1
|
Álvarez-Mínguez A, del Río N, Belén-Blázquez A, Casanova E, Orduña JM, Camarero P, Hurtado-Marcos C, del Águila C, Pérez-Pérez M, Martín-Acebes MA, Agudo R. Development of a luminescence-based method for measuring West Nile Virus MTase activity and its application to screen for antivirals. CURRENT RESEARCH IN MICROBIAL SCIENCES 2024; 7:100282. [PMID: 39445035 PMCID: PMC11497361 DOI: 10.1016/j.crmicr.2024.100282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024] Open
Abstract
West Nile virus (WNV) is a flavivirus responsible for causing febrile illness and severe neurological diseases, with an increasing impact on human health around the world. However, there is still no adequate therapeutic treatment available to struggle WNV infections. Therefore, there is an urgent need to develop new techniques to accelerate the discovery of drugs against this pathogen. The main protein implicated in the replication of WNV is the non-structural protein 5 (NS5). This multifunctional protein contains methyltransferase (MTase) activity involved in the capping formation at the 5'-end of RNA and the methylation of internal viral RNA residues, both functions being essential for viral processes, such as RNA translation and escape from the innate immune response. We have developed a straightforward luminescence-based assay to monitor the MTase activity of the WNV NS5 protein with potential for high-throughput screening. We have validated this method as a sensitive and suitable assay for the identification of WNV MTase inhibitors assessing the inhibitory effect of the broad MTase inhibitor sinefungin, a natural nucleoside analog of the universal methyl donor S-adenosyl methionine (SAM). The screening of a small series of purine derivatives identified an adenosine derivative as a dose-dependent inhibitor of the MTase activity. The antiviral efficacy of this compound was further confirmed in WNV infections, displaying a measurable antiviral effect. This result supports the utility of this novel method for the screening of inhibitors against WNV MTase activity, which can be of special relevance to the discovery and development of therapeutics against WNV.
Collapse
Affiliation(s)
- Alejandra Álvarez-Mínguez
- Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28660, Boadilla del Monte, Spain
| | - Natalia del Río
- Instituto de Quimica Medica (IQM, CSIC) c/ Juan de la Cierva 3, 28006 Madrid, Spain
- Escuela de Doctorado, Universidad Autónoma de Madrid, Spain
| | - Ana Belén-Blázquez
- Departamento de Biotecnología, Instituto Nacional de Investigación y Tecnologia Agraria y Alimentaria (INIA-CSIC) Carretera de A Coruña Km 7.5, 28040 Madrid, Spain
| | - Elena Casanova
- Instituto de Quimica Medica (IQM, CSIC) c/ Juan de la Cierva 3, 28006 Madrid, Spain
| | - José-María Orduña
- Instituto de Quimica Medica (IQM, CSIC) c/ Juan de la Cierva 3, 28006 Madrid, Spain
| | - Patricia Camarero
- Instituto de Quimica Medica (IQM, CSIC) c/ Juan de la Cierva 3, 28006 Madrid, Spain
| | - Carolina Hurtado-Marcos
- Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28660, Boadilla del Monte, Spain
| | - Carmen del Águila
- Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28660, Boadilla del Monte, Spain
| | | | - Miguel A. Martín-Acebes
- Departamento de Biotecnología, Instituto Nacional de Investigación y Tecnologia Agraria y Alimentaria (INIA-CSIC) Carretera de A Coruña Km 7.5, 28040 Madrid, Spain
| | - Rubén Agudo
- Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28660, Boadilla del Monte, Spain
| |
Collapse
|
2
|
Caballero I, Castellví A, Triviño J, Jiménez E, Soler N, Borges R, Usón I. ARCIMBOLDO at low resolution: Verification for coiled coils and globular proteins. Protein Sci 2024; 33:e5136. [PMID: 39150227 PMCID: PMC11328115 DOI: 10.1002/pro.5136] [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: 03/30/2024] [Revised: 07/07/2024] [Accepted: 07/22/2024] [Indexed: 08/17/2024]
Abstract
Crystallography at low resolution must determine the atomic model from less experimental observations, which is challenging in the absence of a model. In addition, model bias is more severe when independent experimental data are scarce. Our methods solve the phase problem by combining the location of accurate model fragments using Phaser with density modification and interpretation of the resulting maps using SHELXE. From a partial, correct structure, the density modification process and the stereochemical constraints draw the rest of the structure, validating the result. This same principle is now exploited at low resolution. Coiled coils are important, ubiquitous structures but notoriously difficult to phase and to predict. Both correct solutions and incorrect ones are poorly discriminated by the crystallographic figures of merit as long as helices are correctly oriented. We incorporate coiled-coil verification, designed to set up competing, incompatible structural hypotheses to probe both the results and establish the power of the data to discriminate them. Efficiency of coiled-coil phasing and validation in test cases from 3 to 4 Å is demonstrated in ARCIMBOLDO_LITE, placing single helices, and in ARCIMBOLDO_SHREDDER, with fragments derived from AlphaFold models. SHELXE tracing at low resolution has been enhanced, maintaining its local character but extending the environment assessment. For non-helical structures, verification is demonstrated in the fragment location process. Its use is exemplified with the solution of the VSR1 structure at 3.5 Å, depending on LLG optimization and the emergence of new features in the electron density. Relying on verification, we have extended the use of the ARCIMBOLDO software to low resolution.
Collapse
Affiliation(s)
- Iracema Caballero
- Instituto de Biología Molecular de Barcelona (IBMB-CSIC), Barcelona Science Park, Barcelona, Spain
| | - Albert Castellví
- Instituto de Biología Molecular de Barcelona (IBMB-CSIC), Barcelona Science Park, Barcelona, Spain
| | - Josep Triviño
- Instituto de Biología Molecular de Barcelona (IBMB-CSIC), Barcelona Science Park, Barcelona, Spain
| | - Elisabet Jiménez
- Instituto de Biología Molecular de Barcelona (IBMB-CSIC), Barcelona Science Park, Barcelona, Spain
| | - Nicolas Soler
- Instituto de Biología Molecular de Barcelona (IBMB-CSIC), Barcelona Science Park, Barcelona, Spain
| | - Rafael Borges
- Instituto de Biología Molecular de Barcelona (IBMB-CSIC), Barcelona Science Park, Barcelona, Spain
| | - Isabel Usón
- Instituto de Biología Molecular de Barcelona (IBMB-CSIC), Barcelona Science Park, Barcelona, Spain
- ICREA: Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain
| |
Collapse
|
3
|
Goh JZH, De Hayr L, Khromykh AA, Slonchak A. The Flavivirus Non-Structural Protein 5 (NS5): Structure, Functions, and Targeting for Development of Vaccines and Therapeutics. Vaccines (Basel) 2024; 12:865. [PMID: 39203991 PMCID: PMC11360482 DOI: 10.3390/vaccines12080865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/20/2024] [Accepted: 07/27/2024] [Indexed: 09/03/2024] Open
Abstract
Flaviviruses, including dengue (DENV), Zika (ZIKV), West Nile (WNV), Japanese encephalitis (JEV), yellow fever (YFV), and tick-borne encephalitis (TBEV) viruses, pose a significant global emerging threat. With their potential to cause widespread outbreaks and severe health complications, the development of effective vaccines and antiviral therapeutics is imperative. The flaviviral non-structural protein 5 (NS5) is a highly conserved and multifunctional protein that is crucial for viral replication, and the NS5 protein of many flaviviruses has been shown to be a potent inhibitor of interferon (IFN) signalling. In this review, we discuss the functions of NS5, diverse NS5-mediated strategies adopted by flaviviruses to evade the host antiviral response, and how NS5 can be a target for the development of vaccines and antiviral therapeutics.
Collapse
Affiliation(s)
| | | | | | - Andrii Slonchak
- Australian Infectious Diseases Research Center, School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (J.Z.H.G.); (L.D.H.); (A.A.K.)
| |
Collapse
|
4
|
Peng NYG, Sng JDJ, Setoh YX, Khromykh AA. Residue K28 of Zika Virus NS5 Protein Is Implicated in Virus Replication and Antagonism of STAT2. Microorganisms 2024; 12:660. [PMID: 38674605 PMCID: PMC11052099 DOI: 10.3390/microorganisms12040660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 03/07/2024] [Accepted: 03/19/2024] [Indexed: 04/28/2024] Open
Abstract
The identification of four potential nonstructural 5 (NS5) residues-K28, K45, V335, and S749-that share the same amino acid preference in STAT2-interacting flaviviruses [Dengue virus (DENV) and Zika virus (ZIKV)], but not in STAT2-non-interacting flaviviruses [West Nile virus (WNV) and/or Yellow fever virus (YFV)] from an alignment of multiple flavivirus NS5 sequences, implied a possible association with the efficiency of ZIKV to antagonize the human signal transducer and activator of transcription factor 2 (STAT2). Through site-directed mutagenesis and reverse genetics, mutational impacts of these residues on ZIKV growth in vitro and STAT2 antagonism were assessed using virus growth kinetics assays and STAT2 immunoblotting. The results showed that mutations at the residue K28 significantly reduced the efficiency of ZIKV to antagonize STAT2. Further investigation involving residue K28 demonstrated its additional effects on the phenotypes of ZIKV-NS5 nuclear bodies. These findings demonstrate that K28, identified from sequence alignment, is an important determinant of replication and STAT2 antagonism by ZIKV.
Collapse
Affiliation(s)
- Nias Y. G. Peng
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (J.D.J.S.); (Y.X.S.)
| | - Julian D. J. Sng
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (J.D.J.S.); (Y.X.S.)
| | - Yin Xiang Setoh
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (J.D.J.S.); (Y.X.S.)
- Infectious Diseases Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
| | - Alexander A. Khromykh
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (J.D.J.S.); (Y.X.S.)
- Australian Infectious Diseases Research Centre, Global Virus Network Centre of Excellence, Brisbane, QLD 4072, Australia
| |
Collapse
|
5
|
Zhang K, Liang J, Zhang B, Huang L, Yu J, Xiao X, He Z, Tao H, Yuan J. A Marine Natural Product, Harzianopyridone, as an Anti-ZIKV Agent by Targeting RNA-Dependent RNA Polymerase. Molecules 2024; 29:978. [PMID: 38474490 DOI: 10.3390/molecules29050978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/15/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
The Zika virus (ZIKV) is a mosquito-borne virus that already poses a danger to worldwide human health. Patients infected with ZIKV generally have mild symptoms like a low-grade fever and joint pain. However, severe symptoms can also occur, such as Guillain-Barré syndrome, neuropathy, and myelitis. Pregnant women infected with ZIKV may also cause microcephaly in newborns. To date, we still lack conventional antiviral drugs to treat ZIKV infections. Marine natural products have novel structures and diverse biological activities. They have been discovered to have antibacterial, antiviral, anticancer, and other therapeutic effects. Therefore, marine products are important resources for compounds for innovative medicines. In this study, we identified a marine natural product, harzianopyridone (HAR), that could inhibit ZIKV replication with EC50 values from 0.46 to 2.63 µM while not showing obvious cytotoxicity in multiple cellular models (CC50 > 45 µM). Further, it also reduced the expression of viral proteins and protected cells from viral infection. More importantly, we found that HAR directly bound to the ZIKV RNA-dependent RNA polymerase (RdRp) and suppressed its polymerase activity. Collectively, our findings provide HAR as an option for the development of anti-ZIKV drugs.
Collapse
Affiliation(s)
- Kexin Zhang
- School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
| | - Jingyao Liang
- School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
| | - Bingzhi Zhang
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Lishan Huang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Jianchen Yu
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Xuhan Xiao
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Zhenjian He
- School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
| | - Huaming Tao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Jie Yuan
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| |
Collapse
|
6
|
Tsukamoto Y, Igarashi M, Kato H. Targeting cap1 RNA methyltransferases as an antiviral strategy. Cell Chem Biol 2024; 31:86-99. [PMID: 38091983 DOI: 10.1016/j.chembiol.2023.11.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 10/30/2023] [Accepted: 11/20/2023] [Indexed: 01/21/2024]
Abstract
Methylation is one of the critical modifications that regulates numerous biological processes. Guanine capping and methylation at the 7th position (m7G) have been shown to mature mRNA for increased RNA stability and translational efficiency. The m7G capped cap0 RNA remains immature and requires additional methylation at the first nucleotide (N1-2'-O-Me), designated as cap1, to achieve full maturation. This cap1 RNA with N1-2'-O-Me prevents its recognition by innate immune sensors as non-self. Viruses have also evolved various strategies to produce self-like capped RNAs with the N1-2'-O-Me that potentially evades the antiviral response and establishes an efficient replication. In this review, we focus on the importance of the presence of N1-2'-O-Me in viral RNAs and discuss the potential for drug development by targeting host and viral N1-2'-O-methyltransferases.
