1
|
Saivish MV, Menezes GDL, da Silva RA, de Assis LR, Teixeira IDS, Fulco UL, Avilla CMS, Eberle RJ, Santos IDA, Korostov K, Webber ML, da Silva GCD, Nogueira ML, Jardim ACG, Regasin LO, Coronado MA, Pacca CC. Acridones as promising drug candidates against Oropouche virus. CURRENT RESEARCH IN MICROBIAL SCIENCES 2023; 6:100217. [PMID: 38234431 PMCID: PMC10792649 DOI: 10.1016/j.crmicr.2023.100217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2024] Open
Abstract
Oropouche virus (OROV) is an emerging vector-borne arbovirus found in South America that causes Oropouche fever, a febrile infection similar to dengue fever. It has a high epidemic potential, causing illness in over 500,000 cases diagnosed since the virus was first discovered in 1955. Currently, the prevention of human viral infection depends on vaccination, but availability for many viruses is limited, and they are classified as neglected viruses. At present, there are no vaccines or antiviral treatments available. An alternative approach to limiting the spread of the virus is to selectively disrupt viral replication mechanisms. Here, we demonstrate the inhibitory effect of acridones, which efficiently inhibited viral replication by 99.9 % in vitro. To evaluate possible mechanisms of action, we conducted tests with dsRNA, an intermediate in virus replication, as well as MD simulations, docking, and binding free energy analysis. The results showed a strong interaction between FAC21 and the OROV endonuclease, which possibly limits the interaction of viral RNA with other proteins. Therefore, our results suggest a dual mechanism of antiviral action, possibly caused by ds-RNA intercalation. In summary, our findings demonstrate that a new generation of antiviral drugs could be developed based on the selective optimization of molecules.
Collapse
Affiliation(s)
- Marielena Vogel Saivish
- Laboratório de Pesquisas em Virologia, Departamento de Doenças Dermatológicas, Infecciosas e Parasitárias, Faculdade de Medicina de São José do Rio Preto, São José do Rio Preto, SP 15090-000, Brazil
| | - Gabriela de Lima Menezes
- Unidade Especial de Ciências Exatas, Universidade Federal de Jataí, Jataí, GO 75801-615, Brazil
- Bioinformatics Multidisciplinary Environment, Programa de Pós-graduação em Bioinformática, Universidade Federal do Rio Grande do Norte, Natal 59078-400, RN, Brazil
| | | | - Leticia Ribeiro de Assis
- Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University, São José do Rio Preto, SP 15054-000, Brazil
| | - Igor da Silva Teixeira
- Laboratório de Pesquisas em Virologia, Departamento de Doenças Dermatológicas, Infecciosas e Parasitárias, Faculdade de Medicina de São José do Rio Preto, São José do Rio Preto, SP 15090-000, Brazil
| | - Umberto Laino Fulco
- Bioinformatics Multidisciplinary Environment, Programa de Pós-graduação em Bioinformática, Universidade Federal do Rio Grande do Norte, Natal 59078-400, RN, Brazil
| | - Clarita Maria Secco Avilla
- Laboratório de Pesquisas em Virologia, Departamento de Doenças Dermatológicas, Infecciosas e Parasitárias, Faculdade de Medicina de São José do Rio Preto, São José do Rio Preto, SP 15090-000, Brazil
- Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University, São José do Rio Preto, SP 15054-000, Brazil
| | - Raphael Josef Eberle
- Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, Jülich 52428, Germany
- Heinrich Heine University Düsseldorf, Faculty of Mathematics and Natural Sciences, Institute of Physical Biology, Universitätsstraße, Düsseldorf 40225, Germany
| | - Igor de Andrade Santos
- Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia-MG 38405-302, Brazil
| | - Karolina Korostov
- Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, Jülich 52428, Germany
| | - Mayara Lucia Webber
- Laboratório de Pesquisas em Virologia, Departamento de Doenças Dermatológicas, Infecciosas e Parasitárias, Faculdade de Medicina de São José do Rio Preto, São José do Rio Preto, SP 15090-000, Brazil
| | - Gislaine Celestino Dutra da Silva
- Laboratório de Pesquisas em Virologia, Departamento de Doenças Dermatológicas, Infecciosas e Parasitárias, Faculdade de Medicina de São José do Rio Preto, São José do Rio Preto, SP 15090-000, Brazil
| | - Maurício Lacerda Nogueira
- Laboratório de Pesquisas em Virologia, Departamento de Doenças Dermatológicas, Infecciosas e Parasitárias, Faculdade de Medicina de São José do Rio Preto, São José do Rio Preto, SP 15090-000, Brazil
| | - Ana Carolina Gomes Jardim
- Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University, São José do Rio Preto, SP 15054-000, Brazil
- Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia-MG 38405-302, Brazil
| | - Luis Octavio Regasin
- Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University, São José do Rio Preto, SP 15054-000, Brazil
| | - Mônika Aparecida Coronado
- Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, Jülich 52428, Germany
| | - Carolina Colombelli Pacca
- Laboratório de Pesquisas em Virologia, Departamento de Doenças Dermatológicas, Infecciosas e Parasitárias, Faculdade de Medicina de São José do Rio Preto, São José do Rio Preto, SP 15090-000, Brazil
- Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University, São José do Rio Preto, SP 15054-000, Brazil
| |
Collapse
|
2
|
Lv M, Wang X, Pan H, Chen J. Direct Observation of Ultrafast Access to a Solvent-Independent Singlet-Triplet Equilibrium State in Acridone Solutions. J Phys Chem B 2021; 125:13291-13297. [PMID: 34841879 DOI: 10.1021/acs.jpcb.1c08844] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Acridone and its derivatives have potential application as emitters for highly efficient blue organic light-emitting diodes (OLEDs). In this paper, we demonstrated ultrafast access of a solvent-independent singlet-triplet equilibrium state in acridone solutions by using femtosecond time-resolved spectroscopy. Our spectral data show that due to highly effective forward and reverse intersystem crossing (both kISC and krISC over 1010 s-1), a singlet-triplet equilibrium state is always populated in acridone in all solvents studied. However, the lifetimes of the equilibrium state varied a lot in different solvent environments and the final decay pathway of this state can switch between high quantum yield fluorescence emission and further internal conversion to the lowest triplet state. These findings provide direct experimental evidence to understand the distinct photophysical behaviors of acridone and also provide guidance for further design of acridone and its derivatives as blue OLED emitters.
Collapse
Affiliation(s)
- Meng Lv
- State Key Laboratory of Precision Spectroscopy, East China Normal University, Shanghai 200062, China
| | - Xueli Wang
- State Key Laboratory of Precision Spectroscopy, East China Normal University, Shanghai 200062, China
| | - Haifeng Pan
- State Key Laboratory of Precision Spectroscopy, East China Normal University, Shanghai 200062, China
| | - Jinquan Chen
- State Key Laboratory of Precision Spectroscopy, East China Normal University, Shanghai 200062, China.,Collaborative Innovation Center of Extreme Optics, Shanxi University, Taiyuan, Shanxi 030006, China
| |
Collapse
|
3
|
Gao Y, Nie J, Li Y, Liao G, Huo Y, Hu X. Rh(III)‐Catalyzed Selective
ortho
‐C−H Amination of Benzoic Acids with Anthranils: A Facile Access to Anthranilic Acid Derivatives (AAs). ChemCatChem 2020. [DOI: 10.1002/cctc.202000052] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Yang Gao
- School of Chemical Engineering and Light Industry Guangdong University of Technology Guangzhou 510006 P. R. China
| | - Jianhong Nie
- School of Chemical Engineering and Light Industry Guangdong University of Technology Guangzhou 510006 P. R. China
| | - Yibiao Li
- School of Biotechnology and Health Sciences Wuyi University Jiangmen Guangdong 529090 P. R. China
| | - Guilan Liao
- School of Chemical Engineering and Light Industry Guangdong University of Technology Guangzhou 510006 P. R. China
| | - Yanping Huo
- School of Chemical Engineering and Light Industry Guangdong University of Technology Guangzhou 510006 P. R. China
| | - Xiao‐Qiang Hu
- Key Laboratory of Catalysis and Energy Materials Chemistry of Ministry of Education & Hubei Key Laboratory of Catalysis and Materials Science School of Chemistry and Materials Science South-Central University for Nationalities Wuhan 430074 P. R. China
| |
Collapse
|
4
|
Kancharla P, Dodean RA, Li Y, Kelly JX. Boron Trifluoride Etherate Promoted Microwave-Assisted Synthesis of Antimalarial Acridones. RSC Adv 2019; 9:42284-42293. [PMID: 35321096 PMCID: PMC8939876 DOI: 10.1039/c9ra09478d] [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] [Indexed: 11/21/2022] Open
Abstract
A microwave-assisted, rapid and efficient method using boron trifluoride etherate (BF3.Et2O) for the synthesis of acridones, via an intramolecular acylation of N-phenylanthranilic acid derivatives, has been developed. The reaction proceeds under solvent-free conditions, tolerates a wide range of functional groups, and provides rapid access to a range of acridones in good to excellent yields. Several of the synthesized acridones exhibited potent antimalarial activities against CQ sensitive and multi-drug resistant (MDR) parasites.
