Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 262
Filter
1.
Front Cell Infect Microbiol ; 14: 1396279, 2024.
Article in English | MEDLINE | ID: mdl-38800832

ABSTRACT

Background: The Chikungunya virus is an Alphavirus that belongs to the Togaviridae family and is primarily transmitted by mosquitoes. It causes acute infection characterized by fever, headache, and arthralgia. Some patients also experience persistent chronic osteoarthritis-like symptoms. Dedicated antiviral treatments are currently unavailable for CHIKV. This study aims to explore the potential anti-CHIKV effect of rosmarinic acid using network pharmacology. Methods: This study employed network pharmacology to predict and verify the molecular targets and pathways associated with ROSA in the context of CHIKV. The analysis outcomes were further validated using molecular docking and in vitro experiments. Results: The analysis of CHIKV targets using the Kyoto Encyclopedia of Genes and Genomes and MCODE identified IL-17 as an important pathogenic pathway in CHIKV infection. Among the 30 targets of ROSA against CHIKV, nearly half were found to be involved in the IL-17 signaling pathway. This suggests that ROSA may help the host in resisting CHIKV invasion by modulating this pathway. Molecular docking validation results showed that ROSA can stably bind to 10 core targets out of the 30 identified targets. In an in vitro CHIKV infection model developed using 293T cells, treatment with 60 µM ROSA significantly improved the survival rate of infected cells, inhibited 50% CHIKV proliferation after CHIKV infection, and reduced the expression of TNF-α in the IL-17 signaling pathway. Conclusion: This study provides the first confirmation of the efficacy of ROSA in suppressing CHIKV infection through the IL-17 signaling pathway. The findings warrant further investigation to facilitate the development of ROSA as a potential treatment for CHIKV infection.


Subject(s)
Antiviral Agents , Chikungunya Fever , Chikungunya virus , Cinnamates , Depsides , Interleukin-17 , Molecular Docking Simulation , Rosmarinic Acid , Signal Transduction , Depsides/pharmacology , Cinnamates/pharmacology , Chikungunya virus/drug effects , Interleukin-17/metabolism , Humans , Antiviral Agents/pharmacology , Signal Transduction/drug effects , Chikungunya Fever/drug therapy , Chikungunya Fever/virology , Network Pharmacology , HEK293 Cells , Virus Replication/drug effects , Animals
2.
ACS Chem Biol ; 19(5): 1093-1105, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38646883

ABSTRACT

Viral macrodomains that can bind to or hydrolyze protein adenosine diphosphate ribosylation (ADP-ribosylation) have emerged as promising targets for antiviral drug development. Many inhibitor development efforts have been directed against the severe acute respiratory syndrome coronavirus 2 macrodomain 1 (SARS-CoV-2 Mac1). However, potent inhibitors for viral macrodomains are still lacking, with the best inhibitors still in the micromolar range. Based on GS-441524, a remdesivir precursor, and our previous studies, we have designed and synthesized potent binders of SARS-CoV-2 Mac1 and other viral macrodomains including those of Middle East respiratory syndrome coronavirus (MERS-CoV), Venezuelan equine encephalitis virus (VEEV), and Chikungunya virus (CHIKV). We show that the 1'-CN group of GS-441524 promotes binding to all four viral macrodomains tested while capping the 1″-OH of GS-441524-diphosphate-ribose with a simple phenyl ring further contributes to binding. Incorporating these two structural features, the best binders show 20- to 6000-fold increases in binding affinity over ADP-ribose for SARS-CoV-2, MERS-CoV, VEEV, and CHIKV macrodomains. Moreover, building on these potent binders, we have developed two highly sensitive fluorescence polarization tracers that only require nanomolar proteins and can effectively resolve the binding affinities of nanomolar inhibitors. Our findings and probes described here will facilitate future development of more potent viral macrodomain inhibitors.


Subject(s)
Antiviral Agents , Fluorescence Polarization , SARS-CoV-2 , Humans , Adenosine Diphosphate Ribose/metabolism , Adenosine Diphosphate Ribose/chemistry , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/chemistry , Adenosine Monophosphate/pharmacology , Adenosine Monophosphate/metabolism , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Antiviral Agents/metabolism , Chikungunya virus/drug effects , COVID-19/virology , COVID-19 Drug Treatment , Encephalitis Virus, Venezuelan Equine/drug effects , Encephalitis Virus, Venezuelan Equine/metabolism , Middle East Respiratory Syndrome Coronavirus , Protein Binding , Protein Domains , SARS-CoV-2/drug effects
3.
Phytomedicine ; 128: 155491, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38489894

