1
|
Jian Y, Dong S, Zhang Q, Pan J, Hu R, Ding Z, Wu H, Ke S, Chen Z. In vitro inhibitory activity of indole alkaloid derivatives against porcine epidemic diarrhea virus. Arch Virol 2025; 170:67. [PMID: 40053140 DOI: 10.1007/s00705-025-06251-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 12/08/2024] [Indexed: 03/29/2025]
Abstract
Porcine epidemic diarrhea virus (PEDV) is an enteric coronavirus that can cause acute diarrhea, vomiting, dehydration, and high mortality of newborn piglets, leading to huge economic losses to the world pig industry. Given the limited efficacy of current PEDV vaccines, there is an urgent need for the development of antiviral drugs. In this study, the antiviral effects of 17 synthesized indole alkaloid derivatives against PEDV were investigated. It was observed that indole alkaloid derivative no. 14 exhibited significant inhibition of PEDV replication in a dose-dependent manner. Furthermore, time-of-addition assays and quantitative real-time PCR (QPCR) showed that delayed administration of this compound resulted in a weaker inhibitory effect on PEDV compared to early treatment. Mechanistic analysis revealed that this compound exerts its inhibitory effects during the entry stage of the PEDV life cycle. This study demonstrates the anti-PEDV effects of indole alkaloid derivative no. 14, suggesting its potential as a candidate drug for treating PEDV infections.
Collapse
Affiliation(s)
- Yaoying Jian
- Department of Preventive Veterinary Medicine, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Siqi Dong
- Department of Preventive Veterinary Medicine, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Qianyi Zhang
- Department of Preventive Veterinary Medicine, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Jiali Pan
- Department of Preventive Veterinary Medicine, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Ruiming Hu
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Zhen Ding
- Department of Preventive Veterinary Medicine, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Huansheng Wu
- Department of Preventive Veterinary Medicine, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Shaoyong Ke
- Hubei Biopesticide Engineering Research Centre, Hubei Academy of Agricultural Sciences, Wuhan, 430064, China.
| | - Zheng Chen
- Department of Preventive Veterinary Medicine, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 330045, China.
| |
Collapse
|
2
|
Mukhtar NA, Suleiman M, Al-Maqtari HM, Theva Das K, Bhat AR, Jamalis J. New Insights into the Modifications and Bioactivities of Indole-3- Carboxaldehyde and its Derivatives as a Potential Scaffold for Drug Design: A Mini-Review. Mini Rev Med Chem 2025; 25:480-503. [PMID: 39781713 DOI: 10.2174/0113895575351704241120060746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 09/27/2024] [Accepted: 10/11/2024] [Indexed: 01/12/2025]
Abstract
Indole, a ubiquitous structural motif in bioactive compounds, has played a pivotal role in drug discovery. Among indole derivatives, indole-3-carboxaldehyde (I3A) has emerged as a particularly promising scaffold for the development of therapeutic agents. This review delves into the recent advancements in the chemical modification of I3A and its derivatives, highlighting their potential applications in various therapeutic areas. I3A derivatives have demonstrated a wide range of biological activities, including anti-inflammatory, anti-leishmanial, anti-cancer, anti-bacterial, antifungal, and anti-HIV properties. The structural modifications introduced to the I3A scaffold, such as substitutions on the indole ring (alkylation/arylation/halogenation), variations in the aldehyde group via condensation (Aldol/Claisen/Knoevenagel), and molecular hybridization with other reputable bioactive compounds like coumarins, chalcones, triazoles, and thiophenes, contribute to these activities. Beyond its therapeutic potential, I3A has also found applications as a ligand for Schiff base synthesis, a polymer, and a chromophore. This review provides a comprehensive overview of the latest research on I3A and its derivatives, focusing on the key reactions, modification pathways, reaction conditions, yields, and associated therapeutic activities. By understanding these advancements, researchers can gain valuable insights into the potential applications and future directions for I3A-based drug discovery.
Collapse
Affiliation(s)
- Nuhu Abdullahi Mukhtar
- Department of Chemistry, Faculty of Science, Universiti Teknologi Malaysia, Johor Bahru, 81310, Johor, Malaysia
- Department of Chemistry, Sa'adatu Rimi University of Education, Kumbotso, Kano State, Nigeria
| | - Mustapha Suleiman
- Department of Chemistry, Faculty of Science, Universiti Teknologi Malaysia, Johor Bahru, 81310, Johor, Malaysia
- Department of Chemistry, Sokoto State University, Sokoto State, Nigeria
| | | | - Kumitaa Theva Das
- Department of Biomedical Sciences, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas, Malaysia
| | - Ajmal R Bhat
- Department of Chemistry, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur, India
| | - Joazaizulfazli Jamalis
- Department of Chemistry, Faculty of Science, Universiti Teknologi Malaysia, Johor Bahru, 81310, Johor, Malaysia
| |
Collapse
|
3
|
Ko HL, Lee DK, Kim Y, Jang HJ, Lee YW, Lee HY, Seok SH, Park JW, Limb JK, On DI, Yun JW, Lyoo KS, Song D, Yeom M, Lee H, Seong JK, Lee S. Development of a neutralization monoclonal antibody with a broad neutralizing effect against SARS-CoV-2 variants. Virol J 2023; 20:285. [PMID: 38041113 PMCID: PMC10693169 DOI: 10.1186/s12985-023-02230-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 11/06/2023] [Indexed: 12/03/2023] Open
Abstract
BACKGROUND The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants has challenged the effectiveness of current therapeutic regimens. Here, we aimed to develop a potent SARS-CoV-2 antibody with broad neutralizing effect by screening a scFv library with the spike protein receptor-binding domain (RBD) via phage display. METHODS SKAI-DS84 was identified through phage display, and we performed pseudovirus neutralization assays, authentic virus neutralization assays, and in vivo neutralization efficacy evaluations. Furthermore, surface plasmon resonance (SPR) analysis was conducted to assess the physical characteristics of the antibody, including binding kinetics and measure its affinity for variant RBDs. RESULTS The selected clones were converted to human IgG, and among them, SKAI-DS84 was selected for further analyses based on its binding affinity with the variant RBDs. Using pseudoviruses, we confirmed that SKAI-DS84 was strongly neutralizing against wild-type, B.1.617.2, B.1.1.529, and subvariants of SARS-CoV-2. We also tested the neutralizing effect of SKAI-DS84 on authentic viruses, in vivo and observed a reduction in viral replication and improved lung pathology. We performed binding and epitope mapping experiments to understand the mechanisms underlying neutralization and identified quaternary epitopes formed by the interaction between RBDs as the target of SKAI-DS84. CONCLUSIONS We identified, produced, and tested the neutralizing effect of SKAI-DS84 antibody. Our results highlight that SKAI-DS84 could be a potential neutralizing antibody against SARS-CoV-2 and its variants.
Collapse
Affiliation(s)
- Hae Li Ko
- Division of Research Program, Scripps Korea Antibody Institute, Chuncheon, 24341, Republic of Korea
- Department of Microbiology, College of Medical Science, Kangwon National University, Chuncheon-si, Gangwon-do, 24341, South Korea
| | - Deuk-Ki Lee
- Division of Research Program, Scripps Korea Antibody Institute, Chuncheon, 24341, Republic of Korea
- Department of Microbiology, College of Medical Science, Kangwon National University, Chuncheon-si, Gangwon-do, 24341, South Korea
| | - Younghyeon Kim
- Division of Research Program, Scripps Korea Antibody Institute, Chuncheon, 24341, Republic of Korea
- Department of Microbiology, College of Medical Science, Kangwon National University, Chuncheon-si, Gangwon-do, 24341, South Korea
| | - Hui Jeong Jang
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, 13620, South Korea
| | - Youn Woo Lee
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, 13620, South Korea
| | - Ho-Young Lee
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, 13620, South Korea
| | - Sang-Hyuk Seok
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, Chuncheon-si, Gangwon-do, 24341, South Korea
| | - Jun Won Park
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, Chuncheon-si, Gangwon-do, 24341, South Korea
| | - Jin-Kyung Limb
- Korea Mouse Phenotyping Center (KMPC), Seoul National University, Seoul, 08826, South Korea
| | - Da In On
- Korea Mouse Phenotyping Center (KMPC), Seoul National University, Seoul, 08826, South Korea
- Laboratory of Developmental Biology and Genomics, Research Institute for Veterinary Science, and BK21 Program for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea
| | - Jun-Won Yun
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea
| | - Kwang-Soo Lyoo
- College of Veterinary Medicine, Jeonbuk National University, Iksan, 54596, Republic of Korea
| | - Daesub Song
- Department of Veterinary Medicine Virology Laboratory, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Minjoo Yeom
- Department of Veterinary Medicine Virology Laboratory, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hanbyeul Lee
- Department of Veterinary Medicine Virology Laboratory, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Je Kyung Seong
- Korea Mouse Phenotyping Center (KMPC), Seoul National University, Seoul, 08826, South Korea.