Collapse
Affiliation(s)
- Papireddy Kancharla
- Department of Chemistry, Portland State University, Portland, Oregon 97201, United States
| | - Rozalia A Dodean
- Department of Chemistry, Portland State University, Portland, Oregon 97201, United States.,Department of Veterans Affairs Medical Center, Portland, Oregon 97239, United States
| | - Yuexin Li
- Department of Chemistry, Portland State University, Portland, Oregon 97201, United States.,Department of Veterans Affairs Medical Center, Portland, Oregon 97239, United States
| | - Jane X Kelly
- Department of Chemistry, Portland State University, Portland, Oregon 97201, United States.,Department of Veterans Affairs Medical Center, Portland, Oregon 97239, United States
| |
Collapse
|
5
|
Xu L, Li S, Liang Z, Lin H, Fu R. Acridone suppresses the proliferation of human breast cancer cells in vitro via ATP-binding cassette subfamily G member 2. Oncol Lett 2018; 15:2651-2654. [PMID: 29434987 DOI: 10.3892/ol.2017.7583] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 10/20/2017] [Indexed: 11/05/2022] Open
Abstract
In the past decades, the tricyclic acridone ring system has become a focus of major research by medicinal chemists due to the biological significance of this moiety in drug design and discovery. Acridone has substantial bio-potential since it performs crucial functions, including antibacterial, antimalarial, antiviral and anti-neoplastic activities. However, the anticancer effect and the underlying mechanisms of acridone on breast cancer cells remains unclear. In the present study, the anti-tumor function and the underlying mechanisms of acridone were evaluated in vitro. Firstly, an MTT assay was used to evaluate the inhibitory effect of acridone. Subsequently, reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was performed to investigate whether ATP binding cassette subfamily G member 2 (ABCG2) was associated with the function of acridone. Finally, western blotting was used to confirm the results of RT-qPCR. The present study demonstrated that acridone may decrease the proliferation of MDA-MB-231 cells dose-dependently. Further experiments revealed that acridone may downregulate the mRNA and protein expression levels of ABCG2, supporting the potential application of acridone in breast cancer treatment. These findings suggested that acridone is a potential agent in the treatment of human breast cancer.
Collapse
Affiliation(s)
- Licheng Xu
- Department of Breast Surgery, Yantaishan Hospital, Yantai, Shandong 264000, P.R. China
| | - Shuyan Li
- Department of Breast Surgery, Yantaishan Hospital, Yantai, Shandong 264000, P.R. China
| | - Zhi Liang
- Department of Breast Surgery, Yantaishan Hospital, Yantai, Shandong 264000, P.R. China
| | - Haixia Lin
- Department of Endocrinology, Yantai Yuhuangding Hospital, Yantai, Shandong 264000, P.R. China
| | - Rongzhan Fu
- Department of Breast Surgery, Qianfoshan Hospital Affiliated to Shandong University, Jinan, Shandong 250014, P.R. China
| |
Collapse
|
6
|
Shimizu JF, Pereira CM, Bittar C, Batista MN, Campos GRF, da Silva S, Cintra ACO, Zothner C, Harris M, Sampaio SV, Aquino VH, Rahal P, Jardim ACG. Multiple effects of toxins isolated from Crotalus durissus terrificus on the hepatitis C virus life cycle. PLoS One 2017; 12:e0187857. [PMID: 29141010 PMCID: PMC5687739 DOI: 10.1371/journal.pone.0187857] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 10/28/2017] [Indexed: 01/12/2023] Open
Abstract
Hepatitis C virus (HCV) is one of the main causes of liver disease and transplantation worldwide. Current therapy is expensive, presents additional side effects and viral resistance has been described. Therefore, studies for developing more efficient antivirals against HCV are needed. Compounds isolated from animal venoms have shown antiviral activity against some viruses such as Dengue virus, Yellow fever virus and Measles virus. In this study, we evaluated the effect of the complex crotoxin (CX) and its subunits crotapotin (CP) and phospholipase A2 (PLA2-CB) isolated from the venom of Crotalus durissus terrificus on HCV life cycle. Huh 7.5 cells were infected with HCVcc JFH-1 strain in the presence or absence of these toxins and virus was titrated by focus formation units assay or by qPCR. Toxins were added to the cells at different time points depending on the stage of virus life cycle to be evaluated. The results showed that treatment with PLA2-CB inhibited HCV entry and replication but no effect on HCV release was observed. CX reduced virus entry and release but not replication. By treating cells with CP, an antiviral effect was observed on HCV release, the only stage inhibited by this compound. Our data demonstrated the multiple antiviral effects of toxins from animal venoms on HCV life cycle.
Collapse
Affiliation(s)
- Jacqueline Farinha Shimizu
- Genomics Study Laboratory, São Paulo State University, IBILCE, S. José do Rio Preto, São Paulo, Brazil
- Laboratory of Virology, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Carina Machado Pereira
- Genomics Study Laboratory, São Paulo State University, IBILCE, S. José do Rio Preto, São Paulo, Brazil
| | - Cintia Bittar
- Genomics Study Laboratory, São Paulo State University, IBILCE, S. José do Rio Preto, São Paulo, Brazil
| | - Mariana Nogueira Batista
- Genomics Study Laboratory, São Paulo State University, IBILCE, S. José do Rio Preto, São Paulo, Brazil
| | | | - Suely da Silva
- Genomics Study Laboratory, São Paulo State University, IBILCE, S. José do Rio Preto, São Paulo, Brazil
- Laboratory of Virology, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | | | - Carsten Zothner
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom
| | - Mark Harris
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom
| | - Suely Vilela Sampaio
- Laboratory of Toxinology, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Victor Hugo Aquino
- Laboratory of Virology, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Paula Rahal
- Genomics Study Laboratory, São Paulo State University, IBILCE, S. José do Rio Preto, São Paulo, Brazil
| | - Ana Carolina Gomes Jardim
- Genomics Study Laboratory, São Paulo State University, IBILCE, S. José do Rio Preto, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
7
|
Campos GRF, Bittar C, Jardim ACG, Shimizu JF, Batista MN, Paganini ER, Ribeiro de Assis L, Bartlett C, Harris M, da Silva Bolzani V, Regasini LO, Rahal P. Hepatitis C virus in vitro replication is efficiently inhibited by acridone Fac4. J Gen Virol 2017; 98:1693-1701. [PMID: 28699869 PMCID: PMC7615702 DOI: 10.1099/jgv.0.000808] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Hepatitis C virus (HCV) affects about 170 million people worldwide. The current treatment has a high cost and variable response rates according to the virus genotype. Acridones, a group of compounds extracted from natural sources, showed potential antiviral actions against HCV. Thus, this study aimed to evaluate the effect of a panel of 14 synthetic acridones on the HCV life cycle. The compounds were screened using an Huh7.5 cell line stably harbouring the HCV genotype 2a subgenomic replicon SGR-Feo-JFH-1. Cells were incubated in the presence or absence of compounds for 72 h and cell viability and replication levels were assessed by MTT and luciferase assays, respectively. At a concentration of 5 µM the acridone Fac4 exhibited a >90 % inhibition of HCV replication with no effect on cell viability. The effects of Fac4 on virus replication, entry and release steps were evaluated in Huh7.5 cells infected with the JFH-1 isolate of HCV (HCVcc). Fac4 inhibited JFH-1 replication to approximately 70 %, while no effect was observed on virus entry. The antiviral activity of Fac4 was also observed on viral release, with almost 80 % of inhibition. No inhibitory effect was observed against genotype 3 replication. Fac4 was able to intercalate into dsRNA, however did not inhibit NS5B polymerase activity or translation driven by the HCV IRES. Although its mode of action is partly understood, Fac4 presents significant inhibition of HCV replication and can therefore be considered as a candidate for the development of a future anti-HCV treatment.