ABSTRACT

BACKGROUND: Dengue and chikungunya, caused by dengue virus (DENV) and chikungunya virus (CHIKV) respectively, are the most common arthropod-borne viral diseases worldwide, for which there are no FDA-approved antivirals or effective vaccines. Arctigenin, a phenylpropanoid lignan from the seeds of Arctium lappa L. is known for its anti-inflammatory, anti-cancer, antibacterial, and immunomodulatory properties. Arctigenin's antimicrobial and immunomodulatory capabilities make it a promising candidate for investigating its potential as an anti-DENV and anti-CHIKV agent. PURPOSE: The aim of the study was to explore the anti-DENV and anti-CHIKV effects of arctigenin and identify the possible mechanisms of action. METHODS: The anti-DENV or anti-CHIKV effects of arctigenin was assessed using various in vitro and in silico approaches. Vero CCL-81 cells were infected with DENV or CHIKV and treated with arctigenin at different concentrations, temperature, and time points to ascertain the effect of the compound on virus entry or replication. In silico molecular docking was performed to identify the interactions of the compound with viral proteins. RESULTS: Arctigenin had no effects on DENV. Various time- and temperature-dependent assays revealed that arctigenin significantly reduced CHIKV RNA copy number and infectious virus particles and affected viral entry. Entry bypass assay revealed that arctigenin inhibited the initial steps of viral replication. In silico docking results revealed the high binding affinity of the compound with the E1 protein and the nsp3 macrodomain of CHIKV. CONCLUSION: This study demonstrates the in-vitro anti-CHIKV potential of arctigenin and suggests that the compound might affect CHIKV entry and replication. Further preclinical and clinical studies are needed to identify its safety and efficacy as an anti-CHIKV drug.


Subject(s)
Antiviral Agents , Arctium , Chikungunya virus , Dengue Virus , Virus Internalization , Virus Replication , Animals , Antiviral Agents/pharmacology , Arctium/chemistry , Chikungunya virus/drug effects , Chikungunya virus/physiology , Chlorocebus aethiops , Dengue Virus/drug effects , Dengue Virus/physiology , Furans/pharmacology , Lignans/pharmacology , Molecular Docking Simulation , Seeds/chemistry , Vero Cells , Virus Internalization/drug effects , Virus Replication/drug effects
4.
Viruses ; 15(2)2023 02 09.
Article in English | MEDLINE | ID: mdl-36851696

ABSTRACT

Ubiquitination and deubiquitination processes are widely involved in modulating the function, activity, localization, and stability of multiple cellular proteins regulating almost every aspect of cellular function. Several virus families have been shown to exploit the cellular ubiquitin-conjugating system to achieve a productive infection: enter the cell, promote genome replication, or assemble and release viral progeny. In this study, we analyzed the role of deubiquitinating enzymes (DUBs) during chikungunya virus (CHIKV) infection. HEK293T, Vero-E6, and Huh-7 cells were treated with two DUB inhibitors (PR619 or WP1130). Then, infected cells were evaluated by flow cytometry, and viral progeny was quantified using the plaque assay method. The changes in viral proteins and viral RNA were analyzed using Western blotting and RT-qPCR, respectively. Results indicate that treatment with DUB inhibitors impairs CHIKV replication due to significant protein and viral RNA synthesis deregulation. Therefore, DUB activity may be a pharmacological target for blocking CHIKV infection.


Subject(s)
Chikungunya Fever , Chikungunya virus , Deubiquitinating Enzymes , Enzyme Inhibitors , Virus Replication , Humans , Chikungunya Fever/drug therapy , Chikungunya virus/drug effects , Deubiquitinating Enzymes/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , HEK293 Cells , RNA, Viral , Virus Replication/drug effects
5.
Acta Trop ; 232: 106497, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35508271

ABSTRACT

Chikungunya virus (CHIKV) is a zoonotic arthropod-borne virus that causes Chikungunya fever (CHIKF), a self-limiting disease characterized by myalgia and acute or chronic arthralgia. CHIKF pathogenesis has an important immunological component since higher levels of pro-inflammatory factors, including cytokines and chemokines, are detected in CHIKV-infected patients. In vitro studies, using monocytes and macrophages have shown that CHIKV infection promotes elevated production of pro-inflammatory cytokines and antiviral response factors. Vitamin D3 (VD3) has been described as an important modulator of immune response and as an antiviral factor for several viruses. Here, we aimed to study the effects of VD3 treatment on viral replication and pro-inflammatory response in CHIKV-infected human monocytes (VD3-Mon) and monocyte-derived macrophages differentiated in the absence (MDMs) or the presence of VD3 (VD3-MDMs). We found that VD3 treatment did not suppress CHIKV replication in either VD3-Mon or VD3-MDMs. However, the expression of VDR, CAMP and CYP24A1 mRNAs was altered by CHIKV infection. Furthermore, VD3 treatment alters TLRs mRNA expression and production of pro-inflammatory cytokines, including TNFα and CXCL8/IL8, but not IL1ß and IL6, in response to CHIKV infection in both VD3-Mon and VD3-MDMs. While a significant decrease in CXCL8/IL8 production was observed in CHIKV-infected VD3-Mon, significantly higher production of CXCL8/IL8 was observed in CHIKV-infected VD3-MDM at 24 hpi. Altogether, our results suggest that vitamin D3 may play an important role in ameliorating pro-inflammatory response during CHIKV infection in human Mon, but not in MDMs. Although further studies are needed to evaluate the efficacy of VD3; nevertheless, this study provides novel insights into its benefits in modulating the inflammatory response elicited by CHIKV infection in humans.


