Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 133
Filter
1.
Int. microbiol ; 26(4): 781-790, Nov. 2023. ilus
Article in English | IBECS | ID: ibc-227469

ABSTRACT

Chikungunya virus (CHIKV) is an arthropod-borne virus that has caused several major epidemics globally, including in Indonesia. Although significant progress has been achieved in understanding the epidemiology and genotype circulation of CHIKV in Indonesia, the evolution of Indonesian CHIKV isolates is poorly understood. Thus, our study aimed to perform phylogenetic and mutation analyses of the orf2 gene encoding its viral structural protein to improve our understanding of CHIKV evolution in Indonesia. Complete orf2 gene sequences encoding the viral structural proteins of Indonesian-derived CHIKV were downloaded from GenBank until August 31, 2022. Various bioinformatics tools were employed to perform phylogenetic and mutation analyses of the orf2 gene. We identified 76 complete sequences of orf2 gene of CHIKV isolates originally derived from Indonesia. Maximum likelihood trees demonstrated that the majority (69/76, 90.8%) of Indonesian-derived CHIKV isolates belonged to the Asian genotype, while seven isolates (9.2%) belonged to the East/Central/South African (ECSA) genotype. The Indonesian-derived CHIKV isolates were calculated to be originated in Indonesia around 95 years ago (1927), with 95% highest posterior density (HPD) ranging from 1910 to 1942 and a nucleotide substitution rate of 5.07 × 10−4 (95% HPD: 3.59 × 10−4 to 6.67 × 10−4). Various synonymous and non-synonymous substitutions were identified in the C, E3, E2, 6K, and E1 genes. Most importantly, the E1-A226V mutation, which has been reported to increase viral adaptation in Aedes albopictus mosquitoes, was present in all ECSA isolates. To our knowledge, our study is the first comprehensive research analyzing the mutation and evolution of Indonesian-derived CHIKV based on complete sequences of the orf2 genes encoding its viral structural proteins. Our results clearly showed a dynamic evolution of CHIKV circulating in Indonesia.(AU)


Subject(s)
Humans , Male , Female , Indonesia/epidemiology , Chikungunya virus/genetics , Phylogeny , Chikungunya Fever/microbiology , DNA Mutational Analysis , Microbiology , Chikungunya virus/growth & development , Chikungunya virus/pathogenicity , Chikungunya Fever/epidemiology
2.
Molecules ; 27(4)2022 Feb 09.
Article in English | MEDLINE | ID: mdl-35208958

ABSTRACT

Chikungunya is an infectious disease caused by mosquito-transmitted chikungunya virus (CHIKV). It was reported that NS1 and E2 siRNAs administration demonstrated CHIKV inhibition in in vitro as well as in vivo systems. Cationic lipids are promising for designing safe non-viral vectors and are beneficial in treating chikungunya. In this study, nanodelivery systems (hybrid polymeric/solid lipid nanoparticles) using cationic lipids (stearylamine, C9 lipid, and dioctadecylamine) and polymers (branched PEI-g-PEG -PEG) were prepared, characterized, and complexed with siRNA. The four developed delivery systems (F1, F2, F3, and F4) were assessed for stability and potential toxicities against CHIKV. In comparison to the other nanodelivery systems, F4 containing stearylamine (Octadecylamine; ODA), with an induced optimum cationic charge of 45.7 mV in the range of 152.1 nm, allowed maximum siRNA complexation, better stability, and higher transfection, with strong inhibition against the E2 and NS1 genes of CHIKV. The study concludes that cationic lipid-like ODA with ease of synthesis and characterization showed maximum complexation by structural condensation of siRNA owing to high transfection alone. Synergistic inhibition of CHIKV along with siRNA was demonstrated in both in vitro and in vivo models. Therefore, ODA-based cationic lipid nanoparticles can be explored as safe, potent, and efficient nonviral vectors overcoming siRNA in vivo complexities against chikungunya.


Subject(s)
Amines , Chikungunya Fever , Chikungunya virus/growth & development , Liposomes , Nanoparticles , RNA, Small Interfering , Amines/chemistry , Amines/pharmacology , Animals , Chikungunya Fever/drug therapy , Chikungunya Fever/metabolism , Chlorocebus aethiops , Liposomes/chemistry , Liposomes/pharmacology , Mice , Nanoparticles/chemistry , RNA, Small Interfering/chemistry , RNA, Small Interfering/pharmacology , Vero Cells
3.
J Gen Virol ; 102(12)2021 12.
Article in English | MEDLINE | ID: mdl-34878970