- Laboratory of Developmental Biology and Genomics, Research Institute for Veterinary Science, and BK21 Program for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea.
- Interdisciplinary Program for Bioinformatics, Program for Cancer Biology and BIO-MAX/N-Bio Institute, Seoul National University, Seoul, 08826, South Korea.
| | - Sungjin Lee
- Division of Research Program, Scripps Korea Antibody Institute, Chuncheon, 24341, Republic of Korea.
| |
Collapse
|
4
|
Souza BGD, Choudhary S, Vilela GG, Passos GFS, Costa CACB, Freitas JDD, Coelho GL, Brandão JDA, Anderson L, Bassi ÊJ, Araújo-Júnior JXD, Tomar S, Silva-Júnior EFD. Design, synthesis, antiviral evaluation, and In silico studies of acrylamides targeting nsP2 from Chikungunya virus. Eur J Med Chem 2023; 258:115572. [PMID: 37364511 DOI: 10.1016/j.ejmech.2023.115572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 06/11/2023] [Accepted: 06/11/2023] [Indexed: 06/28/2023]
Abstract
The Togaviridae family comprises several New- and Old-World Alphaviruses that have been responsible for thousands of human illnesses, including the RNA arbovirus Chikungunya virus (CHIKV). Firstly, it was reported in Tanzania in 1952 but rapidly it spread to several countries from Europe, Asia, and the Americas. Since then, CHIKV has been circulating in diverse countries around the world, leading to increased morbidity rates. Currently, there are no FDA-approved drugs or licensed vaccines to specifically treat CHIKV infections. Thus, there is a lack of alternatives to fight against this viral disease, making it an unmet need. Structurally, CHIKV is composed of five structural proteins (E3, E2, E1, C, and 6k) and four non-structural proteins (nsP1-4), in which nsP2 represents an attractive antiviral target for designing novel inhibitors since it has an essential role in the virus replication and transcription. Herein, we used a rational drug design strategy to select some acrylamide derivatives to be synthesized and evaluated against CHIKV nsP2 and also screened on CHIKV-infected cells. Thus, two regions of modifications were considered for these types of inhibitors, based on a previous study of our group, generating 1560 possible inhibitors. Then, the 24 most promising ones were synthesized and screened by using a FRET-based enzymatic assay protocol targeting CHIKV nsP2, identifying LQM330, 333, 336, and 338 as the most potent inhibitors, with Ki values of 48.6 ± 2.8, 92.3 ± 1.4, 2.3 ± 1.5, and 181.8 ± 2.5 μM, respectively. Still, their Km and Vmax kinetic parameters were also determined, along with their competitive binding modes of CHIKV nsP2 inhibition. Then, ITC analyses revealed KD values of 127, 159, 198, and 218 μM for LQM330, 333, 336, and 338, respectively. Also, their ΔH, ΔS, and ΔG physicochemical parameters were determined. MD simulations demonstrated that these inhibitors present a stable binding mode with nsP2, interacting with important residues of this protease, according to docking analyzes. Moreover, MM/PBSA calculations displayed that van der Waals interactions are mainly responsible for stabilizing the inhibitor-nsP2 complex, and their binding energies corroborated with their Ki values, having -198.7 ± 15.68, -124.8 ± 17.27, -247.4 ± 23.78, and -100.6 ± 19.21 kcal/mol for LQM330, 333, 336, and 338, respectively. Since Sindbis (SINV) nsP2 is similar to CHIKV nsP2, these best inhibitors were screened against SINV-infected cells, and it was verified that LQM330 presented the best result, with an EC50 value of 0.95 ± 0.09 μM. Even at 50 μM concentration, LQM338 was found to be cytotoxic on Vero cells after 48 h. Then, LQM330, 333, and 336 were evaluated against CHIKV-infected cells in antiviral assays, in which LQM330 was found to be the most promising antiviral candidate in this study, exhibiting an EC50 value of 5.2 ± 0.52 μM and SI of 31.78. The intracellular flow cytometry demonstrated that LQM330 is able to reduce the CHIKV cytopathogenic effect on cells, and also reduce the percentage of CHIKV-positive cells from 66.1% ± 7.05 to 35.8% ± 5.78 at 50 μM concentration. Finally, qPCR studies demonstrated that LQM330 was capable of reducing the number of viral RNA copies/μL, suggesting that CHIKV nsP2 is targeted by this inhibitor as its mechanism of action.
Collapse
Affiliation(s)
- Beatriz Gois de Souza
- Laboratory of Medicinal Chemistry, Institute of Pharmaceutical Sciences, Federal University of Alagoas, Lourival Melo Mota Avenue, AC. Simões Campus, 57072-970, Alagoas, Maceió, Brazil
| | - Shweta Choudhary
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, 247667, Uttarakhand, India
| | - Gabriel Gomes Vilela
- Laboratory of Medicinal Chemistry, Institute of Pharmaceutical Sciences, Federal University of Alagoas, Lourival Melo Mota Avenue, AC. Simões Campus, 57072-970, Alagoas, Maceió, Brazil
| | - Gabriel Felipe Silva Passos
- Laboratory of Medicinal Chemistry, Institute of Pharmaceutical Sciences, Federal University of Alagoas, Lourival Melo Mota Avenue, AC. Simões Campus, 57072-970, Alagoas, Maceió, Brazil
| | | | - Johnnatan Duarte de Freitas
- Department of Chemistry, Federal Institute of Alagoas, Maceió Campus, Mizael Domingues Street, 57020-600, Alagoas, Maceió, Brazil
| | - Grazielle Lobo Coelho
- Immunoregulation Research Group, Laboratory of Research in Virology and Immunology, Institute of Biological and Health Sciences, Lourival Melo Mota Avenue, AC. Simões Campus, 57072-970, Alagoas, Maceió, Brazil
| | - Júlia de Andrade Brandão
- Immunoregulation Research Group, Laboratory of Research in Virology and Immunology, Institute of Biological and Health Sciences, Lourival Melo Mota Avenue, AC. Simões Campus, 57072-970, Alagoas, Maceió, Brazil
| | - Leticia Anderson
- Immunoregulation Research Group, Laboratory of Research in Virology and Immunology, Institute of Biological and Health Sciences, Lourival Melo Mota Avenue, AC. Simões Campus, 57072-970, Alagoas, Maceió, Brazil; CESMAC University Center, 57051-160, Alagoas, Maceió, Brazil
| | - Ênio José Bassi
- Immunoregulation Research Group, Laboratory of Research in Virology and Immunology, Institute of Biological and Health Sciences, Lourival Melo Mota Avenue, AC. Simões Campus, 57072-970, Alagoas, Maceió, Brazil
| | - João Xavier de Araújo-Júnior
- Laboratory of Medicinal Chemistry, Institute of Pharmaceutical Sciences, Federal University of Alagoas, Lourival Melo Mota Avenue, AC. Simões Campus, 57072-970, Alagoas, Maceió, Brazil
| | - Shailly Tomar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, 247667, Uttarakhand, India
| | - Edeildo Ferreira da Silva-Júnior
- Laboratory of Medicinal Chemistry, Institute of Pharmaceutical Sciences, Federal University of Alagoas, Lourival Melo Mota Avenue, AC. Simões Campus, 57072-970, Alagoas, Maceió, Brazil; Biological and Molecular Chemistry Research Group, Institute of Chemistry and Biotechnology, Federal University of Alagoas, Lourival Melo Mota Avenue, AC. Simões Campus, 57072-970, Alagoas, Maceió, Brazil.
| |
Collapse
|
5
|
Zhou Z, Zhang J, Zhou E, Ren C, Wang J, Wang Y. Small molecule NS5B RdRp non-nucleoside inhibitors for the treatment of HCV infection: A medicinal chemistry perspective. Eur J Med Chem 2022; 240:114595. [PMID: 35868125 DOI: 10.1016/j.ejmech.2022.114595] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 07/01/2022] [Accepted: 07/05/2022] [Indexed: 02/06/2023]
Abstract
Hepatitis C virus (HCV) infection has become a global health problem with enormous risks. Nonstructural protein 5B (NS5B) RNA-dependent RNA polymerase (RdRp) is a component of HCV, which can promote the formation of the viral RNA replication complex and is also an essential part of the replication complex itself. It plays a vital role in the synthesis of the positive and negative strands of HCV RNA. Therefore, the development of small-molecule inhibitors targeting NS5B RdRp is of great value for treating HCV infection-related diseases. Compared with NS5B RdRp nucleoside inhibitors, non-nucleoside inhibitors have more flexible structures, simpler mechanisms of action, and more predictable efficacy and safety of drugs in humans. Technological advances over the past decade have led to remarkable achievements in developing NS5B RdRp inhibitors. This review will summarize the non-nucleoside inhibitors targeting NS5B RdRp developed in the past decade and describe their structure optimization process and structure-activity relationship.