Subject(s)
Chikungunya Fever , Chikungunya virus , Macrophages , Monocytes , Toll-Like Receptors , Virus Replication , Chikungunya Fever/virology , Chikungunya virus/drug effects , Cholecalciferol/pharmacology , Cytokines/biosynthesis , Humans , Macrophages/drug effects , Macrophages/virology , Monocytes/drug effects , Monocytes/virology , Toll-Like Receptors/biosynthesis , Virus Replication/drug effects , Vitamin D/pharmacology
6.
Viruses ; 14(2)2022 02 18.
Article in English | MEDLINE | ID: mdl-35216015

ABSTRACT

Mayaro virus (MAYV) manipulates cell machinery to successfully replicate. Thus, identifying host proteins implicated in MAYV replication represents an opportunity to discover potential antiviral targets. PIM kinases are enzymes that regulate essential cell functions and also appear to be critical factors in the replication of certain viruses. In this study we explored the consequences of PIM kinase inhibition in the replication of MAYV and other arboviruses. Cytopathic effects or viral titers in samples from MAYV-, Chikungunya-, Una- or Zika-infected cells treated with PIM kinase inhibitors were evaluated using an inverted microscope or plaque-forming assays. The expression of viral proteins E1 and nsP1 in MAYV-infected cells was assessed using an immunofluorescence confocal microscope or Western blot. Our results revealed that PIM kinase inhibition partially prevented MAYV-induced cell damage and also promoted a decrease in viral titers for MAYV, UNAV and ZIKV. The inhibitory effect of PIM kinase blocking was observed for each of the MAYV strains tested and also occurred as late as 8 h post infection (hpi). Finally, PIM kinase inhibition suppressed the expression of MAYV E1 and nsP1 proteins. Taken together, these findings suggest that PIM kinases could represent an antiviral target for MAYV and other arboviruses.


Subject(s)
Alphavirus/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Virus Replication/drug effects , Animals , Antiviral Agents/pharmacology , Cell Line , Chikungunya virus/drug effects , Humans , Zika Virus/drug effects
7.
Antiviral Res ; 197: 105223, 2022 01.
Article in English | MEDLINE | ID: mdl-34856248

ABSTRACT

Repurposing drugs is a promising strategy to identify therapeutic interventions against novel and re-emerging viruses. Posaconazole is an antifungal drug used to treat invasive aspergillosis and candidiasis. Recently, posaconazole and its structural analog, itraconazole were shown to inhibit replication of multiple viruses by modifying intracellular cholesterol homeostasis. Here, we show that posaconazole inhibits replication of the alphaviruses Semliki Forest virus (SFV), Sindbis virus and chikungunya virus with EC50 values ranging from 1.4 µM to 9.5 µM. Posaconazole treatment led to a significant reduction of virus entry in an assay using a temperature-sensitive SFV mutant, but time-of-addition and RNA transfection assays indicated that posaconazole also inhibits post-entry stages of the viral replication cycle. Virus replication in the presence of posaconazole was partially rescued by the addition of exogenous cholesterol. A transferrin uptake assay revealed that posaconazole considerably slowed down cellular endocytosis. A single point mutation in the SFV E2 glycoprotein, H255R, provided partial resistance to posaconazole as well as to methyl-ß-cyclodextrin, corroborating the effect of posaconazole on cholesterol and viral entry. Our results indicate that posaconazole inhibits multiple steps of the alphavirus replication cycle and broaden the spectrum of viruses that can be targeted in vitro by posaconazole, which could be further explored as a therapeutic agent against emerging viruses.


Subject(s)
Alphavirus/drug effects , Antiviral Agents/pharmacology , Drug Repositioning/methods , Triazoles/pharmacology , Virus Replication/drug effects , Alphavirus/classification , Animals , Cell Line , Chikungunya virus/drug effects , Chlorocebus aethiops , Cricetinae , Endocytosis/drug effects , Humans , Semliki forest virus/drug effects , Sindbis Virus/drug effects , Vero Cells , Virus Internalization/drug effects
8.
Indian J Pharmacol ; 53(5): 403-411, 2021.
Article in English | MEDLINE | ID: mdl-34854411

