1
|
Agrawal T, Siddqui G, Dahiya R, Patidar A, Madan U, Das S, Asthana S, Samal S, Awasthi A. Inhibition of early RNA replication in Chikungunya and Dengue virus by lycorine: In vitro and in silico studies. Biochem Biophys Res Commun 2024; 730:150393. [PMID: 39003865 DOI: 10.1016/j.bbrc.2024.150393] [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: 05/23/2024] [Revised: 07/05/2024] [Accepted: 07/10/2024] [Indexed: 07/16/2024]
Abstract
Arboviruses such as chikungunya virus (CHIKV) and dengue virus (DENV) collectively afflict millions of individuals worldwide particularly in endemic countries like India, leading to substantial morbidity and mortality. With the lack of effective vaccines for both CHIKV and DENV in India, the search for antiviral compounds becomes paramount to control these viral infections. In line with this, our investigation was focused on screening natural compounds for their potential antiviral activity against CHIKV and DENV. Using different assays, including plaque assay, immunofluorescence, and reverse transcription-quantitative real-time PCR (qRT-PCR), out of 109 natural compounds tested, we confirmed lycorine's in vitro antiviral activity against CHIKV and DENV at low micromolar concentrations in different cell types. Time of addition assays indicated that lycorine does not impede viral entry. Additionally, qRT-PCR results along with time of addition assay suggested that lycorine interferes with the synthesis of negative strand viral RNA. Molecular docking analysis was done to understand the mode of inhibition of viral replication. The results revealed that the most likely binding site with the highest binding affinity of lycorine, was at the palm and finger domains, in the vicinity of the catalytic site of CHIKV and DENV RNA-dependent RNA polymerase (RdRp). Collectively, our data underscores the potential of lycorine to be developed as a direct acting inhibitor for DENV and CHIKV, addressing the critical need of requirement of an antiviral in regions where these viruses pose significant public health threats.
Collapse
Affiliation(s)
- Tanvi Agrawal
- Centre for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad, 121001, Haryana, India; Centre for Virus Research, Therapeutics and Vaccines, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad, 121001, Haryana, India.
| | - Gazala Siddqui
- Centre for Virus Research, Therapeutics and Vaccines, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad, 121001, Haryana, India
| | - Ridhima Dahiya
- Computational Biophysics and CADD Group, Computational and Mathematical Biology Centre (CMBC), Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad, 121001, Haryana, India
| | - Aanchal Patidar
- Centre for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad, 121001, Haryana, India
| | - Upasna Madan
- Centre for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad, 121001, Haryana, India
| | - Supratik Das
- Centre for Virus Research, Therapeutics and Vaccines, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad, 121001, Haryana, India
| | - Shailendra Asthana
- Computational Biophysics and CADD Group, Computational and Mathematical Biology Centre (CMBC), Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad, 121001, Haryana, India
| | - Sweety Samal
- Centre for Virus Research, Therapeutics and Vaccines, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad, 121001, Haryana, India
| | - Amit Awasthi
- Centre for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad, 121001, Haryana, India.
| |
Collapse
|
2
|
Mehta N, Meng Y, Zare R, Kamenetsky-Goldstein R, Sattely E. A developmental gradient reveals biosynthetic pathways to eukaryotic toxins in monocot geophytes. Cell 2024; 187:5620-5637.e10. [PMID: 39276773 DOI: 10.1016/j.cell.2024.08.027] [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: 04/08/2023] [Revised: 01/23/2024] [Accepted: 08/13/2024] [Indexed: 09/17/2024]
Abstract
Numerous eukaryotic toxins that accumulate in geophytic plants are valuable in the clinic, yet their biosynthetic pathways have remained elusive. A notable example is the >150 Amaryllidaceae alkaloids (AmAs), including galantamine, an FDA-approved treatment for Alzheimer's disease. We show that while AmAs accumulate to high levels in many daffodil tissues, biosynthesis is localized to nascent, growing tissue at the leaf base. A similar trend is found in the production of steroidal alkaloids (e.g., cyclopamine) in corn lily. This model of active biosynthesis enabled the elucidation of a complete set of biosynthetic genes that can be used to produce AmAs. Taken together, our work sheds light on the developmental and enzymatic logic of diverse alkaloid biosynthesis in daffodils. More broadly, it suggests a paradigm for biosynthesis regulation in monocot geophytes, where plants are protected from herbivory through active charging of newly formed cells with eukaryotic toxins that persist as above-ground tissue develops.
Collapse
Affiliation(s)
- Niraj Mehta
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Yifan Meng
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Richard Zare
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | | | - Elizabeth Sattely
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA; HHMI, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
3
|
Shrestha B, Yang S, Griffith L, Ma J, Wang F, Liu H, Zhao Q, Du Y, Zhang J, Chang J, Guo JT. Discovery of hepatitis B virus subviral particle biogenesis inhibitors from a bioactive compound library. Antiviral Res 2024; 228:105955. [PMID: 38964614 DOI: 10.1016/j.antiviral.2024.105955] [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: 05/09/2024] [Revised: 06/30/2024] [Accepted: 07/01/2024] [Indexed: 07/06/2024]
Abstract
High levels of hepatitis B virus (HBV) surface antigen (HBsAg) in the blood of chronic HBV carriers are considered to drive the exhaustion of antigen-specific T and B lymphocytes and thus responsible for the persistence of infection. Accordingly, therapeutic elimination of HBsAg may facilitate the activation of adaptive antiviral immune responses against HBV and achieve a functional cure of chronic hepatitis B. We discovered recently that an amphipathic alpha helix spanning W156 to R169 of HBV small envelope (S) protein plays an essential role in the morphogenesis of subviral particles (SVPs) and metabolism of S protein. We thus hypothesized that pharmacological disruption of SVP morphogenesis may induce intracellular degradation of S protein and reduce HBsAg secretion. To identify inhibitors of SVP biogenesis, we screened 4417 bioactive compounds with a HepG2-derived cell line expressing HBV S protein and efficiently secreting small spherical SVPs. The screen identified 24 compounds that reduced intracellular SVPs and secreted HBsAg in a concentration-dependent manner. However, 18 of those compounds inhibited the secretion of HBsAg and HBeAg in HBV replicon transfected HepG2 cells at similar efficiency, suggesting each of those compounds may disrupt a common cellular function required for the synthesis and/or secretion of these viral proteins. Interestingly, lycorine more efficiently inhibited the secretion of HBsAg in HepG2 cells transfected with HBV replicons, HepG2.2.15 cells and HBV infected - HepG2 cells expressing sodium taurocholate cotransporting polypeptide (NTCP). The structure activity relationship and antiviral mechanism of lycorine against HBV have been determined.
Collapse
Affiliation(s)
| | - Sisi Yang
- Baruch S. Blumberg Institute, Doylestown, PA, USA; Department of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | | | - Julia Ma
- Baruch S. Blumberg Institute, Doylestown, PA, USA
| | - Fuxuan Wang
- Baruch S. Blumberg Institute, Doylestown, PA, USA
| | - Hui Liu
- Baruch S. Blumberg Institute, Doylestown, PA, USA
| | - Qiong Zhao
- Baruch S. Blumberg Institute, Doylestown, PA, USA
| | - Yanming Du
- Baruch S. Blumberg Institute, Doylestown, PA, USA
| | - Jiming Zhang
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | | | - Ju-Tao Guo
- Baruch S. Blumberg Institute, Doylestown, PA, USA.
| |
Collapse
|
4
|
Jayawardena TU, Merindol N, Liyanage NS, Desgagné-Penix I. Unveiling Amaryllidaceae alkaloids: from biosynthesis to antiviral potential - a review. Nat Prod Rep 2024; 41:721-747. [PMID: 38131392 DOI: 10.1039/d3np00044c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
Covering: 2017 to 2023 (now)Amaryllidaceae alkaloids (AAs) are a unique class of specialized metabolites containing heterocyclic nitrogen bridging that play a distinct role in higher plants. Irrespective of their diverse structures, most AAs are biosynthesized via intramolecular oxidative coupling. The complex organization of biosynthetic pathways is constantly enlightened by new insights owing to the advancement of natural product chemistry, synthetic organic chemistry, biochemistry, systems and synthetic biology tools and applications. These promote novel compound identification, trace-level metabolite quantification, synthesis, and characterization of enzymes engaged in AA catalysis, enabling the recognition of biosynthetic pathways. A complete understanding of the pathway benefits biotechnological applications in the long run. This review emphasizes the structural diversity of the AA specialized metabolites involved in biogenesis although the process is not entirely defined yet. Moreover, this work underscores the pivotal role of synthetic and enantioselective studies in justifying biosynthetic conclusions. Their prospective candidacy as lead constituents for antiviral drug discovery has also been established. However, a complete understanding of the pathway requires further interdisciplinary efforts in which antiviral studies address the structure-activity relationship. This review presents current knowledge on the topic.
Collapse
Affiliation(s)
- Thilina U Jayawardena
- Department of Chemistry, Biochemistry, and Physics, Université du Québec à Trois-Rivières, Trois-Rivières, QC, G8Z 4M3, Canada.
| | - Natacha Merindol
- Department of Chemistry, Biochemistry, and Physics, Université du Québec à Trois-Rivières, Trois-Rivières, QC, G8Z 4M3, Canada.
| | - Nuwan Sameera Liyanage
- Department of Chemistry, Biochemistry, and Physics, Université du Québec à Trois-Rivières, Trois-Rivières, QC, G8Z 4M3, Canada.
| | - Isabel Desgagné-Penix
- Department of Chemistry, Biochemistry, and Physics, Université du Québec à Trois-Rivières, Trois-Rivières, QC, G8Z 4M3, Canada.
- Plant Biology Research Group, Université du Québec à Trois-Rivières, Trois-Rivières, QC, Canada
| |
Collapse
|
5
|
Joshi G, Das A, Verma G, Guchhait P. Viral infection and host immune response in diabetes. IUBMB Life 2024; 76:242-266. [PMID: 38063433 DOI: 10.1002/iub.2794] [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: 03/17/2023] [Accepted: 11/05/2023] [Indexed: 04/24/2024]
Abstract
Diabetes, a chronic metabolic disorder disrupting blood sugar regulation, has emerged as a prominent silent pandemic. Uncontrolled diabetes predisposes an individual to develop fatal complications like cardiovascular disorders, kidney damage, and neuropathies and aggravates the severity of treatable infections. Escalating cases of Type 1 and Type 2 diabetes correlate with a global upswing in diabetes-linked mortality. As a growing global concern with limited preventive interventions, diabetes necessitates extensive research to mitigate its healthcare burden and assist ailing patients. An altered immune system exacerbated by chronic hyperinflammation heightens the susceptibility of diabetic individuals to microbial infections, including notable viruses like SARS-CoV-2, dengue, and influenza. Given such a scenario, we scrutinized the literature and compiled molecular pathways and signaling cascades related to immune compartments in diabetics that escalate the severity associated with the above-mentioned viral infections in them as compared to healthy individuals. The pathogenesis of these viral infections that trigger diabetes compromises both innate and adaptive immune functions and pre-existing diabetes also leads to heightened disease severity. Lastly, this review succinctly outlines available treatments for diabetics, which may hold promise as preventive or supportive measures to effectively combat these viral infections in the former.
Collapse
Affiliation(s)
- Garima Joshi
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, India
| | - Anushka Das
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, India
| | - Garima Verma
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, India
| | - Prasenjit Guchhait
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, India
| |
Collapse
|
6
|
Sistrom M, Andrews H, Edwards DL. Comparative genomics of Japanese encephalitis virus shows low rates of recombination and a small subset of codon positions under episodic diversifying selection. PLoS Negl Trop Dis 2024; 18:e0011459. [PMID: 38295106 PMCID: PMC10861042 DOI: 10.1371/journal.pntd.0011459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 02/12/2024] [Accepted: 01/11/2024] [Indexed: 02/02/2024] Open
Abstract
Orthoflavivirus japonicum (JEV) is the dominant cause of viral encephalitis in the Asian region with 100,000 cases and 25,000 deaths reported annually. The genome is comprised of a single polyprotein that encodes three structural and seven non-structural proteins. We collated a dataset of 349 complete genomes from a number of public databases, and analysed the data for recombination, evolutionary selection and phylogenetic structure. There are low rates of recombination in JEV, subsequently recombination is not a major evolutionary force shaping JEV. We found a strong overall signal of purifying selection in the genome, which is the main force affecting the evolutionary dynamics in JEV. There are also a small number of genomic sites under episodic diversifying selection, especially in the envelope protein and non-structural proteins 3 and 5. Overall, these results support previous analyses of JEV evolutionary genomics and provide additional insight into the evolutionary processes shaping the distribution and adaptation of this important pathogenic arbovirus.