Collapse
Affiliation(s)
| | - Cíntia Bittar
- Institute of Bioscience, Language and Exact Science, IBILCE, UNESP – São Paulo State University, São José do Rio Preto, SP, Brazil
| | - Ana Carolina Gomes Jardim
- Institute of Biomedical Science, ICBIM, UFU – Federal University of Uberlândia, Uberlândia, MG, Brazil
| | - Jacqueline Farinha Shimizu
- Institute of Bioscience, Language and Exact Science, IBILCE, UNESP – São Paulo State University, São José do Rio Preto, SP, Brazil
| | - Mariana Nogueira Batista
- Institute of Bioscience, Language and Exact Science, IBILCE, UNESP – São Paulo State University, São José do Rio Preto, SP, Brazil
| | - Eder Ramos Paganini
- Institute of Bioscience, Language and Exact Science, IBILCE, UNESP – São Paulo State University, São José do Rio Preto, SP, Brazil
| | - Letícia Ribeiro de Assis
- Institute of Bioscience, Language and Exact Science, IBILCE, UNESP – São Paulo State University, São José do Rio Preto, SP, Brazil
| | - Christopher Bartlett
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Mark Harris
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | | | - Luis Octavio Regasini
- Institute of Bioscience, Language and Exact Science, IBILCE, UNESP – São Paulo State University, São José do Rio Preto, SP, Brazil
- Institute of Chemistry, São Paulo State University, Araraquara, SP, Brazil
| | - Paula Rahal
- Institute of Bioscience, Language and Exact Science, IBILCE, UNESP – São Paulo State University, São José do Rio Preto, SP, Brazil
| |
Collapse
|
8
|
Wen J, Tang S, Zhang F, Shi R, Lei A. Palladium/Copper Co-catalyzed Oxidative C–H/C–H Carbonylation of Diphenylamines: A Way To Access Acridones. Org Lett 2016; 19:94-97. [DOI: 10.1021/acs.orglett.6b03356] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Jiangwei Wen
- The
Institute for Advanced Studies (IAS), College of Chemistry and Molecular
Sciences, Wuhan University, Wuhan 430072, Hubei, China
| | - Shan Tang
- The
Institute for Advanced Studies (IAS), College of Chemistry and Molecular
Sciences, Wuhan University, Wuhan 430072, Hubei, China
| | - Fan Zhang
- The
Institute for Advanced Studies (IAS), College of Chemistry and Molecular
Sciences, Wuhan University, Wuhan 430072, Hubei, China
| | - Renyi Shi
- The
Institute for Advanced Studies (IAS), College of Chemistry and Molecular
Sciences, Wuhan University, Wuhan 430072, Hubei, China
| | - Aiwen Lei
- The
Institute for Advanced Studies (IAS), College of Chemistry and Molecular
Sciences, Wuhan University, Wuhan 430072, Hubei, China
- Conducted
and State Key Laboratory for Oxo Synthesis and Selective Oxidation, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| |
Collapse
|
9
|
Hassan STS, Berchová-Bímová K, Petráš J. Plumbagin, a Plant-Derived Compound, Exhibits Antifungal Combinatory Effect with Amphotericin B against Candida albicans Clinical Isolates and Anti-hepatitis C Virus Activity. Phytother Res 2016; 30:1487-92. [PMID: 27215409 DOI: 10.1002/ptr.5650] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 03/31/2016] [Accepted: 04/30/2016] [Indexed: 11/09/2022]
Abstract
Plumbagin (5-hydroxy-2-methyl-1,4-naphthoquinone), the major active constituent of Plumbago indica L., has been shown to be effective against a wide range of infectious microbes. In this study, plumbagin has been evaluated in vitro for its antifungal combinatory effect with amphotericin B against Candida albicans (C. albicans) clinical isolates and anti-hepatitis C virus (HCV) activity. Antifungal activity was determined by broth microdilution method, and combinatory effect was evaluated by checkerboard assay according to ΣFIC indices, while cytotoxicity was determined by MTT assay. Anti-HCV activity was determined in infected Huh7.5 cells using quantitative real-time reverse transcription PCR, and cytotoxicity was evaluated by MTT assay. Plumbagin exerted inhibitory effect against all C. albicans strains with minimum inhibitory concentration values ranging from 7.41 to 11.24 µg/mL. The additive effect of plumbagin when combined with amphotericin B at concentrations of (0.12, 0.13 and 0.19, 1.81 µg/mL, respectively) was obtained against five of seven strains tested with ΣFIC ranging from 0.62 to 0.91. In addition, plumbagin was found to be used safely for topical application when combined with amphotericin B at concentrations corresponding to the additive effect. Plumbagin exerted anti-HCV activity compared with that of telaprevir with IC50 values of 0.57 and 0.01 μM/L, respectively, and selectivity indices SI = 53.7 and SI = 2127, respectively. Our results present plumbagin as a potential therapeutic agent in the treatment of C. albicans and HCV infections. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Sherif T S Hassan
- Department of Natural Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences Brno, Palackého tř. 1946/1, 612 42, Brno, Czech Republic.,Department of Applied Ecology, Faculty of Environmental Sciences, Czech University of Life Sciences Prague, Kamýcká 129, 165 21 Praha 6 - Suchdol, Prague, Czech Republic
| | - Kateřina Berchová-Bímová
- Department of Applied Ecology, Faculty of Environmental Sciences, Czech University of Life Sciences Prague, Kamýcká 129, 165 21 Praha 6 - Suchdol, Prague, Czech Republic
| | - Jan Petráš
- Department of Natural Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences Brno, Palackého tř. 1946/1, 612 42, Brno, Czech Republic
| |
Collapse
|
10
|
Wang Y, Gao D, Chu B, Gao C, Cao D, Liu H, Jiang Y. Exposure of CCRF-CEM cells to acridone derivative 8a triggers tumor death via multiple mechanisms. Proteomics 2016; 16:1177-90. [DOI: 10.1002/pmic.201500317] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 02/02/2016] [Accepted: 02/08/2016] [Indexed: 02/06/2023]
Affiliation(s)
- Yini Wang
- Department of Chemistry; Tsinghua University; Beijing P. R. China
- The Key Laboratory of Tumor Metabolomics at Shenzhen; Shenzhen P. R. China
| | - Dan Gao
- The Key Laboratory of Tumor Metabolomics at Shenzhen; Shenzhen P. R. China
- The State Key Laboratory Breeding Base-Shenzhen Key Laboratory of Chemical Biology; Graduate School at Shenzhen; Tsinghua University; Shenzhen P. R. China
| | - Bizhu Chu
- The Key Laboratory of Tumor Metabolomics at Shenzhen; Shenzhen P. R. China
- The State Key Laboratory Breeding Base-Shenzhen Key Laboratory of Chemical Biology; Graduate School at Shenzhen; Tsinghua University; Shenzhen P. R. China
| | - Chunmei Gao
- The State Key Laboratory Breeding Base-Shenzhen Key Laboratory of Chemical Biology; Graduate School at Shenzhen; Tsinghua University; Shenzhen P. R. China
| | - Deliang Cao
- Department of Medical Microbiology; Immunology and Cell Biology; Simmons Cancer Institute; Southern Illinois University School of Medicine, Springfield; IL USA
| | - Hongxia Liu
- The Key Laboratory of Tumor Metabolomics at Shenzhen; Shenzhen P. R. China
- The State Key Laboratory Breeding Base-Shenzhen Key Laboratory of Chemical Biology; Graduate School at Shenzhen; Tsinghua University; Shenzhen P. R. China
| | - Yuyang Jiang
- The State Key Laboratory Breeding Base-Shenzhen Key Laboratory of Chemical Biology; Graduate School at Shenzhen; Tsinghua University; Shenzhen P. R. China
- School of Medicine; Tsinghua University; Beijing P. R. China
| |
Collapse
|
11
|
Feng M, Tang B, Wang N, Xu H, Jiang X. Ligand Controlled Regiodivergent C
1
Insertion on Arynes for Construction of Phenanthridinone and Acridone Alkaloids. Angew Chem Int Ed Engl 2015; 54:14960-4. [DOI: 10.1002/anie.201508340] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Indexed: 12/22/2022]
Affiliation(s)
- Minghao Feng
- Shanghai Key Laboratory of Green Chemistry and Chemical Process, School of Chemistry and Molecular Engineering, East China Normal University, 3663N, Zhongshan Road, Shanghai, 200062 (P.R. China)
| | - Bingqing Tang
- Shanghai Key Laboratory of Green Chemistry and Chemical Process, School of Chemistry and Molecular Engineering, East China Normal University, 3663N, Zhongshan Road, Shanghai, 200062 (P.R. China)
| | - Nengzhong Wang
- Shanghai Key Laboratory of Green Chemistry and Chemical Process, School of Chemistry and Molecular Engineering, East China Normal University, 3663N, Zhongshan Road, Shanghai, 200062 (P.R. China)
| | - Hong‐Xi Xu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Cai Lun Lu 1200, Shanghai, 201203 (P.R. China)
| | - Xuefeng Jiang
- Shanghai Key Laboratory of Green Chemistry and Chemical Process, School of Chemistry and Molecular Engineering, East China Normal University, 3663N, Zhongshan Road, Shanghai, 200062 (P.R. China)
- State Key Laboratory of Elemento‐organic Chemistry, Nankai University, Tianjin, (P.R. China)
- State Key Laboratory of Organometallic Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai (P.R. China)
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Cai Lun Lu 1200, Shanghai, 201203 (P.R. China)
| |
Collapse
|
12
|
Feng M, Tang B, Wang N, Xu H, Jiang X. Ligand Controlled Regiodivergent C
1
Insertion on Arynes for Construction of Phenanthridinone and Acridone Alkaloids. Angew Chem Int Ed Engl 2015. [DOI: 10.1002/ange.201508340] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Minghao Feng
- Shanghai Key Laboratory of Green Chemistry and Chemical Process, School of Chemistry and Molecular Engineering, East China Normal University, 3663N, Zhongshan Road, Shanghai, 200062 (P.R. China)
| | - Bingqing Tang
- Shanghai Key Laboratory of Green Chemistry and Chemical Process, School of Chemistry and Molecular Engineering, East China Normal University, 3663N, Zhongshan Road, Shanghai, 200062 (P.R. China)
| | - Nengzhong Wang
- Shanghai Key Laboratory of Green Chemistry and Chemical Process, School of Chemistry and Molecular Engineering, East China Normal University, 3663N, Zhongshan Road, Shanghai, 200062 (P.R. China)
| | - Hong‐Xi Xu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Cai Lun Lu 1200, Shanghai, 201203 (P.R. China)
| | - Xuefeng Jiang
- Shanghai Key Laboratory of Green Chemistry and Chemical Process, School of Chemistry and Molecular Engineering, East China Normal University, 3663N, Zhongshan Road, Shanghai, 200062 (P.R. China)
- State Key Laboratory of Elemento‐organic Chemistry, Nankai University, Tianjin, (P.R. China)
- State Key Laboratory of Organometallic Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai (P.R. China)
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Cai Lun Lu 1200, Shanghai, 201203 (P.R. China)
| |
Collapse
|
13
|
Jiang Z, Wang H, Li Y, Peng Z, Li Y, Li Z. Synthesis and antiviral activity of a series of novel N-phenylbenzamide and N-phenylacetophenone compounds as anti-HCV and anti-EV71 agents. Acta Pharm Sin B 2015; 5:201-9. [PMID: 26579447 PMCID: PMC4629265 DOI: 10.1016/j.apsb.2015.03.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2014] [Revised: 01/28/2015] [Accepted: 03/09/2015] [Indexed: 11/18/2022] Open
Abstract
A series of novel N-phenylbenzamide and N-phenylacetophenone compounds were synthesized and evaluated for their antiviral activity against HCV and EV71 (strain SZ-98). The biological results showed that three compounds (23, 25 and 41) exhibited considerable anti-HCV activity (IC50=0.57–7.12 μmol/L) and several compounds (23, 28, 29, 30, 31 and 42) displayed potent activity against EV71 with the IC50 values lower than 5.00 μmol/L. The potency of compound 23 (IC50=0.57 μmol/L) was superior to that of reported compounds IMB-1f (IC50=1.90 μmol/L) and IMB-1g (IC50=1.00 μmol/L) as anti-HCV agents, and compound 29 possessed the highest anti-EV71 activity, comparable to the comparator drug pirodavir. The efficacy in vivo and antiviral mechanism of these compounds warrant further investigations.