ABSTRACT

Viral infections are posing a great threat to humanity for the last few years. Among these, Chikungunya which is a mosquito-borne viral infection has produced enormous epidemics around the world after been rebounded. Although this infection shows a low mortality rate, patients suffer from fever, arthralgia, and maculopapular rashes, which reduce the quality of life for several weeks to years. The currently available treatments only provide symptomatic relief based on analgesics, antipyretics, and anti-inflammatory drugs which are nonspecific without satisfactory results. Medicinal plants are a widely accepted source of new molecules for the treatment of infectious diseases including viral infections. The scientific reports, primarily focusing on the anti-chikungunya activity of plant extracts, natural origin pure compounds, and their synthetic analog published from 2011 to 2021, were selected from PubMed, Google Scholar, and Scopus by using related keywords like anti-chikungunya plants, natural antivirals for Chikungunya. The present review decodes scientific reports on medicinal plants against chikungunya virus (CHIKV) infection and demystifies the potential phytoconstituents which reveals that the screening of flavonoids containing plants and phytochemicals showing efficacy against other arbovirus infections, may prove as a potential lead for drug development against CHIKV. The present article also outlines pathogenesis, clinical aspects, molecular virology, and diagnostic approaches of CHIKV infection.


Subject(s)
Antiviral Agents/pharmacology , Chikungunya Fever/drug therapy , Chikungunya virus/drug effects , Plant Extracts/pharmacology , Antiviral Agents/therapeutic use , Humans , Phytotherapy , Plant Extracts/therapeutic use , Plants, Medicinal
9.
PLoS Negl Trop Dis ; 15(11): e0009916, 2021 11.
Article in English | MEDLINE | ID: mdl-34762680

ABSTRACT

Tomatidine, a natural steroidal alkaloid from unripe green tomatoes has been shown to exhibit many health benefits. We recently provided in vitro evidence that tomatidine reduces the infectivity of Dengue virus (DENV) and Chikungunya virus (CHIKV), two medically important arthropod-borne human infections for which no treatment options are available. We observed a potent antiviral effect with EC50 values of 0.82 µM for DENV-2 and 1.3 µM for CHIKV-LR. In this study, we investigated how tomatidine controls CHIKV infectivity. Using mass spectrometry, we identified that tomatidine induces the expression of p62, CD98, metallothionein and thioredoxin-related transmembrane protein 2 in Huh7 cells. The hits p62 and CD98 were validated, yet subsequent analysis revealed that they are not responsible for the observed antiviral effect. In parallel, we sought to identify at which step of the virus replication cycle tomatidine controls virus infectivity. A strong antiviral effect was seen when in vitro transcribed CHIKV RNA was transfected into Huh7 cells treated with tomatidine, thereby excluding a role for tomatidine during CHIKV cell entry. Subsequent determination of the number of intracellular viral RNA copies and viral protein expression levels during natural infection revealed that tomatidine reduces the RNA copy number and viral protein expression levels in infected cells. Once cells are infected, tomatidine is not able to interfere with active RNA replication yet it can reduce viral protein expression. Collectively, the results delineate that tomatidine controls viral protein expression to exert its antiviral activity. Lastly, sequential passaging of CHIKV in presence of tomatidine did not lead to viral resistance. Collectively, these results further emphasize the potential of tomatidine as an antiviral treatment towards CHIKV infection.


Subject(s)
Antiviral Agents/pharmacology , Chikungunya virus/drug effects , Chikungunya virus/genetics , Gene Expression/drug effects , Tomatine/analogs & derivatives , Viral Proteins/genetics , Virus Release/drug effects , Animals , Cell Line , Chlorocebus aethiops , Humans , Proteomics , RNA, Viral/genetics , Tomatine/pharmacology , Vero Cells , Virus Internalization/drug effects , Virus Replication/drug effects
10.
PLoS Pathog ; 17(11): e1009667, 2021 11.
Article in English | MEDLINE | ID: mdl-34780576

ABSTRACT

Chikungunya virus (CHIKV) epidemics around the world have created public health concern with the unavailability of effective drugs and vaccines. This emphasizes the need for molecular understanding of host-virus interactions for developing effective targeted antivirals. Microarray analysis was carried out using CHIKV strain (Prototype and Indian) infected Vero cells and two host isozymes, MAPK activated protein kinase 2 (MK2) and MAPK activated protein kinase 3 (MK3) were selected for further analysis. The substrate spectrum of both enzymes is indistinguishable and covers proteins involved in cytokines production, endocytosis, reorganization of the cytoskeleton, cell migration, cell cycle control, chromatin remodeling and transcriptional regulation. Gene silencing and drug treatment were performed in vitro and in vivo to unravel the role of MK2/MK3 in CHIKV infection. Gene silencing of MK2 and MK3 abrogated around 58% CHIKV progeny release from the host cell and a MK2 activation inhibitor (CMPD1) treatment demonstrated 68% inhibition of viral infection suggesting a major role of MAPKAPKs during late CHIKV infection in vitro. Further, it was observed that the inhibition in viral infection is primarily due to the abrogation of lamellipodium formation through modulation of factors involved in the actin cytoskeleton remodeling pathway. Moreover, CHIKV-infected C57BL/6 mice demonstrated reduction in the viral copy number, lessened disease score and better survivability after CMPD1 treatment. In addition, reduction in expression of key pro-inflammatory mediators such as CXCL13, RAGE, FGF, MMP9 and increase in HGF (a CHIKV infection recovery marker) was observed indicating the effectiveness of the drug against CHIKV. Taken together it can be proposed that MK2 and MK3 are crucial host factors for CHIKV infection and can be considered as important target for developing effective anti-CHIKV strategies.