Collapse
Affiliation(s)
- Yuta Tsukamoto
- Institute of Cardiovascular Immunology, Medical Faculty, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Manabu Igarashi
- Division of Global Epidemiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan; International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Hiroki Kato
- Institute of Cardiovascular Immunology, Medical Faculty, University Hospital Bonn, University of Bonn, Bonn, Germany.
| |
Collapse
|
7
|
Wang L, Zhou R, Liu Y, Guo S, Yi D, Zhao J, Li Q, Zhang Y, Liang C, Wang J, Shan G, Cen S. A cell-based assay to discover inhibitors of Zika virus RNA-dependent RNA polymerase. Virology 2024; 589:109939. [PMID: 37979208 DOI: 10.1016/j.virol.2023.109939] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/01/2023] [Accepted: 11/07/2023] [Indexed: 11/20/2023]
Abstract
Zika virus (ZIKV) belongs to Flaviviridae, the Flavivirus genus. Its infection causes congenital brain abnormalities and Guillain-Barré syndrome. However, there are no effective vaccines, no FDA-approved drugs to manage ZIKV infection. The non-structural protein NS5 of ZIKV has been recognized as a valuable target of antivirals because of its RNA-dependent RNA polymerase (RdRp) and methyltransferase (MTase) activities essential for viral RNA synthesis. Here, we report a cell-based assay for discovering inhibitors of ZIKV NS5 and found that 5-Azacytidine potently inhibits ZIKV NS5, with EC50 of 4.9 μM. Furthermore, 5-Azacytidine suppresses ZIKV replication by inhibiting NS5-mediated viral RNA transcription. Therefore, we have developed a cell-based ZIKV NS5 assay which can be deployed to discover ZIKV NS5 inhibitors and demonstrated the potential of 5-Azacytidine for further development as a ZIKV NS5 inhibitor.
Collapse
Affiliation(s)
- Lidan Wang
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Tiantanxili, Beijing, 100050, China
| | - Rui Zhou
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Tiantanxili, Beijing, 100050, China
| | - Yitong Liu
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Tiantanxili, Beijing, 100050, China
| | - Saisai Guo
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Tiantanxili, Beijing, 100050, China
| | - Dongrong Yi
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Tiantanxili, Beijing, 100050, China
| | - Jianyuan Zhao
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Tiantanxili, Beijing, 100050, China
| | - Quanjie Li
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Tiantanxili, Beijing, 100050, China
| | - Yongxin Zhang
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Tiantanxili, Beijing, 100050, China
| | - Chen Liang
- Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montreal, H3T 1E2, Canada
| | - Jing Wang
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Tiantanxili, Beijing, 100050, China.
| | - Guangzhi Shan
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Tiantanxili, Beijing, 100050, China.
| | - Shan Cen
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Tiantanxili, Beijing, 100050, China.
| |
Collapse
|
8
|
Patar AK, Borah SM, Barman J, Bora A, Baruah TJ. Dronabinol as an answer to flavivirus infections: an in-silico investigation. J Biomol Struct Dyn 2023; 41:11219-11230. [PMID: 36576139 DOI: 10.1080/07391102.2022.2160817] [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: 09/23/2021] [Accepted: 12/15/2022] [Indexed: 12/29/2022]
Abstract
Flavivirus infections are common in several parts of the world. Two major types of flaviviruses are dengue and zika viruses. Both these two viral infections have caused many fatalities around the world. There is an absence of a vaccine and an effective medication against these viruses. In this study, we analyzed the ability of dronabinol to act as a potential cure against these viral infections. We performed the docking of dronabinol with several viral proteins followed by molecular dynamics simulation, MM/PBSA and PCA analysis. We checked the ability of the polyphenol dronabinol to interfere with the binding of viral helicases to their cellular targets. We performed 2 D-QSAR studies, drug likeliness, ADMET and target prediction studies. From our study, we observed that dronabinol had the best docking ability against the helicase proteins of dengue and zika. Molecular dynamics simulation and MM/PBSA investigation confirmed the stability of the binding while PCA investigation showed a lowering of molecular motions in response to dronabinol docking to the helicases. Dronabinol interfered in the binding of the helicases to RNA. 2 D QSAR studies revealed a low IC50 value for dronabinol. Dronabinol showed favorable drug-likeness, ADMET properties and target prediction results. Thus we propose dronabinol be further investigated in-vitro as a cure against dengue and zika virus infections.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Abani Kumar Patar
- Department of Biochemistry, Assam Royal Global University, Guwahati, Assam, India
| | - Sapna Mayuri Borah
- Department of Plant Pathology, Assam Agricultural University, Jorhat, Assam, India
| | - Jitul Barman
- Department of Biochemistry, Assam Royal Global University, Guwahati, Assam, India
| | - Anupam Bora
- Department of Biochemistry, Assam Royal Global University, Guwahati, Assam, India
| | - Taranga Jyoti Baruah
- Department of Biochemistry, Assam Royal Global University, Guwahati, Assam, India
| |
Collapse
|
9
|
Mensah IK, Norvil AB, He M, Lendy E, Hjortland N, Tan H, Pomerantz RT, Mesecar A, Gowher H. Development of a sensitive microplate assay for characterizing RNA methyltransferase activity: Implications for epitranscriptomics and drug development. J Biol Chem 2023; 299:105257. [PMID: 37716702 PMCID: PMC10582764 DOI: 10.1016/j.jbc.2023.105257] [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: 01/25/2023] [Revised: 09/06/2023] [Accepted: 09/08/2023] [Indexed: 09/18/2023] Open
Abstract
RNA methylation is a ubiquitous post-transcriptional modification found in diverse RNA classes and is a critical regulator of gene expression. In this study, we used Zika virus RNA methyltransferase (MTase) to develop a highly sensitive microplate assay that uses a biotinylated RNA substrate and radiolabeled AdoMet coenzyme. The assay is fast, highly reproducible, exhibits linear progress-curve kinetics under multiple turnover conditions, has high sensitivity in competitive inhibition assays, and significantly lower background levels compared with the currently used method. Using our newly developed microplate assay, we observed no significant difference in the catalytic constants of the full-length nonstructural protein 5 enzyme and the truncated MTase domain. These data suggest that, unlike the Zika virus RNA-dependent RNA polymerase activity, the MTase activity is unaffected by RNA-dependent RNA polymerase-MTase interdomain interaction. Given its quantitative nature and accuracy, this method can be used to characterize various RNA MTases, and, therefore, significantly contribute to the field of epitranscriptomics and drug development against infectious diseases.
Collapse
Affiliation(s)
- Isaiah K Mensah
- Department of Biochemistry, Purdue University Center for Cancer Research, Purdue University, West Lafayette, Indiana, USA
| | - Allison B Norvil
- Department of Biochemistry, Purdue University Center for Cancer Research, Purdue University, West Lafayette, Indiana, USA
| | - Ming He
- Department of Biochemistry, Purdue University Center for Cancer Research, Purdue University, West Lafayette, Indiana, USA
| | - Emma Lendy
- Department of Biochemistry, Purdue University Center for Cancer Research, Purdue University, West Lafayette, Indiana, USA
| | - Nicole Hjortland
- Department of Biochemistry, Purdue University Center for Cancer Research, Purdue University, West Lafayette, Indiana, USA
| | - Hern Tan
- Department of Biochemistry, Purdue University Center for Cancer Research, Purdue University, West Lafayette, Indiana, USA
| | - Richard T Pomerantz
- Department Biochemistry and Molecular Biology, Thomas Jefferson University, Sidney Kimmel Cancer Center, Philadelphia, Pennsylvania, USA
| | - Andrew Mesecar
- Department of Biochemistry, Purdue University Center for Cancer Research, Purdue University, West Lafayette, Indiana, USA
| | - Humaira Gowher
- Department of Biochemistry, Purdue University Center for Cancer Research, Purdue University, West Lafayette, Indiana, USA.
| |
Collapse
|
10
|
Samrat SK, Bashir Q, Huang Y, Trieshmann CW, Tharappel AM, Zhang R, Chen K, Geoge Zheng Y, Li Z, Li H. Broad-Spectrum Small-Molecule Inhibitors Targeting the SAM-Binding Site of Flavivirus NS5 Methyltransferase. ACS Infect Dis 2023; 9:1319-1333. [PMID: 37348028 PMCID: PMC10436986 DOI: 10.1021/acsinfecdis.2c00571] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/24/2023]
Abstract
Flavivirus infections, such as those caused by dengue virus (DENV), West Nile virus (WNV), yellow fever virus (YFV), and Zika virus (ZIKV), pose a rising threat to global health. There are no FDA-approved drugs for flaviviruses, although a small number of flaviviruses have vaccines. For flaviviruses or unknown viruses that may appear in the future, it is particularly desirable to identify broad-spectrum inhibitors. The NS5 protein is regarded as one of the most promising flavivirus drug targets because it is conserved across flaviviruses. In this study, we used FL-NAH, a fluorescent analog of the methyl donor S-adenosyl methionine (SAM), to develop a fluorescence polarization (FP)-based high throughput screening (HTS) assay to specifically target methyltransferase (MTase), a vital enzyme for flaviviruses that methylates the N7 and 2'-O positions of the viral 5'-RNA cap. Pilot screening identified two candidate MTase inhibitors, NSC 111552 and 288387. The two compounds inhibited the FL-NAH binding to the DENV3 MTase with low micromolar IC50. Functional assays verified the inhibitory potency of these molecules for the flavivirus MTase activity. Binding studies indicated that these molecules are bound directly to the DENV3 MTase with similar low micromolar affinity. Furthermore, we showed that these compounds greatly reduced ZIKV replication in cell-based experiments at dosages that did not cause cytotoxicity. Finally, docking studies revealed that these molecules bind to the SAM-binding region on the DENV3 MTase, and further mutagenesis studies verified residues important for the binding of these compounds. Overall, these compounds are innovative and attractive candidates for the development of broad-spectrum inhibitors for the treatment of flavivirus infections.
Collapse
Affiliation(s)
- Subodh Kumar Samrat
- Department of Pharmacology and Toxicology, R Ken Coit College of Pharmacy, The University of Arizona, 1703 E Mabel St, Tucson AZ, 85721-0207, USA
| | - Qamar Bashir
- Department of Pharmacology and Toxicology, R Ken Coit College of Pharmacy, The University of Arizona, 1703 E Mabel St, Tucson AZ, 85721-0207, USA
| | - Yiding Huang
- Department of Pharmacology and Toxicology, R Ken Coit College of Pharmacy, The University of Arizona, 1703 E Mabel St, Tucson AZ, 85721-0207, USA
| | - Carl William Trieshmann
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia, 30602, USA
| | - Anil Mathew Tharappel
- Department of Pharmacology and Toxicology, R Ken Coit College of Pharmacy, The University of Arizona, 1703 E Mabel St, Tucson AZ, 85721-0207, USA
| | - Ran Zhang
- Department of Pharmacology and Toxicology, R Ken Coit College of Pharmacy, The University of Arizona, 1703 E Mabel St, Tucson AZ, 85721-0207, USA
| | - Ke Chen
- Department of Pharmacology and Toxicology, R Ken Coit College of Pharmacy, The University of Arizona, 1703 E Mabel St, Tucson AZ, 85721-0207, USA
| | - Y. Geoge Zheng
- Department of Pharmacology and Toxicology, R Ken Coit College of Pharmacy, The University of Arizona, 1703 E Mabel St, Tucson AZ, 85721-0207, USA
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia, 30602, USA
| | - Zhong Li
- Department of Pharmacology and Toxicology, R Ken Coit College of Pharmacy, The University of Arizona, 1703 E Mabel St, Tucson AZ, 85721-0207, USA
| | - Hongmin Li
- Department of Pharmacology and Toxicology, R Ken Coit College of Pharmacy, The University of Arizona, 1703 E Mabel St, Tucson AZ, 85721-0207, USA
- Department of Chemistry and Biochemistry, College of Science & College of Medicine, The University of Arizona, Tucson AZ, 85721, USA
- The BIO5 Institute, The University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
11
|
Hu H, Feng Y, He ML. Targeting Type I Interferon Induction and Signaling: How Zika Virus Escapes from Host Innate Immunity. Int J Biol Sci 2023; 19:3015-3028. [PMID: 37416780 PMCID: PMC10321277 DOI: 10.7150/ijbs.83056] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 05/23/2023] [Indexed: 07/08/2023] Open
Abstract
Zika virus (ZIKV) infection causes neurological disorders and draws great attention. ZIKV infection can elicit a wide range of immune response. Type I interferons (IFNs) as well as its signaling cascade play crucial role in innate immunity against ZIKV infection and in turn ZIKV can antagonize them. ZIKV genome are mainly recognized by Toll-like receptors 3 (TLR3), TLR7/8 and RIG-I-like receptor 1 (RIG-1), which induces the expression of Type I IFNs and interferon-stimulated genes (ISGs). ISGs exert antiviral activity at different stages of the ZIKV life cycle. On the other hand, ZIKV takes multiple strategies to antagonize the Type Ⅰ IFN induction and its signaling pathway to establish a pathogenic infection, especially by using the viral nonstructural (NS) proteins. Most of the NS proteins can directly interact with the factors in the pathways to escape the innate immunity. In addition, structural proteins also participate in the innate immune evasion and activation of antibody-binding of blood dendritic cell antigen 2 (BDCA2) or inflammasome also be used to enhance ZIKV replication. In this review, we summarize the recent findings about the interaction between ZIKV infection and type I IFNs pathways and suggest potential strategies for antiviral drug development.