Collapse
Affiliation(s)
- Mark Sistrom
- Department of Industry, Trade and Tourism, Berrimah Veterinary Laboratories, Darwin, Australia
- Research Institute for the Environment and Livelihoods, Faculty of Science and Technology, Charles Darwin University, Casuarina, Australia
| | - Hannah Andrews
- Department of Industry, Trade and Tourism, Berrimah Veterinary Laboratories, Darwin, Australia
| | - Danielle L. Edwards
- Research Institute for the Environment and Livelihoods, Faculty of Science and Technology, Charles Darwin University, Casuarina, Australia
- Department of Natural Sciences, Museum and Art Gallery of the Northern Territory, Darwin, Australia
| |
Collapse
|
7
|
Akram M, Hameed S, Hassan A, Khan KM. Development in the Inhibition of Dengue Proteases as Drug Targets. Curr Med Chem 2024; 31:2195-2233. [PMID: 37723635 DOI: 10.2174/0929867331666230918110144] [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: 03/20/2023] [Revised: 06/24/2023] [Accepted: 08/04/2023] [Indexed: 09/20/2023]
Abstract
BACKGROUND Viral infections continue to increase morbidity and mortality severely. The flavivirus genus has fifty different species, including the dengue, Zika, and West Nile viruses that can infect 40% of individuals globally, who reside in at least a hundred different countries. Dengue, one of the oldest and most dangerous human infections, was initially documented by the Chinese Medical Encyclopedia in the Jin period. It was referred to as "water poison," connected to flying insects, i.e., Aedes aegypti and Aedes albopictus. DENV causes some medical expressions like dengue hemorrhagic fever, acute febrile illness, and dengue shock syndrome. OBJECTIVE According to the World Health Organization report of 2012, 2500 million people are in danger of contracting dengue fever worldwide. According to a recent study, 96 million of the 390 million dengue infections yearly show some clinical or subclinical severity. There is no antiviral drug or vaccine to treat this severe infection. It can be controlled by getting enough rest, drinking plenty of water, and using painkillers. The first dengue vaccine created by Sanofi, called Dengvaxia, was previously approved by the USFDA in 2019. All four serotypes of the DENV1-4 have shown re-infection in vaccine recipients. However, the usage of Dengvaxia has been constrained by its adverse effects. CONCLUSION Different classes of compounds have been reported against DENV, such as nitrogen-containing heterocycles (i.e., imidazole, pyridine, triazoles quinazolines, quinoline, and indole), oxygen-containing heterocycles (i.e., coumarins), and some are mixed heterocyclic compounds of S, N (thiazole, benzothiazine, and thiazolidinediones), and N, O (i.e., oxadiazole). There have been reports of computationally designed compounds to impede the molecular functions of specific structural and non-structural proteins as potential therapeutic targets. This review summarized the current progress in developing dengue protease inhibitors.
Collapse
Affiliation(s)
- Muhammad Akram
- Department of Chemistry, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Shehryar Hameed
- H.E.J. Research Institute of Chemistry, International Centre for Chemical and Biological Sciences, University of Karachi, Karachi, 75720, Pakistan
| | - Abbas Hassan
- Department of Chemistry, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Khalid Mohammed Khan
- H.E.J. Research Institute of Chemistry, International Centre for Chemical and Biological Sciences, University of Karachi, Karachi, 75720, Pakistan
| |
Collapse
|
8
|
Giri R, Bhardwaj T, Kapuganti SK, Saumya KU, Sharma N, Bhardwaj A, Joshi R, Verma D, Gadhave K. Widespread amyloid aggregates formation by Zika virus proteins and peptides. Protein Sci 2023; 32:e4833. [PMID: 37937856 PMCID: PMC10682691 DOI: 10.1002/pro.4833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 11/01/2023] [Accepted: 11/05/2023] [Indexed: 11/09/2023]
Abstract
Viral pathogenesis typically involves numerous molecular mechanisms. Protein aggregation is a relatively unknown characteristic of viruses, despite the fact that viral proteins have been shown to form terminally misfolded forms. Zika virus (ZIKV) is a neurotropic one with the potential to cause neurodegeneration. Its protein amyloid aggregation may link the neurodegenerative component to the pathogenicity associated with the viral infection. Therefore, we investigated protein aggregation in the ZIKV proteome as a putative pathogenic route and one of the alternate pathways. We discovered that it contains numerous anticipated aggregation-prone regions in this investigation. To validate our prediction, we used a combination of supporting experimental techniques routinely used for morphological characterization and study of amyloid aggregates. Several ZIKV proteins and peptides, including the full-length envelope protein, its domain III (EDIII) and fusion peptide, Pr N-terminal peptide, NS1 β-roll peptide, membrane-embedded signal peptide 2K, and cytosolic region of NS4B protein, were shown to be highly aggregating in our study. Because our findings show that viral proteins can form amyloids in vitro, we need to do a thorough functional study of these anticipated APRs to understand better the role of amyloids in the pathophysiology of ZIKV infection.
Collapse
Affiliation(s)
- Rajanish Giri
- School of Biosciences and BioengineeringIndian Institute of Technology MandiKamandHimachal PradeshIndia
| | - Taniya Bhardwaj
- School of Biosciences and BioengineeringIndian Institute of Technology MandiKamandHimachal PradeshIndia
| | - Shivani K. Kapuganti
- School of Biosciences and BioengineeringIndian Institute of Technology MandiKamandHimachal PradeshIndia
| | - Kumar Udit Saumya
- School of Biosciences and BioengineeringIndian Institute of Technology MandiKamandHimachal PradeshIndia
| | - Nitin Sharma
- Department of Pathology and ImmunologyWashington University School of MedicineSt. LouisMissouriUSA
| | - Aparna Bhardwaj
- School of Biosciences and BioengineeringIndian Institute of Technology MandiKamandHimachal PradeshIndia
| | - Richa Joshi
- School of Biosciences and BioengineeringIndian Institute of Technology MandiKamandHimachal PradeshIndia
| | - Deepanshu Verma
- School of Biosciences and BioengineeringIndian Institute of Technology MandiKamandHimachal PradeshIndia
| | - Kundlik Gadhave
- Department of NeurologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
9
|
Mehta N, Meng Y, Zare R, Kamenetsky-Goldstein R, Sattely E. A developmental gradient reveals biosynthetic pathways to eukaryotic toxins in monocot geophytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.12.540595. [PMID: 37214939 PMCID: PMC10197729 DOI: 10.1101/2023.05.12.540595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Numerous eukaryotic toxins that accumulate in geophytic plants are valuable in the clinic, yet their biosynthetic pathways have remained elusive. A lead example is the >150 Amaryllidaceae alkaloids (AmAs) including galantamine, an FDA-approved treatment for Alzheimer's disease. We show that while AmAs accumulate to high levels in many tissues in daffodils, biosynthesis is localized to nascent, growing tissue at the base of leaves. A similar trend is found for the production of steroidal alkaloids (e.g. cyclopamine) in corn lily. This model of active biosynthesis enabled elucidation of a complete set of biosynthetic genes for the production of AmAs. Taken together, our work sheds light on the developmental and enzymatic logic of diverse alkaloid biosynthesis in daffodil. More broadly, it suggests a paradigm for biosynthesis regulation in monocot geophytes where plants are protected from herbivory through active charging of newly formed cells with eukaryotic toxins that persist as aboveground tissue develops.
Collapse
Affiliation(s)
- Niraj Mehta
- Department of Chemistry, Stanford University, Stanford, CA, 94305, USA
| | - Yifan Meng
- Department of Chemistry, Stanford University, Stanford, CA, 94305, USA
| | - Richard Zare
- Department of Chemistry, Stanford University, Stanford, CA, 94305, USA
| | | | - Elizabeth Sattely
- Department of Chemical Engineering, Stanford University, Stanford, CA, 94305, USA
- HHMI, Stanford University, Stanford, CA 94305
| |
Collapse
|
10
|
Leiva S, Bugnon Valdano M, Gardiol D. Unravelling the epidemiological diversity of Zika virus by analyzing key protein variations. Arch Virol 2023; 168:115. [PMID: 36943525 DOI: 10.1007/s00705-023-05726-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 01/19/2023] [Indexed: 03/23/2023]
Abstract
The consequences of Zika virus (ZIKV) infections were limited to sporadic mild diseases until almost a decade ago, when epidemic outbreaks took place, with quick spread into the Americas. Simultaneously, novel severe neurological manifestations of ZIKV infections were identified, including congenital microcephaly. However, why the epidemic strains behave differently is not yet completely understood, and many questions remain about the actual significance of genetic variations in the epidemiology and biology of ZIKV. In this study, we analysed a large number of viral sequences to identify genes with different levels of variability and patterns of genomic variations that could be associated with ZIKV diversity. We compared numerous epidemic strains with pre-epidemic strains, using the BWA-mem algorithm, and we also examined specific variations among the epidemic ZIKV strains derived from microcephaly cases. We identified several viral genes with dissimilar mutation rates among the ZIKV strain groups and novel protein variation profiles that might be associated with epidemiological particularities. Finally, we assessed the impact of the detected changes on the structure and stability of the NS1, NS5, and E proteins using the I-TASSER, trRosetta, and RaptorX modelling algorithms, and we found some interesting variations that might help to explain the heterogeneous features of the diverse ZIKA strains. This work contributes to the identification of genetic differences in the ZIKV genome that might have a phenotypic impact, providing a basis for future experimental analysis to elucidate the genetic causes of the recent ZIKV emergency.
Collapse
Affiliation(s)
- Santiago Leiva
- Facultad de Ciencias Bioquímicas y Farmacéuticas, Instituto de Biología Molecular y Celular de Rosario-CONICET, Universidad Nacional de Rosario, Suipacha 531, 2000, Rosario, Argentina
| | - Marina Bugnon Valdano
- Facultad de Ciencias Bioquímicas y Farmacéuticas, Instituto de Biología Molecular y Celular de Rosario-CONICET, Universidad Nacional de Rosario, Suipacha 531, 2000, Rosario, Argentina.
| | - Daniela Gardiol
- Facultad de Ciencias Bioquímicas y Farmacéuticas, Instituto de Biología Molecular y Celular de Rosario-CONICET, Universidad Nacional de Rosario, Suipacha 531, 2000, Rosario, Argentina.
| |
Collapse
|
11
|
Nair JJ, van Staden J. Antiviral Effects of the Plant Family Amaryllidaceae. Nat Prod Commun 2023. [DOI: 10.1177/1934578x231162781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023] Open
Abstract
Background In spite of the significant progress in modern medicine, viruses continue to be a formidable challenge to human health. The use of plants for the remediation of viral-borne diseases stretches back to the very dawn of mankind. Among bulbous plants, the Amaryllidaceae is one of the most popular families exploited in the traditional remediation of infectious diseases. Methods This account details the findings from a literature search carried out on the antiviral properties of the Amaryllidaceae. The keyword engaged in the search was “antiviral” in combination with the words “Amaryllidaceae,” “Amaryllidaceae specie,” and “Amaryllidaceae alkaloid.” Results Thirty-six taxa from 13 countries, notably in Africa and parts of Asia, have been cited as traditional remedies for viral diseases. Alcoholic bulb extracts of 18 species were evaluated against 23 different pathogens from 13 viral families. A wide range of activities was observed, with the whole-plant methanol extract of Zephyranthes candida seen to be the most striking (IC50 0.0019 µg/mL against poliovirus). The active principles in the main were isoquinoline alkaloids, of which lycorine impressed against the Avian influenza virus (strain H5N1). The mechanisms underlying the antiviral effects were seen to be related to the inhibition of DNA, RNA, and protein synthesis as well as inhibitory effects toward reverse transcriptase and protease enzymes. Conclusion Amaryllidaceae provides a richly diverse platform for antiviral drug research. Such endeavors have been fortified by the significant amounts of information emerging from indigenous knowledge systems. Ongoing studies will continue to target the active entities, particularly from taxa with verifiable ethnomedicinal backgrounds.
Collapse
Affiliation(s)
- Jerald J Nair
- Research Centre for Plant Growth and Development, School of Life Sciences, University of KwaZulu-Natal Pietermaritzburg, Scottsville, South Africa
| | - Johannes van Staden
- Research Centre for Plant Growth and Development, School of Life Sciences, University of KwaZulu-Natal Pietermaritzburg, Scottsville, South Africa
| |
Collapse
|
12
|
Synthesis and evaluation of anti-yellow fever virus activity of new 6-aryl-3-R-amino-1,2,4-triazin-5(4H)-ones. Eur J Med Chem 2023; 248:115117. [PMID: 36657300 DOI: 10.1016/j.ejmech.2023.115117] [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: 11/08/2022] [Revised: 01/08/2023] [Accepted: 01/10/2023] [Indexed: 01/15/2023]
Abstract
Yellow fever disease is one of public health concerns in the tropics. Despite its significant medicinal and economic impact among large groups of the population, there is a lack of effective treatment against yellow fever. In this regard, here we describe the synthesis of a series of new 6-aryl-3-R-amino-1,2,4-triazin-5(4H)-ones and evaluation of their in vitro inhibitory activity against yellow fever virus. Among all tested compounds 4 derivatives possessing strong inhibitory activity at μM concentrations were identified. All the active compounds revealed a good toxicity profile. These facts make the compounds interesting candidates for further evaluation of their efficacy in the treatment of yellow fever virus infection in vivo.
Collapse
|
13
|
Nair JJ, van Staden J. Antiviral alkaloid principles of the plant family Amaryllidaceae. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 108:154480. [PMID: 36240608 DOI: 10.1016/j.phymed.2022.154480] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 09/18/2022] [Accepted: 09/27/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Viral-borne diseases are amongst the oldest diseases known to mankind. They are responsible for some of the most ravaging effects wrought on human health and well-being. The use of plants against these ailments is entrenched in both traditional and secular medicine around the globe. Their natural abundance and chemical diversity have also boosted their appeal in drug discovery. AIM The plant family Amaryllidaceae is distinguished for its alkaloid principles, some of which are of considerable interest in the clinical arena. This account is the outcome of a literature review undertaken to establish the applicability of these substances as antiviral agents. METHODS The survey utilized the search engines Google Scholar, PubMed, SciFinder, Scopus and Web of Science engaging the word 'antiviral' in conjunction with 'Amaryllidaceae' and 'Amaryllidaceae alkaloid'. The search returned over five hundred hits, of which around eighty were of relevance to the theme of the text. RESULTS Over eighty isoquinoline alkaloids have been screened against nearly fifty pathogens from fourteen viral families, the majority of which were RNA viruses. Potent activities were reported in some instances, such as that of trans-dihydronarciclasine against Yellow fever virus (IC50 0.003 μg/ml), with minimal effects being manifested on host cells. There were also promising results obtained from in vivo studies, in most cases without lethal effects on test subjects. Structure-activity relationship studies afforded useful insight to the antiviral pharmacophore, with the phenanthridone alkaloid nucleus shown to be the most enabling. Although the mechanistic basis to these activities pertained mostly to inhibition of DNA, RNA and protein synthesis, evidence was also forthcoming about the inhibitory action of some of the alkaloids against viral neuraminidase, protease and reverse transcriptase. In silico methods of analysis have offered further perspectives of how some of the alkaloids interact at the active sites of their targets. CONCLUSION The Amaryllidaceae offers a viable platform for plant-based antiviral drug discovery. Its cause is strengthened not only by its wide proliferation and exploitation of its members in alternative forms of medicine, but also by its rich chemical diversity which has already spawned useful antiviral drug leads.
