim, Hyoung Rae, and Kwon, Sunoh
- Abstract
Background: Highly effective novel treatments need to be developed to suppress emerging coronavirus (CoV) infections such as COVID-19. The RNA dependent RNA polymerase (RdRp) among the viral proteins is known as an effective antiviral target. Lycorine is a phenanthridine Amaryllidaceae alkaloid isolated from the bulbs of Lycoris radiata (L'Hér.) Herb. and has various pharmacological bioactivities including antiviral function.Purpose: We investigated the direct-inhibiting action of lycorine on CoV's RdRp, as potential treatment for emerging CoV infections.Methods: We examined the inhibitory effect of lycorine on MERS-CoV, SARS-CoV, and SARS-CoV-2 infections, and then quantitatively measured the inhibitory effect of lycorine on MERS-CoV RdRp activity using a cell-based reporter assay. Finally, we performed the docking simulation with lycorine and SARS-CoV-2 RdRp.Results: Lycorine efficiently inhibited these CoVs with IC50 values of 2.123 ± 0.053, 1.021 ± 0.025, and 0.878 ± 0.022 μM, respectively, comparable with anti-CoV effects of remdesivir. Lycorine directly inhibited MERS-CoV RdRp activity with an IC50 of 1.406 ± 0.260 μM, compared with remdesivir's IC50 value of 6.335 ± 0.731 μM. In addition, docking simulation showed that lycorine interacts with SARS-CoV-2 RdRp at the Asp623, Asn691, and Ser759 residues through hydrogen bonding, at which the binding affinities of lycorine (-6.2 kcal/mol) were higher than those of remdesivir (-4.7 kcal/mol).Conclusions: Lycorine is a potent non-nucleoside direct-acting antiviral against emerging coronavirus infections and acts by inhibiting viral RdRp activity; therefore, lycorine may be a candidate against the current COVID-19 pandemic. [ABSTRACT FROM AUTHOR]- Published
- 2021
- Full Text
- View/download PDF
7. Hepatitis C Virus Attenuates Interferon-Induced Major Histocompatibility Complex Class I Expression and Decreases CD8+ T Cell Effector Functions.
- Author
-
Kang, Wonseok, Sung, Pil Soo, Park, Su-Hyung, Yoon, Sarah, Chang, Dong-Yeop, Kim, Seungtaek, Han, Kwang Hyub, Kim, Ja Kyung, Rehermann, Barbara, Chwae, Yong-Joon, and Shin, Eui-Cheol
- Abstract
Background & Aims: Major histocompatibility complex (MHC) class I−restricted CD8
+ T cells are required for clearance of hepatitis C virus (HCV) infection. MHC class I expression is up-regulated by type I and II interferons (IFNs). However, little is known about the effects of HCV infection on IFN-induced expression of MHC class I. Methods: We used the HCV cell culture system (HCVcc) with the genotype 2a Japanese fulminant hepatitis-1 strain to investigate IFN-induced expression of MHC class I and its regulatory mechanisms. HCVcc-infected Huh-7.5 cells were analyzed by flow cytometry, metabolic labeling, immunoprecipitation, and immunoblotting analyses. Protein kinase R (PKR) was knocked down with lentiviruses that express small hairpin RNAs. The functional effects of MHC class I regulation by HCV were demonstrated in co-culture studies, using HCV-specific CD8+ T cells. Results: Although the baseline level of MHC class I was not affected by HCV infection, IFN-induced expression of MHC class I was notably attenuated in HCV-infected cells. This was associated with replicating HCV RNA, not with viral protein. HCV infection reduced IFN-induced synthesis of MHC class I protein and induced phosphorylation of PKR and eIF2α. IFN-induced MHC class I expression was restored by small hairpin RNA-mediated knockdown of PKR in HCV-infected cells. Co-culture of HCV-specific CD8+ T cells and HCV-infected cells that expressed HLA-A2 demonstrated that HCV infection reduced the effector functions of HCV-specific CD8+ T cells; these functions were restored by small hairpin RNA-mediated knockdown of PKR. Conclusions: IFN-induced expression of MHC class I is attenuated in HCV-infected cells by activation of PKR, which reduces the effector functions of HCV-specific CD8+ T cells. This appears to be an important mechanism by which HCV circumvents antiviral adaptive immune responses. [Copyright &y& Elsevier]- Published