Subject(s)
Actins/metabolism , Anilides/pharmacology , Antiviral Agents/pharmacology , Chikungunya Fever/prevention & control , Chikungunya virus/drug effects , Cytoskeleton/drug effects , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Tetrahydronaphthalenes/pharmacology , Actins/drug effects , Animals , Chikungunya Fever/virology , Chlorocebus aethiops , Male , Mice , Mice, Inbred C57BL , Vero Cells , Virus Release
11.
PLoS One ; 16(9): e0255125, 2021.
Article in English | MEDLINE | ID: mdl-34492036

ABSTRACT

Chikungunya virus (CHIKV) is an arthropod-borne virus that causes large outbreaks world-wide leaving millions of people with severe and debilitating arthritis. Interestingly, clinical presentation of CHIKV arthritides have many overlapping features with rheumatoid arthritis including cellular and cytokine pathways that lead to disease development and progression. Currently, there are no specific treatments or vaccines available to treat CHIKV infections therefore advocating the need for the development of novel therapeutic strategies to treat CHIKV rheumatic disease. Herein, we provide an in-depth analysis of an efficacious new treatment for CHIKV arthritis with a semi-synthetic sulphated polysaccharide, Pentosan Polysulfate Sodium (PPS). Mice treated with PPS showed significant functional improvement as measured by grip strength and a reduction in hind limb foot swelling. Histological analysis of the affected joint showed local inflammation was reduced as seen by a decreased number of infiltrating immune cells. Additionally, joint cartilage was protected as demonstrated by increased proteoglycan staining. Using a multiplex-immunoassay system, we also showed that at peak disease, PPS treatment led to a systemic reduction of the chemokines CXCL1, CCL2 (MCP-1), CCL7 (MCP-3) and CCL12 (MCP-5) which may be associated with the reduction in cellular infiltrates. Further characterisation of the local effect of PPS in its action to reduce joint and muscle inflammation was performed using NanoString™ technology. Results showed that PPS altered the local expression of key functional genes characterised for their involvement in growth factor signalling and lymphocyte activation. Overall, this study shows that PPS is a promising treatment for alphaviral arthritis by reducing inflammation and protecting joint integrity.


Subject(s)
Arthritis, Rheumatoid/drug therapy , Chikungunya Fever/drug therapy , Chikungunya virus/drug effects , Cytokines/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Pentosan Sulfuric Polyester/pharmacology , Animals , Anticoagulants/pharmacology , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/virology , Chikungunya Fever/immunology , Chikungunya Fever/pathology , Chikungunya Fever/virology , Chikungunya virus/immunology , Chikungunya virus/isolation & purification , Disease Models, Animal , Female , Inflammation/drug therapy , Inflammation/immunology , Inflammation/pathology , Inflammation/virology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred C57BL
12.
Microbiol Res ; 252: 126858, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34509708

ABSTRACT

Proteases play an indispensable role in the life cycles of several life-threatening organisms such as the ones causing malaria, cancer and AIDS. A targeted blockade of these enzymes could be an efficient approach for drug modeling against these causative agents. Our study was directed towards the extraction and characterization of a protease inhibitor having activity against Chikungunya virus (CHIKV). A protein-based protease inhibitor (PI) in Streptomyces griseoincarnatus HK12 with anti-viral activity against CHIKV was revealed when screened against two major proteases, papain and trypsin. The PI was efficiently extracted at 60 % ammonium sulfate saturation and purified by ion-exchange chromatography (CM-Sepharose) at 300 mM NaCl elution followed by SDS-PAGE (10 %). The protein was characterized by denaturing SDS-PAGE, reverse zymography, and MALDI-TOF peptide mass fingerprinting. The protein-based PI was studied to have a high molecular weight of 66-70 kDA. The PI was tested to supress the supress cytopathic effects (CPE) exerted by the clinically isolated virus in BHK21 cells. This was used as a measure to determine the antiviral activity. The PI exerted significant effects with an effective concentration calculated as EC50 11.21 µg/mL. The protein was found to be reported as the first of its kind which also stands out to be the first a natural protease inhibitor against the treatment of the chikungunya virus.


Subject(s)
Chikungunya virus , Protease Inhibitors , Streptomyces , Antiviral Agents/chemistry , Antiviral Agents/isolation & purification , Antiviral Agents/pharmacology , Chikungunya virus/drug effects , Electrophoresis, Polyacrylamide Gel , Protease Inhibitors/chemistry , Protease Inhibitors/isolation & purification , Protease Inhibitors/pharmacology , Streptomyces/chemistry
13.
Virol Sin ; 36(6): 1465-1474, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34374926

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.