Collapse
Affiliation(s)
- Jerald J Nair
- Research Centre for Plant Growth and Development, School of Life Sciences, University of KwaZulu-Natal Pietermaritzburg, Private Bag X01, Scottsville 3209, South Africa
| | - Johannes van Staden
- Research Centre for Plant Growth and Development, School of Life Sciences, University of KwaZulu-Natal Pietermaritzburg, Private Bag X01, Scottsville 3209, South Africa.
| |
Collapse
|
14
|
Di Lecce R, Mérindol N, Pérez MG, Karimzadegan V, Berthoux L, Boari A, Zidorn C, Vurro M, Surico G, Desgagné-Penix I, Evidente A. Biochemical Analyses of Bioactive Extracts from Plants Native to Lampedusa, Sicily Minor Island. PLANTS (BASEL, SWITZERLAND) 2022; 11:3447. [PMID: 36559555 PMCID: PMC9788634 DOI: 10.3390/plants11243447] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/29/2022] [Accepted: 12/06/2022] [Indexed: 06/17/2023]
Abstract
Major threats to the human lifespan include cancer, infectious diseases, diabetes, mental degenerative conditions and also reduced agricultural productivity due to climate changes, together with new and more devastating plant diseases. From all of this, the need arises to find new biopesticides and new medicines. Plants and microorganisms are the most important sources for isolating new metabolites. Lampedusa Island host a rich contingent of endemic species and subspecies. Seven plant species spontaneously growing in Lampedusa, i.e., Atriplex halimus L. (Ap), Daucus lopadusanus Tineo (Dl), Echinops spinosus Fiori (Es) Glaucium flavum Crantz (Gf) Hypericum aegypticum L: (Ha), Periploca angustifolia Labill (Pa), and Prasium majus L. (Pm) were collected, assessed for their metabolite content, and evaluated for potential applications in agriculture and medicine. The HPLC-MS analysis of n-hexane (HE) and CH2Cl2 (MC) extracts and the residual aqueous phases (WR) showed the presence of several metabolites in both organic extracts. Crude HE and MC extracts from Dl and He significantly inhibited butyrylcholinesterase, as did WR from the extraction of Dl and Pa. HE and MC extracts showed a significant toxicity towards hepatocarcinoma Huh7, while Dl, Ha and Er HE extracts were the most potently cytotoxic to ileocecal colorectal adenocarcinoma HCT-8 cell lines. Most extracts showed antiviral activity. At the lowest concentration tested (1.56 μg/mL), Dl, Gf and Ap MC extracts inhibited betacoronavirus HCoV-OC43 infection by> 2 fold, while the n-hexane extract of Pm was the most potent. In addition, at 1.56 μg/mL, potent inhibition (>10 fold) of dengue virus was detected for Dl, Er, and Pm HE extracts, while Pa and Ap MC extracts dampened infections to undetectable levels. Regarding to phytotoxicity, MC extracts from Er, Ap and Pm were more effective in inhibiting tomato rootlet elongation; the same first two extracts also inhibited seed cress germination while its radicle elongation, due to high sensitivity, was affected by all the extracts. Es and Gf MC extracts also inhibited seed germination of Phelipanche ramosa. Thus, we have uncovered that many of these Lampedusa plants displayed promising biopesticide, antiviral, and biological properties.
Collapse
Affiliation(s)
- Roberta Di Lecce
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario Monte Sant’Angelo, 80126 Napoli, Italy
| | - Natacha Mérindol
- Département de Chimie, Biochimie et Physique, Université du Québec à Trois-Rivières, Trois-Rivières, QC G8Z 4M3, Canada
| | - Mayra Galarza Pérez
- Pharmazeutisches Institut, Abteilung Pharmazeutische Biologie, Christian-Albrechts-Universität zu Kiel, Gutenbergstraße 76, 24118 Kiel, Germany
| | - Vahid Karimzadegan
- Département de Chimie, Biochimie et Physique, Université du Québec à Trois-Rivières, Trois-Rivières, QC G8Z 4M3, Canada
| | - Lionel Berthoux
- Département de Biologie Médicale, Université du Québec à Trois-Rivières, Trois-Rivières, QC G8Z 4M3, Canada
| | - Angela Boari
- Institute of Sciences of Food Production, National Research Council, Via Amendola 122/O, 70125 Bari, Italy
| | - Christian Zidorn
- Pharmazeutisches Institut, Abteilung Pharmazeutische Biologie, Christian-Albrechts-Universität zu Kiel, Gutenbergstraße 76, 24118 Kiel, Germany
| | - Maurizio Vurro
- Institute of Sciences of Food Production, National Research Council, Via Amendola 122/O, 70125 Bari, Italy
| | - Giuseppe Surico
- Department of Agriculture, Food, Environment, and Forestry (DAGRI), Section of Agricultural Microbiology, Plant Pathology and Enthomology, University of Florence, 50121 Firenze, Italy
| | - Isabel Desgagné-Penix
- Département de Chimie, Biochimie et Physique, Université du Québec à Trois-Rivières, Trois-Rivières, QC G8Z 4M3, Canada
| | - Antonio Evidente
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario Monte Sant’Angelo, 80126 Napoli, Italy
- Institute of Sciences of Food Production, National Research Council, Via Amendola 122/O, 70125 Bari, Italy
| |
Collapse
|
15
|
Dos Santos Nascimento IJ, da Silva Rodrigues ÉE, da Silva MF, de Araújo-Júnior JX, de Moura RO. Advances in Computational Methods to Discover New NS2B-NS3 Inhibitors Useful Against Dengue and Zika Viruses. Curr Top Med Chem 2022; 22:2435-2462. [PMID: 36415099 DOI: 10.2174/1568026623666221122121330] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/20/2022] [Accepted: 09/27/2022] [Indexed: 11/24/2022]
Abstract
The Flaviviridae virus family consists of the genera Hepacivirus, Pestivirus, and Flavivirus, with approximately 70 viral types that use arthropods as vectors. Among these diseases, dengue (DENV) and zika virus (ZIKV) serotypes stand out, responsible for thousands of deaths worldwide. Due to the significant increase in cases, the World Health Organization (WHO) declared DENV a potential threat for 2019 due to being transmitted by infected travelers. Furthermore, ZIKV also has a high rate of transmissibility, highlighted in the outbreak in 2015, generating consequences such as Guillain-Barré syndrome and microcephaly. According to clinical outcomes, those infected with DENV can be asymptomatic, and in other cases, it can be lethal. On the other hand, ZIKV has severe neurological symptoms in newborn babies and adults. More serious symptoms include microcephaly, brain calcifications, intrauterine growth restriction, and fetal death. Despite these worrying data, no drug or vaccine is approved to treat these diseases. In the drug discovery process, one of the targets explored against these diseases is the NS2B-NS3 complex, which presents the catalytic triad His51, Asp75, and Ser135, with the function of cleaving polyproteins, with specificity for basic amino acid residues, Lys- Arg, Arg-Arg, Arg-Lys or Gln-Arg. Since NS3 is highly conserved in all DENV serotypes and plays a vital role in viral replication, this complex is an excellent drug target. In recent years, computer-aided drug discovery (CADD) is increasingly essential in drug discovery campaigns, making the process faster and more cost-effective, mainly explained by discovering new drugs against DENV and ZIKV. Finally, the main advances in computational methods applied to discover new compounds against these diseases will be presented here. In fact, molecular dynamics simulations and virtual screening is the most explored approach, providing several hit and lead compounds that can be used in further optimizations. In addition, fragment-based drug design and quantum chemistry/molecular mechanics (QM/MM) provides new insights for developing anti-DENV/ZIKV drugs. We hope that this review offers further helpful information for researchers worldwide and stimulates the use of computational methods to find a promising drug for treating DENV and ZIKV.
Collapse
Affiliation(s)
- Igor José Dos Santos Nascimento
- Department of Pharmacy, Estácio of Alagoas College, Maceió, Brazil.,Department of Pharmacy, Cesmac University Center, Maceió, Brazil.,Department of Pharmacy, Drug Development and Synthesis Laboratory, State University of Paraíba, Campina Grande, Brazil
| | | | - Manuele Figueiredo da Silva
- Laboratory of Medicinal Chemistry, Pharmaceutical Sciences Institute, Federal University of Alagoas, Maceió, Brazil
| | - João Xavier de Araújo-Júnior
- Laboratory of Medicinal Chemistry, Pharmaceutical Sciences Institute, Federal University of Alagoas, Maceió, Brazil
| | - Ricardo Olimpio de Moura
- Department of Pharmacy, Drug Development and Synthesis Laboratory, State University of Paraíba, Campina Grande, Brazil
| |
Collapse
|
16
|
Chemical Synthesis and Biological Activities of Amaryllidaceae Alkaloid Norbelladine Derivatives and Precursors. Molecules 2022; 27:molecules27175621. [PMID: 36080382 PMCID: PMC9457815 DOI: 10.3390/molecules27175621] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/26/2022] [Accepted: 08/28/2022] [Indexed: 11/22/2022] Open
Abstract
Amaryllidaceae alkaloids (AAs) are a structurally diverse family of alkaloids recognized for their many therapeutic properties, such as antiviral, anti-cholinesterase, and anticancer properties. Norbelladine and its derivatives, whose biological properties are poorly studied, are key intermediates required for the biosynthesis of all ~650 reported AAs. To gain insight into their therapeutic potential, we synthesized a series of O-methylated norbelladine-type alkaloids and evaluated their cytotoxic effects on two types of cancer cell lines, their antiviral effects against the dengue virus (DENV) and the human immunodeficiency virus 1 (HIV-1), and their anti-Alzheimer’s disease (anti-cholinesterase and -prolyl oligopeptidase) properties. In monocytic leukemia cells, norcraugsodine was highly cytotoxic (CC50 = 27.0 μM), while norbelladine was the most cytotoxic to hepatocarcinoma cells (CC50 = 72.6 μM). HIV-1 infection was impaired only at cytotoxic concentrations of the compounds. The 3,4-dihydroxybenzaldehyde (selectivity index (SI) = 7.2), 3′,4′-O-dimethylnorbelladine (SI = 4.8), 4′-O-methylnorbelladine (SI > 4.9), 3′-O-methylnorbelladine (SI > 4.5), and norcraugsodine (SI = 3.2) reduced the number of DENV-infected cells with EC50 values ranging from 24.1 to 44.9 μM. The O-methylation of norcraugsodine abolished its anti-DENV potential. Norbelladine and its O-methylated forms also displayed butyrylcholinesterase-inhibition properties (IC50 values ranging from 26.1 to 91.6 μM). Altogether, the results provided hints of the structure−activity relationship of norbelladine-type alkaloids, which is important knowledge for the development of new inhibitors of DENV and butyrylcholinesterase.
Collapse
|
17
|
Muema JM, Bargul JL, Obonyo MA, Njeru SN, Matoke-Muhia D, Mutunga JM. Contemporary exploitation of natural products for arthropod-borne pathogen transmission-blocking interventions. Parasit Vectors 2022; 15:298. [PMID: 36002857 PMCID: PMC9404607 DOI: 10.1186/s13071-022-05367-8] [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: 02/24/2022] [Accepted: 06/16/2022] [Indexed: 11/26/2022] Open
Abstract
An integrated approach to innovatively counter the transmission of various arthropod-borne diseases to humans would benefit from strategies that sustainably limit onward passage of infective life cycle stages of pathogens and parasites to the insect vectors and vice versa. Aiming to accelerate the impetus towards a disease-free world amid the challenges posed by climate change, discovery, mindful exploitation and integration of active natural products in design of pathogen transmission-blocking interventions is of high priority. Herein, we provide a review of natural compounds endowed with blockade potential against transmissible forms of human pathogens reported in the last 2 decades from 2000 to 2021. Finally, we propose various translational strategies that can exploit these pathogen transmission-blocking natural products into design of novel and sustainable disease control interventions. In summary, tapping these compounds will potentially aid in integrated combat mission to reduce disease transmission trends.
Collapse
Affiliation(s)
- Jackson M Muema
- Department of Biochemistry, Jomo Kenyatta University of Agriculture and Technology (JKUAT), P.O. Box 62000, Nairobi, 00200, Kenya.
| | - Joel L Bargul
- Department of Biochemistry, Jomo Kenyatta University of Agriculture and Technology (JKUAT), P.O. Box 62000, Nairobi, 00200, Kenya.,International Centre of Insect Physiology and Ecology (Icipe), P.O. Box 30772, Nairobi, 00100, Kenya
| | - Meshack A Obonyo
- Department of Biochemistry and Molecular Biology, Egerton University, P.O. Box 536, Egerton, 20115, Kenya
| | - Sospeter N Njeru
- Centre for Traditional Medicine and Drug Research (CTMDR), Kenya Medical Research Institute (KEMRI), P.O. Box 54840, Nairobi, 00200, Kenya
| | - Damaris Matoke-Muhia
- Centre for Biotechnology Research Development (CBRD), Kenya Medical Research Institute (KEMRI), P.O. Box 54840, Nairobi, 00200, Kenya
| | - James M Mutunga
- Department of Biological Sciences, Mount Kenya University (MKU), P.O. Box 54, Thika, 01000, Kenya.,School of Engineering Design, Technology and Professional Programs, Pennsylvania State University, University Park, PA, 16802, USA
| |
Collapse
|
18
|
He J, Huang H, Li B, Li H, Zhao Y, Li Y, Ye W, Qi W, Tang W, Wang L. Identification of cytochrome c oxidase subunit 4 isoform 1 as a positive regulator of influenza virus replication. Front Microbiol 2022; 13:862205. [PMID: 35928150 PMCID: PMC9343726 DOI: 10.3389/fmicb.2022.862205] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 06/28/2022] [Indexed: 11/14/2022] Open
Abstract
Human infection with highly pathogenic H5N1 influenza virus causes severe respiratory diseases. Currently, the drugs against H5N1 are limited to virus-targeted inhibitors. However, drug resistance caused by these inhibitors is becoming a serious threat to global public health. An alternative strategy to reduce the resistance risk is to develop antiviral drugs targeting host cell proteins. In this study, we demonstrated that cytochrome c oxidase subunit 4 isoform 1 (COX41) of host cell plays an important role in H5N1 infection. Overexpression of COX41 promoted viral replication, which was inhibited by silencing or knockout the expression of COX41 in the host cell. The ribonucleoproteins (RNPs) of H5N1 were retained in the cell nucleus after knockout cellular COX41. Strikingly, inhibition of cellular COX41 by lycorine, a small-molecule compound isolated from Amaryllidaceae plants, reduced the levels of COX41-induced ROS and phosphorylation of extracellular signal-regulated kinase (ERK) in cells, thus resulting in the blockage of nuclear export of vRNP and inhibition of viral replication. In H5N1-infected mice that were treated with lycorine, we observed a reduction of viral titers and inhibition of pathological changes in the lung and trachea tissues. Importantly, no resistant virus was generated after culturing the virus with the continuous treatment of lycorine. Collectively, these findings suggest that COX41 is a positive regulator of H5N1 replication and might serve as an alternative target for anti-influenza drug development.
