1
|
Januário MAP, Junior CDOR, Castro-Gamboa I. Indole Derivatives as Promising Anti-Dengue Agents: A Review of Recent Advances. Chem Biodivers 2024:e202402517. [PMID: 39714443 DOI: 10.1002/cbdv.202402517] [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: 10/03/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 12/24/2024]
Abstract
Dengue, a mosquito-borne disease transmitted by Aedes mosquitoes, is a significant global health concern. Despite extensive research, effective treatments remain limited. The indole nucleus, known for its diverse pharmacological properties, has emerged as a promising scaffold for anti-dengue drug discovery. This review comprehensively examines recent advancements in the fields of natural products, medicinal chemistry, and computer-aided drug design focused on discovering indole-based anti-dengue agents. We discuss the rationale for targeting indole frameworks, highlight key structural features associated with anti-dengue activity, and summarize recent research findings. The review aims to provide valuable insights for researchers working on developing novel anti-dengue therapeutics.
Collapse
Affiliation(s)
| | | | - Ian Castro-Gamboa
- Departament of Biochemistry and Organic Chemistry, São Paulo State University-UNESP, Araraquara, Brazil
| |
Collapse
|
2
|
Komarudin AG, Adharis A, Sasmono RT. Natural Compounds and Their Analogs as Antivirals Against Dengue Virus: A Review. Phytother Res 2024. [PMID: 39697048 DOI: 10.1002/ptr.8408] [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: 12/30/2023] [Revised: 09/23/2024] [Accepted: 09/26/2024] [Indexed: 12/20/2024]
Abstract
Dengue virus (DENV) continues to pose a significant global health challenge, causing diseases such as dengue fever, dengue hemorrhagic fever, and dengue shock syndrome. While efforts in vaccine development and antiviral drug discovery are ongoing, effective therapeutic options remain limited. In this review, we highlight natural compounds and the analogs that demonstrated antiviral activity against DENV in in vitro and in vivo studies. Specifically, these studies examine alkaloids, phenolic acids, phenols, flavonoids, terpenoids, and glycosides which have shown potential in inhibiting DENV entry, replication, and reducing the cytokine storm. By focusing on these bioactive compounds and the analogs, a comprehensive overview of their promising roles is provided to advance therapeutic strategies for combating DENV infection.
Collapse
Affiliation(s)
- Amalina Ghaisani Komarudin
- Eijkman Research Center for Molecular Biology, National Research and Innovation Agency (BRIN), Kabupaten Bogor, Jawa Barat, Indonesia
| | - Azis Adharis
- Department of Chemistry, Faculty of Science and Computer Science, Universitas Pertamina (UPER), Jakarta, Indonesia
| | - R Tedjo Sasmono
- Eijkman Research Center for Molecular Biology, National Research and Innovation Agency (BRIN), Kabupaten Bogor, Jawa Barat, Indonesia
| |
Collapse
|
3
|
Apaydın ÇB, Göktaş F, Naesens L, Karalı N. Novel 2-indolinone derivatives as promising agents against respiratory syncytial and yellow fever viruses. Future Med Chem 2024; 16:295-310. [PMID: 38288568 DOI: 10.4155/fmc-2023-0179] [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: 06/16/2023] [Accepted: 01/09/2024] [Indexed: 02/07/2024] Open
Abstract
Background: A vaccine or antiviral drug for respiratory syncytial virus (RSV) infections and a specific antiviral drug for yellow fever virus (YFV) infections has not yet been developed. Method: In this study, 2-indolinone-based N-(4-sulfamoylphenyl)hydrazinecarbothioamides were synthesized. Along with these new compounds, previously synthesized 2-indolinone-based N-(3-sulfamoylphenyl)hydrazinecarbothioamides were evaluated against various DNA and RNA viruses. Results: Some 2-indolinone compounds exhibited nontoxic and selective antiviral activities against RSV and YFV. Halogen substitution at the indole ring increased the anti-RSV activities. Moreover, 1-benzyl and 5-halogen or nitro-substituted compounds were the most effective compounds against YFV. Conclusion: Generally, the 3-sulfonamide-substituted compounds were determined to be more effective than 4-sulfonamide-substituted compounds against RSV and YFV.