Collapse
Affiliation(s)
| | | | | | | | - Yuhuan Li
- Corresponding author. Tel.: +86 10 6301 0984; fax: +86 10 6301 7302.
| | - Zhuorong Li
- Corresponding author. Tel.: +86 10 8315 2017; fax: +86 10 6301 7302.
| |
Collapse
|
14
|
Mazzucco MB, Talarico LB, Vatansever S, Carro AC, Fascio ML, D'Accorso NB, García CC, Damonte EB. Antiviral activity of an N-allyl acridone against dengue virus. J Biomed Sci 2015; 22:29. [PMID: 25908170 PMCID: PMC4407772 DOI: 10.1186/s12929-015-0134-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 04/08/2015] [Indexed: 12/19/2022] Open
Abstract
Background Dengue virus (DENV), a member of the family Flaviviridae, is at present the most widespread causative agent of a human viral disease transmitted by mosquitoes. Despite the increasing incidence of this pathogen, there are no antiviral drugs or vaccines currently available for treatment or prevention. In a previous screening assay, we identified a group of N-allyl acridones as effective virus inhibitors. Here, the antiviral activity and mode of action targeted to viral RNA replication of one of the most active DENV-2 inhibitors was further characterized. Results The compound 10-allyl-7-chloro-9(10H)-acridone, designated 3b, was active to inhibit the in vitro infection of Vero cells with the four DENV serotypes, with effective concentration 50% (EC50) values in the range 12.5-27.1 μM, as determined by virus yield inhibition assays. The compound was also effective in human HeLa cells. No cytotoxicity was detected at 3b concentrations up to 1000 μM. Mechanistic studies demonstrated that virus entry into the host cell was not affected, whereas viral RNA synthesis was strongly inhibited, as quantified by real time RT-PCR. The addition of exogenous guanosine together with 3b rescued only partially the infectivity of DENV-2. Conclusions The acridone derivative 3b selectively inhibits the infection of Vero cells with the four DENV serotypes without a direct interaction with the host cell or the virion but interfering specifically with the intracellular virus multiplication. The mode of antiviral action for this acridone apparently involves the cellular enzyme inosine-monophospahe dehydrogenase together with another still unidentified target related to DENV RNA synthesis.
Collapse
Affiliation(s)
- María B Mazzucco
- Laboratorio de Virología, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, UBA, Ciudad Universitaria, Piso 4, Buenos Aires, Pabellón, 1428, Argentina. .,Present address: Laboratorio de Reproducción y Metabolismo, Facultad de Medicina, CEFYBO-CONICET, UBA, Buenos Aires, 1121, Argentina.
| | - Laura B Talarico
- Laboratorio de Virología, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, UBA, Ciudad Universitaria, Piso 4, Buenos Aires, Pabellón, 1428, Argentina. .,Present address: Fundación Infant, Buenos Aires, 1406, Argentina.
| | - Sezen Vatansever
- Graduate School of Science and Engineering, Koc University, Rumelifener yolu, Istanbul, Sarıyer, 34450, Turke.
| | - Ana C Carro
- Laboratorio de Virología, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, UBA, Ciudad Universitaria, Piso 4, Buenos Aires, Pabellón, 1428, Argentina. .,IQUIBICEN-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.
| | - Mirta L Fascio
- CIHIDECAR (CONICET), Departamento de Química Orgánica, Facultad de Ciencias Exactas y Naturales, UBA, Ciudad Universitaria, Pabellón 2, Piso 3, Buenos Aires, 1428, Argentina.
| | - Norma B D'Accorso
- CIHIDECAR (CONICET), Departamento de Química Orgánica, Facultad de Ciencias Exactas y Naturales, UBA, Ciudad Universitaria, Pabellón 2, Piso 3, Buenos Aires, 1428, Argentina.
| | - Cybele C García
- Laboratorio de Virología, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, UBA, Ciudad Universitaria, Piso 4, Buenos Aires, Pabellón, 1428, Argentina. .,IQUIBICEN-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.
| | - Elsa B Damonte
- Laboratorio de Virología, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, UBA, Ciudad Universitaria, Piso 4, Buenos Aires, Pabellón, 1428, Argentina. .,IQUIBICEN-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.
| |
Collapse
|
15
|
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: 0.7] [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
|
16
|
El-Shenawy R, Tabll A, Bader El Din NG, El Abd Y, Mashaly M, Abdel Malak CA, Dawood R, El-Awady M. Antiviral activity of virocidal peptide derived from NS5A against two different HCV genotypes: an in vitro study. J Immunoassay Immunochem 2015; 36:63-79. [PMID: 24606010 DOI: 10.1080/15321819.2014.896264] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
This study aimed at assessment of the antiviral activity of an amphipathic α-helical peptide derived from the hepatitis C virus NS5A known as C5A virocidal peptide against different HCV genotypes. Two sources of HCV virus for in vitro study: HCV genotype 4 sera samples and JFH-1 infectious culture system genotype 2a were used. Several virocidal peptide concentrations were tested to determine the concentration that inhibits HCV propagation in Huh 7.5 cells according to three different prortocols (pre-infection, coinfection, and post infection). The capacity of the virocidal peptide to block HCV in Huh7.5 cells infected with different 10 individual serum samples was evaluated. In the pre-infection protocol, virocidal concentration (20, 50, and 75 μM) showed no viral RNA. In the co-infection protocol, virocidal concentrations (10, 20, 50, 75 μM) showed no viral RNA while in post-infection protocol, 75 μM was the only concentration that blocked the HCV activity. Results of Huh7.5 cell line transfected with HCV cc J6/JFH and treated with virocidal peptide revealed that only the higher virocidal concentration (75 μM) showed no amplification. The percentage of virocidal blocking in the 10 HCV individual serum samples was 60%. In conclusion, the C5A virocidal peptide has potent antiviral activity against HCV.