Collapse
Affiliation(s)
- Zhilan Zhou
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; State Key Laboratory of Biotherapy and Cancer Center, Department of Respiratory and Critical Care Medicine, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jifa Zhang
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; State Key Laboratory of Biotherapy and Cancer Center, Department of Respiratory and Critical Care Medicine, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Tianfu Jincheng Laboratory, Chengdu, 610041, Sichuan, China
| | - Enda Zhou
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; State Key Laboratory of Biotherapy and Cancer Center, Department of Respiratory and Critical Care Medicine, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Changyu Ren
- Department of Pharmacy, Chengdu Fifth People's Hospital, Chengdu, Sichuan, 611130, China
| | - Jiaxing Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, 38163, Tennessee, United States
| | - Yuxi Wang
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; State Key Laboratory of Biotherapy and Cancer Center, Department of Respiratory and Critical Care Medicine, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Tianfu Jincheng Laboratory, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
6
|
Mohammadi Ziarani G, Hasani S, Mohajer F, Varma RS, Rafiee F. The Molecular Diversity of 1H-Indole-3-Carbaldehyde Derivatives and Their Role in Multicomponent Reactions. Top Curr Chem (Cham) 2022; 380:24. [PMID: 35467226 DOI: 10.1007/s41061-022-00379-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 04/01/2022] [Indexed: 10/18/2022]
Abstract
1H-Indole-3-carbaldehyde and related members of the indole family are ideal precursors for the synthesis of active molecules. 1H-Indole-3-carbaldehyde and its derivatives are essential and efficient chemical precursors for generating biologically active structures. Multicomponent reactions (MCRs) offer access to complex molecules. This review highlights the recent applications of 1H-indole-3-carbaldehyde in such inherently sustainable multicomponent reactions from the period, 2014 to 2021 and provides an overview of the field that awaits further exploitation in the assembly of pharmaceutically interesting scaffolds.
Collapse
Affiliation(s)
- Ghodsi Mohammadi Ziarani
- Department of Chemistry, Faculty of Physics and Chemistry, Alzahra University, PO Box 1993893973, Tehran, Iran.
| | - Samira Hasani
- Department of Chemistry, Faculty of Physics and Chemistry, Alzahra University, PO Box 1993893973, Tehran, Iran
| | - Fatemeh Mohajer
- Department of Chemistry, Faculty of Physics and Chemistry, Alzahra University, PO Box 1993893973, Tehran, Iran
| | - Rajender S Varma
- Regional Centre of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute, Palacky University, Šlechtitelů 27, 783 71, Olomouc, Czech Republic.
| | - Fatemeh Rafiee
- Department of Chemistry, Faculty of Physics and Chemistry, Alzahra University, PO Box 1993893973, Tehran, Iran
| |
Collapse
|
7
|
Design and synthesis of novel substituted indole-acrylamide derivatives and evaluation of their anti-cancer activity as potential tubulin-targeting agents. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.132345] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
8
|
Cebeci YU, Ceylan Ş, Karaoğlu ŞA. Conventional and microwave irradiated synthesis, biological activity evaluation of highly substituted indole-triazole hybrids. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2021.131799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
9
|
Selective Synthesis of 2-Aryl-3-Alkenylindoles and 2-Aryl-3-Alkynylindoles by Palladium-Catalyzed Ligand-Promoted Annulative Coupling of Anilines and Propargyl Alcohols. J Organomet Chem 2021. [DOI: 10.1016/j.jorganchem.2021.121930] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
10
|
Dorababu A. Indole - a promising pharmacophore in recent antiviral drug discovery. RSC Med Chem 2020; 11:1335-1353. [PMID: 34095843 PMCID: PMC8126882 DOI: 10.1039/d0md00288g] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 10/04/2020] [Indexed: 12/17/2022] Open
Abstract
The bicyclic molecule indole has been in the limelight because of its numerous pharmacological potencies. It is used as an excellent scaffold in drug discovery of medicinal drugs such as antimicrobials, anticancer agents, antihypertensives, anti-proliferative agents and anti-inflammatory agents. In spite of its diverse therapeutic activity, it is used as a key pharmacophore in synthesizing the most potent biological agents. Besides, viral infections are ubiquitous and their prevention and cure have become a great challenge. In this regard, the design of indole-containing antiviral drugs is accomplished to combat viral infections. A lot of research is being carried out towards antiviral drug discovery by many researchers round the clock. Herein, the antiviral activity of recently discovered indole scaffolds is compiled and critically evaluated to give a meaningful summary. In addition, the structure-activity relationship of remarkable antiviral agents is discussed. Also, the structural motifs attributed to noteworthy antiviral properties are highlighted to guide future antiviral research.
Collapse
Affiliation(s)
- Atukuri Dorababu
- Department of Chemistry, SRMPP Govt. First Grade College Huvinahadagali-583219 Karnataka India
| |
Collapse
|
11
|
Hawash M, Eid AM, Jaradat N, Abualhasan M, Amer J, Naser Zaid A, Draghmeh S, Daraghmeh D, Daraghmeh H, Shtayeh T, Sawaftah H, Mousa A. Synthesis and Biological Evaluation of Benzodioxole Derivatives as Potential Anticancer and Antioxidant agents. HETEROCYCL COMMUN 2020. [DOI: 10.1515/hc-2020-0105] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Abstracta series of benzodioxole compounds were synthesized and evaluated for their cytotoxic activity against cervical (Hela), colorectal (Caco-2), and liver (Hep3B) cancer cell lines. Compounds 5a, 5b, 6a, 6b, 7a and 7b showed very weak or negligible anticancer activity with IC50 3.94-9.12 mM. On the contrary, carboxamide containing compounds 2a and 2b showed anticancer activity. Both 2a and 2b reduced Hep3B secretions of α-fetoprotein (α-FP) to 1625.8 ng/ml and 2340 ng/ml, respectively, compared to 2519.17 ng/ml in untreated cells. The results also showed that compound 2a has potent anticancer activity against Hep3B cancer cell line. Furthermore, in cell cycle analysis, compound 2a induced arrest in the G2-M phase in value of 8.07% that was very close to the activity of doxorubicin (7.4%). These results indicate that compound 2a has a potent and promising antitumor activity. However, benzodiazepine derivatives (7a and 7b) showed moderate antioxidant activity with IC50 values of 39.85 and 79.95 μM, respectively compared with the potent antioxidant agent Trolox (IC50 = 7.72 μM).
Collapse
Affiliation(s)
- Mohammed Hawash
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, NablusP.O. Box 7, 00970, Palestine
| | - Ahmad M Eid
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, NablusP.O. Box 7, 00970, Palestine
| | - Nidal Jaradat
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, NablusP.O. Box 7, 00970, Palestine
| | - Murad Abualhasan
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, NablusP.O. Box 7, 00970, Palestine
| | - Johnny Amer
- Department of Biomedical Sciences, Physiology, Pharmacology & Toxicology Division. Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Abdel Naser Zaid
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, NablusP.O. Box 7, 00970, Palestine
| | - Saja Draghmeh
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, NablusP.O. Box 7, 00970, Palestine
| | - Donia Daraghmeh
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, NablusP.O. Box 7, 00970, Palestine
| | - Haifa Daraghmeh
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, NablusP.O. Box 7, 00970, Palestine
| | - Tahrir Shtayeh
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, NablusP.O. Box 7, 00970, Palestine
| | - Hadeel Sawaftah
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, NablusP.O. Box 7, 00970, Palestine
| | - Ahmed Mousa
- Department of Biomedical Sciences, Physiology, Pharmacology & Toxicology Division. Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| |
Collapse
|
12
|
Jaradat N, Hawash M, Abualhasan M. Synthesis and Biological Evaluation of Benzodioxol Derivatives as Cyclooxygenase Inhibitors. LETT DRUG DES DISCOV 2020. [DOI: 10.2174/1570180817999200420114402] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Background:
Non-steroidal anti-inflammatory drugs (NSAIDs) are among the most
widely used therapeutics; they are competitive inhibitors of cyclooxygenase (COX), the enzyme
which mediates the conversion of arachidonic acid to inflammatory prostaglandins.