Collapse
Affiliation(s)
- Huan Hu
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Yaxiu Feng
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Ming-Liang He
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
- City University of Hong Kong Shenzhen Research Institute, Shenzhen 518057, China
| |
Collapse
|
12
|
Chen R, Francese R, Wang N, Li F, Sun X, Xu B, Liu J, Liu Z, Donalisio M, Lembo D, Zhou GC. Exploration of novel hexahydropyrrolo[1,2-e]imidazol-1-one derivatives as antiviral agents against ZIKV and USUV. Eur J Med Chem 2023; 248:115081. [PMID: 36623328 DOI: 10.1016/j.ejmech.2022.115081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/28/2022] [Accepted: 12/29/2022] [Indexed: 01/05/2023]
Abstract
Zika virus (ZIKV) and Usutu virus (USUV) are two emerging flaviviruses mostly transmitted by mosquitos. ZIKV is associated with microcephaly in newborns and the less-known USUV, with its reported neurotropism and its extensive spread in Europe, represents a growing concern for human health. There is still no approved vaccine or specific antiviral against ZIKV and USUV infections. The main goal of this study is to investigate the anti-ZIKV and anti-USUV activity of a new library of compounds and to preliminarily investigate the mechanism of action of the selected hit compounds in vitro. Two potent anti-ZIKV and anti-USUV agents, namely ZDL-115 and ZDL-116, were discovered, both presenting low cytotoxicity, cell-line independent antiviral activity in the low micromolar range and ability of reducing viral progeny production. The analysis of the structure-activity relationship (SAR) revealed that introduction of 2-deoxyribose to 3-arene was fundamental to enhance the solubility and improve the antiviral action. Additionally, we demonstrated that ZDL-115 and ZDL-116 are significantly active against both viruses when added on cells for at least 24 h prior to viral inoculation or immediately post-infection. The docking analysis showed that ZDL-116 could target the host vitamin D receptor (VDR) and viral proteins. Future experiments will be focused on compound modification to discover analogues that are more potent and on the clarification of the mechanism of action and the specific drug target. The discovery and the development of a novel anti-flavivirus drug will have a significant impact in a context where there are no fully effective antiviral drugs or vaccines for most flaviviruses.
Collapse
Affiliation(s)
- Ran Chen
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211816, Jiangsu, China; Xitaihu Lake Industrial College, Nanjing Tech University, Changzhou, 213149, Jiangsu, China
| | - Rachele Francese
- Laboratory of Molecular Virology and Antiviral Research, Department of Clinical and Biological Sciences, University of Turin, S. Luigi Gonzaga Hospital, 10043 Orbassano, Turin, Italy
| | - Na Wang
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Feng Li
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211816, Jiangsu, China
| | - Xia Sun
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211816, Jiangsu, China
| | - Bin Xu
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211816, Jiangsu, China
| | - Jinsong Liu
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Zhuyun Liu
- School of Pharmacy, Taizhou Polytechnic College, Taizhou, 225300, Jiangsu, China
| | - Manuela Donalisio
- Laboratory of Molecular Virology and Antiviral Research, Department of Clinical and Biological Sciences, University of Turin, S. Luigi Gonzaga Hospital, 10043 Orbassano, Turin, Italy
| | - David Lembo
- Laboratory of Molecular Virology and Antiviral Research, Department of Clinical and Biological Sciences, University of Turin, S. Luigi Gonzaga Hospital, 10043 Orbassano, Turin, Italy.
| | - Guo-Chun Zhou
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211816, Jiangsu, China; Xitaihu Lake Industrial College, Nanjing Tech University, Changzhou, 213149, Jiangsu, China.
| |
Collapse
|
13
|
Li R, Niu Z, Liu Y, Bai X, Wang D, Chen C. Crystal structure and cap binding analysis of the methyltransferase of langat virus. Antiviral Res 2022; 208:105459. [PMID: 36347437 DOI: 10.1016/j.antiviral.2022.105459] [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: 07/27/2022] [Revised: 10/29/2022] [Accepted: 10/31/2022] [Indexed: 11/08/2022]
Abstract
Tick-borne encephalitis virus (TBEV) is a major dangerous human pathogen, as TBEV infection can cause serious illness that can lead to irreversible neurological sequelae and even death. Langat virus (LGTV), a member of the tick-borne encephalitis virus (TBEV) serogroup, belongs to the family Flaviviridae, genus Flavivirus. Its nonstructural protein 5 (NS5) protein contains a methyltransferase (MTase) domain that can methylate RNA cap structures, which is critical for viral replication. We determined the structure of LGTV NS5 methyltransferase bound to S-adenosyl-L-homocysteine (SAH) at a 1.70 Å resolution. Sequence analysis and structural comparison of homologous MTases suggests that folds and structures are closely conserved throughout Flavivirus species and play important roles. This study provides the key structural information on LGTV MTase and the foundation for research on antiviral drugs targeting LGTV MTase.
Collapse
Affiliation(s)
- Ruixue Li
- School of Life Sciences, Tianjin University, Tianjin, 300072, China
| | - Ziping Niu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Yujie Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Xue Bai
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Deping Wang
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, the Department of Physiology, Shanxi Medical University, Taiyuan, 030001, China.
| | - Chen Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China.
| |
Collapse
|
14
|
Ferrero DS, Albentosa-González L, Mas A, Verdaguer N. Structure and function of the NS5 methyltransferase domain from Usutu virus. Antiviral Res 2022; 208:105460. [DOI: 10.1016/j.antiviral.2022.105460] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/24/2022] [Accepted: 10/31/2022] [Indexed: 11/09/2022]
|
15
|
Valenzuela-Fernández A, Cabrera-Rodriguez R, Ciuffreda L, Perez-Yanes S, Estevez-Herrera J, González-Montelongo R, Alcoba-Florez J, Trujillo-González R, García-Martínez de Artola D, Gil-Campesino H, Díez-Gil O, Lorenzo-Salazar JM, Flores C, Garcia-Luis J. Nanomaterials to combat SARS-CoV-2: Strategies to prevent, diagnose and treat COVID-19. Front Bioeng Biotechnol 2022; 10:1052436. [PMID: 36507266 PMCID: PMC9732709 DOI: 10.3389/fbioe.2022.1052436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 11/09/2022] [Indexed: 11/26/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and the associated coronavirus disease 2019 (COVID-19), which severely affect the respiratory system and several organs and tissues, and may lead to death, have shown how science can respond when challenged by a global emergency, offering as a response a myriad of rapid technological developments. Development of vaccines at lightning speed is one of them. SARS-CoV-2 outbreaks have stressed healthcare systems, questioning patients care by using standard non-adapted therapies and diagnostic tools. In this scenario, nanotechnology has offered new tools, techniques and opportunities for prevention, for rapid, accurate and sensitive diagnosis and treatment of COVID-19. In this review, we focus on the nanotechnological applications and nano-based materials (i.e., personal protective equipment) to combat SARS-CoV-2 transmission, infection, organ damage and for the development of new tools for virosurveillance, diagnose and immune protection by mRNA and other nano-based vaccines. All the nano-based developed tools have allowed a historical, unprecedented, real time epidemiological surveillance and diagnosis of SARS-CoV-2 infection, at community and international levels. The nano-based technology has help to predict and detect how this Sarbecovirus is mutating and the severity of the associated COVID-19 disease, thereby assisting the administration and public health services to make decisions and measures for preparedness against the emerging variants of SARS-CoV-2 and severe or lethal COVID-19.
Collapse
Affiliation(s)
- Agustín Valenzuela-Fernández
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - Romina Cabrera-Rodriguez
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - Laura Ciuffreda
- Research Unit, Hospital Universitario N. S. de Candelaria, Santa Cruz de Tenerife, Spain
| | - Silvia Perez-Yanes
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - Judith Estevez-Herrera
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | | | - Julia Alcoba-Florez
- Servicio de Microbiología, Hospital Universitario N. S. de Candelaria, Santa Cruz de Tenerife, Spain
| | - Rodrigo Trujillo-González
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
- Departamento de Análisis Matemático, Facultad de Ciencias, Universidad de La Laguna, Santa Cruz de Tenerife, Spain
| | | | - Helena Gil-Campesino
- Servicio de Microbiología, Hospital Universitario N. S. de Candelaria, Santa Cruz de Tenerife, Spain
| | - Oscar Díez-Gil
- Servicio de Microbiología, Hospital Universitario N. S. de Candelaria, Santa Cruz de Tenerife, Spain
| | - José M. Lorenzo-Salazar
- Genomics Division, Instituto Tecnológico y de Energías Renovables, Santa Cruz de Tenerife, Spain
| | - Carlos Flores
- Research Unit, Hospital Universitario N. S. de Candelaria, Santa Cruz de Tenerife, Spain
- Genomics Division, Instituto Tecnológico y de Energías Renovables, Santa Cruz de Tenerife, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Faculty of Health Sciences, University of Fernando Pessoa Canarias, Las Palmas de Gran Canaria, Spain
| | - Jonay Garcia-Luis
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| |
Collapse
|
16
|
Li N, Rana TM. Regulation of antiviral innate immunity by chemical modification of viral RNA. WILEY INTERDISCIPLINARY REVIEWS. RNA 2022; 13:e1720. [PMID: 35150188 PMCID: PMC9786758 DOI: 10.1002/wrna.1720] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 01/08/2022] [Accepted: 01/11/2022] [Indexed: 12/30/2022]
Abstract
More than 100 chemical modifications of RNA, termed the epitranscriptome, have been described, most of which occur in prokaryotic and eukaryotic ribosomal, transfer, and noncoding RNA and eukaryotic messenger RNA. DNA and RNA viruses can modify their RNA either directly via genome-encoded enzymes or by hijacking the host enzymatic machinery. Among the many RNA modifications described to date, four play particularly important roles in promoting viral infection by facilitating viral gene expression and replication and by enabling escape from the host innate immune response. Here, we discuss our current understanding of the mechanisms by which the RNA modifications such as N6 -methyladenosine (m6A), N6 ,2'-O-dimethyladenosine (m6Am), 5-methylcytidine (m5C), N4-acetylcytidine (ac4C), and 2'-O-methylation (Nm) promote viral replication and/or suppress recognition by innate sensors and downstream activation of the host antiviral response. This article is categorized under: RNA in Disease and Development > RNA in Disease RNA Structure and Dynamics > Influence of RNA Structure in Biological Systems RNA Evolution and Genomics > RNA and Ribonucleoprotein Evolution.
Collapse
Affiliation(s)
- Na Li
- Division of Genetics, Department of Pediatrics, Program in ImmunologyInstitute for Genomic MedicineLa JollaCaliforniaUSA
| | - Tariq M. Rana
- Division of Genetics, Department of Pediatrics, Program in ImmunologyInstitute for Genomic MedicineLa JollaCaliforniaUSA
| |
Collapse
|
17
|
Ezeh M, Okonkwo OE, Okpoli IN, Orji CE, Modozie BU, Onyema AC, Ezebuo FC. Chemoinformatic Design and Profiling of Derivatives of Dasabuvir, Efavirenz, and Tipranavir as Potential Inhibitors of Zika Virus RNA-Dependent RNA Polymerase and Methyltransferase. ACS OMEGA 2022; 7:33330-33348. [PMID: 36157724 PMCID: PMC9494688 DOI: 10.1021/acsomega.2c03945] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/24/2022] [Indexed: 05/29/2023]
Abstract
Zika virus (ZIKV) infection is one of the mosquito-borne flaviviruses of human importance with more than 2 million suspected cases and more than 1 million people infected in about 30 countries. There are reported inhibitors of the zika virus replication machinery, but no approved effective antiviral therapy including vaccines directed against the virus for treatment or prevention is currently available. The study investigated the chemoinformatic design and profiling of derivatives of dasabuvir, efavirenz, and tipranavir as potential inhibitors of the zika virus RNA-dependent RNA polymerase (RdRP) and/or methyltransferase (MTase). The three-dimensional (3D) coordinates of dasabuvir, efavirenz, and tipranavir were obtained from the PubChem database, and their respective derivatives were designed with DataWarrior-5.2.1 using an evolutionary algorithm. Derivatives that were not mutagenic, tumorigenic, or irritant were selected; docked into RdRP and MTase; and further subjected to absorption, distribution, metabolism, excretion, and toxicity (ADMET) evaluation with Swiss-ADME and pkCSM web tools. Some of the designed compounds are Lipinski's rule-of-five compliant, with good synthetic accessibilities. Compounds 20d, 21d, 22d, and 1e are nontoxic with the only limitation of CYP1A2, CYP2C19, and/or CYP2C9 inhibition. Replacements of -CH3 and -NH- in the methanesulfonamide moiety of dasabuvir with -OH and -CH2- or -CH2CH2-, respectively, improved the safety/toxicity profile. Hepatotoxicity in 5d, 4d, and 18d is likely due to -NH- in their methanesulfonamide/sulfamic acid moieties. These compounds are potent inhibitors of N-7 and 2'-methylation activities of ZIKV methyltransferase and/or RNA synthesis through interactions with amino acid residues in the priming loop/"N-pocket" in the virus RdRP. Synthesis of these compounds and wet laboratory validation against ZIKV are recommended.