Collapse
Affiliation(s)
- Reem El-Shenawy
- a Department of Microbial Biotechnology , National Research Center , Giza , Egypt
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Pourbasheer E, Aalizadeh R, Shokouhi Tabar S, Ganjali MR, Norouzi P, Shadmanesh J. 2D and 3D Quantitative Structure–Activity Relationship Study of Hepatitis C Virus NS5B Polymerase Inhibitors by Comparative Molecular Field Analysis and Comparative Molecular Similarity Indices Analysis Methods. J Chem Inf Model 2014; 54:2902-14. [DOI: 10.1021/ci500216c] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Eslam Pourbasheer
- Department
of Chemistry, Payame Noor University (PNU), P. O. Box 19395-3697, Tehran, Iran
| | | | - Samira Shokouhi Tabar
- Department
of Chemistry, Payame Noor University (PNU), P. O. Box 19395-3697, Tehran, Iran
| | - Mohammad Reza Ganjali
- Center
of Excellence in Electrochemistry, Faculty of Chemistry, University of Tehran, P.O. Box 143981-7435, Tehran, Iran
- Biosensor
Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences
Institute, Tehran University of Medical Sciences, P. O. Box,
14114-13137, Tehran, Iran
| | - Parviz Norouzi
- Center
of Excellence in Electrochemistry, Faculty of Chemistry, University of Tehran, P.O. Box 143981-7435, Tehran, Iran
- Biosensor
Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences
Institute, Tehran University of Medical Sciences, P. O. Box,
14114-13137, Tehran, Iran
| | | |
Collapse
|
18
|
Docking studies of Pakistani HCV NS3 helicase: a possible antiviral drug target. PLoS One 2014; 9:e106339. [PMID: 25188400 PMCID: PMC4154687 DOI: 10.1371/journal.pone.0106339] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2014] [Accepted: 07/19/2014] [Indexed: 01/02/2023] Open
Abstract
The nonstructural protein 3 (NS3) of hepatitis C virus (HCV) helicase is believed to be essential for viral replication and has become an attractive target for the development of antiviral drugs. The study of helicase is useful for elucidating its involvement in positive sense single-stranded RNA virus replication and to serve as templates for the design of novel antiviral drugs. In recent years, several models have been proposed on the conformational change leading to protein movement and RNA unwinding. Some compounds have been recently reported to inhibit the helicase and these include small molecules, RNA aptamers and antibodies. The current study is designed to help gain insights for the consideration of potential inhibitors for Pakistani HCV NS3 helicase protein. We have cloned, expressed and purified HCV NS3 helicase from Pakistani HCV serum samples and determined its 3D structure and employed it further in computational docking analysis to identify inhibitors against HCV genotype 3a (GT3a),including six antiviral key molecules such as quercetin, beta-carotene, resveratrol, catechins, lycopene and lutein. The conformation obtained after docking showed good hydrogen bond (HBond) interactions with best docking energy for quercetin and catechins followed by resveratrol and lutein. These anti-helicase key molecules will offer an alternative attraction to target the viral helicase, due to the current limitation with the interferon resistance treatment and presences of high rate of resistance in anti-protease inhibitor classes.
Collapse
|
19
|
Silva VL, Silva AM. New synthetic methods for 2,3-diarylacridin-9(10H)-one and (E)-2-aryl-4-styrylfuro[3,2-c]quinoline derivatives. Tetrahedron 2014. [DOI: 10.1016/j.tet.2014.05.040] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
20
|
Ng FN, Zhou Z, Yu WY. [RhIII(Cp*)]-Catalyzedortho-Selective Direct C(sp2)H Bond Amidation/Amination of Benzoic Acids byN-Chlorocarbamates andN-Chloromorpholines. A Versatile Synthesis of Functionalized Anthranilic Acids. Chemistry 2014; 20:4474-80. [DOI: 10.1002/chem.201304855] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Indexed: 11/06/2022]
|
21
|
Di Fusco M, Quintavalla A, Trombini C, Lombardo M, Roda A, Guardigli M, Mirasoli M. Preparation and Characterization of Thermochemiluminescent Acridine-Containing 1,2-Dioxetanes as Promising Ultrasensitive Labels in Bioanalysis. J Org Chem 2013; 78:11238-46. [DOI: 10.1021/jo401683r] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Massimo Di Fusco
- Advanced
Applications in Mechanical Engineering and Materials Technology Interdepartmental
Center for Industrial Research, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Arianna Quintavalla
- Department
of Chemistry ‘‘G. Ciamician’’, Alma Mater
Studiorum, University of Bologna, Bologna, Italy
| | - Claudio Trombini
- Department
of Chemistry ‘‘G. Ciamician’’, Alma Mater
Studiorum, University of Bologna, Bologna, Italy
| | - Marco Lombardo
- Department
of Chemistry ‘‘G. Ciamician’’, Alma Mater
Studiorum, University of Bologna, Bologna, Italy
| | - Aldo Roda
- Department
of Chemistry ‘‘G. Ciamician’’, Alma Mater
Studiorum, University of Bologna, Bologna, Italy
| | - Massimo Guardigli
- Department
of Chemistry ‘‘G. Ciamician’’, Alma Mater
Studiorum, University of Bologna, Bologna, Italy
| | - Mara Mirasoli
- Department
of Chemistry ‘‘G. Ciamician’’, Alma Mater
Studiorum, University of Bologna, Bologna, Italy
| |
Collapse
|
22
|
Hepatitis C virus NS3 inhibitors: current and future perspectives. BIOMED RESEARCH INTERNATIONAL 2013; 2013:467869. [PMID: 24282816 PMCID: PMC3825274 DOI: 10.1155/2013/467869] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 09/08/2013] [Indexed: 12/24/2022]
Abstract
Currently, hepatitis C virus (HCV) infection is considered a serious health-care problem all over the world. A good number of direct-acting antivirals (DAAs) against HCV infection are in clinical progress including NS3-4A protease inhibitors, RNA-dependent RNA polymerase inhibitors, and NS5A inhibitors as well as host targeted inhibitors. Two NS3-4A protease inhibitors (telaprevir and boceprevir) have been recently approved for the treatment of hepatitis C in combination with standard of care (pegylated interferon plus ribavirin). The new therapy has significantly improved sustained virologic response (SVR); however, the adverse effects associated with this therapy are still the main concern. In addition to the emergence of viral resistance, other targets must be continually developed. One such underdeveloped target is the helicase portion of the HCV NS3 protein. This review article summarizes our current understanding of HCV treatment, particularly with those of NS3 inhibitors.
Collapse
|
23
|
Li C, Ge LL, Li PP, Wang Y, Sun MX, Huang L, Ishag H, Di DD, Shen ZQ, Fan WX, Mao X. The DEAD-box RNA helicase DDX5 acts as a positive regulator of Japanese encephalitis virus replication by binding to viral 3' UTR. Antiviral Res 2013; 100:487-99. [PMID: 24035833 PMCID: PMC7113685 DOI: 10.1016/j.antiviral.2013.09.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 08/26/2013] [Accepted: 09/01/2013] [Indexed: 12/29/2022]
Abstract
Japanese encephalitis virus (JEV), one of the causes for epidemic encephalitis, belongs to the family of Flaviviridae. In this study, we demonstrated that cellular DEAD-box RNA helicase DDX5 plays an important role in JEV replication. The knockdown of DDX5 was able to decrease JEV replication, and overexpression of DDX5 mutants lacking the helicase activity also reduced JEV replication, suggesting the helicase activity is essential for JEV replication. DDX5 knockdown did not affect virus assembly and release. GST-pulldown and co-immunoprecipitation experiments demonstrated that DDX5 could interact with JEV core protein, non-structural protein 3 (NS3) and 5 (NS5-MTase and NS5-RdRp domains). Meanwhile, we also confirmed that DDX5 interacts with these viral proteins during JEV infection. Confocal microscopy analysis showed that endogenous DDX5 is recruited to the cytoplasm and colocalizes with these viral proteins and viral RNA. RNA-pulldown experiment showed that DDX5 only binds to the JEV 3' untranslated region (UTR). Finally, we confirmed the role of DDX5 in JEV RNA replication using JEV-replicon system. In conclusion, we identified DDX5 as a positive regulator for JEV replication.