Collapse
Affiliation(s)
- Çağla Begüm Apaydın
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Istanbul University, 34126, Istanbul, Turkey
| | - Füsun Göktaş
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Istanbul University, 34126, Istanbul, Turkey
| | - Lieve Naesens
- Rega Institute, KU Leuven, Department of Microbiology, Immunology & Transplantation, B-3000, Leuven, Belgium
| | - Nilgün Karalı
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Istanbul University, 34126, Istanbul, Turkey
| |
Collapse
|
4
|
Lu JW, Huang CK, Chen YC, Lee GC, Ho YJ. Virucidal activity of trehalose 6-monolaurate against dengue virus in vitro. Drug Dev Res 2023; 84:1699-1708. [PMID: 37688413 DOI: 10.1002/ddr.22112] [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: 04/08/2023] [Revised: 08/05/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023]
Abstract
Dengue fever is an acute febrile disease caused by dengue virus (DENV) infection. Over the past 60 years, DENV has spread throughout tropical regions and currently affects more than 50% of the world's population; however, there are as of yet no effective anti-DENV drugs for clinical treatment. A number of research teams have investigated derivatives of glycolipids as possible agents for the inhibition of DENV. Our objective in this research was to study the antiviral effects of trehalose 6-caprate (TMC), trehalose 6-monolaurate (TML), and trehalose 6-monooleate (TMO), based on reports that the corresponding glycosyl, trehalose, reduces the transmission of Zika virus (ZIKV). We also sought to elucidate the molecular mechanisms underlying inhibition using the RNA isolation and reverse transcription-quantitative polymerase chain reaction, western blot analysis, median tissue culture infectious dose (TCID50 ) assay, and immunofluorescence assay and immunochemistry staining, in vitro. This is the first study to demonstrate the TML-induced inhibition of DENV serotype 2 (DENV-2) in a dose-dependent manner. The inhibitory effects of TML in the pretreated, cotreated, and full-treated groups were confirmed using time of addition assays. We determined that TML restricted viral binding, entry, replication, and release. We also confirmed the efficacy of TML against three clinical isolates of DENV serotypes 1, 3, and 4 (DENV-1, DENV-3, and DENV-4). The findings obtained in this study identify TML as a promising candidate for the development of drugs to treat DENV infection.
Collapse
Affiliation(s)
- Jeng-Wei Lu
- Biotech Research and Innovation Centre, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- The Finsen Laboratory, Rigshospitalet/National University Hospital, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Chin-Kai Huang
- School of Pharmacy, National Defense Medical Center, Taipei, Taiwan, ROC
- Department of Pharmacy, Tri-Service General Hospital Penghu Branch, National Defense Medical Center, Magong City, Taiwan, ROC
| | - Yen-Chen Chen
- School of Pharmacy, National Defense Medical Center, Taipei, Taiwan, ROC
- Institute of Preventive Medicine, National Defense Medical Center, New Taipei, Taiwan, ROC
| | - Guan-Chiun Lee
- School of Life Science, National Taiwan Normal University, Taipei, Taiwan, ROC
| | - Yi-Jung Ho
- School of Pharmacy, National Defense Medical Center, Taipei, Taiwan, ROC
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan, ROC
| |
Collapse
|
5
|
García-Ariza LL, González-Rivillas N, Díaz-Aguirre CJ, Rocha-Roa C, Padilla-Sanabria L, Castaño-Osorio JC. Antiviral Activity of an Indole-Type Compound Derived from Natural Products, Identified by Virtual Screening by Interaction on Dengue Virus NS5 Protein. Viruses 2023; 15:1563. [PMID: 37515249 PMCID: PMC10384440 DOI: 10.3390/v15071563] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 07/07/2023] [Accepted: 07/08/2023] [Indexed: 07/30/2023] Open
Abstract
Dengue is an acute febrile illness caused by the Dengue virus (DENV), with a high number of cases worldwide. There is no available treatment that directly affects the virus or the viral cycle. The objective of this study was to identify a compound derived from natural products that interacts with the NS5 protein of the dengue virus through virtual screening and evaluate its in vitro antiviral effect on DENV-2. Molecular docking was performed on NS5 using AutoDock Vina software, and compounds with physicochemical and pharmacological properties of interest were selected. The preliminary antiviral effect was evaluated by the expression of the NS1 protein. The effect on viral genome replication and/or translation was determined by NS5 production using DENV-2 Huh-7 replicon through ELISA and viral RNA quantification using RT-qPCR. The in silico strategy proved effective in finding a compound (M78) with an indole-like structure and with an effect on the replication cycle of DENV-2. Treatment at 50 µM reduced the expression of the NS5 protein by 70% and decreased viral RNA by 1.7 times. M78 is involved in the replication and/or translation of the viral genome.