Collapse
Affiliation(s)
- Jun He
- Center for Bioactive Natural Molecules and Innovative Drugs Research, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
- Institute of Laboratory Animal Science, Jinan University, Guangzhou, China
| | - Huibin Huang
- Center for Bioactive Natural Molecules and Innovative Drugs Research, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
- Pharmacy Department, Wenzhou People’s Hospital, Wenzhou, China
| | - Bo Li
- National Avian Influenza Professional Laboratory, Key Laboratory of Zoonoses, Ministry of Agriculture, South China Agricultural University, Guangzhou, China
- Chongqing Academy of Animal Sciences, Chongqing, China
| | - Huanan Li
- National Avian Influenza Professional Laboratory, Key Laboratory of Zoonoses, Ministry of Agriculture, South China Agricultural University, Guangzhou, China
| | - Yue Zhao
- Institute of Laboratory Animal Science, Jinan University, Guangzhou, China
| | - Yaolan Li
- Center for Bioactive Natural Molecules and Innovative Drugs Research, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Wencai Ye
- Center for Bioactive Natural Molecules and Innovative Drugs Research, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Wenbao Qi
- National Avian Influenza Professional Laboratory, Key Laboratory of Zoonoses, Ministry of Agriculture, South China Agricultural University, Guangzhou, China
| | - Wei Tang
- Center for Bioactive Natural Molecules and Innovative Drugs Research, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
- *Correspondence: Lei Wang, Wei Tang,
| | - Lei Wang
- Center for Bioactive Natural Molecules and Innovative Drugs Research, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
- *Correspondence: Lei Wang, Wei Tang,
| |
Collapse
|
19
|
Qian X, Qi Z. Mosquito-Borne Flaviviruses and Current Therapeutic Advances. Viruses 2022; 14:v14061226. [PMID: 35746697 PMCID: PMC9229039 DOI: 10.3390/v14061226] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/25/2022] [Accepted: 06/02/2022] [Indexed: 12/10/2022] Open
Abstract
Mosquito-borne flavivirus infections affect approximately 400 million people worldwide each year and are global threats to public health. The common diseases caused by such flaviviruses include West Nile, yellow fever, dengue, Zika infection and Japanese encephalitis, which may result in severe symptoms and disorders of multiple organs or even fatal outcomes. Till now, no specific antiviral agents are commercially available for the treatment of the diseases. Numerous strategies have been adopted to develop novel and promising inhibitors against mosquito-borne flaviviruses, including drugs targeting the critical viral components or essential host factors during infection. Research advances in antiflaviviral therapy might optimize and widen the treatment options for flavivirus infection. This review summarizes the current developmental progresses and involved molecular mechanisms of antiviral agents against mosquito-borne flaviviruses.
Collapse
|
20
|
Narayanan A, Narwal M, Majowicz SA, Varricchio C, Toner SA, Ballatore C, Brancale A, Murakami KS, Jose J. Identification of SARS-CoV-2 inhibitors targeting Mpro and PLpro using in-cell-protease assay. Commun Biol 2022; 5:169. [PMID: 35217718 PMCID: PMC8881501 DOI: 10.1038/s42003-022-03090-9] [Citation(s) in RCA: 110] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 02/01/2022] [Indexed: 01/08/2023] Open
Abstract
SARS-CoV-2 proteases Mpro and PLpro are promising targets for antiviral drug development. In this study, we present an antiviral screening strategy involving a novel in-cell protease assay, antiviral and biochemical activity assessments, as well as structural determinations for rapid identification of protease inhibitors with low cytotoxicity. We identified eight compounds with anti-SARS-CoV-2 activity from a library of 64 repurposed drugs and modeled at protease active sites by in silico docking. We demonstrate that Sitagliptin and Daclatasvir inhibit PLpro, and MG-101, Lycorine HCl, and Nelfinavir mesylate inhibit Mpro of SARS-CoV-2. The X-ray crystal structure of Mpro in complex with MG-101 shows a covalent bond formation between the inhibitor and the active site Cys145 residue indicating its mechanism of inhibition is by blocking the substrate binding at the active site. Thus, we provide methods for rapid and effective screening and development of inhibitors for blocking virus polyprotein processing as SARS-CoV-2 antivirals. Additionally, we show that the combined inhibition of Mpro and PLpro is more effective in inhibiting SARS-CoV-2 and the delta variant.
Collapse
Affiliation(s)
- Anoop Narayanan
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, USA
| | - Manju Narwal
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, USA
| | - Sydney A Majowicz
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Carmine Varricchio
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, CF10 3NB, Cardiff, UK
| | - Shay A Toner
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, USA
| | - Carlo Ballatore
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Andrea Brancale
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, CF10 3NB, Cardiff, UK
| | - Katsuhiko S Murakami
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, USA
| | - Joyce Jose
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, USA.
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, USA.
| |
Collapse
|
21
|
Ren PX, Shang WJ, Yin WC, Ge H, Wang L, Zhang XL, Li BQ, Li HL, Xu YC, Xu EH, Jiang HL, Zhu LL, Zhang LK, Bai F. A multi-targeting drug design strategy for identifying potent anti-SARS-CoV-2 inhibitors. Acta Pharmacol Sin 2022; 43:483-493. [PMID: 33907306 PMCID: PMC8076879 DOI: 10.1038/s41401-021-00668-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/22/2021] [Indexed: 02/02/2023] Open
Abstract
The COVID-19, caused by SARS-CoV-2, is threatening public health, and there is no effective treatment. In this study, we have implemented a multi-targeted anti-viral drug design strategy to discover highly potent SARS-CoV-2 inhibitors, which simultaneously act on the host ribosome, viral RNA as well as RNA-dependent RNA polymerases, and nucleocapsid protein of the virus, to impair viral translation, frameshifting, replication, and assembly. Driven by this strategy, three alkaloids, including lycorine, emetine, and cephaeline, were discovered to inhibit SARS-CoV-2 with EC50 values of low nanomolar levels potently. The findings in this work demonstrate the feasibility of this multi-targeting drug design strategy and provide a rationale for designing more potent anti-virus drugs.
Collapse
Affiliation(s)
- Peng-Xuan Ren
- School of Life Science and Technology, and Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| | - Wei-Juan Shang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Wan-Chao Yin
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Huan Ge
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Lin Wang
- School of Life Science and Technology, and Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| | - Xiang-Lei Zhang
- School of Life Science and Technology, and Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| | - Bing-Qian Li
- School of Life Science and Technology, and Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
- Department of Chemistry, Imperial College London, London, United Kingdom
| | - Hong-Lin Li
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Ye-Chun Xu
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Eric H Xu
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hua-Liang Jiang
- School of Life Science and Technology, and Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Li-Li Zhu
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China.
| | - Lei-Ke Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China.
| | - Fang Bai
- School of Life Science and Technology, and Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China.
| |
Collapse
|
22
|
Tan S, Banwell MG, Ye WC, Lan P, White LV. The Inhibition of RNA Viruses by Amaryllidaceae Alkaloids: Opportunities for the Development of Broad-Spectrum Anti-Coronavirus Drugs. Chem Asian J 2022; 17:e202101215. [PMID: 35032358 DOI: 10.1002/asia.202101215] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 12/06/2021] [Indexed: 12/16/2022]
Abstract
The global COVID-19 pandemic has claimed the lives of millions and disrupted nearly every aspect of human society. Currently, vaccines remain the only widely available medical means to address the cause of the pandemic, the SARS-CoV-2 virus. Unfortunately, current scientific consensus deems the emergence of vaccine-resistant SARS-CoV-2 variants highly likely. In this context, the design and development of broad-spectrum, small-molecule based antiviral drugs has been described as a potentially effective, alternative medical strategy to address circulating and re-emerging CoVs. Small molecules are well-suited to target the least-rapidly evolving structures within CoVs such as highly conserved RNA replication enzymes, and this renders them less vulnerable to evolved drug resistance. Examination of the vast literature describing the inhibition of RNA viruses by Amaryllidaceae alkaloids suggests that future, broad-spectrum anti-CoV drugs may be derived from this family of natural products.
Collapse
Affiliation(s)
- Shen Tan
- The Institute for Advanced and Applied Chemical Synthesis, Jinan University, Guangzhou, 510632, P. R. China
| | - Martin G Banwell
- The Institute for Advanced and Applied Chemical Synthesis, Jinan University, Guangzhou, 510632, P. R. China
| | - Wen-Cai Ye
- College of Pharmacy, Jinan University, Guangzhou, 510632, P. R. China
| | - Ping Lan
- The Institute for Advanced and Applied Chemical Synthesis, Jinan University, Guangzhou, 510632, P. R. China
| | - Lorenzo V White
- The Institute for Advanced and Applied Chemical Synthesis, Jinan University, Guangzhou, 510632, P. R. China
| |
Collapse
|
23
|
Biological Investigation of Amaryllidaceae Alkaloid Extracts from the Bulbs of Pancratium trianthum Collected in the Senegalese Flora. Molecules 2021; 26:molecules26237382. [PMID: 34885964 PMCID: PMC8659059 DOI: 10.3390/molecules26237382] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/01/2021] [Accepted: 12/03/2021] [Indexed: 11/29/2022] Open
Abstract
Amaryllidaceae plants are rich in alkaloids with biological properties. Pancratium trianthum is an Amaryllidaceae species widely used in African folk medicine to treat several diseases such as central nervous system disorders, tumors, and microbial infections, and it is used to heal wounds. The current investigation explored the biological properties of alkaloid extracts from bulbs of P. trianthum collected in the Senegalese flora. Alkaloid extracts were analyzed and identified by chromatography and mass spectrometry. Alkaloid extracts from P. trianthum displayed pleiotropic biological properties. Cytotoxic activity of the extracts was determined on hepatocarcinoma Huh7 cells and on acute monocytic leukemia THP-1 cells, while agar diffusion and microdilution assays were used to evaluate antibacterial activity. Antiviral activity was measured by infection of extract-treated cells with dengue virus (DENVGFP) and human immunodeficiency virus-1 (HIV-1GFP) reporter vectors. Cytotoxicity and viral inhibition were the most striking of P. trianthum’s extract activities. Importantly, non-cytotoxic concentrations were highly effective in completely preventing DENVGFP replication and in reducing pseudotyped HIV-1GFP infection levels. Our results show that P. trianthum is a rich source of molecules for the potential discovery of new treatments against various diseases. Herein, we provide scientific evidence to rationalize the traditional uses of P. trianthum for wound treatment as an anti-dermatosis and antiseptic agent.
Collapse
|
24
|
Antiviral and virucidal activities of lycorine on duck tembusu virus in vitro by blocking viral internalization and entry. Poult Sci 2021; 100:101404. [PMID: 34478911 PMCID: PMC8414183 DOI: 10.1016/j.psj.2021.101404] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 07/14/2021] [Accepted: 07/21/2021] [Indexed: 11/21/2022] Open
Abstract
Duck tembusu virus (DTMUV) was firstly identified in 2010 in China; since then, it has caused enormous economic loss to breeding industry. Great efforts have been made to develop drugs and vaccines against DTMUV. However, current available vaccines or anti-DTMUV drugs are consistently inefficient. Hence, various more broadly effective drugs have become important for the treatment of DTMUV infection; among these, lycorine, one of the important sources of active alkaloids, is a promising example. Nevertheless, it is not known whether lycorine has any antiviral activities against DTMUV. Therefore, the purpose of the present study is to investigate the anti-DTMUV abilities of lycorine. The cytotoxicity of lycorine was evaluated on BHK-21 cells by CCK-8 assay, and its antiviral effect against DTMUV was examined by real-time PCR assays, virus titer determination, Western blot and immunofluorescence (IFA) assays, respectively. Furthermore, the underlying mechanisms of the anti-DTMUV effects of lycorine were also investigated. The results indicated that the highest nontoxicity concentration of lycorine on BHK-21 cells was 5 µM. Lycorine possessed the antiviral ability against DTMUV on BHK-21 cells, as demonstrated by the reduction of virus titers and copy numbers in vitro. Western blot and IFA analysis showed the inhibitory effect of lycorine on DTMUV envelope (E) protein expression. Moreover, using time-of-addition assays, we found that lycorine displays its antivirus and virucidal activities through blocking viral internalization and entry in vitro. Taken together, our findings firstly demonstrate the antiviral activities of lycorine against DTMUV, suggesting that lycorine can be a potential drug for the treatment of DTMUV infection.