Objective:
In this study, new benzodioxol derivatives with different core cycles and functional
groups (i.e., aryl acetate, aryl acetic acid and diazepine) were designed, synthesized, identified and
evaluated for their analgesic and anti-inflammatory activity, as a preliminary screening study to
identify the most potent and more selective groups.
Methods:
The synthesized compounds were identified using FTIR, HRMS, 1H-NMR and 13C-NMR,
and evaluated for their inhibitory activity against ovine COX-1 and COX-2 using an in vitro cyclooxygenase
(COX) inhibition assay kit.
Results:
Six compounds were synthesized as a preliminary screening study to identify
which was the most potent and more selective group towards COX-2 versus COX-1, compared
to ketoprofen as non-selective NSAIDs. The compounds have three different groups: aryl acetate,
aryl acetic acid and diazepine. The results showed that the most potent compound against the COX-
1 enzyme was 4b (which has diazepine and 2-chlorophenyl) with IC50 = 0.363 μM, and the selectivity
ratio of 4b was found to be better than ketoprofen. In contrast, compound 4a (which has diazepine
and 3-chlorophenyl) was the most selective with a COX-1/COX-2 ratio value of 0.85 in comparison
with a ketoprofen ratio value of 0.20.
Conclusion:
In general, the synthesized library has moderate activity against both enzymes (i.e.,
COX-1 and COX-2). Moreover, all six compounds have better COX-2 inhibition selectivity compared
to the commercial drug ketoprofen.
Collapse
Affiliation(s)
- Nidal Jaradat
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus 00970, Palestinian Territory, Occupied
| | - Mohammed Hawash
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus 00970, Palestinian Territory, Occupied
| | - Murad Abualhasan
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus 00970, Palestinian Territory, Occupied
| |
Collapse
|
13
|
Hawash M, Jaradat N, Hameedi S, Mousa A. Design, synthesis and biological evaluation of novel benzodioxole derivatives as COX inhibitors and cytotoxic agents. BMC Chem 2020; 14:54. [PMID: 32944715 PMCID: PMC7487730 DOI: 10.1186/s13065-020-00706-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 09/02/2020] [Indexed: 02/06/2023] Open
Abstract
Non-steroidal anti-inflammatory drugs are among the most used drugs. They are competitive inhibitors of cyclooxygenase (COX). Twelve novel compounds (aryl acetate and aryl acetic acid groups) were synthesized in this work in order to identify which one was the most potent and which group was most selective towards COX1 and COX2 by using an in vitro COX inhibition assay kit. The cytotoxicity was evaluated for these compounds utilizing MTS assay against cervical carcinoma cells line (HeLa). The synthesized compounds were identified using FTIR, HRMS, 1H-NMR, and 13C-NMR techniques. The results showed that the most potent compound against the COX1 enzyme was 4f with IC50 = 0.725 µM. The compound 3b showed potent activity against both COX1 and COX2 with IC50 = 1.12 and 1.3 µM, respectively, and its selectivity ratio (0.862) was found to be better than Ketoprofen (0.196). In contrast, compound 4d was the most selective with a COX1/COX2 ratio value of 1.809 in comparison with the Ketoprofen ratio. All compounds showed cytotoxic activity against the HeLa Cervical cancer cell line at a higher concentration ranges (0.219–1.94 mM), and the most cytotoxic compound was 3e with a CC50 value of 219 µM. This was tenfold more than its IC50 values of 2.36 and 2.73 µM against COX1 and COX2, respectively. In general, the synthesized library has moderate activity against both enzymes (i.e., COX1 and COX2) and ortho halogenated compounds were more potent than the meta ones.![]()
Collapse
Affiliation(s)
- Mohammed Hawash
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, P.O. Box 7, Nablus, 00970 Palestine
| | - Nidal Jaradat
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, P.O. Box 7, Nablus, 00970 Palestine
| | - Saba Hameedi
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, P.O. Box 7, Nablus, 00970 Palestine
| | - Ahmed Mousa
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, 00970 Palestine
| |
Collapse
|
14
|
Taha M, Rahim F, Khan AA, Anouar EH, Ahmed N, Shah SAA, Ibrahim M, Zakari ZA. Synthesis of diindolylmethane (DIM) bearing thiadiazole derivatives as a potent urease inhibitor. Sci Rep 2020; 10:7969. [PMID: 32409737 PMCID: PMC7224224 DOI: 10.1038/s41598-020-64729-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/26/2020] [Indexed: 11/30/2022] Open
Abstract
The current study describes synthesis of diindolylmethane (DIM) derivatives based-thiadiazole as a new class of urease inhibitors. Diindolylmethane is natural product alkaloid reported to use in medicinal chemistry extensively. Diindolylmethane-based-thiadiazole analogs (1–18) were synthesized and characterized by various spectroscopic techniques 1HNMR, 13C-NMR, EI-MS and evaluated for urease (jack bean urease) inhibitory potential. All compounds showed excellent to moderate inhibitory potential having IC50 value within the range of 0.50 ± 0.01 to 33.20 ± 1.20 µM compared with the standard thiourea (21.60 ± 0.70 µM). Compound 8 (IC50 = 0.50 ± 0.01 µM) was the most potent inhibitor amongst all derivatives. Structure-activity relationships have been established for all compounds. The key binding interactions of most active compounds with enzyme were confirmed through molecular docking studies.
Collapse
Affiliation(s)
- Muhammad Taha
- Department of clinical pharmacy, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 31441, Dammam, Saudi Arabia.
| | - Fazal Rahim
- Department of Chemistry, Hazara University, Mansehra, 21300, Khyber Pakhtunkhwa, Pakistan
| | - Aftab Ahmad Khan
- Department of Chemistry, Hazara University, Mansehra, 21300, Khyber Pakhtunkhwa, Pakistan
| | - El Hassane Anouar
- Department of Chemistry, College of Science and Humanities in Al-Kharj, Prince Sattam bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia
| | - Naveed Ahmed
- Department of Pharmacy, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Syed Adnan Ali Shah
- Atta-ur-Rahman Institute for Natural Product Discovery (AuRIns), Universiti Teknologi MARA Cawangan Selangor Kampus Puncak Alam, 42300 Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia, 42300, D. E., Selangor, Malaysia.,Faculty of Pharmacy, Universiti Teknologi MARA Cawangan Selangor Kampus Puncak Alam, 42300 Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia, 42300, Darul Ehsan, Selangor, Malaysia
| | - Mohamed Ibrahim
- Department of clinical pharmacy, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 31441, Dammam, Saudi Arabia
| | - Zainul Amiruddin Zakari
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia. .,Halal Institute Research Institute, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.
| |
Collapse
|
15
|
Herrera-Arozamena C, Estrada-Valencia M, Pérez C, Lagartera L, Morales-García JA, Pérez-Castillo A, Franco-Gonzalez JF, Michalska P, Duarte P, León R, López MG, Mills A, Gago F, García-Yagüe ÁJ, Fernández-Ginés R, Cuadrado A, Rodríguez-Franco MI. Tuning melatonin receptor subtype selectivity in oxadiazolone-based analogues: Discovery of QR2 ligands and NRF2 activators with neurogenic properties. Eur J Med Chem 2020; 190:112090. [DOI: 10.1016/j.ejmech.2020.112090] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 01/20/2020] [Accepted: 01/21/2020] [Indexed: 12/18/2022]
|
16
|
Paul A, Chatterjee D, Banerjee S, Yadav S. Synthesis of 3-alkenylindoles through regioselective C-H alkenylation of indoles by a ruthenium nanocatalyst. Beilstein J Org Chem 2020; 16:140-148. [PMID: 32082433 PMCID: PMC7006491 DOI: 10.3762/bjoc.16.16] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 01/21/2020] [Indexed: 01/18/2023] Open
Abstract
3-Alkenylindoles are biologically and medicinally very important compounds, and their syntheses have received considerable attention. Herein, we report the synthesis of 3-alkenylindoles via a regioselective alkenylation of indoles, catalysed by a ruthenium nanocatalyst (RuNC). The reaction tolerates several electron-withdrawing and electron-donating groups on the indole moiety. Additionally, a "robustness screen" has also been employed to demonstrate the tolerance of several functional groups relevant to medicinal chemistry. With respect to the Ru nanocatalyst, it has been demonstrated that it is recoverable and recyclable up to four cycles. Also, the catalyst acts through a heterogeneous mechanism, which has been proven by various techniques, such as ICPMS and three-phase tests. The nature of the Ru nanocatalyst surface has also been thoroughly examined by various techniques, and it has been found that the oxides on the surface are responsible for the high catalytic efficiency of the Ru nanocatalyst.