Collapse
Affiliation(s)
| | | | | | - Cristian Rocha-Roa
- Grupo de Parasitología Molecular GEPAMOL, Universidad del Quindío, Armenia 630001, Quindío, Colombia
| | | | | |
Collapse
|
6
|
Celegato M, Sturlese M, Vasconcelos Costa V, Trevisan M, Lallo Dias AS, Souza Passos IB, Queiroz-Junior CM, Messa L, Favaro A, Moro S, Teixeira MM, Loregian A, Mercorelli B. Small-Molecule Inhibitor of Flaviviral NS3-NS5 Interaction with Broad-Spectrum Activity and Efficacy In Vivo. mBio 2023; 14:e0309722. [PMID: 36622141 PMCID: PMC9973282 DOI: 10.1128/mbio.03097-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 11/30/2022] [Indexed: 01/10/2023] Open
Abstract
Every year, dengue virus (DENV) causes one hundred million infections worldwide that can result in dengue disease and severe dengue. Two other mosquito-borne flaviviruses, i.e., Zika virus (ZIKV) and West Nile virus (WNV), are responsible of prolonged outbreaks and are associated with severe neurological diseases, congenital defects, and eventually death. These three viruses, despite their importance for global public health, still lack specific drug treatments. Here, we describe the structure-guided discovery of small molecules with pan-flavivirus antiviral potential by a virtual screening of ~1 million structures targeting the NS3-NS5 interaction surface of different flaviviruses. Two molecules inhibited the interaction between DENV NS3 and NS5 in vitro and the replication of all DENV serotypes as well as ZIKV and WNV and exhibited low propensity to select resistant viruses. Remarkably, one molecule demonstrated efficacy in a mouse model of dengue by reducing peak viremia, viral load in target organs, and associated tissue pathology. This study provides the proof of concept that targeting the flaviviral NS3-NS5 interaction is an effective therapeutic strategy able to reduce virus replication in vivo and discloses new chemical scaffolds that could be further developed, thus providing a significant milestone in the development of much awaited broad-spectrum antiflaviviral drugs. IMPORTANCE More than one-third of the human population is at risk of infection by different mosquito-borne flaviviruses. Despite this, no specific antiviral drug is currently available. In this work, using a computational approach based on molecular dynamics simulation and virtual screening of ~1 million small-molecule structures, we identified a compound that targets the interaction between the two sole flaviviral enzymes, i.e., NS3 and NS5. This compound demonstrated pan-serotype anti-DENV activity and pan-flavivirus potential in infected cells, low propensity to select viral resistant mutant viruses, and efficacy in a mouse model of dengue. Broad-spectrum antivirals are much awaited, and this work represents a significant advance toward the development of therapeutic molecules with extended antiflavivirus potential that act by an innovative mechanism and could be used alone or in combination with other antivirals.