Collapse
|
25
|
Amaryllidaceae Alkaloid Cherylline Inhibits the Replication of Dengue and Zika Viruses. Antimicrob Agents Chemother 2021; 65:e0039821. [PMID: 34152811 PMCID: PMC8370201 DOI: 10.1128/aac.00398-21] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Dengue fever, caused by dengue virus (DENV), is the most prevalent arthropod-borne viral disease and is endemic in many tropical and subtropical parts of the world, with an increasing incidence in temperate regions. The closely related flavivirus Zika virus (ZIKV) can be transmitted vertically in utero and causes congenital Zika syndrome and other birth defects. In adults, ZIKV is associated with Guillain-Barré syndrome. There are no approved antiviral therapies against either virus. Effective antiviral compounds are urgently needed. Amaryllidaceae alkaloids (AAs) are a specific class of nitrogen-containing compounds produced by plants of the Amaryllidaceae family with numerous biological activities. Recently, the AA lycorine was shown to present strong antiflaviviral properties. Previously, we demonstrated that Crinum jagus contained lycorine and several alkaloids of the cherylline, crinine, and galanthamine types with unknown antiviral potential. In this study, we explored their biological activities. We show that C. jagus crude alkaloid extract inhibited DENV infection. Among the purified AAs, cherylline efficiently inhibited both DENV (50% effective concentration [EC50], 8.8 μM) and ZIKV replication (EC50, 20.3 μM) but had no effect on HIV-1 infection. Time-of-drug-addition and -removal experiments identified a postentry step as the one targeted by cherylline. Consistently, using subgenomic replicons and replication-defective genomes, we demonstrate that cherylline specifically hinders the viral RNA synthesis step but not viral translation. In conclusion, AAs are an underestimated source of antiflavivirus compounds, including the effective inhibitor cherylline, which could be optimized for new therapeutic approaches.
Collapse
|
26
|
Li N, Wang Z, Wang R, Zhang ZR, Zhang YN, Deng CL, Zhang B, Shang LQ, Ye HQ. In Vitro Inhibition of Alphaviruses by Lycorine. Virol Sin 2021; 36:1465-1474. [PMID: 34374926 PMCID: PMC8353614 DOI: 10.1007/s12250-021-00438-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 06/08/2021] [Indexed: 11/25/2022] Open
Abstract
Chikungunya virus (CHIKV) is a mosquito-borne alphavirus. As an emerging virus, CHIKV imposes a threat to public health. Currently, there are no vaccines or antivirals available for the prevention of CHIKV infection. Lycorine, an alkaloid from Amaryllidaceae plants, has antiviral activity against a number of viruses such as coronavirus, flavivirus and enterovirus. In this study, we found that lycorine could inhibit CHIKV in cell culture at a concentration of 10 μmol/L without apparent cytotoxicity. In addition, it exhibited broad-spectrum anti-alphavirus activity, including Sindbis virus (SINV), Semliki Forest virus (SFV), and Venezuelan equine encephalomyelitis virus (VEEV). The time of addition studies indicated that lycorine functions at an early post-entry stage of CHIKV life cycle. The results based on two different CHIKV replicons provided further evidence that lycorine exerts its antiviral activity mainly by inhibiting CHIKV translation. Overall, our study extends the antiviral spectrum of lycorine.
Collapse
Affiliation(s)
- Na Li
- Key Laboratory of Special Pathogens and Biosafety, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Zhen Wang
- Key Laboratory of Special Pathogens and Biosafety, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Rui Wang
- College of Pharmacy and Drug Discovery Center for Infectious Diseases, Nankai University, Tianjin, 300350, China
| | - Zhe-Rui Zhang
- Key Laboratory of Special Pathogens and Biosafety, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Ya-Nan Zhang
- Key Laboratory of Special Pathogens and Biosafety, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Cheng-Lin Deng
- Key Laboratory of Special Pathogens and Biosafety, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Bo Zhang
- Key Laboratory of Special Pathogens and Biosafety, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China. .,College of Pharmacy and Drug Discovery Center for Infectious Diseases, Nankai University, Tianjin, 300350, China.
| | - Lu-Qing Shang
- College of Pharmacy and Drug Discovery Center for Infectious Diseases, Nankai University, Tianjin, 300350, China.
| | - Han-Qing Ye
- Key Laboratory of Special Pathogens and Biosafety, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.
| |
Collapse
|
27
|
Insights on Dengue and Zika NS5 RNA-dependent RNA polymerase (RdRp) inhibitors. Eur J Med Chem 2021; 224:113698. [PMID: 34274831 DOI: 10.1016/j.ejmech.2021.113698] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/09/2021] [Accepted: 07/10/2021] [Indexed: 11/20/2022]
Abstract
Over recent years, many outbreaks caused by (re)emerging RNA viruses have been reported worldwide, including life-threatening Flaviviruses, such as Dengue (DENV) and Zika (ZIKV). Currently, there is only one licensed vaccine against Dengue, Dengvaxia®. However, its administration is not recommended for children under nine years. Still, there are no specific inhibitors available to treat these infectious diseases. Among the flaviviral proteins, NS5 RNA-dependent RNA polymerase (RdRp) is a metalloenzyme essential for viral replication, suggesting that it is a promising macromolecular target since it has no human homolog. Nowadays, several NS5 RdRp inhibitors have been reported, while none inhibitors are currently in clinical development. In this context, this review constitutes a comprehensive work focused on RdRp inhibitors from natural, synthetic, and even repurposing sources. Furthermore, their main aspects associated with the structure-activity relationship (SAR), proposed mechanisms of action, computational studies, and other topics will be discussed in detail.
Collapse
|
28
|
Zhang W, Zeng M, Jiang B, Lu T, Guo J, Hu T, Wang M, Jia R, Zhu D, Liu M, Zhao X, Yang Q, Wu Y, Zhang S, Ou X, Liu Y, Zhang L, Yu Y, Pan L, Cheng A, Chen S. Amelioration of Beta Interferon Inhibition by NS4B Contributes to Attenuating Tembusu Virus Virulence in Ducks. Front Immunol 2021; 12:671471. [PMID: 34079553 PMCID: PMC8165282 DOI: 10.3389/fimmu.2021.671471] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 05/04/2021] [Indexed: 11/13/2022] Open
Abstract
Our previous studies reported that duck Tembusu virus nonstructural protein 2A (NS2A) is a major inhibitor of the IFNβ signaling pathway through competitively binding to STING with TBK1, leading to a reduction in TBK1 phosphorylation. Duck TMUV NS2B3 could cleave and bind STING to subvert the IFNβ signaling pathway. Here, we found that overexpression of duck TMUV NS4B could compete with TBK1 in binding to STING, reducing TBK1 phosphorylation and inhibiting the IFNβ signaling pathway by using the Dual-Glo® Luciferase Assay System and the NanoBiT protein-protein interaction (PPI) assay. We further identified the E2, M3, G4, W5, K10 and D34 residues in NS4B that were important for its interaction with STING and its inhibition of IFNβ induction, which were subsequently introduced into a duck TMUV replicon and an infectious cDNA clone. We found that the NS4B M3A mutant enhanced RNA replication and exhibited significantly higher titer levels than WT at 48-72 hpi but significantly decreased mortality (80%) in duck embryos compared to WT (100%); the NS4B G4A and R36A mutants slightly reduced RNA replication but exhibited the same titer levels as WT. However, the NS4B R36A mutant did not attenuate the virulence in duck embryos, whereas the G4A mutant significantly decreased the mortality (70%) of duck embryos. In addition, the NS4B W5A mutant did not affect viral replication, whereas the D34A mutant slightly reduced RNA replication, and both mutants exhibited significantly lower titer levels than the WT and significantly decreased mortality (90% and 70%, respectively) in duck embryos. Hence, our findings provide new insight into the development of attenuated flaviviruses by targeting the disabling viral strategies used to evade the innate defense mechanisms.
Collapse
Affiliation(s)
- Wei Zhang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
| | - Miao Zeng
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
| | - Bowen Jiang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
| | - Tong Lu
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
| | - Jiaqi Guo
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
| | - Tao Hu
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
| | - Mingshu Wang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China.,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu City, China
| | - Renyong Jia
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China.,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu City, China
| | - Dekang Zhu
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China.,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu City, China
| | - Mafeng Liu
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China.,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu City, China
| | - Xinxin Zhao
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China.,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu City, China
| | - Qiao Yang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China.,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu City, China
| | - Ying Wu
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China.,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu City, China
| | - Shaqiu Zhang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China.,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu City, China
| | - Xumin Ou
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China.,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu City, China
| | - Yunya Liu
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
| | - Ling Zhang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
| | - Yanling Yu
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
| | - Leichang Pan
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
| | - Anchun Cheng
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China.,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu City, China
| | - Shun Chen
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China.,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu City, China
| |
Collapse
|
29
|
Bakoa F, Préhaud C, Beauclair G, Chazal M, Mantel N, Lafon M, Jouvenet N. Genomic diversity contributes to the neuroinvasiveness of the Yellow fever French neurotropic vaccine. NPJ Vaccines 2021; 6:64. [PMID: 33903598 PMCID: PMC8076279 DOI: 10.1038/s41541-021-00318-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 03/15/2021] [Indexed: 02/02/2023] Open
Abstract
Mass vaccination with the live attenuated vaccine YF-17D is the current way to prevent infection with Yellow fever virus (YFV). However, 0.000012-0.00002% of vaccinated patients develop post-vaccination neurological syndrome (YEL-AND). Understanding the factors responsible for neuroinvasion, neurotropism, and neurovirulence of the vaccine is critical for improving its biosafety. The YF-FNV vaccine strain, known to be associated with a higher frequency of YEL-AND (0.3-0.4%) than YF-17D, is an excellent model to study vaccine neuroinvasiveness. We determined that neuroinvasiveness of YF-FNV occured both via infection and passage through human brain endothelial cells. Plaque purification and next generation sequencing (NGS) identified several neuroinvasive variants. Their neuroinvasiveness was not higher than that of YF-FNV. However, rebuilding the YF-FNV population diversity from a set of isolated YF-FNV-N variants restored the original neuroinvasive phenotype of YF-FNV. Therefore, we conclude that viral population diversity is a critical factor for YFV vaccine neuroinvasiveness.
Collapse
Affiliation(s)
- Florian Bakoa
- Unité de Neuroimmunologie Virale, Institut Pasteur, Paris, France
- Research and External Innovation Department, Sanofi Pasteur, Marcy L'Etoile, France
- Sorbonne Université, Collège doctoral, Paris, France
- Unité de Signalisation Antivirale, CNRS UMR 3569, Institut Pasteur, Paris, France
| | | | - Guillaume Beauclair
- Unité de Signalisation Antivirale, CNRS UMR 3569, Institut Pasteur, Paris, France
- Institut de Biologie Intégrative de la Cellule, UMR9198, Équipe Autophagie et Immunité Antivirale, Faculté de Pharmacie, Châtenay-Malabry, France
| | - Maxime Chazal
- Unité de Signalisation Antivirale, CNRS UMR 3569, Institut Pasteur, Paris, France
| | - Nathalie Mantel
- Research and External Innovation Department, Sanofi Pasteur, Marcy L'Etoile, France
| | - Monique Lafon
- Unité de Neuroimmunologie Virale, Institut Pasteur, Paris, France.
| | - Nolwenn Jouvenet
- Unité de Signalisation Antivirale, CNRS UMR 3569, Institut Pasteur, Paris, France.
| |
Collapse
|
30
|
|
31
|
Mandal A, Jha AK, Hazra B. Plant Products as Inhibitors of Coronavirus 3CL Protease. Front Pharmacol 2021; 12:583387. [PMID: 33767619 PMCID: PMC7985176 DOI: 10.3389/fphar.2021.583387] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 01/19/2021] [Indexed: 12/23/2022] Open
Abstract
Background: The ongoing COVID-19 pandemic has created an alarming situation due to extensive loss of human lives and economy, posing enormous threat to global health security. Till date, no antiviral drug or vaccine against SARS-CoV-2 has reached the market, although a number of clinical trials are under way. The viral 3-chymotrypsin-like cysteine protease (3CLpro), playing pivotal roles in coronavirus replication and polyprotein processing, is essential for its life cycle. In fact, 3CLpro is already a proven drug discovery target for SARS- and MERS-CoVs. This underlines the importance of 3CL protease in the design of potent drugs against COVID-19. Methods: We have collected one hundred twenty-seven relevant literatures to prepare the review article. PubMed, Google Scholar and other scientific search engines were used to collect the literature based on keywords, like "SARS-CoVs-3CL protease," "medicinal plant and anti-SARS-CoVs-3CL protease" published during 2003-2020. However, earlier publications related to this topic are also cited for necessary illustration and discussion. Repetitive articles and non-English studies were excluded. Results: From the literature search, we have enlisted medicinal plants reported to inhibit coronavirus 3CL protease. Some of the plants like Isatis tinctoria L. (syn. Isatis indigotica Fort.), Torreya nucifera (L.) Siebold and Zucc., Psoralea corylifolia L., and Rheum palmatum L. have exhibited strong anti-3CLpro activity. We have also discussed about the phytochemicals with encouraging antiviral activity, such as, bavachinin, psoralidin, betulinic acid, curcumin and hinokinin, isolated from traditional medicinal plants. Conclusion: Currently, searching for a plant-derived novel drug with better therapeutic index is highly desirable due to lack of specific treatment for SARS-CoV-2. It is expected that in-depth evaluation of medicinally important plants would reveal new molecules with significant potential to inhibit coronavirus 3CL protease for development into approved antiviral drug against COVID-19 in future.