Collapse
Affiliation(s)
- Abhijit Paul
- Department of Chemistry, Indian Institute of Technology (ISM), Dhanbad, 826004, Jharkhand, India
| | - Debnath Chatterjee
- Department of Chemistry, Indian Institute of Technology (ISM), Dhanbad, 826004, Jharkhand, India
| | - Srirupa Banerjee
- Department of Chemistry, Bethune College, Bidhan Sarani, Kolkata, 700006, West Bengal, India
| | - Somnath Yadav
- Department of Chemistry, Indian Institute of Technology (ISM), Dhanbad, 826004, Jharkhand, India
| |
Collapse
|
17
|
Synthesis, characterization and cytotoxic investigations of novel bis(indole) analogues besides antimicrobial study. ARAB J CHEM 2019. [DOI: 10.1016/j.arabjc.2015.05.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
|
18
|
Kumari A, Singh RK. Medicinal chemistry of indole derivatives: Current to future therapeutic prospectives. Bioorg Chem 2019; 89:103021. [PMID: 31176854 DOI: 10.1016/j.bioorg.2019.103021] [Citation(s) in RCA: 303] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 05/22/2019] [Accepted: 05/29/2019] [Indexed: 12/21/2022]
Abstract
Indole is a versatile pharmacophore, a privileged scaffold and an outstanding heterocyclic compound with wide ranges of pharmacological activities due to different mechanisms of action. It is an superlative moiety in drug discovery with the sole property of resembling different structures of the protein. Plenty of research has been taking place in recent years to synthesize and explore the various therapeutic prospectives of this moiety. This review summarizes some of the recent effective chemical synthesis (2014-2018) for indole ring. This review also emphasized on the structure-activity relationship (SAR) to reveal the active pharmacophores of various indole analogues accountable for anticancer, anticonvulsant, antimicrobial, antitubercular, antimalarial, antiviral, antidiabetic and other miscellaneous activities which have been investigated in the last five years. The precise features with motives and framework of each research topic is introduced for helping the medicinal chemists to understand the perspective of the context in a better way. This review will definitely offer the platform for researchers to strategically design diverse novel indole derivatives having different promising pharmacological activities with reduced toxicity and side effects.
Collapse
Affiliation(s)
- Archana Kumari
- Rayat-Bahra Institute of Pharmacy, Dist. Hoshiarpur, 146104 Punjab, India
| | - Rajesh K Singh
- Department of Pharmaceutical Chemistry, Shivalik College of Pharmacy, Nangal, Dist. Rupnagar, 140126 Punjab, India.
| |
Collapse
|
19
|
Brahmachari G, Nurjamal K, Begam S, Mandal M, Nayek N, Karmakar I, Mandal B. Alum (KAl(SO4)2.12H2O) - An Eco-friendly and Versatile Acid-catalyst in Organic Transformations: A Recent Update. CURRENT GREEN CHEMISTRY 2019. [DOI: 10.2174/2213346106666190307160332] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Potassium alum (KAl(SO4)2.12H2O), commonly known as ‘alum’, has recently drawn the attention of synthetic chemists as an efficient, safe and eco-friendly acid catalyst in implementing a large number of organic transformations, thereby generating interesting molecular frameworks. The present review article offers an overview of the potent catalytic applications of this commercially available and low-cost inorganic sulfate salt in organic reactions reported during the period of 2014 to 2018.
Collapse
Affiliation(s)
- Goutam Brahmachari
- Laboratory of Natural Products & Organic Synthesis, Department of Chemistry, Visva-Bharati (Central University), Santiniketan-731 235, West Bengal, India
| | - Khondekar Nurjamal
- Laboratory of Natural Products & Organic Synthesis, Department of Chemistry, Visva-Bharati (Central University), Santiniketan-731 235, West Bengal, India
| | - Sanchari Begam
- Laboratory of Natural Products & Organic Synthesis, Department of Chemistry, Visva-Bharati (Central University), Santiniketan-731 235, West Bengal, India
| | - Mullicka Mandal
- Laboratory of Natural Products & Organic Synthesis, Department of Chemistry, Visva-Bharati (Central University), Santiniketan-731 235, West Bengal, India
| | - Nayana Nayek
- Laboratory of Natural Products & Organic Synthesis, Department of Chemistry, Visva-Bharati (Central University), Santiniketan-731 235, West Bengal, India
| | - Indrajit Karmakar
- Laboratory of Natural Products & Organic Synthesis, Department of Chemistry, Visva-Bharati (Central University), Santiniketan-731 235, West Bengal, India
| | - Bhagirath Mandal
- Laboratory of Natural Products & Organic Synthesis, Department of Chemistry, Visva-Bharati (Central University), Santiniketan-731 235, West Bengal, India
| |
Collapse
|
20
|
Ganta NM, Gedda G, Rathnakar B, Satyanarayana M, Yamajala B, Ahsan MJ, Jadav SS, Balaraju T. A review on HCV inhibitors: Significance of non-structural polyproteins. Eur J Med Chem 2018; 164:576-601. [PMID: 30639895 PMCID: PMC7185800 DOI: 10.1016/j.ejmech.2018.12.045] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 12/17/2018] [Accepted: 12/17/2018] [Indexed: 12/19/2022]
Abstract
Hepatitis C virus (HCV) mortality and morbidity is a world health misery with an approximate 130–150 million chronically HCV tainted and suffering individuals and it initiate critical liver malfunction like cirrhosis, hepatocellular carcinoma or liver HCV cancer. HCV NS5B protein one of the best studied therapeutic target for the identification of new drug candidates to be added to the combination or multiple combination medication recently approved. During the past few years, NS5B has thus been an important object of attractive medicinal chemistry endeavors, which induced to the surfacing of betrothal preclinical drug molecules. In this scenario, the current review set limit to discuss research published on NS5B and few other therapeutic functional inhibitors concentrating on hit investigation, hit to lead optimization, ADME parameters evaluation, and the SAR data which was out for each compound type and similarity taken into consideration. The discussion outlined in this specific review will surly helpful and vital tool for those medicinal chemists investigators working with HCV research programs mainly pointing on NS5B and set broad spectrum identification of creative anti HCV compounds. This mini review also tells each and every individual compound ability related how much they are active against NS5B and few other targets. Hepatitis C infection causes severe liver cirrhosis and carcinoma. The new acute HCV infections are raising every year and mortality rate become serious concern. The plausible list of anti-HCV drugs and clinical agents were listed in this review. The divergent medicinal scaffolds as anti-HCV agents were presented as per their targets.
Collapse
Affiliation(s)
- Narayana Murthy Ganta
- Department of Pharmaceutical Chemistry, Vishnu Institute of Pharmaceutical Education and Research, Narsapur, Medak, Telangana, 502313, India
| | - Gangaraju Gedda
- Department of Chemistry, School of Science, GITAM deemed to be University, Rudraram, Patancheru Mandal, Hyderabad, Telangana, Sangareddy Dist. 502329, India
| | - Bethi Rathnakar
- Department of Pharmaceutical Chemistry, Telangana University, Nizamabad, Telangana, 503322, India
| | - Mavurapu Satyanarayana
- Department of Pharmaceutical Chemistry, Telangana University, Nizamabad, Telangana, 503322, India
| | - Bhaskar Yamajala
- Department of Chemistry, School of Science, GITAM deemed to be University, Rudraram, Patancheru Mandal, Hyderabad, Telangana, Sangareddy Dist. 502329, India
| | - Mohamed Jawed Ahsan
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Abha, 62529, Saudi Arabia
| | - Surender Singh Jadav
- CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad, 500007, India.
| | - Tuniki Balaraju
- Deapartment of Chemistry, Material Science Centre, Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia, WB, 741 246, India.