- 2014
- Full Text
- View/download PDF
8. Discovery of 2-aminoquinolone acid derivatives as potent inhibitors of SARS-CoV-2.
- Author
-
Shin, Young Sup, Lee, Jun Young, Jeon, Sangeun, Myung, Subeen, Gong, Hyun June, Kim, Seungtaek, Kim, Hyoung Rae, Jeong, Lak Shin, and Park, Chul Min
- Subjects
- *
SARS-CoV-2 - Abstract
[Display omitted] The COVID-19 pandemic caused by the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) continues to threaten human health and create socioeconomic problems worldwide. A library of 200,000 small molecules from the Korea Chemical Bank (KCB) were evaluated for their inhibitory activities against SARS-CoV-2 in a phenotypic-based screening assay to discover new therapeutics to combat COVID-19. A primary hit of this screen was the quinolone structure-containing compound 1. Based on the structure of compound 1 and enoxacin, which is a quinolone-based antibiotic previously reported to have weak activity against SARS-CoV-2, we designed and synthesized 2-aminoquinolone acid derivatives. Among them, compound 9b exhibited potent antiviral activity against SARS-CoV-2 (EC 50 = 1.5 µM) without causing toxicity, while having satisfactory in vitro PK profiles. This study shows that 2-aminoquinolone acid 9b provides a promising new template for developing anti-SARS-CoV-2 entry inhibitors. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
9. Design, synthesis and biological evaluation of 2-aminoquinazolin-4(3H)-one derivatives as potential SARS-CoV-2 and MERS-CoV treatments.
- Author
-
Lee, Jun Young, Shin, Young Sup, Jeon, Sangeun, Lee, Se In, Noh, Soojin, Cho, Jung-Eun, Jang, Min Seong, Kim, Seungtaek, Song, Jong Hwan, Kim, Hyoung Rae, and Park, Chul Min
- Subjects
- *
SARS-CoV-2 , *BIOSYNTHESIS , *MERS coronavirus , *CORONAVIRUSES , *COVID-19 - Abstract
[Display omitted] Despite the rising threat of fatal coronaviruses, there are no general proven effective antivirals to treat them. 2-Aminoquinazolin-4(3 H)-one derivatives were newly designed, synthesized, and investigated to show the inhibitory effects on SARS-CoV-2 and MERS-CoV. Among the synthesized derivatives, 7-chloro-2-((3,5-dichlorophenyl)amino)quinazolin-4(3 H)-one (9g) and 2-((3,5-dichlorophenyl)amino)-5-hydroxyquinazolin-4 (3 H)-one (11e) showed the most potent anti-SARS-CoV-2 activities (IC 50 < 0.25 μM) and anti-MERS-CoV activities (IC 50 < 1.1 μM) with no cytotoxicity (CC 50 > 25 μM). In addition, both compounds showed acceptable results in metabolic stabilities, hERG binding affinities, CYP inhibitions, and preliminary PK studies. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
10. Discovery of cyclic sulfonamide derivatives as potent inhibitors of SARS-CoV-2.
- Author
-
Shin, Young Sup, Lee, Jun Young, Noh, Soojin, Kwak, Yoonna, Jeon, Sangeun, Kwon, Sunoh, Jin, Young-hee, Jang, Min Seong, Kim, Seungtaek, Song, Jong Hwan, Kim, Hyoung Rae, and Park, Chul Min
- Subjects
- *
SARS-CoV-2 , *CHEMICAL synthesis , *SULFONAMIDES - Abstract
Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) continues to spread worldwide, with 25 million confirmed cases and 800 thousand deaths. Effective treatments to target SARS-CoV-2 are urgently needed. In the present study, we have identified a class of cyclic sulfonamide derivatives as novel SARS-CoV-2 inhibitors. Compound 13c of the synthesized compounds exhibited robust inhibitory activity (IC 50 = 0.88 μM) against SARS-CoV-2 without cytotoxicity (CC 50 > 25 μM), with a selectivity index (SI) of 30.7. In addition, compound 13c exhibited high oral bioavailability (77%) and metabolic stability with good safety profiles in hERG and cytotoxicity studies. The present study identified that cyclic sulfonamide derivatives are a promising new template for the development of anti-SARS-CoV-2 agents. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