Subject(s)
Alphavirus/drug effects , Amaryllidaceae Alkaloids/pharmacology , Chikungunya virus/drug effects , Phenanthridines/pharmacology , Virus Replication , Alphavirus/physiology , Animals , Cell Line , Chikungunya virus/physiology , Semliki forest virus , Sindbis Virus
14.
Cells ; 10(7)2021 07 20.
Article in English | MEDLINE | ID: mdl-34359995

ABSTRACT

Chikungunya virus (CHIKV) is a re-emerging, mosquito-transmitted, enveloped positive stranded RNA virus. Chikungunya fever is characterized by acute and chronic debilitating arthritis. Although multiple host factors have been shown to enhance CHIKV infection, the molecular mechanisms of cell entry and entry factors remain poorly understood. The phosphatidylserine-dependent receptors, T-cell immunoglobulin and mucin domain 1 (TIM-1) and Axl receptor tyrosine kinase (Axl), are transmembrane proteins that can serve as entry factors for enveloped viruses. Previous studies used pseudoviruses to delineate the role of TIM-1 and Axl in CHIKV entry. Conversely, here, we use the authentic CHIKV and cells ectopically expressing TIM-1 or Axl and demonstrate a role for TIM-1 in CHIKV infection. To further characterize TIM-1-dependent CHIKV infection, we generated cells expressing domain mutants of TIM-1. We show that point mutations in the phosphatidylserine binding site of TIM-1 lead to reduced cell binding, entry, and infection of CHIKV. Ectopic expression of TIM-1 renders immortalized keratinocytes permissive to CHIKV, whereas silencing of endogenously expressed TIM-1 in human hepatoma cells reduces CHIKV infection. Altogether, our findings indicate that, unlike Axl, TIM-1 readily promotes the productive entry of authentic CHIKV into target cells.


Subject(s)
Chikungunya virus/genetics , Hepatitis A Virus Cellular Receptor 1/genetics , Host-Pathogen Interactions/genetics , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Virus/genetics , Virus Internalization , Animals , Antibodies, Monoclonal/pharmacology , CHO Cells , Cell Line , Cell Line, Tumor , Chikungunya virus/drug effects , Chikungunya virus/growth & development , Chikungunya virus/immunology , Chlorocebus aethiops , Cricetulus , Endosomes/drug effects , Endosomes/immunology , Endosomes/metabolism , Epithelial Cells/immunology , Epithelial Cells/virology , Fibroblasts/immunology , Fibroblasts/virology , Gene Expression , HEK293 Cells , Hepatitis A Virus Cellular Receptor 1/antagonists & inhibitors , Hepatitis A Virus Cellular Receptor 1/immunology , Hepatocytes/immunology , Hepatocytes/virology , Host-Pathogen Interactions/immunology , Humans , Keratinocytes/immunology , Keratinocytes/virology , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/immunology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/immunology , Receptors, Virus/antagonists & inhibitors , Receptors, Virus/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Transgenes , Vero Cells , Virus Internalization/drug effects , Axl Receptor Tyrosine Kinase
15.
Int J Mol Sci ; 22(15)2021 Jul 23.
Article in English | MEDLINE | ID: mdl-34360656

ABSTRACT

Chikungunya virus (CHIKV) is a mosquito-transmitted infectious agent that causes an endemic or epidemic outbreak(s) of Chikungunya fever that is reported in almost all countries. This virus is an intense global threat, due to its high rate of contagion and the lack of effective remedies. In this study, we developed two baculovirus expression vector system (BEVS)-based approaches for the screening of anti-CHIKV drugs in Spodoptera frugiperda insect (Sf21) cells and U-2OS cells. First, structural protein of CHIKV was co-expressed through BEVS and thereby induced cell fusion in Sf21 cells. We used an internal ribosome entry site (IRES) to co-express the green fluorescent protein (EGFP) for identifying these fusion events. The EGFP-positive Sf21 cells fused with each other and with uninfected cells to form syncytia. We identified that ursolic acid has potential anti-CHIKV activity in vitro, by using this approach. Second, BacMam virus-based gene delivery has been successfully applied for the transient expression of non-structural proteins with a subgenomic promoter-EGFP (SP-EGFP) cassette in U-2OS cells to act as an in vitro CHIKV replicon system. Our BacMam-based screening system has identified that the potential effects of baicalin and baicalein phytocompounds can inhibit the replicon activity of CHIKV in U-2OS cells. In conclusion, our results suggested that BEVS can be a potential tool for screening drugs against CHIKV.