| |
Collapse
|
21
|
Halawa AH, El-Gilil SMA, Bedair AH, Shaaban M, Frese M, Sewald N, Eliwa EM, El-Agrody AM. Synthesis, biological activity and molecular modeling study of new Schiff bases incorporated with indole moiety. ACTA ACUST UNITED AC 2017; 72:467-475. [PMID: 28525356 DOI: 10.1515/znc-2017-0025] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 04/25/2017] [Indexed: 11/15/2022]
Abstract
A new series of heterocyclic Schiff bases 2-9 containing indole moiety were synthesized by facile and efficient condensation of indole-3/2/5-carboxaldehyde (1a/1b/1c) with different aromatic and heterocyclic primary amines using conventional and/or microwave irradiation methods. The structures of the obtained compounds were assigned by sophisticated spectroscopic and spectrometric techniques (1D-NMR, 2D-NMR and MS). The synthesized compounds were screened for their cytotoxicity and antibacterial activities. In vitro cytotoxicity screening revealed that compound 5 exhibited moderate activity against KB-3-1 cell line (IC50=57.7 μM) while 5-indolylimino derivative 7 indicated close to the activity (IC50=19.6 μM) in comparison with the positive control (+)-Griseofulvin (IC50=19.2 μM), while the tested compounds 5, 6b, 7 and 9 revealed good or moderate antibacterial activity. In addition, molecular docking study of Schiff bases 2-9 was performed by Molecular Operating Environment (MOE 2014.09) program on the matrix metalloproteinase-8 (MMP-8) (Protein Data Bank (PDB) ID: 1MNC) in an attempt to explore their mode of action as anticancer drugs.
Collapse
|
22
|
Jin G, Lee J, Lee K. Chemical genetics-based development of small molecules targeting hepatitis C virus. Arch Pharm Res 2017; 40:1021-1036. [PMID: 28856597 DOI: 10.1007/s12272-017-0949-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 08/20/2017] [Indexed: 12/21/2022]
Abstract
Hepatitis C virus (HCV) infection is a major worldwide problem that has emerged as one of the most significant diseases affecting humans. There are currently no vaccines or efficient therapies without side effects, despite today's advanced medical technology. Currently, the common therapy for most patients (i.e. genotype 1) is combination of HCV-specific direct-acting antivirals (DAAs). Up to 2011, the standard of care (SOC) was a combination of peg-IFNα with ribavirin (RBV). After approval of NS3/4A protease inhibitor, SOC was peg-IFNα and RBV with either the first-generation DAAs boceprevir or telaprevir. In the past several years, various novel small molecules have been discovered and some of them (i.e., HCV polymerase, protease, helicase and entry inhibitors) have undergone clinical trials. Between 2013 and 2016, the second-generation DAA drugs simeprevir, asunaprevir, daclatasvir, dasabuvir, sofosbuvir, and elbasvir were approved, as well as the combinational drugs Harvoni®, Zepatier®, Technivie®, and Epclusa®. A number of reviews have been recently published describing the structure-activity relationship (SAR) in the development of HCV inhibitors and outlining current therapeutic approaches to hepatitis C infection. Target identification involves studying a drug's mechanism of action (MOA), and a variety of target identification methods have been developed in the past few years. Chemical biology has emerged as a powerful tool for studying biological processes using small molecules. The use of chemical genetic methods is a valuable strategy for studying the molecular mechanisms of the viral lifecycle and screening for anti-viral agents. Two general screening approaches have been employed: forward and reverse chemical genetics. This review reveals information on the small molecules in HCV drug discovery by using chemical genetics for targeting the HCV protein and describes successful examples of targets identified with these methods.
Collapse
Affiliation(s)
- Guanghai Jin
- College of Pharmacy, Dongguk University-Seoul, Goyang, 10326, Republic of Korea
| | - Jisu Lee
- College of Pharmacy, Dongguk University-Seoul, Goyang, 10326, Republic of Korea
| | - Kyeong Lee
- College of Pharmacy, Dongguk University-Seoul, Goyang, 10326, Republic of Korea.
| |
Collapse
|
23
|
He X, Chen X, Lin S, Mo X, Zhou P, Zhang Z, Lu Y, Yang Y, Gu H, Shang Z, Lou Y, Wu J. Diversity-Oriented Synthesis of Natural-Product-like Libraries Containing a 3-Methylbenzofuran Moiety for the Discovery of New Chemical Elicitors. ChemistryOpen 2017; 6:102-111. [PMID: 28168155 PMCID: PMC5288756 DOI: 10.1002/open.201600118] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Indexed: 12/26/2022] Open
Abstract
Natural products are a major source of biological molecules. The 3-methylfuran scaffold is found in a variety of plant secondary metabolite chemical elicitors that confer host-plant resistance against insect pests. Herein, the diversity-oriented synthesis of a natural-product-like library is reported, in which the 3-methylfuran core is fused in an angular attachment to six common natural product scaffolds-coumarin, chalcone, flavone, flavonol, isoflavone and isoquinolinone. The structural diversity of this library is assessed computationally using cheminformatic analysis. Phenotypic high-throughput screening of β-glucuronidase activity uncovers several hits. Further in vivo screening confirms that these hits can induce resistance in rice to nymphs of the brown planthopper Nilaparvata lugens. This work validates the combination of diversity-oriented synthesis and high-throughput screening of β-glucuronidase activity as a strategy for discovering new chemical elicitors.
Collapse
Affiliation(s)
- Xingrui He
- Department of ChemistryZhejiang UniversityHangzhou310027P. R. China
| | - Xia Chen
- State Key Laboratory of Rice BiologyInstitute of Insect ScienceZhejiang UniversityHangzhou310058P. R. China
| | - Songbo Lin
- Department of ChemistryZhejiang UniversityHangzhou310027P. R. China
| | - Xiaochang Mo
- State Key Laboratory of Rice BiologyInstitute of Insect ScienceZhejiang UniversityHangzhou310058P. R. China
| | - Pengyong Zhou
- State Key Laboratory of Rice BiologyInstitute of Insect ScienceZhejiang UniversityHangzhou310058P. R. China
| | - Zhihao Zhang
- Department of ChemistryZhejiang UniversityHangzhou310027P. R. China
| | - Yaoyao Lu
- School of Biological and Chemical EngineeringZhejiang University of Science and TechnologyHangzhou310023P. R. China
| | - Yu Yang
- School of Biological and Chemical EngineeringZhejiang University of Science and TechnologyHangzhou310023P. R. China
| | - Haining Gu
- Department of ChemistryZhejiang UniversityHangzhou310027P. R. China
| | - Zhicai Shang
- Department of ChemistryZhejiang UniversityHangzhou310027P. R. China
| | - Yonggen Lou
- State Key Laboratory of Rice BiologyInstitute of Insect ScienceZhejiang UniversityHangzhou310058P. R. China
| | - Jun Wu
- Department of ChemistryZhejiang UniversityHangzhou310027P. R. China
| |
Collapse
|
24
|
The discovery of indole derivatives as novel hepatitis C virus inhibitors. Eur J Med Chem 2016; 116:147-155. [DOI: 10.1016/j.ejmech.2016.03.062] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 03/22/2016] [Accepted: 03/23/2016] [Indexed: 12/16/2022]
|
25
|
Applications of Replicating-Competent Reporter-Expressing Viruses in Diagnostic and Molecular Virology. Viruses 2016; 8:v8050127. [PMID: 27164126 PMCID: PMC4885082 DOI: 10.3390/v8050127] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 03/31/2016] [Accepted: 04/29/2016] [Indexed: 12/13/2022] Open
Abstract
Commonly used tests based on wild-type viruses, such as immunostaining, cannot meet the demands for rapid detection of viral replication, high-throughput screening for antivirals, as well as for tracking viral proteins or virus transport in real time. Notably, the development of replicating-competent reporter-expressing viruses (RCREVs) has provided an excellent option to detect directly viral replication without the use of secondary labeling, which represents a significant advance in virology. This article reviews the applications of RCREVs in diagnostic and molecular virology, including rapid neutralization tests, high-throughput screening systems, identification of viral receptors and virus-host interactions, dynamics of viral infections in vitro and in vivo, vaccination approaches and others. However, there remain various challenges associated with RCREVs, including pathogenicity alterations due to the insertion of a reporter gene, instability or loss of the reporter gene expression, or attenuation of reporter signals in vivo. Despite all these limitations, RCREVs have become powerful tools for both basic and applied virology with the development of new technologies for generating RCREVs, the inventions of novel reporters and the better understanding of regulation of viral replication.