Collapse
Affiliation(s)
- Chen Li
- College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing, Jiangsu Province 210095, China; Shandong Binzhou Animal Science and Veterinary Medicine Institute, Binzhou 256600, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Synthesis of novel substituted N-aryl benzamides as hA3G stabilizers and their inhibitory activities against hepatitis C virus replication. Acta Pharm Sin B 2013. [DOI: 10.1016/j.apsb.2013.07.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
25
|
Aromatase and glycosyl transferase inhibiting acridone alkaloids from fruits of Cameroonian Zanthoxylum species. Chem Cent J 2013; 7:125. [PMID: 23866063 PMCID: PMC3750308 DOI: 10.1186/1752-153x-7-125] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2013] [Accepted: 06/27/2013] [Indexed: 11/23/2022] Open
Abstract
Background Zanthoxylum zanthoxyloides and Z. leprieurii fruits are commonly used in traditional system of medicine for diarrhea, pain, wound healing, etc. in Cameroon, Africa. Z. leprieurii fruits have been chemically studied for its bioactive compounds whereas the investigation on Z. zanthoxyloides fruits is lacking. Results After a detailed chemical analysis of the fruits of Z. leprieurii and Z. zanthoxyloides, a series of new acridone alkaloids, namely, 3-hydroxy-1,5,6-trimethoxy-9-acridone (1), 1,6-dihydroxy-3-methoxy-9-acridone (2), 3,4,5,7-tetrahydroxy-1-methoxy-10-methyl-9-acridone (3), 4-methoxyzanthacridone (8), 4-hydroxyzanthacridone (9), 4-hydroxyzanthacridone oxide (2,4’) (10) have been isolated. The known acridones which have been characterized are, helebelicine A (4), 1-hydroxy-3-methoxy-10-methyl-9-acridone (5), 1,3-dihydroxy-4-methoxy-10-methyl-9-acridone (6) and tegerrardin A (7). The in vitro antibacterial and cytotoxic screening of these acridones reveal that compound 3 has a moderate antibacterial activity (MIC 125 μg/mL) against Micrococcus luteus and Pseudomonas aeruginosa while compound 1 shows a moderate cytotoxic effect (IC50 of 86 μM) against WRL-68 (liver cancer cell line). Furthermore, the molecular modeling of these acridones predicted the structural basis for their mode of action and binding affinity for aromatase, quinone reductase and WAAG, a glycosyltransferase involved in bacterial lipopolysaccharide synthesis. Computational approaches, quantitative SAR and modeling studies predicted that acridones 1, 2, 3, 4, 9 and 10 were the inhibitors of glycosyltransferase while 1, 8, 9 and 10, the inhibitors of aromatase. Conclusions A total of 10 acridones have been isolated out of which 6 are new (1, 2, 3, 8, 9 and 10). Alkaloids 8, 9 and 10, having novel tetracyclic acridone structure with new carbon skeleton, have now been named as zanthacridone. The quantitative SAR and molecular modeling studies suggested that the compounds 1, 9 and 10 are inhibitors of both aromatase and glycosyltransferase.
Collapse
|
26
|
Wang Y, Gao D, Chen Z, Li S, Gao C, Cao D, Liu F, Liu H, Jiang Y. Acridone derivative 8a induces oxidative stress-mediated apoptosis in CCRF-CEM leukemia cells: application of metabolomics in mechanistic studies of antitumor agents. PLoS One 2013; 8:e63572. [PMID: 23667640 PMCID: PMC3646819 DOI: 10.1371/journal.pone.0063572] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Accepted: 04/03/2013] [Indexed: 01/13/2023] Open
Abstract
A new acridone derivative, 2-aminoacetamido-10-(3, 5-dimethoxy)-benzyl-9(10H)-acridone hydrochloride (named 8a) synthesized in our lab shows potent antitumor activity, but the mechanism of action remains unclear. Herein, we report the use of an UPLC/Q-TOF MS metabolomic approach to study the effects of three compounds with structures optimized step-by-step, 9(10H)-acridone (A), 10-(3,5-dimethoxy)benzyl-9(10H)-acridone (I), and 8a, on CCRF-CEM leukemia cells and to shed new light on the probable antitumor mechanism of 8a. Acquired data were processed by principal component analysis (PCA) and orthogonal partial least squares discriminant analysis (OPLS-DA) to identify potential biomarkers. Comparing 8a-treated CCRF-CEM leukemia cells with vehicle control (DMSO), 23 distinct metabolites involved in five metabolic pathways were identified. Metabolites from glutathione (GSH) and glycerophospholipid metabolism were investigated in detail, and results showed that GSH level and the reduced/oxidized glutathione (GSH/GSSG) ratio were significantly decreased in 8a-treated cells, while L-cysteinyl-glycine (L-Cys-Gly) and glutamate were greatly increased. In glycerophospholipid metabolism, cell membrane components phosphatidylcholines (PCs) were decreased in 8a-treated cells, while the oxidative products lysophosphatidylcholines (LPCs) were significantly increased. We further found that in 8a-treated cells, the reactive oxygen species (ROS) and lipid peroxidation product malondialdehyde (MDA) were notably increased, accompanied with decrease of mitochondrial transmembrane potential, release of cytochrome C and activation of caspase-3. Taken together our results suggest that the acridone derivative 8a induces oxidative stress-mediated apoptosis in CCRF-CEM leukemia cells. The UPLC/Q-TOF MS based metabolomic approach provides novel insights into the mechanistic studies of antitumor drugs from a point distinct from traditional biological investigations.
Collapse
Affiliation(s)
- Yini Wang
- Department of Chemistry, Tsinghua University, Beijing, China
- The Key Laboratory of Tumor Metabolomics at Shenzhen, Shenzhen, China
| | - Dan Gao
- The Key Laboratory of Tumor Metabolomics at Shenzhen, Shenzhen, China
- The State Key Laboratory Breeding Base-Shenzhen Key Laboratory of Chemical Biology, Graduate School at Shenzhen, Tsinghua University, Shenzhen, China
| | - Zhe Chen
- The State Key Laboratory Breeding Base-Shenzhen Key Laboratory of Chemical Biology, Graduate School at Shenzhen, Tsinghua University, Shenzhen, China
| | - Shangfu Li
- The State Key Laboratory Breeding Base-Shenzhen Key Laboratory of Chemical Biology, Graduate School at Shenzhen, Tsinghua University, Shenzhen, China
| | - Chunmei Gao
- The State Key Laboratory Breeding Base-Shenzhen Key Laboratory of Chemical Biology, Graduate School at Shenzhen, Tsinghua University, Shenzhen, China
| | - Deliang Cao
- The State Key Laboratory Breeding Base-Shenzhen Key Laboratory of Chemical Biology, Graduate School at Shenzhen, Tsinghua University, Shenzhen, China
| | - Feng Liu
- The State Key Laboratory Breeding Base-Shenzhen Key Laboratory of Chemical Biology, Graduate School at Shenzhen, Tsinghua University, Shenzhen, China
| | - Hongxia Liu
- The Key Laboratory of Tumor Metabolomics at Shenzhen, Shenzhen, China
- The State Key Laboratory Breeding Base-Shenzhen Key Laboratory of Chemical Biology, Graduate School at Shenzhen, Tsinghua University, Shenzhen, China
- * E-mail: (HXL); (YYJ)
| | - Yuyang Jiang
- The State Key Laboratory Breeding Base-Shenzhen Key Laboratory of Chemical Biology, Graduate School at Shenzhen, Tsinghua University, Shenzhen, China
- School of Medicine, Tsinghua University, Beijing, China
- * E-mail: (HXL); (YYJ)
| |
Collapse
|
27
|
Huang J, Wan C, Xu MF, Zhu Q. Synthesis of 10-Methylacridin-9(10H)-ones through Cu-Catalyzed Intramolecular Oxidative C(sp2)-H Amination of 2-(Methylamino)benzophenones. European J Org Chem 2013. [DOI: 10.1002/ejoc.201201748] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
28
|
VERMA RAMANK, KUMAR VIJAY, GHOSH PRITHWISH, WADHWA LALITK. Heterocyclyl linked anilines and benzaldehydes as precursors for biologically significant new chemical entities. J CHEM SCI 2012. [DOI: 10.1007/s12039-012-0298-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
29
|
Linero FN, Sepúlveda CS, Giovannoni F, Castilla V, García CC, Scolaro LA, Damonte EB. Host cell factors as antiviral targets in arenavirus infection. Viruses 2012; 4:1569-91. [PMID: 23170173 PMCID: PMC3499820 DOI: 10.3390/v4091569] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Revised: 08/31/2012] [Accepted: 09/04/2012] [Indexed: 12/11/2022] Open
Abstract
Among the members of the Arenaviridae family, Lassa virus and Junin virus generate periodic annual outbreaks of severe human hemorrhagic fever (HF) in endemic areas of West Africa and Argentina, respectively. Given the human health threat that arenaviruses represent and the lack of a specific and safe chemotherapy, the search for effective antiviral compounds is a continuous demanding effort. Since diverse host cell pathways and enzymes are used by RNA viruses to fulfill their replicative cycle, the targeting of a host process has turned an attractive antiviral approach in the last years for many unrelated virus types. This strategy has the additional benefit to reduce the serious challenge for therapy of RNA viruses to escape from drug effects through selection of resistant variants triggered by their high mutation rate. This article focuses on novel strategies to identify inhibitors for arenavirus therapy, analyzing the potential for antiviral developments of diverse host factors essential for virus infection.
Collapse
Affiliation(s)
- Florencia N Linero
- Laboratorio de Virología, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires/IQUIBICEN (CONICET), Ciudad Universitaria, Pabellón 2, Piso 4, Buenos Aires 1428, Argentina.
| | | | | | | | | | | | | |
Collapse
|
30
|
Xu Z, Hobman TC. The helicase activity of DDX56 is required for its role in assembly of infectious West Nile virus particles. Virology 2012; 433:226-35. [PMID: 22925334 PMCID: PMC7119007 DOI: 10.1016/j.virol.2012.08.011] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Revised: 06/01/2012] [Accepted: 08/03/2012] [Indexed: 01/05/2023]
Abstract
Although flaviviruses encode their own helicases, evidence suggests that cellular helicases are also required for replication and/or assembly of these viruses. By and large, the mechanisms of action for viral and cellular helicases are not known. Moreover, in some cases, enzymatic activity is not even required for their roles in virus biology. Recently, we showed that expression of the host nucleolar helicase DDX56 is important for infectivity of West Nile virus (WNV) particles. In the present study, we demonstrate that the helicase activity of this enzyme is essential for its role in assembly of infectious WNV virions. Over-expression of the capsid-binding region of DDX56 also reduces infectivity of WNV suggesting that interaction of DDX56 and capsid protein is an important step in the virion assembly pathway. To our knowledge, this is the first study showing that enzymatic activity of a cellular helicase is critical for infectivity of flaviviruses.