Collapse
Affiliation(s)
- Madeleine
I. Ezeh
- Department
of Pharmaceutical and Medicinal Chemistry, Faculty of Pharmaceutical
Sciences, Nnamdi Azikiwe University, PMB 5025, Awka 420110, Anambra
State, Nigeria
| | - Onyinyechi E. Okonkwo
- Department
of Pharmaceutical and Medicinal Chemistry, Faculty of Pharmaceutical
Sciences, Nnamdi Azikiwe University, PMB 5025, Awka 420110, Anambra
State, Nigeria
| | - Innocent N. Okpoli
- Department
of Pharmaceutical and Medicinal Chemistry, Faculty of Pharmaceutical
Sciences, Nnamdi Azikiwe University, PMB 5025, Awka 420110, Anambra
State, Nigeria
- Drug
Design and Informatics Group, Faculty of Pharmaceutical Sciences, Nnamdi Azikiwe University, PMB 5025, Awka 420110, Anambra State, Nigeria
| | - Chima E. Orji
- Department
of Pharmacology and Toxicology, Faculty of Pharmaceutical Sciences, Nnamdi Azikiwe University, PMB 5025, Awka 420110, Anambra State, Nigeria
| | - Benjamin U. Modozie
- Department
of Pharmaceutical and Medicinal Chemistry, Faculty of Pharmaceutical
Sciences, Nnamdi Azikiwe University, PMB 5025, Awka 420110, Anambra
State, Nigeria
| | - Augustine C. Onyema
- Department
of Biochemistry, Graduate Center, City University
of New York (CUNY), New York, New York 10016, United States
| | - Fortunatus C. Ezebuo
- Department
of Pharmaceutical and Medicinal Chemistry, Faculty of Pharmaceutical
Sciences, Nnamdi Azikiwe University, PMB 5025, Awka 420110, Anambra
State, Nigeria
- Drug
Design and Informatics Group, Faculty of Pharmaceutical Sciences, Nnamdi Azikiwe University, PMB 5025, Awka 420110, Anambra State, Nigeria
| |
Collapse
|
18
|
Wu X, Zhang Y, Wang M, Chen S, Liu M, Zhu D, Zhao X, Wu Y, Yang Q, Zhang S, Huang J, Ou X, Zhang L, Liu Y, Yu Y, Gao Q, Mao S, Sun D, Tian B, Yin Z, Jing B, Cheng A, Jia R. Methyltransferase-Deficient Avian Flaviviruses Are Attenuated Due to Suppression of Viral RNA Translation and Induction of a Higher Innate Immunity. Front Immunol 2021; 12:751688. [PMID: 34691066 PMCID: PMC8526935 DOI: 10.3389/fimmu.2021.751688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 09/20/2021] [Indexed: 11/13/2022] Open
Abstract
The 5' end of the flavivirus genome contains a type 1 cap structure formed by sequential N-7 and 2'-O methylations by viral methyltransferase (MTase). Cap methylation of flavivirus genome is an essential structural modification to ensure the normal proliferation of the virus. Tembusu virus (TMUV) (genus Flavivirus) is a causative agent of duck egg drop syndrome and has zoonotic potential. Here, we identified the in vitro activity of TMUV MTase and determined the effect of K61-D146-K182-E218 enzymatic tetrad on N-7 and 2'-O methylation. The entire K61-D146-K182-E218 motif is essential for 2'-O MTase activity, whereas N-7 MTase activity requires only D146. To investigate its phenotype, the single point mutation (K61A, D146A, K182A or E218A) was introduced into TMUV replicon (pCMV-Rep-NanoLuc) and TMUV infectious cDNA clone (pACYC-TMUV). K-D-K-E mutations reduced the replication ability of replicon. K61A, K182A and E218A viruses were genetically stable, whereas D146A virus was unstable and reverted to WT virus. Mutant viruses were replication and virulence impaired, showing reduced growth and attenuated cytopathic effects and reduced mortality of duck embryos. Molecular mechanism studies showed that the translation efficiency of mutant viruses was inhibited and a higher host innate immunity was induced. Furthermore, we found that the translation inhibition of MTase-deficient viruses was caused by a defect in N-7 methylation, whereas the absence of 2'-O methylation did not affect viral translation. Taken together, our data validate the debilitating mechanism of MTase-deficient avian flavivirus and reveal an important role for cap-methylation in viral translation, proliferation, and escape from innate immunity.
Collapse
Affiliation(s)
- Xuedong Wu
- Research Centre of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yuetian Zhang
- Research Centre of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Mingshu Wang
- Research Centre of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Shun Chen
- Research Centre of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Mafeng Liu
- Research Centre of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Dekang Zhu
- Research Centre of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Xinxin Zhao
- Research Centre of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Ying Wu
- Research Centre of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Qiao Yang
- Research Centre of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Shaqiu Zhang
- Research Centre of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Juan Huang
- Research Centre of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Xumin Ou
- Research Centre of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Ling Zhang
- Research Centre of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yunya Liu
- Research Centre of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yanling Yu
- Research Centre of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Qun Gao
- Research Centre of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Sai Mao
- Research Centre of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Di Sun
- Research Centre of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Bin Tian
- Research Centre of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Zhongqiong Yin
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Bo Jing
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Anchun Cheng
- Research Centre of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Renyong Jia
- Research Centre of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
19
|
Fernandes PO, Chagas MA, Rocha WR, Moraes AH. Non-structural protein 5 (NS5) as a target for antiviral development against established and emergent flaviviruses. Curr Opin Virol 2021; 50:30-39. [PMID: 34340199 DOI: 10.1016/j.coviro.2021.07.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 06/18/2021] [Accepted: 07/02/2021] [Indexed: 11/19/2022]
Abstract
Flaviviruses are among the most critical pathogens in tropical regions and cause a growing number of severe diseases in developing countries. The development of antiviral therapeutics is crucial for managing flavivirus outbreaks. Among the ten proteins encoded in the flavivirus RNA, non-structural protein 5, NS5, is a promising drug target. NS5 plays a fundamental role in flavivirus replication, viral RNA methylation, RNA polymerization, and host immune system evasion. Most of the NS5 inhibitor candidates target NS5 active sites. However, the similarity of NS5 activity sites with human enzymes can cause side effects. Identifying new allosteric sites in NS5 can contribute enormously to antiviral development. The NS5 structural characterization enabled exploring new regions, such as the residues involved in MTase-RdRp interaction, NS5 oligomerization, and NS5 interaction with other viral and host-cell proteins. Targeting NS5 critical interactions might lead to new compounds and overcomes the toxicity of current NS5-inhibitor candidates.
Collapse
Affiliation(s)
- Philipe O Fernandes
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, Brazil
| | - Marcelo A Chagas
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, Brazil
| | - Willian R Rocha
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, Brazil
| | - Adolfo H Moraes
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, Brazil; Department of NMR-based Structural Biology, Max-Planck Institute for Biophysical Chemistry, Am Fassberg 11, D-37077 Göttingen, Germany.
| |
Collapse
|
20
|
Molecular Insights into the Flavivirus Replication Complex. Viruses 2021; 13:v13060956. [PMID: 34064113 PMCID: PMC8224304 DOI: 10.3390/v13060956] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 05/17/2021] [Accepted: 05/17/2021] [Indexed: 12/11/2022] Open
Abstract
Flaviviruses are vector-borne RNA viruses, many of which are clinically relevant human viral pathogens, such as dengue, Zika, Japanese encephalitis, West Nile and yellow fever viruses. Millions of people are infected with these viruses around the world each year. Vaccines are only available for some members of this large virus family, and there are no effective antiviral drugs to treat flavivirus infections. The unmet need for vaccines and therapies against these flaviviral infections drives research towards a better understanding of the epidemiology, biology and immunology of flaviviruses. In this review, we discuss the basic biology of the flavivirus replication process and focus on the molecular aspects of viral genome replication. Within the virus-induced intracellular membranous compartments, flaviviral RNA genome replication takes place, starting from viral poly protein expression and processing to the assembly of the virus RNA replication complex, followed by the delivery of the progeny viral RNA to the viral particle assembly sites. We attempt to update the latest understanding of the key molecular events during this process and highlight knowledge gaps for future studies.
Collapse
|
21
|
Kumar N, Sarma H, Sastry GN. Repurposing of approved drug molecules for viral infectious diseases: a molecular modelling approach. J Biomol Struct Dyn 2021; 40:8056-8072. [PMID: 33810775 DOI: 10.1080/07391102.2021.1905558] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The identification of new viral drugs has become a task of paramount significance due to the frequent occurrence of viral infections and especially during the current pandemic. Despite the recent advancements, the development of antiviral drugs has not made parallel progress. Reduction of time frame and cost of the drug development process is the major advantage of drug repurposing. Therefore, in this study, a drug repurposing strategy using molecular modelling techniques, i.e. biological activity prediction, virtual screening, and molecular dynamics simulation was employed to find promising repurposing candidates for viral infectious diseases. The biological activities of non-redundant (4171) drug molecules were predicted using PASS analysis, and 1401 drug molecules were selected which showed antiviral activities in the analysis. These drug molecules were subjected to virtual screening against the selected non-structural viral proteins. A series of filters, i.e. top 10 drug molecules based on binding affinity, mean value of binding affinity, visual inspection of protein-drug complexes, and number of H-bond between protein and drug molecules were used to narrow down the drug molecules. Molecular dynamics simulation analysis was carried out to validate the intrinsic atomic interactions and binding conformations of protein-drug complexes. The binding free energies of drug molecules were assessed by employing MMPBSA analysis. Finally, nine drug molecules were prioritized, as promising repurposing candidates with the potential to inhibit the selected non-structural viral proteins.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Nandan Kumar
- Centre for Molecular Modelling, CSIR-Indian Institute of Chemical Technology, Hyderabad, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Himakshi Sarma
- Advanced Computation and Data Sciences Division, CSIR-North East Institute of Science and Technology, Jorhat, India
| | - G Narahari Sastry
- Centre for Molecular Modelling, CSIR-Indian Institute of Chemical Technology, Hyderabad, India.,Advanced Computation and Data Sciences Division, CSIR-North East Institute of Science and Technology, Jorhat, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
22
|
Zika Virus Pathogenesis: A Battle for Immune Evasion. Vaccines (Basel) 2021; 9:vaccines9030294. [PMID: 33810028 PMCID: PMC8005041 DOI: 10.3390/vaccines9030294] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 03/12/2021] [Accepted: 03/13/2021] [Indexed: 12/13/2022] Open
Abstract
Zika virus (ZIKV) infection and its associated congenital and other neurological disorders, particularly microcephaly and other fetal developmental abnormalities, constitute a World Health Organization (WHO) Zika Virus Research Agenda within the WHO’s R&D Blueprint for Action to Prevent Epidemics, and continue to be a Public Health Emergency of International Concern (PHEIC) today. ZIKV pathogenicity is initiated by viral infection and propagation across multiple placental and fetal tissue barriers, and is critically strengthened by subverting host immunity. ZIKV immune evasion involves viral non-structural proteins, genomic and non-coding RNA and microRNA (miRNA) to modulate interferon (IFN) signaling and production, interfering with intracellular signal pathways and autophagy, and promoting cellular environment changes together with secretion of cellular components to escape innate and adaptive immunity and further infect privileged immune organs/tissues such as the placenta and eyes. This review includes a description of recent advances in the understanding of the mechanisms underlying ZIKV immune modulation and evasion that strongly condition viral pathogenesis, which would certainly contribute to the development of anti-ZIKV strategies, drugs, and vaccines.
Collapse
|
23
|
Elrefaey AME, Abdelnabi R, Rosales Rosas AL, Wang L, Basu S, Delang L. Understanding the Mechanisms Underlying Host Restriction of Insect-Specific Viruses. Viruses 2020; 12:E964. [PMID: 32878245 PMCID: PMC7552076 DOI: 10.3390/v12090964] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 08/26/2020] [Accepted: 08/27/2020] [Indexed: 12/13/2022] Open
Abstract
Arthropod-borne viruses contribute significantly to global mortality and morbidity in humans and animals. These viruses are mainly transmitted between susceptible vertebrate hosts by hematophagous arthropod vectors, especially mosquitoes. Recently, there has been substantial attention for a novel group of viruses, referred to as insect-specific viruses (ISVs) which are exclusively maintained in mosquito populations. Recent discoveries of novel insect-specific viruses over the past years generated a great interest not only in their potential use as vaccine and diagnostic platforms but also as novel biological control agents due to their ability to modulate arbovirus transmission. While arboviruses infect both vertebrate and invertebrate hosts, the replication of insect-specific viruses is restricted in vertebrates at multiple stages of virus replication. The vertebrate restriction factors include the genetic elements of ISVs (structural and non-structural genes and the untranslated terminal regions), vertebrate host factors (agonists and antagonists), and the temperature-dependent microenvironment. A better understanding of these bottlenecks is thus warranted. In this review, we explore these factors and the complex interplay between ISVs and their hosts contributing to this host restriction phenomenon.
Collapse
Affiliation(s)
| | - Rana Abdelnabi
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, 3000 Leuven, Belgium; (R.A.); (A.L.R.R.); (L.W.)
| | - Ana Lucia Rosales Rosas
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, 3000 Leuven, Belgium; (R.A.); (A.L.R.R.); (L.W.)
| | - Lanjiao Wang
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, 3000 Leuven, Belgium; (R.A.); (A.L.R.R.); (L.W.)
| | - Sanjay Basu
- The Pirbright Institute, Pirbright, Woking GU24 0NF, UK;
| | - Leen Delang
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, 3000 Leuven, Belgium; (R.A.); (A.L.R.R.); (L.W.)
| |
Collapse
|
24
|
Viswanathan T, Arya S, Chan SH, Qi S, Dai N, Misra A, Park JG, Oladunni F, Kovalskyy D, Hromas RA, Martinez-Sobrido L, Gupta YK. Structural basis of RNA cap modification by SARS-CoV-2. Nat Commun 2020; 11:3718. [PMID: 32709886 PMCID: PMC7381649 DOI: 10.1038/s41467-020-17496-8] [Citation(s) in RCA: 166] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 07/04/2020] [Indexed: 02/06/2023] Open
Abstract
The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), the causative agent of COVID-19 illness, has caused millions of infections worldwide. In SARS coronaviruses, the non-structural protein 16 (nsp16), in conjunction with nsp10, methylates the 5'-end of virally encoded mRNAs to mimic cellular mRNAs, thus protecting the virus from host innate immune restriction. We report here the high-resolution structure of a ternary complex of SARS-CoV-2 nsp16 and nsp10 in the presence of cognate RNA substrate analogue and methyl donor, S-adenosyl methionine (SAM). The nsp16/nsp10 heterodimer is captured in the act of 2'-O methylation of the ribose sugar of the first nucleotide of SARS-CoV-2 mRNA. We observe large conformational changes associated with substrate binding as the enzyme transitions from a binary to a ternary state. This induced fit model provides mechanistic insights into the 2'-O methylation of the viral mRNA cap. We also discover a distant (25 Å) ligand-binding site unique to SARS-CoV-2, which can alternatively be targeted, in addition to RNA cap and SAM pockets, for antiviral development.