Collapse
|
26
|
Camarasa M, Puig de la Bellacasa R, González ÀL, Ondoño R, Estrada R, Franco S, Badia R, Esté J, Martínez MÁ, Teixidó J, Clotet B, Borrell JI. Design, synthesis and biological evaluation of pyrido[2,3-d]pyrimidin-7-(8H)-ones as HCV inhibitors. Eur J Med Chem 2016; 115:463-83. [PMID: 27054294 DOI: 10.1016/j.ejmech.2016.03.055] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 03/03/2016] [Accepted: 03/18/2016] [Indexed: 11/26/2022]
Abstract
The design and selection of a combinatorial library of pyrido[2,3-d]pyrimidin-7(8H)-ones (4) has allowed the synthesis of 121 compounds, using known and new synthetic methodologies, and the evaluation of the inhibitory activity against hepatitis C virus (HCV) genotype 1b replicon. Among these compounds, 21{4,10} and 24{2,10} presented very high activities [EC50 = 0.027 μM (CC50 = 5.3 μM) and EC50 = 0.034 μM (CC50 = 13.5 μM), respectively] and high selectivity indexes, 196 and 397. These values are similar to the EC50 reported for sofosbuvir (2) (0.048 μM) using a similar methodological approach and the same virus subtype. 21{4,10} and 24{2,10} are obtained through shorter synthetic itineraries than sofosbuvir and 24{2,10} is achiral contrary to sofosbuvir which presents 4 stereogenic centers. In silico studies suggest that 21{4,10} and 24{2,10} inhibits NS5B polymerase through allosteric site binding.
Collapse
Affiliation(s)
- Marta Camarasa
- Grup d'Enginyeria Molecular, Institut Químic de Sarrià, Universitat Ramon Llull, Via Augusta 390, E-08017 Barcelona, Spain.
| | - Raimon Puig de la Bellacasa
- Grup d'Enginyeria Molecular, Institut Químic de Sarrià, Universitat Ramon Llull, Via Augusta 390, E-08017 Barcelona, Spain.
| | - Àlex L González
- Grup d'Enginyeria Molecular, Institut Químic de Sarrià, Universitat Ramon Llull, Via Augusta 390, E-08017 Barcelona, Spain.
| | - Raül Ondoño
- Grup d'Enginyeria Molecular, Institut Químic de Sarrià, Universitat Ramon Llull, Via Augusta 390, E-08017 Barcelona, Spain.
| | - Roger Estrada
- Grup d'Enginyeria Molecular, Institut Químic de Sarrià, Universitat Ramon Llull, Via Augusta 390, E-08017 Barcelona, Spain.
| | - Sandra Franco
- Retrovirology Laboratory IrsiCaixa, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, 08916 Badalona, Spain.
| | - Roger Badia
- Retrovirology Laboratory IrsiCaixa, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, 08916 Badalona, Spain.
| | - José Esté
- Retrovirology Laboratory IrsiCaixa, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, 08916 Badalona, Spain.
| | - Miguel Ángel Martínez
- Retrovirology Laboratory IrsiCaixa, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, 08916 Badalona, Spain.
| | - Jordi Teixidó
- Grup d'Enginyeria Molecular, Institut Químic de Sarrià, Universitat Ramon Llull, Via Augusta 390, E-08017 Barcelona, Spain.
| | - Bonaventura Clotet
- Retrovirology Laboratory IrsiCaixa, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, 08916 Badalona, Spain.
| | - José I Borrell
- Grup d'Enginyeria Molecular, Institut Químic de Sarrià, Universitat Ramon Llull, Via Augusta 390, E-08017 Barcelona, Spain.
| |
Collapse
|
27
|
Wei Y, Li J, Qing J, Huang M, Wu M, Gao F, Li D, Hong Z, Kong L, Huang W, Lin J. Discovery of Novel Hepatitis C Virus NS5B Polymerase Inhibitors by Combining Random Forest, Multiple e-Pharmacophore Modeling and Docking. PLoS One 2016; 11:e0148181. [PMID: 26845440 PMCID: PMC4742222 DOI: 10.1371/journal.pone.0148181] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 01/14/2016] [Indexed: 02/07/2023] Open
Abstract
The NS5B polymerase is one of the most attractive targets for developing new drugs to block Hepatitis C virus (HCV) infection. We describe the discovery of novel potent HCV NS5B polymerase inhibitors by employing a virtual screening (VS) approach, which is based on random forest (RB-VS), e-pharmacophore (PB-VS), and docking (DB-VS) methods. In the RB-VS stage, after feature selection, a model with 16 descriptors was used. In the PB-VS stage, six energy-based pharmacophore (e-pharmacophore) models from different crystal structures of the NS5B polymerase with ligands binding at the palm I, thumb I and thumb II regions were used. In the DB-VS stage, the Glide SP and XP docking protocols with default parameters were employed. In the virtual screening approach, the RB-VS, PB-VS and DB-VS methods were applied in increasing order of complexity to screen the InterBioScreen database. From the final hits, we selected 5 compounds for further anti-HCV activity and cellular cytotoxicity assay. All 5 compounds were found to inhibit NS5B polymerase with IC50 values of 2.01-23.84 μM and displayed anti-HCV activities with EC50 values ranging from 1.61 to 21.88 μM, and all compounds displayed no cellular cytotoxicity (CC50 > 100 μM) except compound N2, which displayed weak cytotoxicity with a CC50 value of 51.3 μM. The hit compound N2 had the best antiviral activity against HCV, with a selective index of 32.1. The 5 hit compounds with new scaffolds could potentially serve as NS5B polymerase inhibitors through further optimization and development.
Collapse
Affiliation(s)
- Yu Wei
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300071, China
| | - Jinlong Li
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300071, China
- High-Throughput Molecular Drug Discovery Center, Tianjin Joint Academy of Biomedicine and Technology, Tianjin, 300457, China
| | - Jie Qing
- Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Mingjie Huang
- College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Ming Wu
- Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Fenghua Gao
- Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Dongmei Li
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300071, China
| | - Zhangyong Hong
- College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Lingbao Kong
- College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, 330045, China
- * E-mail: (JPL); (WH); (LK)
| | - Weiqiang Huang
- PracticaChem-China, Tianjin, 300192, PR China
- * E-mail: (JPL); (WH); (LK)
| | - Jianping Lin
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300071, China
- High-Throughput Molecular Drug Discovery Center, Tianjin Joint Academy of Biomedicine and Technology, Tianjin, 300457, China
- * E-mail: (JPL); (WH); (LK)
| |
Collapse
|
28
|
Lee S, Yoon KD, Lee M, Cho Y, Choi G, Jang H, Kim B, Jung D, Oh J, Kim G, Oh J, Jeong Y, Kwon HJ, Bae SK, Min D, Windisch MP, Heo T, Lee C. Identification of a resveratrol tetramer as a potent inhibitor of hepatitis C virus helicase. Br J Pharmacol 2016; 173:191-211. [PMID: 26445091 PMCID: PMC4813382 DOI: 10.1111/bph.13358] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 09/16/2015] [Accepted: 10/02/2015] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND AND PURPOSE Hepatitis C virus (HCV) infection is responsible for various chronic inflammatory liver diseases. Here, we have identified a naturally occurring compound with anti-HCV activity and have elucidated its mode of antiviral action. EXPERIMENTAL APPROACH Luciferase reporter and real-time RT-PCR assays were used to measure HCV replication. Western blot, fluorescence-labelled HCV replicons and infectious clones were employed to quantitate expression levels of viral proteins. Resistant HCV mutant mapping, in vitro NS3 protease, helicase, NS5B polymerase and drug affinity responsive target stability assays were also used to study the antiviral mechanism. KEY RESULTS A resveratrol tetramer, vitisin B from grapevine root extract showed high potency against HCV replication (EC50 = 6 nM) with relatively low cytotoxicity (EC50 >10 μM). Combined treatment of vitisin B with an NS5B polymerase inhibitor (sofosbuvir) exhibited a synergistic or at least additive antiviral activity. Analysis of a number of vitisin B-resistant HCV variants suggested an NS3 helicase as its potential target. We confirmed a direct binding between vitisin B and a purified NS3 helicase in vitro. Vitisin B was a potent inhibitor of a HCV NS3 helicase (IC50 = 3 nM). In vivo, Finally, we observed a preferred tissue distribution of vitisin B in the liver after i.p. injection in rats, at clinically attainable concentrations. Conclusion and Implications Vitisin B is one of the most potent HCV helicase inhibitors identified so far. Vitisin B is thus a prime candidate to be developed as the first HCV drug derived from natural products.