Collapse
Affiliation(s)
- Anirban Mandal
- Department of Microbiology, Mrinalini Datta Mahavidyapith, Kolkata, India
| | - Ajeet Kumar Jha
- Animal Health Research Division, Nepal Agricultural Research Council, Kathmandu, Nepal
| | - Banasri Hazra
- Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| |
Collapse
|
32
|
Parthasarathy A, Borrego EJ, Savka MA, Dobson RCJ, Hudson AO. Amino acid-derived defense metabolites from plants: A potential source to facilitate novel antimicrobial development. J Biol Chem 2021; 296:100438. [PMID: 33610552 PMCID: PMC8024917 DOI: 10.1016/j.jbc.2021.100438] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 02/16/2021] [Accepted: 02/17/2021] [Indexed: 12/23/2022] Open
Abstract
For millennia, humanity has relied on plants for its medicines, and modern pharmacology continues to reexamine and mine plant metabolites for novel compounds and to guide improvements in biological activity, bioavailability, and chemical stability. The critical problem of antibiotic resistance and increasing exposure to viral and parasitic diseases has spurred renewed interest into drug treatments for infectious diseases. In this context, an urgent revival of natural product discovery is globally underway with special attention directed toward the numerous and chemically diverse plant defensive compounds such as phytoalexins and phytoanticipins that combat herbivores, microbial pathogens, or competing plants. Moreover, advancements in “omics,” chemistry, and heterologous expression systems have facilitated the purification and characterization of plant metabolites and the identification of possible therapeutic targets. In this review, we describe several important amino acid–derived classes of plant defensive compounds, including antimicrobial peptides (e.g., defensins, thionins, and knottins), alkaloids, nonproteogenic amino acids, and phenylpropanoids as potential drug leads, examining their mechanisms of action, therapeutic targets, and structure–function relationships. Given their potent antibacterial, antifungal, antiparasitic, and antiviral properties, which can be superior to existing drugs, phytoalexins and phytoanticipins are an excellent resource to facilitate the rational design and development of antimicrobial drugs.
Collapse
Affiliation(s)
- Anutthaman Parthasarathy
- Rochester Institute of Technology, Thomas H. Gosnell School of Life Sciences, Rochester, New York, USA
| | - Eli J Borrego
- Rochester Institute of Technology, Thomas H. Gosnell School of Life Sciences, Rochester, New York, USA
| | - Michael A Savka
- Rochester Institute of Technology, Thomas H. Gosnell School of Life Sciences, Rochester, New York, USA
| | - Renwick C J Dobson
- Biomolecular Interaction Centre and School of Biological Sciences, University of Canterbury, Christchurch, New Zealand; Bio21 Molecular Science and Biotechnology Institute, Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria, Australia
| | - André O Hudson
- Rochester Institute of Technology, Thomas H. Gosnell School of Life Sciences, Rochester, New York, USA.
| |
Collapse
|
33
|
Scroggs SLP, Gass JT, Chinnasamy R, Widen SG, Azar SR, Rossi SL, Arterburn JB, Vasilakis N, Hanley KA. Evolution of resistance to fluoroquinolones by dengue virus serotype 4 provides insight into mechanism of action and consequences for viral fitness. Virology 2021; 552:94-106. [PMID: 33120225 PMCID: PMC7528753 DOI: 10.1016/j.virol.2020.09.004] [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] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 08/30/2020] [Accepted: 09/08/2020] [Indexed: 02/07/2023]
Abstract
Drugs against flaviviruses such as dengue (DENV) and Zika (ZIKV) virus are urgently needed. We previously demonstrated that three fluoroquinolones, ciprofloxacin, enoxacin, and difloxacin, suppress replication of six flaviviruses. To investigate the barrier to resistance and mechanism(s) of action of these drugs, DENV-4 was passaged in triplicate in HEK-293 cells in the presence or absence of each drug. Resistance to ciprofloxacin was detected by the seventh passage and to difloxacin by the tenth, whereas resistance to enoxacin did not occur within ten passages. Two putative resistance-conferring mutations were detected in the envelope gene of ciprofloxacin and difloxacin-resistant DENV-4. In the absence of ciprofloxacin, ciprofloxacin-resistant viruses sustained a significantly higher viral titer than control viruses in HEK-293 and HuH-7 cells and resistant viruses were more stable than control viruses at 37 °C. These results suggest that the mechanism of action of ciprofloxacin and difloxacin involves interference with virus binding or entry.
Collapse
Affiliation(s)
- Stacey L P Scroggs
- Department of Biology, New Mexico State University, Las Cruces, NM, USA.
| | - Jordan T Gass
- Department of Biology, New Mexico State University, Las Cruces, NM, USA
| | - Ramesh Chinnasamy
- Department of Chemistry and Biochemistry, New Mexico State University, Las Cruces, NM, USA
| | - Steven G Widen
- Department of Biochemistry & Molecular Biology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Sasha R Azar
- Department of Pathology, The University of University of Texas Medical Branch, Galveston, TX, USA
| | - Shannan L Rossi
- Department of Pathology, The University of University of Texas Medical Branch, Galveston, TX, USA; Institute for Human Infection and Immunity, The University of University of Texas Medical Branch, Galveston, TX, USA
| | - Jeffrey B Arterburn
- Department of Chemistry and Biochemistry, New Mexico State University, Las Cruces, NM, USA
| | - Nikos Vasilakis
- Department of Pathology, The University of University of Texas Medical Branch, Galveston, TX, USA; Center for Biodefense and Emerging Infectious Diseases, The University of University of Texas Medical Branch, Galveston, TX, USA; Center for Tropical Diseases, The University of University of Texas Medical Branch, Galveston, TX, USA; Institute for Human Infection and Immunity, The University of University of Texas Medical Branch, Galveston, TX, USA
| | - Kathryn A Hanley
- Department of Biology, New Mexico State University, Las Cruces, NM, USA
| |
Collapse
|
34
|
Laureti M, Paradkar PN, Fazakerley JK, Rodriguez-Andres J. Superinfection Exclusion in Mosquitoes and Its Potential as an Arbovirus Control Strategy. Viruses 2020; 12:v12111259. [PMID: 33167513 PMCID: PMC7694488 DOI: 10.3390/v12111259] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/27/2020] [Accepted: 10/28/2020] [Indexed: 12/19/2022] Open
Abstract
The continuing emergence of arbovirus disease outbreaks around the world, despite the use of vector control strategies, warrants the development of new strategies to reduce arbovirus transmission. Superinfection exclusion, a phenomenon whereby a primary virus infection prevents the replication of a second closely related virus, has potential to control arbovirus disease emergence and outbreaks. This phenomenon has been observed for many years in plants, insects and mammalian cells. In this review, we discuss the significance of identifying novel vector control strategies, summarize studies exploring arbovirus superinfection exclusion and consider the potential for this phenomenon to be the basis for novel arbovirus control strategies.
Collapse
Affiliation(s)
- Mathilde Laureti
- Peter Doherty Institute for Infection and Immunity and Faculty of Veterinary and Agricultural Sciences, University of Melbourne, VIC 3000 Melbourne, Australia;
- CSIRO Health & Biosecurity, Australian Centre for Diseases Preparedness, VIC 3220 Geelong, Australia;
- Correspondence: (M.L.); (J.R.-A.)
| | - Prasad N. Paradkar
- CSIRO Health & Biosecurity, Australian Centre for Diseases Preparedness, VIC 3220 Geelong, Australia;
| | - John K. Fazakerley
- Peter Doherty Institute for Infection and Immunity and Faculty of Veterinary and Agricultural Sciences, University of Melbourne, VIC 3000 Melbourne, Australia;
| | - Julio Rodriguez-Andres
- Peter Doherty Institute for Infection and Immunity and Faculty of Veterinary and Agricultural Sciences, University of Melbourne, VIC 3000 Melbourne, Australia;
- Correspondence: (M.L.); (J.R.-A.)
| |
Collapse
|
35
|
N'Tamon AD, Okpekon AT, Bony NF, Bernadat G, Gallard JF, Kouamé T, Séon-Méniel B, Leblanc K, Rharrabti S, Mouray E, Grellier P, Ake M, Amin NC, Champy P, Beniddir MA, Le Pogam P. Streamlined targeting of Amaryllidaceae alkaloids from the bulbs of Crinum scillifolium using spectrometric and taxonomically-informed scoring metabolite annotations. PHYTOCHEMISTRY 2020; 179:112485. [PMID: 32861139 DOI: 10.1016/j.phytochem.2020.112485] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 07/07/2020] [Accepted: 08/07/2020] [Indexed: 06/11/2023]
Abstract
Four undescribed alkaloids have been isolated from the bulbs of the previously unstudied Crinum scillifolium. These compounds were targeted following a state-of-the-art molecular networking strategy comprising a dereplication against in silico databases and re-ranking of the candidate structures based on taxonomically informed scoring. The unreported structures span across a variety of Amaryllidaceae alkaloids appendages. Their structures were unambiguously elucidated by thorough interpretation of their HRESIMS and 1D and 2D NMR data, and comparison to literature data. DFT-NMR calculations were performed to support the determined relative configurations of scillitazettine and scilli-N-desmethylpretazettine and their absolute configurations were mitigated by comparison between experimental and theoretically calculated ECD spectra. The lack of a methyl group on the nitrogen atom in the structure of scilli-N-desmethylpretazettine series is highly unusual in the pretazettine/tazettine series but the most original structural feature in it lies in its 11α disposed hydrogen, which is new to pretazettines. The antiplasmodial as well as the cytotoxic activities against the human colon cancer cell line HCT116 were evaluated, revealing mild to null activities.
Collapse
Affiliation(s)
- Amon Diane N'Tamon
- Université Paris-Saclay, CNRS, BioCIS, 92290, Châtenay-Malabry, France; Département de Chimie Analytique, Minérale et Générale, Technologie Alimentaire, UFR Sciences Pharmaceutiques et Biologiques, Univ. FHB, 06 B. P. 2256, Abidjan 06, Cote d'Ivoire
| | - Aboua Timothée Okpekon
- Laboratoire de Chimie Organique et de Substances Naturelles (LCOSN), UFR Sciences des Structures de la Matière et Technologie, Univ. FHB, 22 BP 582, Abidjan 22, Cote d'Ivoire
| | - Nicaise F Bony
- Département de Chimie Analytique, Minérale et Générale, Technologie Alimentaire, UFR Sciences Pharmaceutiques et Biologiques, Univ. FHB, 06 B. P. 2256, Abidjan 06, Cote d'Ivoire
| | | | - Jean-François Gallard
- Institut de Chimie des Substances Naturelles, CNRS, ICSN UPR 2301, Université Paris-Saclay, 21 Avenue de la Terrasse, 91198, Gif-sur-Yvette, France
| | - Tapé Kouamé
- Université Paris-Saclay, CNRS, BioCIS, 92290, Châtenay-Malabry, France; Laboratoire de Chimie Organique et de Substances Naturelles (LCOSN), UFR Sciences des Structures de la Matière et Technologie, Univ. FHB, 22 BP 582, Abidjan 22, Cote d'Ivoire
| | | | - Karine Leblanc
- Université Paris-Saclay, CNRS, BioCIS, 92290, Châtenay-Malabry, France
| | - Somia Rharrabti
- Université Paris-Saclay, CNRS, BioCIS, 92290, Châtenay-Malabry, France
| | - Elisabeth Mouray
- Muséum National d'Histoire Naturelle, Unité Molécules de Communication et Adaptation des Micro-organismes, UMR7245, CP54, 57, Rue Cuvier, 75005, Paris, France
| | - Philippe Grellier
- Muséum National d'Histoire Naturelle, Unité Molécules de Communication et Adaptation des Micro-organismes, UMR7245, CP54, 57, Rue Cuvier, 75005, Paris, France
| | - Michèle Ake
- Département de Chimie Analytique, Minérale et Générale, Technologie Alimentaire, UFR Sciences Pharmaceutiques et Biologiques, Univ. FHB, 06 B. P. 2256, Abidjan 06, Cote d'Ivoire
| | - N'Cho Christophe Amin
- Département de Chimie Analytique, Minérale et Générale, Technologie Alimentaire, UFR Sciences Pharmaceutiques et Biologiques, Univ. FHB, 06 B. P. 2256, Abidjan 06, Cote d'Ivoire
| | - Pierre Champy
- Université Paris-Saclay, CNRS, BioCIS, 92290, Châtenay-Malabry, France
| | - Mehdi A Beniddir
- Université Paris-Saclay, CNRS, BioCIS, 92290, Châtenay-Malabry, France.
| | - Pierre Le Pogam
- Université Paris-Saclay, CNRS, BioCIS, 92290, Châtenay-Malabry, France.
| |
Collapse
|
36
|
Li LH, Kaptein SJF, Schmid MA, Zmurko J, Leyssen P, Neyts J, Dallmeier K. A dengue type 2 reporter virus assay amenable to high-throughput screening. Antiviral Res 2020; 183:104929. [PMID: 32898584 DOI: 10.1016/j.antiviral.2020.104929] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 08/27/2020] [Accepted: 09/02/2020] [Indexed: 10/23/2022]
Abstract
Dengue virus (DV) is an important mosquito-borne flavivirus threatening almost half of the world's population. Prophylaxis and potent anti-DV drugs are urgently needed. Here, we developed a high content imaging-based (HCI) assay with DV type 2 expressing the fluorescent protein mCherry (DV2/mCherry) to improve the efficiency and robustness of the drug discovery process. For the construction of the reporter virus, the mCherry gene followed by the ribosome-skipping 2A sequence of the Thosea asigna virus (T2A) was placed upstream of the full DV2 open reading frame. The biological characteristics including mCherry expression, virus replication rate, and plaque phenotype was examined and validated in BHK-21, Vero and C6/36 cells. A robust image-based antiviral assay combined with an automated robotic system was then developed, with a Z' factor of 0.73. To validate the image-based antiviral assay, a panel of reference compounds with different molecular mechanisms of anti-DV activity was assessed: (i) the glycosylation inhibitor, Celgosivir, (ii) two NS4b-targeting compounds: a 3-Acyl-indole derivative and NITD618, and (iii) two nucleoside viral polymerase inhibitors, 2'CMC and 7DMA. The inhibition profiles were quantified and obtained by means of HCI and RT-qPCR. Both methods resulted in very comparable inhibition profiles. In conclusion, a powerful and robust assay was developed with a fully automated data generation and processing pipeline. It makes the new reporter virus assay amenable to high-throughput screening of large libraries of small molecules.