Collapse
Affiliation(s)
- Thiruselvam Viswanathan
- Greehey Children's Cancer Research Institute, University of Texas Health at San Antonio, 8403 Floyd Curl Drive, San Antonio, TX, 78229, USA
- Department of Biochemistry and Structural Biology, University of Texas Health at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA
| | - Shailee Arya
- Greehey Children's Cancer Research Institute, University of Texas Health at San Antonio, 8403 Floyd Curl Drive, San Antonio, TX, 78229, USA
- Department of Biochemistry and Structural Biology, University of Texas Health at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA
| | - Siu-Hong Chan
- New England Biolabs, 240 County Road, Ipswich, MA, 01938, USA
| | - Shan Qi
- Greehey Children's Cancer Research Institute, University of Texas Health at San Antonio, 8403 Floyd Curl Drive, San Antonio, TX, 78229, USA
- Department of Biochemistry and Structural Biology, University of Texas Health at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA
| | - Nan Dai
- New England Biolabs, 240 County Road, Ipswich, MA, 01938, USA
| | - Anurag Misra
- Greehey Children's Cancer Research Institute, University of Texas Health at San Antonio, 8403 Floyd Curl Drive, San Antonio, TX, 78229, USA
- Department of Biochemistry and Structural Biology, University of Texas Health at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA
| | - Jun-Gyu Park
- Texas Biomedical Research Institute, San Antonio, TX, 78227, USA
| | - Fatai Oladunni
- Texas Biomedical Research Institute, San Antonio, TX, 78227, USA
| | - Dmytro Kovalskyy
- Greehey Children's Cancer Research Institute, University of Texas Health at San Antonio, 8403 Floyd Curl Drive, San Antonio, TX, 78229, USA
| | - Robert A Hromas
- Division of Hematology and Oncology, Department of Medicine, University of Texas Health at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA
| | | | - Yogesh K Gupta
- Greehey Children's Cancer Research Institute, University of Texas Health at San Antonio, 8403 Floyd Curl Drive, San Antonio, TX, 78229, USA.
- Department of Biochemistry and Structural Biology, University of Texas Health at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA.
| |
Collapse
|
25
|
Chagas M, Rocha W, Moraes A. Dynamics and allostery of Zika virus non-structural protein 5 methyltransferase. J Biomol Struct Dyn 2020; 39:5526-5538. [DOI: 10.1080/07391102.2020.1792343] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Marcelo Chagas
- Departamento de Química, ICEx, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Willian Rocha
- Departamento de Química, ICEx, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Adolfo Moraes
- Departamento de Química, ICEx, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Department of NMR-based Structural Biology, Max-Planck Institute for Biophysical Chemistry, Göttingen, Germany
| |
Collapse
|
26
|
Yun SI, Song BH, Woolley ME, Frank JC, Julander JG, Lee YM. Development, Characterization, and Application of Two Reporter-Expressing Recombinant Zika Viruses. Viruses 2020; 12:v12050572. [PMID: 32456014 PMCID: PMC7290298 DOI: 10.3390/v12050572] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 05/16/2020] [Accepted: 05/18/2020] [Indexed: 12/20/2022] Open
Abstract
Zika virus (ZIKV), a mosquito-borne transplacentally transmissible flavivirus, is an enveloped virus with an ~10.8 kb plus-strand RNA genome that can cause neurological disease. To facilitate the identification of potential antivirals, we developed two reporter-expressing ZIKVs, each capable of expressing an enhanced green fluorescent protein or an improved luminescent NanoLuc luciferase. First, a full-length functional ZIKV cDNA clone was engineered as a bacterial artificial chromosome, with each reporter gene under the cap-independent translational control of a cardiovirus-derived internal ribosome entry site inserted downstream of the single open reading frame of the viral genome. Two reporter-expressing ZIKVs were then generated by transfection of ZIKV-susceptible BHK-21 cells with infectious RNAs derived by in vitro run-off transcription from the respective cDNAs. As compared to the parental virus, the two reporter-expressing ZIKVs grew to lower titers with slower growth kinetics and formed smaller foci; however, they displayed a genome-wide viral protein expression profile identical to that of the parental virus, except for two previously unrecognized larger forms of the C and NS1 proteins. We then used the NanoLuc-expressing ZIKV to assess the in vitro antiviral activity of three inhibitors (T-705, NITD-008, and ribavirin). Altogether, our reporter-expressing ZIKVs represent an excellent molecular tool for the discovery of novel antivirals.
Collapse
Affiliation(s)
- Sang-Im Yun
- Department of Animal, Dairy, and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA; (S.-I.Y.); (B.-H.S.); (M.E.W.); (J.C.F.); (J.G.J.)
| | - Byung-Hak Song
- Department of Animal, Dairy, and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA; (S.-I.Y.); (B.-H.S.); (M.E.W.); (J.C.F.); (J.G.J.)
| | - Michael E. Woolley
- Department of Animal, Dairy, and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA; (S.-I.Y.); (B.-H.S.); (M.E.W.); (J.C.F.); (J.G.J.)
| | - Jordan C. Frank
- Department of Animal, Dairy, and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA; (S.-I.Y.); (B.-H.S.); (M.E.W.); (J.C.F.); (J.G.J.)
| | - Justin G. Julander
- Department of Animal, Dairy, and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA; (S.-I.Y.); (B.-H.S.); (M.E.W.); (J.C.F.); (J.G.J.)
- Institute for Antiviral Research, Utah State University, Logan, UT 84322, USA
| | - Young-Min Lee
- Department of Animal, Dairy, and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA; (S.-I.Y.); (B.-H.S.); (M.E.W.); (J.C.F.); (J.G.J.)
- Veterinary Diagnostics and Infectious Diseases, Utah Science Technology and Research, Utah State University, Logan, UT 84341, USA
- Correspondence: ; Tel.: +1-435-797-9667
| |
Collapse
|
27
|
Viswanathan T, Arya S, Chan SH, Qi S, Dai N, Hromas RA, Park JG, Oladunni F, Martinez-Sobrido L, Gupta YK. Structural Basis of RNA Cap Modification by SARS-CoV-2 Coronavirus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.04.26.061705. [PMID: 32511383 PMCID: PMC7263512 DOI: 10.1101/2020.04.26.061705] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The novel severe acute respiratory syndrome coronoavirus-2 (SARS-CoV-2), the causative agent of COVID-19 illness, has caused over 2 million infections worldwide in four months. In SARS coronaviruses, the non-structural protein 16 (nsp16) methylates the 5'-end of virally encoded mRNAs to mimic cellular mRNAs, thus protecting the virus from host innate immune restriction. We report here the high-resolution structure of a ternary complex of full-length nsp16 and nsp10 of SARS-CoV-2 in the presence of cognate RNA substrate and a methyl donor, S-adenosyl methionine. The nsp16/nsp10 heterodimer was captured in the act of 2'-O methylation of the ribose sugar of the first nucleotide of SARS-CoV-2 mRNA. We reveal large conformational changes associated with substrate binding as the enzyme transitions from a binary to a ternary state. This structure provides new mechanistic insights into the 2'-O methylation of the viral mRNA cap. We also discovered a distantly located ligand-binding site unique to SARS-CoV-2 that may serve as an alternative target site for antiviral development.
Collapse
Affiliation(s)
- Thiruselvam Viswanathan
- Greehey Children’s Cancer Research Institute, University of Texas Health San Antonio, 8403 Floyd Curl Drive, San Antonio, TX 78229, USA
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, 8403 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Shailee Arya
- Greehey Children’s Cancer Research Institute, University of Texas Health San Antonio, 8403 Floyd Curl Drive, San Antonio, TX 78229, USA
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, 8403 Floyd Curl Drive, San Antonio, TX 78229, USA
| | | | - Shan Qi
- Greehey Children’s Cancer Research Institute, University of Texas Health San Antonio, 8403 Floyd Curl Drive, San Antonio, TX 78229, USA
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, 8403 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Nan Dai
- New England Biolabs, Ipswich, MA
| | - Robert A. Hromas
- Division of Hematology and Oncology, Department of Medicine, University of Texas Health San Antonio, 8403 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Jun-Gyu Park
- Texas Biomedical Research Institute, San Antonio, TX
| | | | | | - Yogesh K. Gupta
- Greehey Children’s Cancer Research Institute, University of Texas Health San Antonio, 8403 Floyd Curl Drive, San Antonio, TX 78229, USA
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, 8403 Floyd Curl Drive, San Antonio, TX 78229, USA
- Lead contact
| |
Collapse
|
28
|
Afaq S, Atiya A, Malik A, Alwabli AS, Alzahrani DA, Al-Solami HM, Alzahrani O, Alam Q, Kamal MA, Abulfaraj AA, Alhebshi AM, Tarique M. Analysis of methyltransferase (MTase) domain from Zika virus (ZIKV). Bioinformation 2020; 16:229-235. [PMID: 32308265 PMCID: PMC7147495 DOI: 10.6026/97320630016229] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 02/15/2020] [Accepted: 02/20/2020] [Indexed: 11/23/2022] Open
Abstract
A comprehensive analysis of methyltransferase (MTase) from Zika virus (ZIKV) is of interest in the development of drugs and biomarkers in the combat and care of ZIKA fever with impulsive joint pain and conjunctivitis. MTase sequence is homologous in several viral species. We analyzed the MTase domain from ZIKV using Bioinformatics tools such as SMART, PROSITE, PFAM, PANTHER, and InterProScan to glean insights on the sequence to structure to function data. We document inclusive information on MTase from ZIKV for application in the design of drugs and biomarkers to fight against the disease.
Collapse
Affiliation(s)
- Sarah Afaq
- Department of Clinical Biochemistry, College of Medicine, King Khalid University, Abha, Kingdom of Saudi Arabia
| | - Akhtar Atiya
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha, Kingdom of Saudi Arabia
| | - Arshi Malik
- Department of Clinical Biochemistry, College of Medicine, King Khalid University, Abha, Kingdom of Saudi Arabia
| | - Afaf S Alwabli
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Kingdom of Saudi Arabia
| | - Dhafer A Alzahrani
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Kingdom of Saudi Arabia
| | - Habeeb M Al-Solami
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Kingdom of Saudi Arabia
| | - Othman Alzahrani
- Department of Biology, Faculty of Science, University of Tabuk, Tabuk, Kingdom of Saudi Arabia
| | - Qamre Alam
- Medical Genomics Research Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Kingdom of Saudi Arabia
| | - Mohammad Azhar Kamal
- University of Jeddah, Faculty of Science, Department of Biochemistry, Jeddah, Kingdom of Saudi Arabia
- University of Jeddah Center for Science and Medical Research (UJC-SMR), Jeddah, Kingdom of Saudi Arabia
| | - Aala A Abulfaraj
- Department of Biological Sciences, College of Sciences and Arts-Rabigh Campus, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Alawiah M Alhebshi
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Kingdom of Saudi Arabia
| | - Mohammed Tarique
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi-110025, India
| |
Collapse
|
29
|
Kesari AS, Heintz VJ, Poudyal S, Miller AS, Kuhn RJ, LaCount DJ. Zika virus NS5 localizes at centrosomes during cell division. Virology 2020; 541:52-62. [PMID: 32056715 DOI: 10.1016/j.virol.2019.11.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Revised: 11/04/2019] [Accepted: 11/18/2019] [Indexed: 12/15/2022]
Abstract
Zika virus (ZIKV) nonstructural protein 5 (NS5) plays a critical role in viral RNA replication and mediates key virus-host cell interactions. As with other flavivirus NS5 proteins, ZIKV NS5 is primarily found in the nucleus. We previously reported that the NS5 protein of dengue virus, another flavivirus, localized to centrosomes during cell division. Here we show that ZIKV NS5 also relocalizes from the nucleus to centrosomes during mitosis. In infected cells with supernumerary centrosomes, NS5 was present at all centrosomes. Transient expression of NS5 in uninfected cells confirmed that centrosomal localization was independent of other viral proteins. Live-cell imaging demonstrated that NS5-GFP accumulated at centrosomes shortly after break down of nuclear membrane and remained there through mitosis. Cells expressing NS5-GFP took longer to complete mitosis than control cells. Finally, an analysis of ZIKV NS5 binding partners revealed several centrosomal proteins, providing potential direct links between NS5 and centrosomes.