Collapse
Affiliation(s)
- Sungjin Lee
- College of PharmacyDongguk UniversityGoyangKorea
| | - Kee Dong Yoon
- College of Pharmacy and Integrated Research Institute of Pharmaceutical SciencesThe Catholic University of KoreaBucheonKorea
| | - Myungeun Lee
- Hepatitis Research LaboratoryInstitut Pasteur KoreaSeongnamKorea
| | - Yoojin Cho
- Hepatitis Research LaboratoryInstitut Pasteur KoreaSeongnamKorea
| | - Gahee Choi
- Hepatitis Research LaboratoryInstitut Pasteur KoreaSeongnamKorea
| | - Hongje Jang
- Department of ChemistrySeoul National UniversitySeoulKorea
| | - BeomSeok Kim
- Translational Research Center for Protein Function Control, Department of Biotechnology, College of Life Science and BiotechnologyYonsei UniversitySeoulKorea
| | - Da‐Hee Jung
- Department of Bio and Nano ChemistryKookmin UniversitySeoulKorea
| | - Jin‐Gyo Oh
- College of Pharmacy and Integrated Research Institute of Pharmaceutical SciencesThe Catholic University of KoreaBucheonKorea
| | - Geon‐Woo Kim
- Department of BiotechnologyYonsei UniversitySeoulKorea
| | - Jong‐Won Oh
- Department of BiotechnologyYonsei UniversitySeoulKorea
| | - Yong‐Joo Jeong
- Department of Bio and Nano ChemistryKookmin UniversitySeoulKorea
| | - Ho Jeong Kwon
- Translational Research Center for Protein Function Control, Department of Biotechnology, College of Life Science and BiotechnologyYonsei UniversitySeoulKorea
| | - Soo Kyung Bae
- College of Pharmacy and Integrated Research Institute of Pharmaceutical SciencesThe Catholic University of KoreaBucheonKorea
| | - Dal‐Hee Min
- Department of ChemistrySeoul National UniversitySeoulKorea
| | - Marc P Windisch
- Hepatitis Research LaboratoryInstitut Pasteur KoreaSeongnamKorea
| | - Tae‐Hwe Heo
- College of Pharmacy and Integrated Research Institute of Pharmaceutical SciencesThe Catholic University of KoreaBucheonKorea
| | - Choongho Lee
- College of PharmacyDongguk UniversityGoyangKorea
| |
Collapse
|
29
|
He X, Yu Z, Jiang S, Zhang P, Shang Z, Lou Y, Wu J. Finding new elicitors that induce resistance in rice to the white-backed planthopper Sogatella furcifera. Bioorg Med Chem Lett 2015; 25:5601-3. [PMID: 26508551 DOI: 10.1016/j.bmcl.2015.10.041] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 10/12/2015] [Accepted: 10/15/2015] [Indexed: 11/18/2022]
Abstract
Herein we report a new way to identify chemical elicitors that induce resistance in rice to herbivores. Using this method, by quantifying the induction of chemicals for GUS activity in a specific screening system that we established previously, 5 candidate elicitors were selected from the 29 designed and synthesized phenoxyalkanoic acid derivatives. Bioassays confirmed that these candidate elicitors could induce plant defense and then repel feeding of white-backed planthopper Sogatella furcifera.
Collapse
Affiliation(s)
- Xingrui He
- Department of Chemistry, Zhejiang University, Hangzhou 310027, China
| | - Zhaonan Yu
- National Key Laboratory of Rice Biology, Institute of Insect Science, Zhejiang University, Hangzhou 310058, China
| | - Shaojie Jiang
- Department of Chemistry, Zhejiang University, Hangzhou 310027, China
| | - Peizhi Zhang
- School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou 310023, China
| | - Zhicai Shang
- Department of Chemistry, Zhejiang University, Hangzhou 310027, China
| | - Yonggen Lou
- National Key Laboratory of Rice Biology, Institute of Insect Science, Zhejiang University, Hangzhou 310058, China.
| | - Jun Wu
- Department of Chemistry, Zhejiang University, Hangzhou 310027, China.
| |
Collapse
|
30
|
Zhao C, Wang Y, Ma S. Recent advances on the synthesis of hepatitis C virus NS5B RNA-dependent RNA-polymerase inhibitors. Eur J Med Chem 2015; 102:188-214. [PMID: 26276434 DOI: 10.1016/j.ejmech.2015.07.046] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 07/23/2015] [Accepted: 07/28/2015] [Indexed: 02/07/2023]
Abstract
Hepatitis C is a viral liver infection considered as the major cause of cirrhosis and hepatocellular carcinoma (HCC). The HCV NS5B polymerase, an RNA-dependent RNA polymerase, is essential for HCV replication, which is able to catalyze the synthesis of positive (genomic) and negative (template) strand HCV RNA, but has no functional equivalent in mammalian cells. Therefore, the NS5B polymerase has emerged as an attractive target for the development of specifically targeted antiviral therapy for HCV (DAA, for direct-acting antivirals). Recently, a growing number of compounds have been reported as the NS5B polymerase inhibitors, some of which especially have been licensed in clinical trials. This review describes recent advances on the synthesis of the NS5B polymerase inhibitors, focusing on the merits and demerits of their synthetic methods. In particular, inspiration from the synthesis and the future direction of the NS5B polymerase inhibitors are highlighted.
Collapse
Affiliation(s)
- Can Zhao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, Jinan 250012, PR China
| | - Yinhu Wang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, Jinan 250012, PR China
| | - Shutao Ma
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, Jinan 250012, PR China.
| |
Collapse
|
31
|
Son S, Kim D, Lee S, Jin G, Park JA, Han HK, Lee K, Lee C. Synthesis and Structure-Activity Relationship of Novel Indole Acrylamide Derivatives as HCV Replication Inhibitors. B KOREAN CHEM SOC 2015. [DOI: 10.1002/bkcs.10021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Seohyun Son
- College of Pharmacy; Dongguk University-Seoul; Goyang 410-820 Ilsandong-gu Korea
| | - Dahee Kim
- College of Pharmacy; Dongguk University-Seoul; Goyang 410-820 Ilsandong-gu Korea
| | - Sungjin Lee
- College of Pharmacy; Dongguk University-Seoul; Goyang 410-820 Ilsandong-gu Korea
| | - Guanghai Jin
- College of Pharmacy; Dongguk University-Seoul; Goyang 410-820 Ilsandong-gu Korea
| | - Jin-Ah Park
- College of Pharmacy; Dongguk University-Seoul; Goyang 410-820 Ilsandong-gu Korea
| | - Hyo-Kyung Han
- College of Pharmacy; Dongguk University-Seoul; Goyang 410-820 Ilsandong-gu Korea
| | - Kyeong Lee
- College of Pharmacy; Dongguk University-Seoul; Goyang 410-820 Ilsandong-gu Korea
| | - Choongho Lee
- College of Pharmacy; Dongguk University-Seoul; Goyang 410-820 Ilsandong-gu Korea
| |
Collapse
|
32
|
Choi M, Kim YM, Lee S, Chin YW, Lee C. Mangosteen xanthones suppress hepatitis C virus genome replication. Virus Genes 2014; 49:208-22. [PMID: 24986787 DOI: 10.1007/s11262-014-1098-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 06/16/2014] [Indexed: 02/06/2023]
Abstract
Hepatitis C virus (HCV) is a hepatotropic single-stranded RNA virus. HCV infection is causally linked with development of liver cirrhosis and hepatocellular carcinoma. Enhanced production of reactive oxygen species by HCV has been implicated to play an important role in HCV-induced pathogenesis. Mangosteen has been widely used as a traditional medicine as well as a dietary supplement ,thanks to its powerful anti-oxidant effect. In the present study, we demonstrated that the ethanol extract from mangosteen fruit peels (MG-EtOH) is able to block HCV genome replication using HCV genotype 1b Bart79I subgenomic (EC50 5.1 μg/mL) and genotype 2a J6/JFH-1 infectious replicon systems (EC50 3.8 μg/mL). We found that inhibition of HCV replication by MG-EtOH led to subsequent down-regulation of expression of HCV proteins. Interestingly, MG-EtOH exhibited a modest inhibitory effect on in vitro RNA polymerase activity of NS5B. Among a number of xanthones compounds identified within this MG-EtOH, we discovered α-MG (EC50 6.3 μM) and γ-MG (EC50 2.7 μM) as two major single molecules responsible for suppression of HCV replication. This finding will provide a valuable molecular basis to further develop mangosteen as an important dietary supplement to combat HCV-induced liver diseases.
Collapse
Affiliation(s)
- Moonju Choi
- College of Pharmacy, Dongguk University-Seoul, Goyang, 410-050, South Korea
| | | | | | | | | |
Collapse
|