11. Antiviral activity against Middle East Respiratory Syndrome coronavirus by Montelukast, an anti-asthma drug.
- Author
-
Gan, Hanjie Jonathan, Harikishore, Amaravadhi, Lee, Jihye, Jeon, Sangeun, Rajan, Sreekanth, Chen, Ming Wei, Neo, Jun Long, Kim, Seungtaek, and Yoon, Ho Sup
- Subjects
- *
MERS coronavirus , *ANTIASTHMATIC agents , *MIDDLE East respiratory syndrome , *ANTIVIRAL agents , *CD26 antigen , *MONTELUKAST - Abstract
Middle East Respiratory Syndrome (MERS) is a respiratory disease caused by a coronavirus (MERS-CoV). Since its emergence in 2012, nosocomial amplifications have led to its high epidemic potential and mortality rate of 34.5%. To date, there is an unmet need for vaccines and specific therapeutics for this disease. Available treatments are either supportive medications in use for other diseases or those lacking specificity requiring higher doses. The viral infection mode is initiated by the attachment of the viral spike glycoprotein to the human Dipeptidyl Peptidase IV (DPP4). Our attempts to screen antivirals against MERS led us to identify montelukast sodium hydrate (MSH), an FDA-approved anti-asthma drug, as an agent attenuating MERS-CoV infection. We showed that MSH directly binds to MERS-CoV-Receptor-Binding Domain (RBD) and inhibits its molecular interaction with DPP4 in a dose-dependent manner. Our cell-based inhibition assays using MERS pseudovirions demonstrated that viral infection was significantly inhibited by MSH and was further validated using infectious MERS-CoV culture. Thus, we propose MSH as a potential candidate for therapeutic developments against MERS-CoV infections. • Montelukast (MSH) directly binds to MERS-CoV-Receptor-Binding Domain (RBD). • MSH inhibits MERS-Spike pseudovirion (PV) entry. • MSH attenuates live MERS-CoV infection in host cells. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
12. Identification of 4-anilino-6-aminoquinazoline derivatives as potential MERS-CoV inhibitors.
- Author
-
Lee, Jun Young, Shin, Young Sup, Lee, Jihye, Kwon, Sunoh, Jin, Young-hee, Jang, Min Seong, Kim, Seungtaek, Song, Jong Hwan, Kim, Hyoung Rae, and Park, Chul Min
- Subjects
- *
MERS coronavirus , *CORONAVIRUSES , *COVID-19 , *MIDDLE East respiratory syndrome - Abstract
New therapies for treating coronaviruses are urgently needed. A series of 4-anilino-6-aminoquinazoline derivatives were synthesized and evaluated to show high anti-MERS-CoV activities. N4 -(3-Chloro-4-fluorophenyl)- N6 -(3-methoxybenzyl)quinazoline-4,6-diamine (1) has been identified in a random screen as a hit compound for inhibiting MERS-CoV infection. Throughout optimization process, compound 20 was found to exhibit high inhibitory effect (IC 50 = 0.157 μM, SI = 25) with no cytotoxicity and moderate in vivo PK properties. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
13. Synthesis and biological evaluation of 3-acyl-2-phenylamino-1,4-dihydroquinolin-4(1H)-one derivatives as potential MERS-CoV inhibitors.
- Author
-
Yoon, Ji Hye, Lee, Jun Young, Lee, Jihye, Shin, Young Sup, Jeon, Sangeun, Kim, Dong Eon, Min, Jung Sun, Song, Jong Hwan, Kim, Seungtaek, Kwon, Sunoh, Jin, Young-hee, Jang, Min Seong, Kim, Hyoung Rae, and Park, Chul Min
- Subjects
- *
BIOSYNTHESIS , *MERS coronavirus , *RNA viruses - Abstract
3-Acyl-2-phenylamino-1,4-dihydroquinolin-4(1 H)-one derivatives were synthesized and evaluated to show high anti-MERS-CoV inhibitory activities. Among them, 6,8-difluoro-3-isobutyryl-2-((2,3,4-trifluorophenyl)amino)quinolin-4(1 H)-one (6u) exhibits high inhibitory effect (IC 50 = 86 nM) and low toxicity (CC 50 > 25 μM). Moreover, it shows good metabolic stability, low hERG binding affinity, no cytotoxicity, and good in vivo PK properties. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.