Subject(s)
Antiviral Agents/pharmacology , Baculoviridae/genetics , Cell Fusion , Chikungunya Fever/drug therapy , Chikungunya virus/drug effects , Viral Envelope Proteins/metabolism , Virus Replication , Animals , Chikungunya Fever/virology , Genetic Vectors/genetics , Green Fluorescent Proteins , High-Throughput Screening Assays , Mosquito Vectors , Sf9 Cells , Viral Envelope Proteins/genetics
16.
Viruses ; 13(7)2021 07 05.
Article in English | MEDLINE | ID: mdl-34372513

ABSTRACT

Chikungunya virus (CHIKV) is a mosquito-transmitted alphavirus that has re-emerged in recent decades, causing large-scale epidemics in many parts of the world. CHIKV infection leads to a febrile disease known as chikungunya fever (CHIKF), which is characterised by severe joint pain and myalgia. As many patients develop a painful chronic stage and neither antiviral drugs nor vaccines are available, the development of a potent CHIKV inhibiting drug is crucial for CHIKF treatment. A comprehensive summary of current antiviral research and development of small-molecule inhibitor against CHIKV is presented in this review. We highlight different approaches used for the identification of such compounds and further discuss the identification and application of promising viral and host targets.


Subject(s)
Antiviral Agents/pharmacology , Chikungunya Fever/virology , Chikungunya virus/drug effects , Virus Replication/drug effects , Animals , Chikungunya Fever/drug therapy , Computer Simulation , Culicidae , Disease Models, Animal , Drug Development , Humans , Mice
17.
Viruses ; 13(8)2021 08 12.
Article in English | MEDLINE | ID: mdl-34452467

ABSTRACT

Type III interferons (lambda IFNs) are a quite new, small family of three closely related cytokines with interferon-like activity. Attention to IFN-λ is mainly focused on direct antiviral activity in which, as with IFN-α, viral genome replication is inhibited without the participation of immune system cells. The heterodimeric receptor for lambda interferons is exposed mainly on epithelial cells, which limits its possible action on other cells, thus reducing the likelihood of developing undesirable side effects compared to type I IFN. In this study, we examined the antiviral potential of exogenous human IFN-λ1 in cellular models of viral infection. To study the protective effects of IFN-λ1, three administration schemes were used: 'preventive' (pretreatment); 'preventive/therapeutic' (pre/post); and 'therapeutic' (post). Three IFN-λ1 concentrations (from 10 to 500 ng/mL) were used. We have shown that human IFN-λ1 restricts SARS-CoV-2 replication in Vero cells with all three treatment schemes. In addition, we have shown a decrease in the viral loads of CHIKV and IVA with the 'preventive' and 'preventive/therapeutic' regimes. No significant antiviral effect of IFN-λ1 against AdV was detected. Our study highlights the potential for using IFN-λ as a broad-spectrum therapeutic agent against respiratory RNA viruses.


Subject(s)
Adenoviruses, Human/drug effects , Chikungunya virus/drug effects , Influenza A virus/drug effects , Interferons/pharmacology , SARS-CoV-2/drug effects , A549 Cells , Adenoviruses, Human/physiology , Animals , Chikungunya virus/physiology , Chlorocebus aethiops , Dose-Response Relationship, Drug , Gene Expression Regulation , Humans , Influenza A virus/physiology , Interferons/therapeutic use , Interleukins , RNA Virus Infections/drug therapy , RNA Virus Infections/prevention & control , Recombinant Proteins/pharmacology , SARS-CoV-2/physiology , Vero Cells , Viral Load/drug effects , Virus Replication/drug effects , Interferon Lambda
18.
Int Immunopharmacol ; 99: 108045, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34435582

ABSTRACT

AIM: Andrographolide, the major bioactive compound of the plant Andrographis paniculata, exerts anti-inflammatory, cyto-, neuro- and hepato-protective effects. Traditional remedies for infectious diseases include A. paniculata for maladies like fever, pain, rashes which are associated with chikungunya and other arboviral diseases. Since andrographolide and A. paniculata have potent antiviral properties, the present review aims to provide a comprehensive report of symptoms and immunological molecules involved in chikungunya virus (CHIKV) infection and the therapeutic role of andrographolide in the mitigation of chikungunya and associated symptoms. MATERIALS AND METHODS: Studies on the therapeutic role of A. paniculata and andrographolide in chikungunya and other viral infections published between 1991 and 2021 were searched on various databases. RESULTS AND DISCUSSION: The havoc created by chikungunya is due to the associated debilitating symptoms including arthralgia and myalgia which sometimes remains for years. The authors reviewed and summarized the various symptoms and immunological molecules related to CHIKV replication and associated inflammation, oxidative and unfolded protein stress, apoptosis and arthritis. Additionally, the authors suggested andrographolide as a remedy for chikungunya and other arboviral infections by highlighting its role in the regulation of molecules involved in unfolded protein response pathway, immunomodulation, inflammation, virus multiplication, oxidative stress, apoptosis and arthritis. CONCLUSION: The present review demonstrated the major complications associated with chikungunya and the role of andrographolide in alleviating the chikungunya associated symptoms to encourage further investigations using this promising compound towards early development of an anti-CHIKV drug. Chemical Compound studied: andrographolide (PubChem CID: 5318517).