Collapse
Affiliation(s)
- Aditi S Kesari
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, USA; Department of Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, USA; Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN, 47907, USA
| | - Veronica J Heintz
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, USA; Department of Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, USA; Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN, 47907, USA
| | - Shishir Poudyal
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA; Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN, 47907, USA
| | - Andrew S Miller
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA; Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN, 47907, USA
| | - Richard J Kuhn
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA; Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN, 47907, USA
| | - Douglas J LaCount
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, USA; Department of Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, USA; Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN, 47907, USA.
| |
Collapse
|
30
|
Santos FRS, Lima WG, Maia EHB, Assis LC, Davyt D, Taranto AG, Ferreira JMS. Identification of a Potential Zika Virus Inhibitor Targeting NS5 Methyltransferase Using Virtual Screening and Molecular Dynamics Simulations. J Chem Inf Model 2020; 60:562-568. [PMID: 31985225 DOI: 10.1021/acs.jcim.9b00809] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The NS5 methyltransferase (MTase) has been reported as an attractive molecular target for antivirals discovery against the Zika virus (ZIKV). Here, we report structure-based virtual screening of 42 390 structures from the Development Therapeutics Program (DTP) AIDS Antiviral Screen Database. Among the docked compounds, ZINC1652386 stood out due to its high affinity for MTase in comparison to the cocrystallized ligand MS2042, which interacts with the Asp146 residue in the MTase binding site by hydrogen bonding. Subsequent molecular dynamics simulations predicted that this compound forms a stable complex with MTase within 50 ns. Thus, ZINC1652386 may represent a promising ZIKV methyltransferase inhibitor.
Collapse
Affiliation(s)
- Felipe R S Santos
- Laboratório de Microbiologia Médica , Universidade Federal de São João Del-Rei , Divinópolis , Minas Gerais , Brasil
| | - William G Lima
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia , Universidade Federal de Minas Gerais , Belo Horizonte , Minas Gerais , Brasil
| | - Eduardo H B Maia
- Laboratório de Quı́mica Farmacêutica Medicinal , Universidade Federal de São João Del-Rei , Minas Gerais , Divinópolis , Brasil
| | - Letícia C Assis
- Laboratório de Quı́mica Farmacêutica Medicinal , Universidade Federal de São João Del-Rei , Minas Gerais , Divinópolis , Brasil
| | - Danilo Davyt
- Departamento de Quı́mica Orgánica , Universidad de la República , Montevideo , Uruguay
| | - Alex Gutterres Taranto
- Laboratório de Quı́mica Farmacêutica Medicinal , Universidade Federal de São João Del-Rei , Minas Gerais , Divinópolis , Brasil
| | - Jaqueline M S Ferreira
- Laboratório de Microbiologia Médica , Universidade Federal de São João Del-Rei , Divinópolis , Minas Gerais , Brasil
| |
Collapse
|
31
|
Pramanik S, Thaker M, Perumal AG, Ekambaram R, Poondla N, Schmidt M, Kim PS, Kutzner A, Heese K. Proteomic Atomics Reveals a Distinctive Uracil-5-Methyltransferase. Mol Inform 2020; 39:e1900135. [PMID: 31943843 DOI: 10.1002/minf.201900135] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 01/14/2020] [Indexed: 12/20/2022]
Abstract
Carbon (C), hydrogen (H), nitrogen (N), oxygen (O), and sulfur (S) atoms intrigue as they are the foundation for amino acid (AA) composition and the folding and functions of proteins and thus define and control the survival of a cell, the smallest unit of life. Here, we calculated the proteomic atom distribution in >1500 randomly selected species across the entire current phylogenetic tree and identified uracil-5-methyltransferase (U5MTase) of the protozoan parasite Plasmodium falciparum (Pf, strain Pf3D7), with a distinct atom and AA distribution pattern. We determined its apicoplast location and in silico 3D protein structure to refocus attention exclusively on U5MTase with tremendous potential for therapeutic intervention in malaria. Around 300 million clinical cases of malaria occur each year in tropical and subtropical regions of the world, resulting in over one million deaths annually, placing malaria among the most serious infectious diseases. Genomic and proteomic research of the clades of parasites containing Pf is progressing slowly and the functions of most of the ∼5300 genes are still unknown. We applied a 'bottom-up' comparative proteomic atomics analysis across the phylogenetic tree to visualize a protein molecule on its actual basis - i. e., its atomic level. We identified a protruding Pf3D7-specific U5MTase, determined its 3D protein structure, and identified potential inhibitory drug molecules through in silico drug screening that might serve as possible remedies for the treatment of malaria. Besides, this atomic-based proteome map provides a unique approach for the identification of parasite-specific proteins that could be considered as novel therapeutic targets.
Collapse
Affiliation(s)
- Subrata Pramanik
- Graduate School of Biomedical Science and Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 1, 33-791, Republic of Korea.,Institute of Biotechnology, RWTH Aachen University, Worringerweg 3, Aachen, 52074, Germany
| | - Manisha Thaker
- Department of Medicine, Harvard Medical School, 3 Blackfan Circle, Boston, MA 02115, USA
| | - Ananda Gopu Perumal
- Technology Business Incubator, Periyar Maniammai Institute of Science and Technology, Vallam, Thanjavur, 613403, Tamil Nadu, India
| | - Rajasekaran Ekambaram
- Department of Chemistry, V.S.B. Engineering College, 67 Covai Road, Karudayampalayam Post, Karur, 639111, Tamil Nadu, India
| | - Naresh Poondla
- Graduate School of Biomedical Science and Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 1, 33-791, Republic of Korea
| | - Markus Schmidt
- Department of Information Systems, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 133-791, Republic of Korea
| | - Pok-Son Kim
- Department of Mathematics, Kookmin University, 77 Jeongneung-ro, Seongbuk-gu, Seoul 1, 36-702, Republic of Korea
| | - Arne Kutzner
- Department of Information Systems, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 133-791, Republic of Korea
| | - Klaus Heese
- Graduate School of Biomedical Science and Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 1, 33-791, Republic of Korea
| |
Collapse
|
32
|
Silva S, Shimizu JF, Oliveira DMD, Assis LRD, Bittar C, Mottin M, Sousa BKDP, Mesquita NCDMR, Regasini LO, Rahal P, Oliva G, Perryman AL, Ekins S, Andrade CH, Goulart LR, Sabino-Silva R, Merits A, Harris M, Jardim ACG. A diarylamine derived from anthranilic acid inhibits ZIKV replication. Sci Rep 2019; 9:17703. [PMID: 31776405 PMCID: PMC6881388 DOI: 10.1038/s41598-019-54169-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 10/29/2019] [Indexed: 12/13/2022] Open
Abstract
Zika virus (ZIKV) is a mosquito-transmitted Flavivirus, originally identified in Uganda in 1947 and recently associated with a large outbreak in South America. Despite extensive efforts there are currently no approved antiviral compounds for treatment of ZIKV infection. Here we describe the antiviral activity of diarylamines derived from anthranilic acid (FAMs) against ZIKV. A synthetic FAM (E3) demonstrated anti-ZIKV potential by reducing viral replication up to 86%. We analyzed the possible mechanisms of action of FAM E3 by evaluating the intercalation of this compound into the viral dsRNA and its interaction with the RNA polymerase of bacteriophage SP6. However, FAM E3 did not act by these mechanisms. In silico results predicted that FAM E3 might bind to the ZIKV NS3 helicase suggesting that this protein could be one possible target of this compound. To test this, the thermal stability and the ATPase activity of the ZIKV NS3 helicase domain (NS3Hel) were investigated in vitro and we demonstrated that FAM E3 could indeed bind to and stabilize NS3Hel.
Collapse
Affiliation(s)
- Suely Silva
- Laboratory of Virology, Institute of Biomedical Science, ICBIM, Federal University of Uberlandia, Uberlândia, MG, Brazil
- São Paulo State University, IBILCE, S. José do Rio Preto, SP, Brazil
| | - Jacqueline Farinha Shimizu
- Laboratory of Virology, Institute of Biomedical Science, ICBIM, Federal University of Uberlandia, Uberlândia, MG, Brazil
- São Paulo State University, IBILCE, S. José do Rio Preto, SP, Brazil
| | - Débora Moraes de Oliveira
- Laboratory of Virology, Institute of Biomedical Science, ICBIM, Federal University of Uberlandia, Uberlândia, MG, Brazil
| | - Leticia Ribeiro de Assis
- Laboratory of Antibiotics and Chemotherapeutics, São Paulo State University, IBILCE, S. José do Rio Preto, SP, Brazil
| | - Cintia Bittar
- São Paulo State University, IBILCE, S. José do Rio Preto, SP, Brazil
| | - Melina Mottin
- LabMol - Laboratory of Molecular Modeling and Drug Design, Faculdade de Farmácia, Universidade Federal de Goias, Goiânia, GO, 74605- 170, Brazil
| | - Bruna Katiele de Paula Sousa
- LabMol - Laboratory of Molecular Modeling and Drug Design, Faculdade de Farmácia, Universidade Federal de Goias, Goiânia, GO, 74605- 170, Brazil
| | | | - Luis Octávio Regasini
- Laboratory of Antibiotics and Chemotherapeutics, São Paulo State University, IBILCE, S. José do Rio Preto, SP, Brazil
| | - Paula Rahal
- São Paulo State University, IBILCE, S. José do Rio Preto, SP, Brazil
| | - Glaucius Oliva
- Institute of Physics of São Carlos, University of São Paulo, São Carlos, Brazil
| | - Alexander Luke Perryman
- Department of Pharmacology, Physiology and Neuroscience, Rutgers University-New Jersey Medical School, Newark, NJ 07103, United States
- Repare Therapeutics, 7210 Rue Frederick-Banting, Suite 100, Montreal, QC, H4S 2A1, Canada
| | - Sean Ekins
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, 27606, United States
| | - Carolina Horta Andrade
- LabMol - Laboratory of Molecular Modeling and Drug Design, Faculdade de Farmácia, Universidade Federal de Goias, Goiânia, GO, 74605- 170, Brazil
| | - Luiz Ricardo Goulart
- Laboratory of Nanobiotechnology, Federal University of Uberlandia, Uberlândia, MG, Brasil
| | - Robinson Sabino-Silva
- Integrative Physiology and Salivary Nanobiotechnology Group, Federal University of Uberlandia, Uberlândia, MG, Brasil
| | - Andres Merits
- Institute of Technology, University of Tartu, Nooruse 1, 50411, Tartu, Estonia
| | - 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, United Kingdom
| | - Ana Carolina Gomes Jardim
- Laboratory of Virology, Institute of Biomedical Science, ICBIM, Federal University of Uberlandia, Uberlândia, MG, Brazil.
- São Paulo State University, IBILCE, S. José do Rio Preto, SP, Brazil.
| |
Collapse
|
33
|
Liu X, Zhao Y, Zhang JZ. Molecular mechanism of ligand bindings to Zika virus at SAM site. Chem Phys Lett 2019. [DOI: 10.1016/j.cplett.2019.136771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
34
|
Molecular signatures associated with prostate cancer cell line (PC-3) exposure to inactivated Zika virus. Sci Rep 2019; 9:15351. [PMID: 31653965 PMCID: PMC6814752 DOI: 10.1038/s41598-019-51954-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 10/02/2019] [Indexed: 02/07/2023] Open
Abstract
The recent outbreak of Zika virus (ZIKV) infection associated with microcephaly cases has elicited much research on the mechanisms involved in ZIKV-host cell interactions. It has been described that Zika virus impairs cell growth, raising a hypothesis about its oncolytic potential against cancer cells. ZIKV tumor cell growth inhibition was later confirmed for glioblastoma. It was also demonstrated that an inactivated ZIKV prototype (ZVp) based on bacterial outer membrane vesicles has antiproliferative activity upon other cancer cell lines, such as PC-3 prostate cancer cell. This study aims at understanding the pathways that might be involved with the antiproliferative effect of Zika virus against prostate cancer cells. A metabolomic approach based on high-resolution mass spectrometry analysis led to the identification of 21 statistically relevant markers of PC-3 cells treated with ZVp. The markers were associated with metabolic alterations that trigger lipid remodeling, endoplasmic reticulum stress, inflammatory mediators, as well as disrupted porphyrin and folate metabolism. These findings highlight molecular signatures of ZVp-induced response that may be involved on cellular pathways triggered by its antiproliferative effect. To our knowledge, this is the first reported metabolomic assessment of ZIKV effect on prostate cancer cells, a promising topic for further research.
Collapse
|
35
|
Sariyer IK, Gordon J, Burdo TH, Wollebo HS, Gianti E, Donadoni M, Bellizzi A, Cicalese S, Loomis R, Robinson JA, Carnevale V, Steiner J, Ozdener MH, Miller AD, Amini S, Klein ML, Khalili K. Suppression of Zika Virus Infection in the Brain by the Antiretroviral Drug Rilpivirine. Mol Ther 2019; 27:2067-2079. [PMID: 31653397 DOI: 10.1016/j.ymthe.2019.10.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 10/03/2019] [Accepted: 10/04/2019] [Indexed: 01/02/2023] Open
Abstract
Zika virus (ZIKV) infection is associated with microcephaly in neonates and Guillain-Barré syndrome in adults. ZIKV produces a class of nonstructural (NS) regulatory proteins that play a critical role in viral transcription and replication, including NS5, which possesses RNA-dependent RNA polymerase (RdRp) activity. Here we demonstrate that rilpivirine (RPV), a non-nucleoside reverse transcriptase inhibitor (NNRTI) used in the treatment of HIV-1 infection, inhibits the enzymatic activity of NS5 and suppresses ZIKV infection and replication in primary human astrocytes. Similarly, other members of the NNRTI family, including etravirine and efavirenz, showed inhibitory effects on viral infection of brain cells. Site-directed mutagenesis identified 14 amino acid residues within the NS5 RdRp domain (AA265-903), which are important for the RPV interaction and the inhibition of NS5 polymerase activity. Administration of RPV to ZIKV-infected interferon-alpha/beta receptor (IFN-A/R) knockout mice improved the clinical outcome and prevented ZIKV-induced mortality. Histopathological examination of the brains from infected animals revealed that RPV reduced ZIKV RNA levels in the hippocampus, frontal cortex, thalamus, and cerebellum. Repurposing of NNRTIs, such as RPV, for the inhibition of ZIKV replication offers a possible therapeutic strategy for the prevention and treatment of ZIKV-associated disease.