Collapse
Affiliation(s)
- Zaikun Xu
- Department of Cell Biology, University of Alberta, Edmonton, Canada T6G 2H7
| | | |
Collapse
|
31
|
Mukherjee S, Hanson AM, Shadrick WR, Ndjomou J, Sweeney NL, Hernandez JJ, Bartczak D, Li K, Frankowski KJ, Heck JA, Arnold LA, Schoenen FJ, Frick DN. Identification and analysis of hepatitis C virus NS3 helicase inhibitors using nucleic acid binding assays. Nucleic Acids Res 2012; 40:8607-21. [PMID: 22740655 PMCID: PMC3458564 DOI: 10.1093/nar/gks623] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Typical assays used to discover and analyze small molecules that inhibit the hepatitis C virus (HCV) NS3 helicase yield few hits and are often confounded by compound interference. Oligonucleotide binding assays are examined here as an alternative. After comparing fluorescence polarization (FP), homogeneous time-resolved fluorescence (HTRF®; Cisbio) and AlphaScreen® (Perkin Elmer) assays, an FP-based assay was chosen to screen Sigma’s Library of Pharmacologically Active Compounds (LOPAC) for compounds that inhibit NS3-DNA complex formation. Four LOPAC compounds inhibited the FP-based assay: aurintricarboxylic acid (ATA) (IC50 = 1.4 μM), suramin sodium salt (IC50 = 3.6 μM), NF 023 hydrate (IC50 = 6.2 μM) and tyrphostin AG 538 (IC50 = 3.6 μM). All but AG 538 inhibited helicase-catalyzed strand separation, and all but NF 023 inhibited replication of subgenomic HCV replicons. A counterscreen using Escherichia coli single-stranded DNA binding protein (SSB) revealed that none of the new HCV helicase inhibitors were specific for NS3h. However, when the SSB-based assay was used to analyze derivatives of another non-specific helicase inhibitor, the main component of the dye primuline, it revealed that some primuline derivatives (e.g. PubChem CID50930730) are up to 30-fold more specific for HCV NS3h than similarly potent HCV helicase inhibitors.
Collapse
Affiliation(s)
- Sourav Mukherjee
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Todorov AR, Nieger M, Helaja J. Tautomeric switching and metal-cation sensing of ligand-equipped 4-hydroxy-/4-oxo-1,4-dihydroquinolines. Chemistry 2012; 18:7269-77. [PMID: 22517539 DOI: 10.1002/chem.201103868] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2011] [Revised: 02/21/2012] [Indexed: 11/11/2022]
Abstract
Novel 4-hydroxyquinoline (4HQ) based tautomeric switches are reported. 4HQs equipped with coordinative side arms (8-arylimino and 3-piperidin-1-ylmethyl groups) were synthesized to access O- or N-selective chelation of Zn(2+) and Cd(2+) ions by 4HQ. In the case of the monodentate arylimino group, O chelation of metal ions induces concomitant switching of phenol tautomer to the keto form in nonpolar or aprotic media. This change is accompanied by selective and highly sensitive fluorometric sensing of Zn(2+) ions. In the case of the bidentate 8-(quinolin-8-ylimino)methyl side arm, NMR studies in CD(3) OD indicated that both Cd(2+) and Zn(2+) ions afford N chelation for 4HQ, coexisting with tautomeric switching from quinolin-4(1H)-one to quinolin-4-olate. In corroboration, UV/Vis-monitored metal-ion titrations in toluene and methanol implied similar structural changes. Additionally, fluorescence measurements indicated that the metal-triggered tautomeric switching is associated with compound signaling properties. The results are supported by DFT calculations at the B3LYP 6-31G* level. Several X-ray structures of metal-free and metal-chelating 4HQ are presented to support the solution studies.
Collapse
Affiliation(s)
- Aleksandar R Todorov
- Laboratory of Organic Chemistry, Department of Chemistry, University of Helsinki, A.I.Virtasen aukio 1, P.O. Box. 55, University of Helsinki, 00014 Helsinki, Finland
| | | | | |
Collapse
|
33
|
Li K, Frankowski KJ, Belon CA, Neuenswander B, Ndjomou J, Hanson AM, Shanahan MA, Schoenen FJ, Blagg BSJ, Aubé J, Frick DN. Optimization of potent hepatitis C virus NS3 helicase inhibitors isolated from the yellow dyes thioflavine S and primuline. J Med Chem 2012; 55:3319-30. [PMID: 22409723 DOI: 10.1021/jm300021v] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A screen for hepatitis C virus (HCV) NS3 helicase inhibitors revealed that the commercial dye thioflavine S was the most potent inhibitor of NS3-catalyzed DNA and RNA unwinding in the 827-compound National Cancer Institute Mechanistic Set. Thioflavine S and the related dye primuline were separated here into their pure components, all of which were oligomers of substituted benzothiazoles. The most potent compound (P4), a benzothiazole tetramer, inhibited unwinding >50% at 2 ± 1 μM, inhibited the subgenomic HCV replicon at 10 μM, and was not toxic at 100 μM. Because P4 also interacted with DNA, more specific analogues were synthesized from the abundant dimeric component of primuline. Some of the 32 analogues prepared retained ability to inhibit HCV helicase but did not appear to interact with DNA. The most potent of these specific helicase inhibitors (compound 17) was active against the replicon and inhibited the helicase more than 50% at 2.6 ± 1 μM.
Collapse
Affiliation(s)
- Kelin Li
- University of Kansas Specialized Chemistry Center, University of Kansas, 2034 Becker Drive, Lawrence, Kansas 66047, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Peng HK, Lin CK, Yang SY, Tseng CK, Tzeng CC, Lee JC, Yang SC. Synthesis and anti-HCV activity evaluation of anilinoquinoline derivatives. Bioorg Med Chem Lett 2012; 22:1107-10. [DOI: 10.1016/j.bmcl.2011.11.121] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Revised: 11/25/2011] [Accepted: 11/28/2011] [Indexed: 12/12/2022]
|
35
|
Sepúlveda CS, García CC, Fascio ML, D'Accorso NB, Docampo Palacios ML, Pellón RF, Damonte EB. Inhibition of Junin virus RNA synthesis by an antiviral acridone derivative. Antiviral Res 2011; 93:16-22. [PMID: 22027649 DOI: 10.1016/j.antiviral.2011.10.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Revised: 09/06/2011] [Accepted: 10/07/2011] [Indexed: 01/31/2023]
Abstract
There are no specific approved drugs for the treatment of agents of viral hemorrhagic fevers (HF) and antiviral therapies against these viruses are urgently needed. The present study characterizes the potent and selective antiviral activity against the HF causing arenavirus Junin virus (JUNV) of the compound 10-allyl-6-chloro-4-methoxy-9(10H)-acridone, designated 3f. The effectiveness of 3f to inhibit JUNV multiplication was not importantly affected by the initial multiplicity of infection, with similar effective concentration 50% (EC(50)) values in virus yield inhibition assays performed in Vero cells in the range of 0.2-40 plaque forming units (PFU)/cell. Mechanistic studies demonstrated that 3f did not affect the initial steps of adsorption and internalization. The subsequent process of viral RNA synthesis was strongly inhibited, as quantified by real time RT-PCR in compound-treated cells relative to non-treated cells. The addition of exogenous guanosine rescued the infectivity and RNA synthesis of JUNV in 3f-treated cells in a dose-dependent manner, but the reversal was partial, suggesting that the reduction of the GTP pool contributed to the antiviral activity of 3f, but it was not the main operative mechanism. The comparison of 3f with two other viral RNA inhibitors, ribavirin and mycophenolic acid, showed that ribavirin did not act against JUNV through the cellular enzyme inosine monophosphate dehydrogenase (IMPDH) inhibition whereas the anti-JUNV activity of mycophenolic acid was mainly targeted at this enzyme.