Collapse
Affiliation(s)
- Marta Celegato
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Mattia Sturlese
- Molecular Modeling Section (MMS), Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | | | - Marta Trevisan
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Angélica Samer Lallo Dias
- Departamento de Bioquímica e Imunologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | | | - Lorenzo Messa
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Annagiulia Favaro
- Molecular Modeling Section (MMS), Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | - Stefano Moro
- Molecular Modeling Section (MMS), Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | - Mauro Martins Teixeira
- Departamento de Bioquímica e Imunologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Arianna Loregian
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | | |
Collapse
|
7
|
Faisal S, Badshah SL, Kubra B, Emwas AH, Jaremko M. Alkaloids as potential antivirals. A comprehensive review. NATURAL PRODUCTS AND BIOPROSPECTING 2023; 13:4. [PMID: 36598588 PMCID: PMC9812014 DOI: 10.1007/s13659-022-00366-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 12/01/2022] [Indexed: 05/26/2023]
Abstract
Alkaloids are a diverse group of natural phytochemicals. These phytochemicals in plants provide them protection against pests, and herbivorous organisms and also control their development. Numerous of these alkaloids have a variety of biological effects, and some have even been developed into medications with different medicinal properties. This review aims to provide a broad overview of the numerous naturally occurring alkaloids (isolated from both terrestrial and aquatic species) along with synthetically produced alkaloid compounds having prominent antiviral properties. Previous reviews on this subject have focused on the biological actions of both natural and synthetic alkaloids, but they have not gone into comprehensive detail about their antiviral properties. We reviewed here several antiviral alkaloids that have been described in the literature in different investigational environments i.e. (in-vivo, in-ovo, in-vitro, and in-silico), and found that these alkaloid compounds have significant antiviral properties against several infectious viruses. These alkaloids repressed and targeted various important stages of viral infection at non-toxic doses while some of the alkaloids reported here also exhibited comparable inhibitory activities to commercially used drugs. Overall, these anti-viral effects of alkaloids point to a high degree of specificity, implying that they could serve as effective and safe antiviral medicines if further pursued in medicinal and pharmacological investigations.
Collapse
Affiliation(s)
- Shah Faisal
- Department of Chemistry, Islamia College University Peshawar, Peshawar, 25120, Pakistan
| | - Syed Lal Badshah
- Department of Chemistry, Islamia College University Peshawar, Peshawar, 25120, Pakistan.
| | - Bibi Kubra
- Department of Chemistry, Islamia College University Peshawar, Peshawar, 25120, Pakistan
| | - Abdul-Hamid Emwas
- Core Labs, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Mariusz Jaremko
- Division of Biological and Environmental Sciences and Engineering (BESE), Smart-Health Initiative (SHI) and Red Sea Research Center (RSRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia.
| |
Collapse
|
8
|
Gao S, Song L, Xu H, Fikatas A, Oeyen M, De Jonghe S, Zhao F, Jing L, Jochmans D, Vangeel L, Cheng Y, Kang D, Neyts J, Herdewijn P, Schols D, Zhan P, Liu X. Identification of Polyphenol Derivatives as Novel SARS-CoV-2 and DENV Non-Nucleoside RdRp Inhibitors. MOLECULES (BASEL, SWITZERLAND) 2022; 28:molecules28010160. [PMID: 36615354 PMCID: PMC9822497 DOI: 10.3390/molecules28010160] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/19/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022]
Abstract
The Coronavirus Disease 2019 (COVID-19) and dengue fever (DF) pandemics both remain to be significant public health concerns in the foreseeable future. Anti-SARS-CoV-2 drugs and vaccines are both indispensable to eliminate the epidemic situation. Here, two piperazine-based polyphenol derivatives DF-47 and DF-51 were identified as potential inhibitors directly blocking the active site of SARS-CoV-2 and DENV RdRp. Data through RdRp inhibition screening of an in-house library and in vitro antiviral study selected DF-47 and DF-51 as effective inhibitors of SARS-CoV-2/DENV polymerase. Moreover, in silico simulation revealed stable binding modes between the DF-47/DF-51 and SARS-CoV-2/DENV RdRp, respectively, including chelating with Mg2+ near polymerase active site. This work discovered the inhibitory effect of two polyphenols on distinct viral RdRp, which are expected to be developed into broad-spectrum, non-nucleoside RdRp inhibitors with new scaffold.