Collapse
Affiliation(s)
- Li-Hsin Li
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Leuven, Belgium; Molecular Vaccinology and Vaccine Discovery Group, Canada; GVN, Global Virus Network, USA
| | - Suzanne J F Kaptein
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Leuven, Belgium; GVN, Global Virus Network, USA
| | - Michael A Schmid
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Joanna Zmurko
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Leuven, Belgium; Molecular Vaccinology and Vaccine Discovery Group, Canada
| | - Pieter Leyssen
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Leuven, Belgium; GVN, Global Virus Network, USA
| | - Johan Neyts
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Leuven, Belgium; Molecular Vaccinology and Vaccine Discovery Group, Canada; GVN, Global Virus Network, USA
| | - Kai Dallmeier
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Leuven, Belgium; Molecular Vaccinology and Vaccine Discovery Group, Canada; GVN, Global Virus Network, USA.
| |
Collapse
|
37
|
Felicetti T, Manfroni G, Cecchetti V, Cannalire R. Broad-Spectrum Flavivirus Inhibitors: a Medicinal Chemistry Point of View. ChemMedChem 2020; 15:2391-2419. [PMID: 32961008 DOI: 10.1002/cmdc.202000464] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/16/2020] [Indexed: 12/16/2022]
Abstract
Infections by flaviviruses, such as Dengue, West Nile, Yellow Fever and Zika viruses, represent a growing risk for global health. There are vaccines only for few flaviviruses while no effective treatments are available. Flaviviruses share epidemiological, structural, and ecologic features and often different viruses can co-infect the same host. Therefore, the identification of broad-spectrum inhibitors is highly desirable either for known flaviviruses or for viruses that likely will emerge in the future. Strategies targeting both virus and host factors have been pursued to identify broad-spectrum antiflaviviral agents. In this review, we describe the most promising and best characterized targets and their relative broad-spectrum inhibitors, identified by drug repurposing/libraries screenings and by focused medicinal chemistry campaigns. Finally, we discuss about future strategies to identify new broad-spectrum antiflavivirus agents.
Collapse
Affiliation(s)
- Tommaso Felicetti
- Department of Pharmaceutical Sciences, University of Perugia, via del Liceo 1, 06123, Perugia, Italy
| | - Giuseppe Manfroni
- Department of Pharmaceutical Sciences, University of Perugia, via del Liceo 1, 06123, Perugia, Italy
| | - Violetta Cecchetti
- Department of Pharmaceutical Sciences, University of Perugia, via del Liceo 1, 06123, Perugia, Italy
| | - Rolando Cannalire
- Department of Pharmacy, University of Napoli "Federico II", via D. Montesano 49, 80131, Napoli, Italy
| |
Collapse
|
38
|
Troost B, Smit JM. Recent advances in antiviral drug development towards dengue virus. Curr Opin Virol 2020; 43:9-21. [PMID: 32795907 DOI: 10.1016/j.coviro.2020.07.009] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 07/09/2020] [Indexed: 01/29/2023]
Abstract
Despite the high disease burden of dengue virus, there is no approved antiviral treatment or broadly applicable vaccine to treat or prevent dengue virus infection. In the last decade, many antiviral compounds have been identified but only few have been further evaluated in pre-clinical or clinical trials. This review will give an overview of the direct-acting and host-directed antivirals identified to date. Furthermore, important parameters for further development that is, drug properties including efficacy, specificity and stability, pre-clinical animal testing, and combinational drug therapy will be discussed.
Collapse
Affiliation(s)
- Berit Troost
- Department of Medical Microbiology and Infection Prevention, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jolanda M Smit
- Department of Medical Microbiology and Infection Prevention, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
39
|
Liang Q, Cai W, Zhao Y, Xu H, Tang H, Chen D, Qian F, Sun L. Lycorine ameliorates bleomycin-induced pulmonary fibrosis via inhibiting NLRP3 inflammasome activation and pyroptosis. Pharmacol Res 2020; 158:104884. [DOI: 10.1016/j.phrs.2020.104884] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 04/10/2020] [Accepted: 05/01/2020] [Indexed: 01/17/2023]
|
40
|
Hosseini S, Muñoz-Soto RB, Oliva-Ramírez J, Vázquez-Villegas P, Aghamohammadi N, Rodriguez-Garcia A, Martinez-Chapa SO. Latest Updates in Dengue Fever Therapeutics: Natural, Marine and Synthetic Drugs. Curr Med Chem 2020; 27:719-744. [DOI: 10.2174/0929867325666180629124709] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 05/25/2018] [Accepted: 06/01/2018] [Indexed: 11/22/2022]
Abstract
In this paper, we review the history of Dengue, the mechanism of infection, the
molecular characteristics and components of Dengue, the mechanism of entry to the target
cells, cyclization of the genome and replication process, as well as translation of the proteins
for virus assembly. The major emphasis of this work is on natural products and plant extracts,
which were used for as palliative or adjuvant treatment of Dengue. This review article also
summarizes the latest findings in regards to the marine products as effective drugs to target
different symptoms of Dengue. Furthermore, an update on synthetic drugs for treating Dengue
is provided in this review. As a novel alternative, we describe monoclonal antibody therapy
for Dengue management and treatment.
Collapse
Affiliation(s)
- Samira Hosseini
- Tecnologico de Monterrey, School of Engineering and Sciences, Monterrey, N.L. 64849, Mexico
| | - Rodrigo B. Muñoz-Soto
- Tecnologico de Monterrey, Campus Ciudad de México, Escuela de Ingeniería y Ciencias, Calle del Puente 222, Mexico City, Mexico
| | - Jacqueline Oliva-Ramírez
- Tecnologico de Monterrey, Escuela de Ingenieria y Ciencias, Ave. Lago de Guadalupe Km 3.5, Cd Lopez Mateos, Atizapan, Estado de Mexico, Mexico
| | | | - Nasrin Aghamohammadi
- Centre for Occupational and Environmental Health, Department of Social and Preventive Medicine, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Aida Rodriguez-Garcia
- Universidad Autonoma de Nuevo Leon, Facultad de Ciencias Biologicas, Instituto de Biotecnología. Ave. Pedro de Alba S/N, Ciudad Universitaria, San Nicolás de los Garza, N.L. 66455, Mexico
| | | |
Collapse
|
41
|
Screening of Natural Extracts for Inhibitors against Japanese Encephalitis Virus Infection. Antimicrob Agents Chemother 2020; 64:AAC.02373-19. [PMID: 31871089 PMCID: PMC7038234 DOI: 10.1128/aac.02373-19] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 12/18/2019] [Indexed: 12/17/2022] Open
Abstract
The mosquito-borne Japanese encephalitis virus (JEV) causes serious illness worldwide that is associated with high morbidity and mortality. Currently, there are no effective drugs approved for the treatment of JEV infection. Drug-repurposing screening is an alternative approach to discover potential antiviral agents. In this study, high-content screening (HCS) of a natural extracts library was performed, and two hit FDA-approved Na+/K+-ATPase inhibitors, ouabain and digoxin, were identified as having robust efficiency against JEV infection with the selectivity indexes over 1,000. The results indicated that ouabain and digoxin blocked the JEV infection at the replication stage by targeting the Na+/K+-ATPase. Furthermore, it was proven that ouabain significantly reduced the morbidity and mortality caused by JEV in a BALB/c mouse model. This work demonstrated that Na+/K+-ATPase could serve as the target of treatment of JEV infection, and ouabain has the potential to be developed as an effective anti-JEV drug.
Collapse
|
42
|
Sinigaglia A, Peta E, Riccetti S, Barzon L. New avenues for therapeutic discovery against West Nile virus. Expert Opin Drug Discov 2020; 15:333-348. [DOI: 10.1080/17460441.2020.1714586] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
| | - Elektra Peta
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Silvia Riccetti
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Luisa Barzon
- Department of Molecular Medicine, University of Padova, Padova, Italy
| |
Collapse
|
43
|
Bozkurt B, Kaya GI, Somer NU. Chemical Composition and Enzyme Inhibitory Activities of Turkish Pancratium maritimum Bulbs. Nat Prod Commun 2019. [DOI: 10.1177/1934578x19872905] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The chemical composition of the bulbs of Pancratium maritimum L. (Amaryllidaceae) from Turkey (Pamucak, Aydın) has been determined by gas chromatography-mass spectrometry. A total of 29 compounds belonging to different skeletal types of Amaryllidaceae alkaloids were identified. Lycorine, galanthamine, crinine, and pancracine were found as major constituents. Interestingly, indole alkaloids (1-acetyl-β -carboline and galanthindole) were also detected. Acetylcholinesterase, butyrylcholinesterase, and prolyl oligopeptidase inhibitory activity potentials of the alkaloidal extract were also determined. The results of the present study show that Turkish P. maritimum comprises a rich repository for bioactive alkaloids with intriguing structural diversities.
Collapse
Affiliation(s)
- Buket Bozkurt
- Department of Pharmacognosy, Faculty of Pharmacy, Ege University, Izmir, Turkey
| | - Gulen I. Kaya
- Department of Pharmacognosy, Faculty of Pharmacy, Ege University, Izmir, Turkey
| | - Nehir U. Somer
- Department of Pharmacognosy, Faculty of Pharmacy, Ege University, Izmir, Turkey
| |
Collapse
|
44
|
Duggal NK, Langwig KE, Ebel GD, Brault AC. On the Fly: Interactions Between Birds, Mosquitoes, and Environment That Have Molded West Nile Virus Genomic Structure Over Two Decades. JOURNAL OF MEDICAL ENTOMOLOGY 2019; 56:1467-1474. [PMID: 31549720 PMCID: PMC7182917 DOI: 10.1093/jme/tjz112] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Indexed: 05/15/2023]
Abstract
West Nile virus (WNV) was first identified in North America almost 20 yr ago. In that time, WNV has crossed the continent and established enzootic transmission cycles, resulting in intermittent outbreaks of human disease that have largely been linked with climatic variables and waning avian seroprevalence. During the transcontinental dissemination of WNV, the original genotype has been displaced by two principal extant genotypes which contain an envelope mutation that has been associated with enhanced vector competence by Culex pipiens L. (Diptera: Culicidae) and Culex tarsalis Coquillett vectors. Analyses of retrospective avian host competence data generated using the founding NY99 genotype strain have demonstrated a steady reduction in viremias of house sparrows over time. Reciprocally, the current genotype strains WN02 and SW03 have demonstrated an inverse correlation between house sparrow viremia magnitude and the time since isolation. These data collectively indicate that WNV has evolved for increased avian viremia while house sparrows have evolved resistance to the virus such that the relative host competence has remained constant. Intrahost analyses of WNV evolution demonstrate that selection pressures are avian species-specific and purifying selection is greater in individual birds compared with individual mosquitoes, suggesting that the avian adaptive and/or innate immune response may impose a selection pressure on WNV. Phylogenomic, experimental evolutionary systems, and models that link viral evolution with climate, host, and vector competence studies will be needed to identify the relative effect of different selective and stochastic mechanisms on viral phenotypes and the capacity of newly evolved WNV genotypes for transmission in continuously changing landscapes.
Collapse
Affiliation(s)
- Nisha K Duggal
- Department of Biomedical Sciences and Pathobiology, Virginia Polytechnic Institute and State University, Blacksburg, VA
| | - Kate E Langwig
- Department of Biological Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA
| | - Gregory D Ebel
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO
| | - Aaron C Brault
- Division of Vector-borne Diseases, Centers for Disease Control and Prevention, Fort Collins, CO
- Corresponding author, e-mail:
| |
Collapse
|
45
|
Yang L, Zhang JH, Zhang XL, Lao GJ, Su GM, Wang L, Li YL, Ye WC, He J. Tandem mass tag-based quantitative proteomic analysis of lycorine treatment in highly pathogenic avian influenza H5N1 virus infection. PeerJ 2019; 7:e7697. [PMID: 31592345 PMCID: PMC6778435 DOI: 10.7717/peerj.7697] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 08/19/2019] [Indexed: 12/14/2022] Open
Abstract
Highly pathogenic H5N1 influenza viruses (HPAIV) cause rapid systemic illness and death in susceptible animals, leading to a disease with high morbidity and mortality rates. Although vaccines and drugs are the best solution to prevent this threat, a more effective treatment for H5 strains of influenza has yet to be developed. Therefore, the development of therapeutics/drugs that combat H5N1 influenza virus infection is becoming increasingly important. Lycorine, the major component of Amaryllidaceae alkaloids, exhibits better protective effects against A/CK/GD/178/04 (H5N1) (GD178) viruses than the commercial neuraminidase (NA) inhibitor oseltamivir in our prior study. Lycorine demonstrates outstanding antiviral activity because of its inhibitory activity against the export of viral ribonucleoprotein complexes (vRNPs) from the nucleus. However, how lycorine affects the proteome of AIV infected cells is unknown. Therefore, we performed a comparative proteomic analysis to identify changes in protein expression in AIV-infected Madin-Darby Canine Kidney cells treated with lycorine. Three groups were designed: mock infection group (M), virus infection group (V), and virus infection and lycorine-treated after virus infection group (L). The multiplexed tandem mass tag (TMT) approach was employed to analyze protein level in this study. In total, 5,786 proteins were identified from the three groups of cells by using TMT proteomic analysis. In the V/M group, 1,101 proteins were identified, of which 340 differentially expressed proteins (DEPs) were determined during HPAIV infection; among the 1,059 proteins identified from the lycorine-treated group, 258 proteins presented significant change. Here, 71 proteins showed significant upregulation or downregulation of expression in the virus-infected/mock and virus-infected/lycorine-treated comparisons, and the proteins in each fraction were functionally classified further. Interestingly, lycorine treatment decreased the levels of the nuclear pore complex protein 93 (Nup93, E2RSV7), which is associated with nuclear–cytoplasmic transport. In addition, Western blot experiments confirmed that the expression of Nup93 was significantly downregulated in lycorine treatment but induced after viral infection. Our results may provide new insights into how lycorine may trap vRNPs in the nucleus and suggest new potential therapeutic targets for influenza virus.