Collapse
Affiliation(s)
- Claudia S Sepúlveda
- Laboratorio de Virología, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Pabellón 2, Piso 4, 1428 Buenos Aires, Argentina
| | | | | | | | | | | | | |
Collapse
|
36
|
Hwu JR, Lin SY, Tsay SC, Singha R, Pal BK, Leyssen P, Neyts J. Development of New Sulfur-Containing Conjugated Compounds as Anti-HCV Agents. PHOSPHORUS SULFUR 2011. [DOI: 10.1080/10426507.2010.520284] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Jih Ru Hwu
- a Department of Chemistry , National Tsing Hua University , Hsinchu, Taiwan, R.O.C
- b Department of Chemistry , National Central University , Jhongli City, Taiwan, R.O.C
| | - Shu-Yu Lin
- a Department of Chemistry , National Tsing Hua University , Hsinchu, Taiwan, R.O.C
| | - Shwu-Chen Tsay
- a Department of Chemistry , National Tsing Hua University , Hsinchu, Taiwan, R.O.C
| | - Raghunath Singha
- a Department of Chemistry , National Tsing Hua University , Hsinchu, Taiwan, R.O.C
| | - Benoy Kumar Pal
- a Department of Chemistry , National Tsing Hua University , Hsinchu, Taiwan, R.O.C
| | - Pieter Leyssen
- c Rega Institute for Medical Research , Katholieke Universiteit Leuven , Leuven, Belgium
| | - Johan Neyts
- c Rega Institute for Medical Research , Katholieke Universiteit Leuven , Leuven, Belgium
| |
Collapse
|
37
|
Dörner B, Kuntner C, Bankstahl JP, Wanek T, Bankstahl M, Stanek J, Müllauer J, Bauer F, Mairinger S, Löscher W, Miller DW, Chiba P, Müller M, Erker T, Langer O. Radiosynthesis and in vivo evaluation of 1-[18F]fluoroelacridar as a positron emission tomography tracer for P-glycoprotein and breast cancer resistance protein. Bioorg Med Chem 2011; 19:2190-8. [PMID: 21419632 DOI: 10.1016/j.bmc.2011.02.039] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2010] [Revised: 02/18/2011] [Accepted: 02/19/2011] [Indexed: 11/29/2022]
Abstract
Aim of this study was to label the potent dual P-glycoprotein (Pgp) and breast cancer resistance protein (BCRP) inhibitor elacridar (1) with (18)F to provide a positron emission tomography (PET) radiotracer to visualize Pgp and BCRP. A series of new 1- and 2-halogen- and nitro-substituted derivatives of 1 (4a-e) was synthesized as precursor molecules and reference compounds for radiolabelling and shown to display comparable in vitro potency to 1 in increasing rhodamine 123 accumulation in a cell line overexpressing human Pgp (MDCKII-MDR1). 1-[(18)F]fluoroelacridar ([(18)F]4b) was synthesized in a decay-corrected radiochemical yield of 1.7±0.9% by a 1-step no-carrier added nucleophilic aromatic (18)F-substitution of 1-nitro precursor 4c. Small-animal PET imaging of [(18)F]4b was performed in naïve rats, before and after administration of unlabelled 1 (5 mg/kg, n=3), as well as in wild-type and Mdr1a/b((-/-))Bcrp1((-/-)) mice (n=3). In PET experiments in rats, administration of unlabelled 1 increased brain activity uptake by a factor of 9.5 (p=0.0002, 2-tailed Student's t-test), whereas blood activity levels remained unchanged. In Mdr1a/b((-/-))Bcrp1((-/-)) mice, the mean brain-to-blood ratio of activity at 60 min after tracer injection was 7.6 times higher as compared to wild-type animals (p=0.0002). HPLC analysis of rat brain tissue extracts collected at 40 min after injection of [(18)F]4b revealed that 93±7% of total radioactivity in brain was in the form of unchanged [(18)F]4b. In conclusion, the in vivo behavior of [(18)F]4b was found to be similar to previously described [(11)C]1 suggesting transport of [(18)F]4b by Pgp and/or BCRP at the rodent BBB. However, low radiochemical yields and a significant degree of in vivo defluorination will limit the utility of [(18)F]4b as a PET tracer.
Collapse
Affiliation(s)
- Bernd Dörner
- Department of Medicinal Chemistry, University of Vienna, Austria
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Briguglio I, Piras S, Corona P, Carta A. Inhibition of RNA Helicases of ssRNA(+) Virus Belonging to Flaviviridae, Coronaviridae and Picornaviridae Families. INTERNATIONAL JOURNAL OF MEDICINAL CHEMISTRY 2010; 2011:213135. [PMID: 27516903 PMCID: PMC4970650 DOI: 10.1155/2011/213135] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2010] [Revised: 10/03/2010] [Accepted: 10/25/2010] [Indexed: 01/13/2023]
Abstract
Many viral pathogens encode the motor proteins named RNA helicases which display various functions in genome replication. General strategies to design specific and selective drugs targeting helicase for the treatment of viral infections could act via one or more of the following mechanisms: inhibition of the NTPase activity, by interferences with ATP binding and therefore by limiting the energy required for the unwinding and translocation, or by allosteric mechanism and therefore by stabilizing the conformation of the enzyme in low helicase activity state; inhibition of nucleic acids binding to the helicase; inhibition of coupling of ATP hydrolysis to unwinding; inhibition of unwinding by sterically blocking helicase translocation. Recently, by in vitro screening studies, it has been reported that several benzotriazole, imidazole, imidazodiazepine, phenothiazine, quinoline, anthracycline, triphenylmethane, tropolone, pyrrole, acridone, small peptide, and Bananin derivatives are endowed with helicase inhibition of pathogen viruses belonging to Flaviviridae, Coronaviridae, and Picornaviridae families.
Collapse
Affiliation(s)
- Irene Briguglio
- Department of Medicinal and Toxicological Chemistry, University of Sassari, Via Muroni 23/a, 07100 Sassari, Italy
| | - Sandra Piras
- Department of Medicinal and Toxicological Chemistry, University of Sassari, Via Muroni 23/a, 07100 Sassari, Italy
| | - Paola Corona
- Department of Medicinal and Toxicological Chemistry, University of Sassari, Via Muroni 23/a, 07100 Sassari, Italy
| | - Antonio Carta
- Department of Medicinal and Toxicological Chemistry, University of Sassari, Via Muroni 23/a, 07100 Sassari, Italy
| |
Collapse
|
39
|
Ranji A, Boris-Lawrie K. RNA helicases: emerging roles in viral replication and the host innate response. RNA Biol 2010; 7:775-87. [PMID: 21173576 DOI: 10.4161/rna.7.6.14249] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
RNA helicases serve multiple roles at the virus-host interface. In some situations, RNA helicases are essential host factors to promote viral replication; however, in other cases they serve as a cellular sensor to trigger the antiviral state in response to viral infection. All family members share the conserved ATP-dependent catalytic core linked to different substrate recognition and protein-protein interaction domains. These flanking domains can be shuffled between different helicases to achieve functional diversity. This review summarizes recent studies, which have revealed two types of activity by RNA helicases. First, RNA helicases are catalysts of progressive RNA-protein rearrangements that begin at gene transcription and culminate in mRNA translation. Second, RNA helicases can act as a scaffold for alternative protein-protein interactions that can defeat the antiviral state. The mounting fundamental understanding of RNA helicases is being used to develop selective and efficacious drugs against human and animal pathogens. The analysis of RNA helicases in virus model systems continues to provide insights into virology, cell biology and immunology, and has provided fresh perspective to continue unraveling the complexity of virus-host interactions.
Collapse
Affiliation(s)
- Arnaz Ranji
- Department of Veterinary Biosciences, Ohio State University, Columbus, OH, USA
| | | |
Collapse
|
40
|
Gao C, Liu F, Luan X, Tan C, Liu H, Xie Y, Jin Y, Jiang Y. Novel synthetic 2-amino-10-(3,5-dimethoxy)benzyl-9(10H)-acridinone derivatives as potent DNA-binding antiproliferative agents. Bioorg Med Chem 2010; 18:7507-14. [PMID: 20863710 DOI: 10.1016/j.bmc.2010.08.058] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2010] [Revised: 08/26/2010] [Accepted: 08/28/2010] [Indexed: 12/16/2022]
Abstract
A series of novel 9(10H)-acridinone derivatives with terminal amino substituents at C2 position on the acridinone ring were synthesized and studied for their antiproliferative activity and underlying mechanisms. These compounds demonstrated promising cytotoxicity to leukemia cells CCRF-CEM, displaying IC(50) values in the low micromolar range. Structure-activity relationships (SAR) indicated that the compound 6d bearing a pyrrolidine substituent and 8a with a methyl ammonium side chain displayed higher cytotoxicity to CCRF-CEM cells and also solid tumor cells A549, HepG2, and MCF7. Furthermore, the compounds 6d and 8a had strong binding activity to calf thymus DNA (ct DNA), as detected by UV absorption and fluorescence quenching assays, but limited inhibitory activity to human topoisomerase 1 (topo 1). Taken together, this study discovered a series of new synthetic 9(10H)-acridinone derivatives with potent DNA binding and anticancer activity.
Collapse
Affiliation(s)
- Chunmei Gao
- The Guangdong Province Key Laboratory of Chemical Biology, The Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, PR China
| | | | | | | | | | | | | | | |
Collapse
|