Collapse
Affiliation(s)
- Ilker Kudret Sariyer
- Department of Neuroscience, Center for Neurovirology, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, USA
| | - Jennifer Gordon
- Department of Neuroscience, Center for Neurovirology, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, USA
| | - Tricia H Burdo
- Department of Neuroscience, Center for Neurovirology, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, USA
| | - Hassen S Wollebo
- Department of Neuroscience, Center for Neurovirology, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, USA
| | - Eleonora Gianti
- Department of Chemistry, Institute for Computational Molecular Science, College of Science and Technology, Temple University, 1901 N. 12(th) Street, Philadelphia, PA 19122, USA
| | - Martina Donadoni
- Department of Neuroscience, Center for Neurovirology, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, USA
| | - Anna Bellizzi
- Department of Neuroscience, Center for Neurovirology, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, USA
| | - Stephanie Cicalese
- Department of Neuroscience, Center for Neurovirology, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, USA
| | - Regina Loomis
- Department of Neuroscience, Center for Neurovirology, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, USA
| | - Jake A Robinson
- Department of Neuroscience, Center for Neurovirology, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, USA
| | - Vincenzo Carnevale
- Department of Chemistry, Institute for Computational Molecular Science, College of Science and Technology, Temple University, 1901 N. 12(th) Street, Philadelphia, PA 19122, USA
| | - Joseph Steiner
- Translational Neuroscience Center, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA
| | - Mehmet H Ozdener
- Monell Chemical Senses Center, 3500 Market Street, Philadelphia, PA 19104, USA
| | - Andrew D Miller
- College of Veterinary Medicine, Cornell University, T5-006A Veterinary Research Tower, Ithaca, NY 14853, USA
| | - Shohreh Amini
- Department of Neuroscience, Center for Neurovirology, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, USA
| | - Michael L Klein
- Department of Chemistry, Institute for Computational Molecular Science, College of Science and Technology, Temple University, 1901 N. 12(th) Street, Philadelphia, PA 19122, USA
| | - Kamel Khalili
- Department of Neuroscience, Center for Neurovirology, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, USA.
| |
Collapse
|
36
|
Elshahawi H, Syed Hassan S, Balasubramaniam V. Importance of Zika Virus NS5 Protein for Viral Replication. Pathogens 2019; 8:pathogens8040169. [PMID: 31574966 PMCID: PMC6963216 DOI: 10.3390/pathogens8040169] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 09/27/2019] [Accepted: 09/27/2019] [Indexed: 01/19/2023] Open
Abstract
Zika virus is the latest addition to an ever-growing list of arboviruses that are causing outbreaks with serious consequences. A few mild cases were recorded between 1960 and 1980 until the first major outbreak in 2007 on Yap Island. This was followed by more severe outbreaks in French Polynesia (2013) and Brazil (2015), which significantly increased both Guillain-Barre syndrome and microcephaly cases. No current vaccines or treatments are available, however, recent studies have taken interest in the NS5 protein which encodes both the viral methyltransferase and RNA-dependent RNA polymerase. This makes it important for viral replication alongside other important functions such as inhibiting the innate immune system thus ensuring virus survival and replication. Structural studies can help design inhibitors, while biochemical studies can help understand the various mechanisms utilized by NS5 thus counteracting them might inhibit or abolish the viral infection. Drug repurposing targeting the NS5 protein has also proven to be an effective tool since hundreds of thousands of compounds can be screened therefore saving time and resources, moreover information on these compounds might already be available especially if they are used to treat other ailments.
Collapse
Affiliation(s)
- Hesham Elshahawi
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Subang Jaya 47500, Selangor, Malaysia.
| | - Sharifah Syed Hassan
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Subang Jaya 47500, Selangor, Malaysia.
- Tropical Medicine & Biology Multidisciplinary Platform, Monash University Malaysia, Jalan Lagoon Selatan, Subang Jaya 47500, Selangor, Malaysia.
| | - Vinod Balasubramaniam
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Subang Jaya 47500, Selangor, Malaysia.
| |
Collapse
|
37
|
Valente AP, Moraes AH. Zika virus proteins at an atomic scale: how does structural biology help us to understand and develop vaccines and drugs against Zika virus infection? J Venom Anim Toxins Incl Trop Dis 2019; 25:e20190013. [PMID: 31523227 PMCID: PMC6727858 DOI: 10.1590/1678-9199-jvatitd-2019-0013] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
In Brazil and in other tropical areas Zika virus infection was directly associated with clinical complications as microcephaly in newborn children whose mothers were infected during pregnancy and the Guillain-Barré syndrome in adults. Recently, research has been focused on developing new vaccines and drug candidates against Zika virus infection since none of those are available. In order to contribute to vaccine and drug development efforts, it becomes important the understanding of the molecular basis of the Zika virus recognition, infection and blockade. To this purpose, it is essential the structural determination of the Zika virus proteins. The genome sequencing of the Zika virus identified ten proteins, being three structural (protein E, protein C and protein prM) and seven non-structural proteins (NS1, NS2A, NS2B, NS3, NS4A, NS4B and NS5). Together, these proteins are the main targets for drugs and antibody recognition. Here we examine new discoveries on high-resolution structural biology of Zika virus, observing the interactions and functions of its proteins identified via state-of-art structural methodologies as X-ray crystallography, nuclear magnetic resonance spectroscopy and cryogenic electronic microscopy. The aim of the present study is to contribute to the understanding of the structural basis of Zika virus infection at an atomic level and to point out similarities and differences to others flaviviruses.
Collapse
Affiliation(s)
- Ana Paula Valente
- National Center of Magnetic Resonance, Leopoldo de Meis Institute of Medical Biochemistry, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | - Adolfo Henrique Moraes
- Department of Chemistry, Institute of Exact Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| |
Collapse
|
38
|
Palmatine inhibits Zika virus infection by disrupting virus binding, entry, and stability. Biochem Biophys Res Commun 2019; 518:732-738. [PMID: 31472967 DOI: 10.1016/j.bbrc.2019.08.120] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 08/23/2019] [Indexed: 12/15/2022]
Abstract
Zika virus (ZIKV) is an emerging vector-borne virus that is associated with severe congenital cerebral anomalies in fetuses and paralytic Guillain-Barré syndrome in adults. In the current global health crisis, there are no vaccines or therapeutics available for the treatment of ZIKV infection. In the present study, we evaluated the efficacy of the protoberberine alkaloid, palmatine, in inhibiting ZIKV and Japanese encephalitis virus (JEV). Palmatine was shown to bind to restricted viruses, inhibit ZIKV infection, and resist ZIKV-induced cytopathic effects. Palmatine was also shown to inhibit JEV infection in multiple cell lines. Overall, the effects of palmatine in disrupting ZIKV binding, entry, and stability indicate that this small molecule would be a good starting point for the development of treatments aimed at inhibiting ZIKV infection.
Collapse
|
39
|
Hu Y, Sun L. Systematic Analysis of Structure Similarity between Zika Virus and Other Flaviviruses. ACS Infect Dis 2019; 5:1070-1080. [PMID: 31038920 DOI: 10.1021/acsinfecdis.9b00047] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Zika virus (ZIKV) infection has caused global concern because of its association with fetal microcephaly and serious neurological complications in adults since 2016. Currently, no specific anti-ZIKV therapy is available to control ZIKV infection. During the last couple of years, the intensive investigation of ZIKV structure has provided significant information for structure-based vaccine and drug design. In this review, we summarized the research progress on the structures of ZIKV and its component proteins. We analyzed the structure identity and the differences between ZIKV and other flaviviruses. This information is crucial to guiding structure-based anti-ZIKV inhibitors and vaccine discovery.
Collapse
Affiliation(s)
- Yuxia Hu
- The Fifth People’s Hospital of Shanghai and Institutes of Biomedical Sciences, Fudan University, 131 Dongan Road, Shanghai 20032, China
| | - Lei Sun
- The Fifth People’s Hospital of Shanghai and Institutes of Biomedical Sciences, Fudan University, 131 Dongan Road, Shanghai 20032, China
| |
Collapse
|
40
|
Saw WG, Chan KWK, Vasudevan SG, Grüber G. Zika virus nonstructural protein 5 residue R681 is critical for dimer formation and enzymatic activity. FEBS Lett 2019; 593:1272-1291. [PMID: 31090058 DOI: 10.1002/1873-3468.13437] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/08/2019] [Accepted: 05/09/2019] [Indexed: 11/07/2022]
Abstract
Zika virus (ZIKV) relies on its nonstructural protein 5 (NS5) for capping and synthesis of the viral RNA. Recent small-angle X-ray scattering (SAXS) data of recombinant ZIKV NS5 protein showed that it is dimeric in solution. Here, we present insights into the critical residues responsible for its dimer formation. SAXS studies of the engineered ZIKV NS5 mutants revealed that R681A mutation on NS5 (NS5R681A ) disrupts the dimer formation and affects its RNA-dependent RNA polymerase activity as well as the subcellular localization of NS5R681A in mammalian cells. The critical residues involved in the dimer arrangement of ZIKV NS5 are discussed, and the data provide further insights into the diversity of flaviviral NS5 proteins in terms of their propensity for oligomerization.
Collapse
Affiliation(s)
- Wuan-Geok Saw
- Nanyang Technological University, School of Biological Sciences, Singapore
| | - Kitti Wing-Ki Chan
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore.,Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Subhash G Vasudevan
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore.,Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Gerhard Grüber
- Nanyang Technological University, School of Biological Sciences, Singapore
| |
Collapse
|
41
|
Discovery and Computational Analyses of Novel Small Molecule Zika Virus Inhibitors. Molecules 2019; 24:molecules24081465. [PMID: 31013906 PMCID: PMC6514826 DOI: 10.3390/molecules24081465] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 04/04/2019] [Accepted: 04/12/2019] [Indexed: 01/19/2023] Open
Abstract
Zika virus (ZIKV), one of the flaviviruses, has attracted worldwide attention since its large epidemics around Brazil. Association of ZIKV infection with microcephaly and neurological problems such as Guillain–Barré syndrome has prompted intensive pathological investigations. However, there is still a long way to go on the discovery of effective anti-ZIKV therapeutics. In this study, an in silico screening of the National Cancer Institute (NCI) diversity set based on ZIKV NS3 helicase was performed using a molecular docking approach. Selected compounds with drug-like properties were subjected to cell-based antiviral assays resulting in the identification of two novel lead compounds (named Compounds 1 and 2). They inhibited ZIKV infection with IC50 values at the micro-molar level (8.5 μM and 15.2 μM, respectively). Binding mode analysis, absolute binding free energy calculation, and structure–activity relationship studies of these two compounds revealed their possible interactions with ZIKV NS3 helicase, suggesting a mechanistic basis for further optimization. These two novel small molecules may represent new leads for the development of inhibitory drugs against ZIKV.
Collapse
|
42
|
Ferrero DS, Ruiz-Arroyo VM, Soler N, Usón I, Guarné A, Verdaguer N. Supramolecular arrangement of the full-length Zika virus NS5. PLoS Pathog 2019; 15:e1007656. [PMID: 30951555 PMCID: PMC6469808 DOI: 10.1371/journal.ppat.1007656] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 04/17/2019] [Accepted: 02/23/2019] [Indexed: 12/12/2022] Open
Abstract
Zika virus (ZIKV), a member of the Flaviviridae family, has emerged as a major public health threat, since ZIKV infection has been connected to microcephaly and other neurological disorders. Flavivirus genome replication is driven by NS5, an RNA-dependent RNA polymerase (RdRP) that also contains a N-terminal methyltransferase domain essential for viral mRNA capping. Given its crucial roles, ZIKV NS5 has become an attractive antiviral target. Here, we have used integrated structural biology approaches to characterize the supramolecular arrangement of the full-length ZIKV NS5, highlighting the assembly and interfaces between NS5 monomers within a dimeric structure, as well as the dimer-dimer interactions to form higher order fibril-like structures. The relative orientation of each monomer within the dimer provides a model to explain the coordination between MTase and RdRP domains across neighboring NS5 molecules and mutational studies underscore the crucial role of the MTase residues Y25, K28 and K29 in NS5 dimerization. The basic residue K28 also participates in GTP binding and competition experiments indicate that NS5 dimerization is disrupted at high GTP concentrations. This competition represents a first glimpse at a molecular level explaining how dimerization might regulate the capping process. The lack of vaccine or antiviral drugs to combat Zika virus (ZIKV) infection has encouraged scientists to characterize in depth potential drug targets. One attractive candidate is NS5, responsible for the catalysis of the 5’-RNA capping, methylation and RNA synthesis, during flavivirus genome replication. To fulfill these activities, the methyltransferase and RNA-dependent RNA polymerase domains of NS5 need to cooperate with each other. The structural and biophysical data presented in this work demonstrate that the ZIKV NS5 protein has the ability to form dimers, as well as higher order oligomers that may participate in the fine-tuning regulation of the multiple enzyme functions in the replication complex. In addition, we have found that NS5 dimerization is disrupted at high GTP concentrations, explaining how dimerization might regulate the capping process.