Collapse
Affiliation(s)
- Li Yang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou, China.,College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Jia Hao Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xiao Li Zhang
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Guang Jie Lao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Guan Ming Su
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Lei Wang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou, China
| | - Yao Lan Li
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou, China
| | - Wen Cai Ye
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou, China
| | - Jun He
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou, China.,Institute of Laboratory Animal Science, Jinan University, Guangzhou, China
| |
Collapse
|
46
|
Bonica MB, Goenaga S, Martin ML, Feroci M, Luppo V, Muttis E, Fabbri C, Morales MA, Enria D, Micieli MV, Levis S. Vector competence of Aedes aegypti for different strains of Zika virus in Argentina. PLoS Negl Trop Dis 2019; 13:e0007433. [PMID: 31188869 PMCID: PMC6561534 DOI: 10.1371/journal.pntd.0007433] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 04/15/2019] [Indexed: 12/20/2022] Open
Abstract
The importance of Zika virus (ZIKV) has increased noticeably since the outbreak in the Americas in 2015, when the illness was associated with congenital disorders. Although there is evidence of sexual transmission of the virus, the mosquito Aedes aegypti is believed to be the main vector for transmission to humans. This species of mosquito has not only been found naturally infected with ZIKV, but also has been the subject of study in many vector competence assays that employ different strains of ZIKV around the world. In Argentina, the first case was reported in February 2016 and a total of 278 autochthonous cases have since been confirmed, however, ZIKV virus has not been isolated from any mosquito species yet in Argentina. In order to elucidate if Argentinian Ae. aegypti populations could be a possible vector of ZIKV, we conducted vector competence studies that involved a local strain of ZIKV from Chaco province, and a Venezuelan strain obtained from an imported case. For this purpose, Ae. aegypti adults from the temperate area of Argentina (Buenos Aires province) were fed with infected blood. Body, legs and saliva were harvested and tested by plaque titration on plates of Vero cells for ZIKV at 7, 11 and 14 days post infection (DPI) in order to calculate infection, transmission, and dissemination rates, respectively. Both strains were able to infect mosquitoes at all DPIs, whereas dissemination and transmission were observed at all DPIs for the Argentinian strain but only at 14 DPI for the Venezuelan strain. This study proves the ability of Ae. aegypti mosquitoes from Argentina to become infected with two different strains of ZIKV, both belonging to the Asian lineage, and that the virus can disseminate to the legs and salivary glands. Zika virus is a flavivirus transmitted by mosquitoes, isolated for the first time in the Ziika Forest in Uganda in 1947 from a rhesus macaque monkey. The disease is usually asymptomatic, but sometimes it causes a mild illness that comes with fever, rash, joint pain, and conjunctivitis. The World Health Organization focused the attention on this virus after the outbreak in the Americas, when the virus was linked to microcephaly and serious neurological diseases, including Guillain-Barré syndrome. Aedes aegypti was incriminated as the main vector of the virus as it was found both naturally and experimentally infected. This mosquito species was declared eradicated in Argentina by 1970 but re-emerged in 1989. Recent studies found a peculiarity in the genetics of Argentinian Ae. aegypti populations that consists in a combination between both subspecies: Ae. aegypti formosus and Ae. aegypti aegypti. Our study tries to elucidate if Ae. aegypti from Argentina are able to transmit the virus in order to add these mosquitoes to the list of possible vectors of ZIKV and, in future prospect, orient to fight the virus by controlling the vector.
Collapse
Affiliation(s)
- Melisa Berenice Bonica
- Centro de Estudios Parasitológicos y de Vectores (CEPAVE-CONICET), Universidad Nacional de La Plata, La Plata, Buenos Aires, Argentina
- * E-mail: (MBB); (SG)
| | - Silvina Goenaga
- Instituto Nacional de Enfermedades Virales Humanas “Dr. Julio Maiztegui” (INEVH-ANLIS), Pergamino, Buenos Aires, Argentina
- * E-mail: (MBB); (SG)
| | - María Laura Martin
- Instituto Nacional de Enfermedades Virales Humanas “Dr. Julio Maiztegui” (INEVH-ANLIS), Pergamino, Buenos Aires, Argentina
| | - Mariel Feroci
- Instituto Nacional de Enfermedades Virales Humanas “Dr. Julio Maiztegui” (INEVH-ANLIS), Pergamino, Buenos Aires, Argentina
| | - Victoria Luppo
- Instituto Nacional de Enfermedades Virales Humanas “Dr. Julio Maiztegui” (INEVH-ANLIS), Pergamino, Buenos Aires, Argentina
| | - Evangelina Muttis
- Centro de Estudios Parasitológicos y de Vectores (CEPAVE-CONICET), Universidad Nacional de La Plata, La Plata, Buenos Aires, Argentina
| | - Cintia Fabbri
- Instituto Nacional de Enfermedades Virales Humanas “Dr. Julio Maiztegui” (INEVH-ANLIS), Pergamino, Buenos Aires, Argentina
| | - María Alejandra Morales
- Instituto Nacional de Enfermedades Virales Humanas “Dr. Julio Maiztegui” (INEVH-ANLIS), Pergamino, Buenos Aires, Argentina
| | - Delia Enria
- Instituto Nacional de Enfermedades Virales Humanas “Dr. Julio Maiztegui” (INEVH-ANLIS), Pergamino, Buenos Aires, Argentina
| | - María Victoria Micieli
- Centro de Estudios Parasitológicos y de Vectores (CEPAVE-CONICET), Universidad Nacional de La Plata, La Plata, Buenos Aires, Argentina
| | - Silvana Levis
- Instituto Nacional de Enfermedades Virales Humanas “Dr. Julio Maiztegui” (INEVH-ANLIS), Pergamino, Buenos Aires, Argentina
| |
Collapse
|
47
|
Dighe SN, Ekwudu O, Dua K, Chellappan DK, Katavic PL, Collet TA. Recent update on anti-dengue drug discovery. Eur J Med Chem 2019; 176:431-455. [PMID: 31128447 DOI: 10.1016/j.ejmech.2019.05.010] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 04/12/2019] [Accepted: 05/06/2019] [Indexed: 01/27/2023]
Abstract
Dengue is the most important arthropod-borne viral disease of humans, with more than half of the global population living in at-risk areas. Despite the negative impact on public health, there are no antiviral therapies available, and the only licensed vaccine, Dengvaxia®, has been contraindicated in children below nine years of age. In an effort to combat dengue, several small molecules have entered into human clinical trials. Here, we review anti-DENV molecules and their drug targets that have been published within the past five years (2014-2018). Further, we discuss their probable mechanisms of action and describe a role for classes of clinically approved drugs and also an unclassified class of anti-DENV agents. This review aims to enhance our understanding of novel agents and their cognate targets in furthering innovations in the use of small molecules for dengue drug therapies.
Collapse
Affiliation(s)
- Satish N Dighe
- Innovative Medicines Group, Institute of Health & Biomedical Innovation, School of Clinical Sciences, Queensland University of Technology, Brisbane, Australia.
| | - O'mezie Ekwudu
- Innovative Medicines Group, Institute of Health & Biomedical Innovation, School of Clinical Sciences, Queensland University of Technology, Brisbane, Australia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, Australia
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University (IMU), Bukit Jalil, Kuala Lumpur, 57000, Malaysia
| | - Peter L Katavic
- Innovative Medicines Group, Institute of Health & Biomedical Innovation, School of Clinical Sciences, Queensland University of Technology, Brisbane, Australia
| | - Trudi A Collet
- Innovative Medicines Group, Institute of Health & Biomedical Innovation, School of Clinical Sciences, Queensland University of Technology, Brisbane, Australia
| |
Collapse
|
48
|
Brown C, Kong T, Britten JF, Werstiuk NH, McNulty J, D’Aiuto L, Demers M, Nimgaonkar VL. Asymmetric Entry into 10 b-aza-Analogues of Amaryllidaceae Alkaloids Reveals a Pronounced Electronic Effect on Antiviral Activity. ACS OMEGA 2018; 3:11469-11476. [PMID: 30320263 PMCID: PMC6173499 DOI: 10.1021/acsomega.8b01987] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 09/06/2018] [Indexed: 06/08/2023]
Abstract
Development of a chiral pool-based synthesis of 10b-aza-analogues of biologically active Amaryllidaceae alkaloids is described, involving a concise reductive amination and condensation sequence, leading to ring-B/C-modified, fully functionalized ring-C derivatives. Differentiated anticancer and antiviral activities of these analogues are presented. Despite complete conformational and functional group overlap, the 10b-aza-analogues have diminished anticancer activity and no antiviral activity. These unprecedented electronic effects suggest a possible role for π-type secondary orbital interactions with the biological target.
Collapse
Affiliation(s)
- Carla
E. Brown
- Department
of Chemistry & Chemical Biology, McMaster
University, 1280 Main Street West, Hamilton, Ontario L8S 4M1, Canada
| | - Tiffany Kong
- Department
of Chemistry & Chemical Biology, McMaster
University, 1280 Main Street West, Hamilton, Ontario L8S 4M1, Canada
| | - James F. Britten
- Department
of Chemistry & Chemical Biology, McMaster
University, 1280 Main Street West, Hamilton, Ontario L8S 4M1, Canada
| | - Nick H. Werstiuk
- Department
of Chemistry & Chemical Biology, McMaster
University, 1280 Main Street West, Hamilton, Ontario L8S 4M1, Canada
| | - James McNulty
- Department
of Chemistry & Chemical Biology, McMaster
University, 1280 Main Street West, Hamilton, Ontario L8S 4M1, Canada
| | - Leonardo D’Aiuto
- Department
of Psychiatry, University of Pittsburgh
School of Medicine, 3811
O’Hara Street, Pittsburgh, Pennsylvania 15213, United States
| | - Matthew Demers
- Department
of Psychiatry, University of Pittsburgh
School of Medicine, 3811
O’Hara Street, Pittsburgh, Pennsylvania 15213, United States
| | - Vishwajit L. Nimgaonkar
- Department
of Psychiatry, University of Pittsburgh
School of Medicine, 3811
O’Hara Street, Pittsburgh, Pennsylvania 15213, United States
| |
Collapse
|
49
|
Li B, Liao HM, Liu H, Tsai S, Zhang J, Hung GC, Chin PJ, Gao Y, Lo SC. Comparative genomics, infectivity and cytopathogenicity of Zika viruses produced by acutely and persistently infected human hematopoietic cell lines. PLoS One 2018; 13:e0203331. [PMID: 30192813 PMCID: PMC6128475 DOI: 10.1371/journal.pone.0203331] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 08/17/2018] [Indexed: 12/17/2022] Open
Abstract
Zika virus (ZIKV), an arthropod-borne virus, has emerged as a major human pathogen. Prolonged or persistent ZIKV infection of human cells and tissues may serve as a reservoir for the virus and present serious challenges to the safety of public health. Human hematopoietic cell lines with different developmental properties revealed differences in susceptibility and outcomes to ZIKV infection. In three separate studies involving the prototypic MR 766 ZIKV strain and the human monocytic leukemia U937 cell line, ZIKV initially developed only a low-grade infection at a slow rate. After continuous culture for several months, persistently ZIKV-infected cell lines were observed with most, if not all, cells testing positive for ZIKV antigen. The infected cultures produced ZIKV RNA (v-RNA) and infectious ZIKVs persistently (“persistent ZIKVs”) with distinct infectivity and pathogenicity when tested using various kinds of host cells. When the genomes of ZIKVs from the three persistently infected cell lines were compared with the genome of the prototypic MR 766 ZIKV strain, distinct sets of mutations specific to each cell line were found. Significantly, all three “persistent ZIKVs” were capable of infecting fresh U937 cells with high efficiency at rapid rates, resulting in the development of a new set of persistently ZIKV-infected U937 cell lines. The genomes of ZIKVs from the new set of persistently ZIKV-infected U937 cell lines were further analyzed for their different mutations. The 2nd generation of persistent ZIKVs continued to possess most of the distinct sets of mutations specific to the respective 1st generation of persistent ZIKVs. We anticipate that the study will contribute to the understanding of the fundamental biology of adaptive mutations and selection during viral persistence. The persistently ZIKV-infected human cell lines that we developed will also be useful to investigate critical molecular pathways of ZIKV persistence and to study drugs or countermeasures against ZIKV infections and transmission.
Collapse
Affiliation(s)
- Bingjie Li
- Tissue Microbiology Laboratory, Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Hsiao-Mei Liao
- Tissue Microbiology Laboratory, Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Hebing Liu
- Tissue Microbiology Laboratory, Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Shien Tsai
- Tissue Microbiology Laboratory, Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Jing Zhang
- Tissue Microbiology Laboratory, Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Guo-Chiuan Hung
- Tissue Microbiology Laboratory, Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Pei-Ju Chin
- Tissue Microbiology Laboratory, Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Yamei Gao
- Lab of Pediatric and Respiratory Viral Diseases, Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Shyh-Ching Lo
- Tissue Microbiology Laboratory, Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
- * E-mail:
| |
Collapse
|
50
|
Roy M, Liang L, Xiao X, Feng P, Ye M, Liu J. Lycorine: A prospective natural lead for anticancer drug discovery. Biomed Pharmacother 2018; 107:615-624. [PMID: 30114645 PMCID: PMC7127747 DOI: 10.1016/j.biopha.2018.07.147] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Revised: 07/18/2018] [Accepted: 07/30/2018] [Indexed: 02/07/2023] Open
Abstract
Outline of the anticancer properties and associated molecular mechanism mediated by lycorine. Comprehensive analysis of the structure activity relationship associated with anticancer activity of lycorine. Summary of the pharmacological aspects and implications for future directions with this compound.
Nature is the most abundant source for novel drug discovery. Lycorine is a natural alkaloid with immense therapeutic potential. Lycorine is active in a very low concentration and with high specificity against a number of cancers both in vivo and in vitro and against various drug-resistant cancer cells. This review summarized the therapeutic effect and the anticancer mechanisms of lycorine. At the same time, we have discussed the pharmacology and comparative structure-activity relationship for the anticancer activity of this compound. The researches outlined in this paper serve as a foundation to explain lycorine as an important lead compound for new generation anticancer drug design and provide the principle for the development of biological strategies to utilize lycorine in the treatment of cancers.
Collapse
Affiliation(s)
- Mridul Roy
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China; Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China
| | - Long Liang
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China
| | - Xiaojuan Xiao
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China
| | - Peifu Feng
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Mao Ye
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China.
| | - Jing Liu
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China.
| |
Collapse
|