Subject(s)
Antiviral Agents/pharmacology , Arthritis, Infectious/drug therapy , Chikungunya Fever/drug therapy , Diterpenes/pharmacology , Animals , Antiviral Agents/therapeutic use , Arthritis, Infectious/virology , Chikungunya Fever/transmission , Chikungunya Fever/virology , Chikungunya virus/drug effects , Diterpenes/therapeutic use , Host Microbial Interactions , Humans
19.
J Gen Virol ; 102(7)2021 07.
Article in English | MEDLINE | ID: mdl-34328830

ABSTRACT

The 5' capped, message-sense RNA genome of Chikungunya virus (CHIKV) utilizes the host cell machinery for translation. Translation is regulated by eIF2 alpha at the initiation phase and by eIF4F at cap recognition. Translational suppression by eIF2 alpha phosphorylation occurs as an early event in many alphavirus infections. We observe that in CHIKV-infected HEK293 cells, this occurs as a late event, by which time the viral replication has reached an exponential phase, implying its minimal role in virus restriction. The regulation by eIF4F is mediated through the PI3K-Akt-mTOR, p38 MAPK and RAS-RAF-MEK-ERK pathways. A kinetic analysis revealed that CHIKV infection did not modulate AKT phosphorylation, but caused a significant reduction in p38 MAPK phosphorylation. It caused degradation of phospho-ERK 1/2 by increased autophagy, leaving the PI3K-Akt-mTOR and p38 MAPK pathways for pharmacological targeting. mTOR inhibition resulted in moderate reduction in viral titre, but had no effect on CHIKV E2 protein expression, indicating a minimal role of the mTOR complex in virus replication. Inhibition of p38 MAPK using SB202190 caused a significant reduction in viral titre and CHIKV E2 and nsP3 protein expression. Furthermore, inhibiting the two pathways together did not offer any synergism, indicating that inhibiting the p38 MAPK pathway alone is sufficient to cause restriction of CHIKV replication. Meanwhile, in uninfected cells the fully functional RAS-RAF-MEK-ERK pathway can circumvent the effect of p38 MAPK inhibition on cap-dependent translation. Thus, our results show that host-directed antiviral strategies targeting cellular p38 MAPK are worth exploring against Chikungunya as they could be selective against CHIKV-infected cells with minimal effects on uninfected host cells.


Subject(s)
Autophagy , Chikungunya virus/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Imidazoles/pharmacology , Protein Biosynthesis , Pyridines/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Apoptosis , Cell Line, Tumor , Chikungunya virus/genetics , Chikungunya virus/physiology , Down-Regulation , Enzyme Inhibitors/pharmacology , HEK293 Cells , Humans , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , RNA Caps , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Virus Replication/drug effects
20.
Molecules ; 26(11)2021 Jun 05.
Article in English | MEDLINE | ID: mdl-34198817

ABSTRACT

Despite the serious public health problem represented by the diseases caused by dengue (DENV), Zika (ZIKV) and chikungunya (CHIKV) viruses, there are still no specific licensed antivirals available for their treatment. Here, we examined the potential anti-arbovirus activity of ten di-halogenated compounds derived from L-tyrosine with modifications in amine and carboxyl groups. The activity of compounds on VERO cell line infection and the possible mechanism of action of the most promising compounds were evaluated. Finally, molecular docking between the compounds and viral and cellular proteins was evaluated in silico with Autodock Vina®, and the molecular dynamic with Gromacs®. Only two compounds (TDC-2M-ME and TDB-2M-ME) inhibited both ZIKV and CHIKV. Within the possible mechanism, in CHIKV, the two compounds decreased the number of genome copies and in the pre-treatment strategy the infectious viral particles. In the ZIKV model, only TDB-2M-ME inhibited the viral protein and demonstrate a virucidal effect. Moreover, in the U937 cell line infected with CHIKV, both compounds inhibited the viral protein and TDB-2M-ME inhibited the viral genome too. Finally, the in silico results showed a favorable binding energy between the compounds and the helicases of both viral models, the NSP3 of CHIKV and cellular proteins DDC and ß2 adrenoreceptor.


Subject(s)
Antiviral Agents/chemical synthesis , Chikungunya virus/drug effects , Dengue Virus/drug effects , Phenols/chemical synthesis , Tyrosine/analogs & derivatives , Zika Virus/drug effects , Animals , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Cell Line , Chikungunya virus/genetics , Chikungunya virus/metabolism , Chlorocebus aethiops , Dengue Virus/genetics , Genome, Viral/drug effects , Halogenation , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Structure , Phenols/chemistry , Phenols/pharmacology , Vero Cells , Zika Virus/genetics , Zika Virus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...