Collapse
Affiliation(s)
- Shenghua Gao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
- Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
- Shenzhen Research Institute of Shandong University, A301 Virtual University Park in South District of Shenzhen, Shenzhen 518057, China
| | - Letian Song
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
- Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Hongtao Xu
- Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
- Correspondence: (H.X.); (D.S.); (P.Z.); (X.L.)
| | - Antonios Fikatas
- Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
| | - Merel Oeyen
- Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
| | - Steven De Jonghe
- Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
| | - Fabao Zhao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
- Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Lanlan Jing
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
- Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Dirk Jochmans
- Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
| | - Laura Vangeel
- Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
| | - Yusen Cheng
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
- Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Dongwei Kang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
- Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Johan Neyts
- Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
| | - Piet Herdewijn
- Laboratory of Medicinal Chemistry, Rega Institute for Medical Research, KU Leuven, 3000 Leuven, Belgium
| | - Dominique Schols
- Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
- Correspondence: (H.X.); (D.S.); (P.Z.); (X.L.)
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
- Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
- Correspondence: (H.X.); (D.S.); (P.Z.); (X.L.)
| | - Xinyong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
- Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
- Correspondence: (H.X.); (D.S.); (P.Z.); (X.L.)
| |
Collapse
|
9
|
Li Q, Kang C. Dengue virus NS4B protein as a target for developing antivirals. Front Cell Infect Microbiol 2022; 12:959727. [PMID: 36017362 PMCID: PMC9398000 DOI: 10.3389/fcimb.2022.959727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 07/21/2022] [Indexed: 12/04/2022] Open
Abstract
Dengue virus is an important pathogen affecting global population while no specific treatment is available against this virus. Effort has been made to develop inhibitors through targeting viral nonstructural proteins such as NS3 and NS5 with enzymatic activities. No potent inhibitors entering clinical studies have been developed so far due to many challenges. The genome of dengue virus encodes four membrane-bound nonstructural proteins which do not possess any enzymatic activities. Studies have shown that the membrane protein-NS4B is a validated target for drug discovery and several NS4B inhibitors exhibited antiviral activities in various assays and entered preclinical studies.. Here, we summarize the recent studies on dengue NS4B protein. The structure and membrane topology of dengue NS4B derived from biochemical and biophysical studies are described. Function of NS4B through protein-protein interactions and some available NS4B inhibitors are summarized. Accumulated studies demonstrated that cell-based assays play important roles in developing NS4B inhibitors. Although the atomic structure of NS4B is not obtained, target-based drug discovery approach become feasible to develop NS4B inhibitors as recombinant NS4B protein is available.
Collapse
Affiliation(s)
- Qingxin Li
- Guangdong Provincial Engineering Laboratory of Biomass High Value Utilization, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
| | - Congbao Kang
- Experimental Drug Development Centre, Agency for Science, Technology and Research, Singapore, Singapore
| |
Collapse
|
10
|
Randhawa V, Pathania S, Kumar M. Computational Identification of Potential Multitarget Inhibitors of Nipah Virus by Molecular Docking and Molecular Dynamics. Microorganisms 2022; 10:microorganisms10061181. [PMID: 35744699 PMCID: PMC9227315 DOI: 10.3390/microorganisms10061181] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/28/2022] [Accepted: 05/11/2022] [Indexed: 02/04/2023] Open
Abstract
Nipah virus (NiV) is a recently emerged paramyxovirus that causes severe encephalitis and respiratory diseases in humans. Despite the severe pathogenicity of this virus and its pandemic potential, not even a single type of molecular therapeutics has been approved for human use. Considering the role of NiV attachment glycoprotein G (NiV-G), fusion glycoprotein (NiV-F), and nucleoprotein (NiV-N) in virus replication and spread, these are the most attractive targets for anti-NiV drug discovery. Therefore, to prospect for potential multitarget chemical/phytochemical inhibitor(s) against NiV, a sequential molecular docking and molecular-dynamics-based approach was implemented by simultaneously targeting NiV-G, NiV-F, and NiV-N. Information on potential NiV inhibitors was compiled from the literature, and their 3D structures were drawn manually, while the information and 3D structures of phytochemicals were retrieved from the established structural databases. Molecules were docked against NiV-G (PDB ID:2VSM), NiV-F (PDB ID:5EVM), and NiV-N (PDB ID:4CO6) and then prioritized based on (1) strong protein-binding affinity, (2) interactions with critically important binding-site residues, (3) ADME and pharmacokinetic properties, and (4) structural stability within the binding site. The molecules that bind to all the three viral proteins (NiV-G ∩ NiV-F ∩ NiV-N) were considered multitarget inhibitors. This study identified phytochemical molecules RASE0125 (17-O-Acetyl-nortetraphyllicine) and CARS0358 (NA) as distinct multitarget inhibitors of all three viral proteins, and chemical molecule ND_nw_193 (RSV604) as an inhibitor of NiV-G and NiV-N. We expect the identified compounds to be potential candidates for in vitro and in vivo antiviral studies, followed by clinical treatment of NiV.