Collapse
Affiliation(s)
- Diego S. Ferrero
- Structural Biology Unit, Institut de Biología Molecular de Barcelona CSIC, Barcelona, Spain
| | - Victor M. Ruiz-Arroyo
- Structural Biology Unit, Institut de Biología Molecular de Barcelona CSIC, Barcelona, Spain
| | - Nicolas Soler
- Structural Biology Unit, Institut de Biología Molecular de Barcelona CSIC, Barcelona, Spain
| | - Isabel Usón
- Structural Biology Unit, Institut de Biología Molecular de Barcelona CSIC, Barcelona, Spain
- ICREA Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain
| | - Alba Guarné
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Núria Verdaguer
- Structural Biology Unit, Institut de Biología Molecular de Barcelona CSIC, Barcelona, Spain
- * E-mail:
| |
Collapse
|
43
|
Optimization of a fragment linking hit toward Dengue and Zika virus NS5 methyltransferases inhibitors. Eur J Med Chem 2019; 161:323-333. [DOI: 10.1016/j.ejmech.2018.09.056] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 09/15/2018] [Accepted: 09/23/2018] [Indexed: 12/12/2022]
|
44
|
Rusanov T, Kent T, Saeed M, Hoang TM, Thomas C, Rice CM, Pomerantz RT. Identification of a Small Interface between the Methyltransferase and RNA Polymerase of NS5 that is Essential for Zika Virus Replication. Sci Rep 2018; 8:17384. [PMID: 30478404 PMCID: PMC6255901 DOI: 10.1038/s41598-018-35511-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 11/06/2018] [Indexed: 12/16/2022] Open
Abstract
The spread of Zika virus (ZIKV) has caused an international health emergency due to its ability to cause microcephaly in infants. Yet, our knowledge of how ZIKV replicates at the molecular level is limited. For example, how the non-structural protein 5 (NS5) performs replication, and in particular whether the N-terminal methytransferase (MTase) domain is essential for the function of the C-terminal RNA-dependent RNA polymerase (RdRp) remains unclear. In contrast to previous reports, we find that MTase is absolutely essential for all activities of RdRp in vitro. For instance, the MTase domain confers stability onto the RdRp elongation complex (EC) and and is required for de novo RNA synthesis and nucleotide incorporation by RdRp. Finally, structure function analyses identify key conserved residues at the MTase-RdRp interface that specifically activate RdRp elongation and are essential for ZIKV replication in Huh-7.5 cells. These data demonstrate the requirement for the MTase-RdRp interface in ZIKV replication and identify a specific site within this region as a potential site for therapeutic development.
Collapse
Affiliation(s)
- Timur Rusanov
- Fels Institute for Cancer Research, Department of Medical Genetics and Molecular Biochemistry, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA
| | - Tatiana Kent
- Fels Institute for Cancer Research, Department of Medical Genetics and Molecular Biochemistry, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA
| | - Mohsan Saeed
- Laboratory of Virology and Infectious Disease, Center for the Study of Hepatitis C, The Rockefeller University, New York, NY, USA
| | - Trung M Hoang
- Fels Institute for Cancer Research, Department of Medical Genetics and Molecular Biochemistry, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA
| | - Crystal Thomas
- Fels Institute for Cancer Research, Department of Medical Genetics and Molecular Biochemistry, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA
| | - Charles M Rice
- Laboratory of Virology and Infectious Disease, Center for the Study of Hepatitis C, The Rockefeller University, New York, NY, USA
| | - Richard T Pomerantz
- Fels Institute for Cancer Research, Department of Medical Genetics and Molecular Biochemistry, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
45
|
Sinigaglia A, Riccetti S, Trevisan M, Barzon L. In silico approaches to Zika virus drug discovery. Expert Opin Drug Discov 2018; 13:825-835. [PMID: 30160181 DOI: 10.1080/17460441.2018.1515909] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
INTRODUCTION After the WHO declared Zika virus (ZIKV) as a public health emergency of international concern, intense research for the development of vaccines and drugs has been undertaken, leading to the development of several candidates. Areas covered: This review discusses the developments achieved so far by computational methods in the discovery of candidate compounds targeting ZIKV proteins, i.e. the envelope and capsid structural proteins, the NS3 helicase/protease, and the NS5 methyltransferase/RNA-dependent RNA polymerase. Expert opinion: Research for effective drugs against ZIKV is still in a very early discovery phase. Notwithstanding the intense efforts for the development of new drugs and the identification of several promising candidates by using different approaches, including computational methods, so far only a few candidates have been experimentally tested. An important caveat of anti-flavivirus drug development is represented by the difficult of reproducing the in vivo microenvironment of the replication complex, which may lead to discrepancies between in vitro results and experimental evaluation in vivo. Moreover, anti-ZIKV drugs have the additional requirement of an excellent safety profile in pregnancy and ability to diffuse to different tissues, including the central nervous system, the testis, and the placenta.
Collapse
Affiliation(s)
| | - Silvia Riccetti
- a Department of Molecular Medicine , University of Padova , Padova , Italy
| | - Marta Trevisan
- a Department of Molecular Medicine , University of Padova , Padova , Italy
| | - Luisa Barzon
- a Department of Molecular Medicine , University of Padova , Padova , Italy
| |
Collapse
|
46
|
Saw WG, Pan A, Subramanian Manimekalai MS, Grüber A, Grüber G. Structure and flexibility of non-structural proteins 3 and -5 of Dengue- and Zika viruses in solution. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2018; 143:67-77. [PMID: 30171868 DOI: 10.1016/j.pbiomolbio.2018.08.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 08/16/2018] [Accepted: 08/24/2018] [Indexed: 01/17/2023]
Abstract
Dengue- (DENV) and Zika viruses (ZIKV) rely on their non-structural protein 5 (NS5) including a methyl-transferase (MTase) and a RNA-dependent RNA polymerase (RdRp) for capping and synthesis of the viral RNA, and the non-structural protein 3 (NS3) with its protease and helicase domain for polyprotein possessing, unwinding dsRNA proceeding replication, and NTPase/RTPase activities. Accumulation of data for DENV- and ZIKV NS3 and NS5 in solution during recent years provides information about their overall shape, substrate-induced alterations, oligomeric forms and flexibility, with the latter being essential for domain-domain crosstalk. The importance and differences of the linker regions that connect the two domains of NS3 or NS5 are highlighted in particular with respect to the different DENV serotypes (DENV-1 to -4) as well as to the sequence diversities between the DENV and ZIKV proteins. Novel mutants of the French Polynesia ZIKV NS3 linker presented, identify critical residues in protein stability and enzymatic activity.
Collapse
Affiliation(s)
- Wuan Geok Saw
- Nanyang Technological University, School of Biological Sciences, 60 Nanyang Drive, 637551, Singapore
| | - Ankita Pan
- Nanyang Technological University, School of Biological Sciences, 60 Nanyang Drive, 637551, Singapore
| | | | - Ardina Grüber
- Nanyang Technological University, School of Biological Sciences, 60 Nanyang Drive, 637551, Singapore
| | - Gerhard Grüber
- Nanyang Technological University, School of Biological Sciences, 60 Nanyang Drive, 637551, Singapore.
| |
Collapse
|
47
|
Saiz JC, Oya NJD, Blázquez AB, Escribano-Romero E, Martín-Acebes MA. Host-Directed Antivirals: A Realistic Alternative to Fight Zika Virus. Viruses 2018; 10:v10090453. [PMID: 30149598 PMCID: PMC6163279 DOI: 10.3390/v10090453] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 08/17/2018] [Accepted: 08/22/2018] [Indexed: 12/21/2022] Open
Abstract
Zika virus (ZIKV), a mosquito-borne flavivirus, was an almost neglected pathogen until its introduction in the Americas in 2015, where it has been responsible for a threat to global health, causing a great social and sanitary alarm due to its increased virulence, rapid spread, and an association with severe neurological and ophthalmological complications. Currently, no specific antiviral therapy against ZIKV is available, and treatments are palliative and mainly directed toward the relief of symptoms, such as fever and rash, by administering antipyretics, anti-histamines, and fluids for dehydration. Nevertheless, lately, search for antivirals has been a major aim in ZIKV investigations. To do so, screening of libraries from different sources, testing of natural compounds, and repurposing of drugs with known antiviral activity have allowed the identification of several antiviral candidates directed to both viral (structural proteins and enzymes) and cellular elements. Here, we present an updated review of current knowledge about anti-ZIKV strategies, focusing on host-directed antivirals as a realistic alternative to combat ZIKV infection.
Collapse
Affiliation(s)
- Juan-Carlos Saiz
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), 28040 Madrid, Spain.
| | - Nereida Jiménez de Oya
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), 28040 Madrid, Spain.
| | - Ana-Belén Blázquez
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), 28040 Madrid, Spain.
| | - Estela Escribano-Romero
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), 28040 Madrid, Spain.
| | - Miguel A Martín-Acebes
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), 28040 Madrid, Spain.
| |
Collapse
|
48
|
Qadir A, Riaz M, Saeed M, Shahzad-Ul-Hussan S. Potential targets for therapeutic intervention and structure based vaccine design against Zika virus. Eur J Med Chem 2018; 156:444-460. [PMID: 30015077 DOI: 10.1016/j.ejmech.2018.07.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 06/28/2018] [Accepted: 07/06/2018] [Indexed: 01/01/2023]
Abstract
Continuously increasing number of reports of Zika virus (ZIKV) infections and associated severe clinical manifestations, including autoimmune abnormalities and neurological disorders such as neonatal microcephaly and Guillain-Barré syndrome have created alarming situation in various countries. To date, no specific antiviral therapy or vaccine is available against ZIKV. This review provides a comprehensive insight into the potential therapeutic targets and describes viral epitopes of broadly neutralizing antibodies (bNAbs) in vaccine design perspective. Interactions between ZIKV envelope glycoprotein E and cellular receptors mediate the viral fusion and entry to the target cell. Blocking these interactions by targeting cellular receptors or viral structural proteins mediating these interactions or viral surface glycans can inhibit viral entry to the cell. Similarly, different non-structural proteins of ZIKV and un-translated regions (UTRs) of its RNA play essential roles in viral replication cycle and potentiate for therapeutic interventions. Structure based vaccine design requires identity and structural description of the epitopes of bNAbs. We have described different conserved bNAb epitopes present in the ZIKV envelope as potential targets for structure based vaccine design. This review also highlights successes, unanswered questions and future perspectives in relation to therapeutic and vaccine development against ZIKV.
Collapse
Affiliation(s)
- Amina Qadir
- Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, 54792, Pakistan
| | - Muhammad Riaz
- Department of Chemistry, University of Azad Jammu & Kashmir, Muzaffarabad, Pakistan
| | - Muhammad Saeed
- Department of Chemistry and Chemical Engineering, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, 54792, Pakistan.
| | - Syed Shahzad-Ul-Hussan
- Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, 54792, Pakistan.
| |
Collapse
|
49
|
Wang B, Thurmond S, Hai R, Song J. Structure and function of Zika virus NS5 protein: perspectives for drug design. Cell Mol Life Sci 2018; 75:1723-1736. [PMID: 29423529 PMCID: PMC5911220 DOI: 10.1007/s00018-018-2751-x] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 01/02/2018] [Accepted: 01/11/2018] [Indexed: 12/29/2022]
Abstract
Zika virus (ZIKV) belongs to the positive-sense single-stranded RNA-containing Flaviviridae family. Its recent outbreak and association with human diseases (e.g. neurological disorders) have raised global health concerns, and an urgency to develop a therapeutic strategy against ZIKV infection. However, there is no currently approved antiviral against ZIKV. Here we present a comprehensive overview on recent progress in structure-function investigation of ZIKV NS5 protein, the largest non-structural protein of ZIKV, which is responsible for replication of the viral genome, RNA capping and suppression of host interferon responses. Structural comparison of the N-terminal methyltransferase domain and C-terminal RNA-dependent RNA polymerase domain of ZIKV NS5 with their counterparts from related viruses provides mechanistic insights into ZIKV NS5-mediated RNA replication, and identifies residues critical for its enzymatic activities. Finally, a collection of recently identified small molecule inhibitors against ZIKV NS5 or its closely related flavivirus homologues are also discussed.
Collapse
Affiliation(s)
- Boxiao Wang
- Department of Biochemistry, University of California, Riverside, CA, 92521, USA
| | - Stephanie Thurmond
- Department of Microbiology and Plant Pathology , University of California, Riverside, CA, 92521, USA
| | - Rong Hai
- Department of Microbiology and Plant Pathology , University of California, Riverside, CA, 92521, USA.
| | - Jikui Song
- Department of Biochemistry, University of California, Riverside, CA, 92521, USA.
| |
Collapse
|
50
|
Panwar U, Singh SK. An Overview on Zika Virus and the Importance of Computational Drug Discovery. JOURNAL OF EXPLORATORY RESEARCH IN PHARMACOLOGY 2018; 3:43-51. [DOI: 10.14218/jerp.2017.00025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|