Collapse
Affiliation(s)
- Vinay Randhawa
- Virology Discovery Unit and Bioinformatics Centre, CSIR-Institute of Microbial Technology, Council of Scientific and Industrial Research, Chandigarh 160036, India; (V.R.); (S.P.)
| | - Shivalika Pathania
- Virology Discovery Unit and Bioinformatics Centre, CSIR-Institute of Microbial Technology, Council of Scientific and Industrial Research, Chandigarh 160036, India; (V.R.); (S.P.)
| | - Manoj Kumar
- Virology Discovery Unit and Bioinformatics Centre, CSIR-Institute of Microbial Technology, Council of Scientific and Industrial Research, Chandigarh 160036, India; (V.R.); (S.P.)
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
- Correspondence: ; Tel.: +91-172-6665453
| |
Collapse
|
11
|
da Silva PG, Fonseca AH, Ribeiro MP, Silva TD, Grael CFF, Pena LJ, Silva TMS, Oliveira EDJ. Bisbenzylisoquinoline Alkaloids of Cissampelos Sympodialis With in Vitro Antiviral Activity Against Zika Virus. Front Pharmacol 2021; 12:743541. [PMID: 34603056 PMCID: PMC8482128 DOI: 10.3389/fphar.2021.743541] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 08/31/2021] [Indexed: 12/13/2022] Open
Abstract
In search of new antiviral compounds against Zika virus we conducted a bioassay-guided fractionation of bisbenzyilisoquinoline alkaloids isolated from Cissampelos sympodialis (Menispermaceae), a medicinal plant species endemic to Brazil. Six subfractions were obtained from a tertiary alkaloidal fraction of the rhizomes (TAFrz) using preparative high-performance liquid chromatography. All the subfractions were tested against Zika virus-infected Vero cells as the cellular model to evaluate cytotoxicity and antiviral effective concentrations. The results showed that three of the six TAFrz subfractions tested were active. The most active ones were the subfraction 6 (that consisted of the alkaloids methylwarifteine and warifteine present as a mixture at a ratio of 8.8:1.2 respectively) and the subfraction 5, that was later identified as warifteine, the major tertiary alkaloid of this species. Warifteine was able to significantly reduce virus titer in Zika virus-infected Vero cells with an IC50 of 2.2 μg/ml and this effect was selective (selectivity index, SI = 68.3). Subfraction 6 had an IC50 = 3.5 μg/ml and was more cytotoxic than pure warifteine, with SI = 6.14. Fraction 5 and fraction 6 were more potent in decreasing the viral titer of Zika virus-infected Vero cells than 6-methylmercaptopurine riboside (IC50 = 24.5 μg/ml and SI = 11.9), a mercaptopurine riboside with ZIKV antiviral activity used as a positive control. Our data demonstrate that alkaloids of the bisbenzylisoquinoline type may be explored as new antiviral agents or as an useful pharmacophore for investigating ZIKV antiviral activity.
Collapse
Affiliation(s)
| | - Aventino H Fonseca
- Department of Pharmacy, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil
| | - Malu P Ribeiro
- Department of Pharmacy, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil
| | - Taizia D Silva
- Department of Pharmacy, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil
| | - Cristiane F F Grael
- Department of Pharmacy, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil
| | - Lindomar J Pena
- Department of Virology, Aggeu Magalhães Institute (IAM), Fiocruz, Recife, Brazil
| | - Tania M S Silva
- Laboratório de Bioprospecção Fitoquímica, Department of Chemistry, Federal Rural University of Pernambuco, Recife, Brazil
| | | |
Collapse
|