ABSTRACT

Although RNA viruses have high mutation rates, host cells and organisms work as selective environments, maintaining the viability of virus populations by eliminating deleterious genotypes. In serial passages of RNA viruses in a single cell line, most of these selective bottlenecks are absent, with no virus circulation and replication in different tissues or host alternation. In this work, Aedes aegypti Aag-2 cells were accidentally infected with Chikungunya virus (CHIKV) and Mayaro virus (MAYV). After numerous passages to achieve infection persistency, the infectivity of these viruses was evaluated in Ae. albopictus C6/36 cells, African green monkey Vero cells and primary-cultured human fibroblasts. While these CHIKV and MAYV isolates were still infectious to mosquito cells, they lost their ability to infect mammalian cells. After genome sequencing, it was observed that CHIKV accumulated many nonsynonymous mutations and a significant deletion in the coding sequence of the hypervariable domain in the nsP3 gene. Since MAYV showed very low titres, it was not sequenced successfully. Persistently infected Aag-2 cells also accumulated high loads of short and recombinant CHIKV RNAs, which seemed to have been originated from virus-derived DNAs. In conclusion, the genome of this CHIKV isolate could guide mutagenesis strategies for the production of attenuated or non-infectious (to mammals) CHIKV vaccine candidates. Our results also reinforce that a paradox is expected during passages of cells persistently infected by RNA viruses: more loosening for the development of more diverse virus genotypes and more pressure for virus specialization to this constant cellular environment.


Subject(s)
Chikungunya virus/growth & development , Chikungunya virus/genetics , Genome, Viral/genetics , Alphavirus/genetics , Alphavirus/growth & development , Animals , Cell Line , Culicidae , Host Specificity , Humans , Mammals , Mutation , RNA, Viral/genetics , Viral Load/genetics , Viral Nonstructural Proteins/genetics , Virus Replication/genetics
4.
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
5.
Virology ; 560: 34-42, 2021 08.
Article in English | MEDLINE | ID: mdl-34023723

ABSTRACT

Alphaviruses are positive sense, RNA viruses commonly transmitted by an arthropod vector to a mammalian or avian host. In recent years, a number of the Alphavirus members have reemerged as public health concerns. Transmission from mosquito vector to vertebrate hosts requires an understanding of the interaction between the virus and both vertebrate and insect hosts to develop rational intervention strategies. The current study uncovers a novel role for capsid protein during Chikungunya virus replication whereby the interaction with viral RNA in the E1 coding region regulates protein synthesis processes early in infection. Studies done in both the mammalian and mosquito cells indicate that interactions between viral RNA and capsid protein have functional consequences that are host species specific. Our data support a vertebrate-specific role for capsid:vRNA interaction in temporally regulating viral translation in a manner dependent on the PI3K-AKT-mTOR pathway.


Subject(s)
Capsid Proteins/metabolism , Chikungunya virus/growth & development , Protein Biosynthesis/genetics , RNA, Viral/metabolism , Virus Replication/physiology , Aedes/virology , Animals , Capsid/metabolism , Cell Line , Chikungunya Fever/pathology , Chikungunya virus/genetics , Cricetinae , Gene Expression Regulation, Viral/genetics , Mosquito Vectors/virology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Viral/genetics , TOR Serine-Threonine Kinases/metabolism
6.
PLoS Pathog ; 17(2): e1009110, 2021 02.
Article in English | MEDLINE | ID: mdl-33556143

ABSTRACT

Defective viral genomes (DVGs) are truncated and/or rearranged viral genomes produced during virus replication. Described in many RNA virus families, some of them have interfering activity on their parental virus and/or strong immunostimulatory potential, and are being considered in antiviral approaches. Chikungunya virus (CHIKV) is an alphavirus transmitted by Aedes spp. that infected millions of humans in the last 15 years. Here, we describe the DVGs arising during CHIKV infection in vitro in mammalian and mosquito cells, and in vivo in experimentally infected Aedes aegypti mosquitoes. We combined experimental and computational approaches to select DVG candidates most likely to have inhibitory activity and showed that, indeed, they strongly interfere with CHIKV replication both in mammalian and mosquito cells. We further demonstrated that some DVGs present broad-spectrum activity, inhibiting several CHIKV strains and other alphaviruses. Finally, we showed that pre-treating Aedes aegypti with DVGs prevented viral dissemination in vivo.


Subject(s)
Aedes/virology , Antiviral Agents/pharmacology , Chikungunya Fever/transmission , Chikungunya virus/genetics , Defective Viruses/genetics , Genome, Viral , Virus Replication , Animals , Chikungunya Fever/immunology , Chikungunya Fever/virology , Chikungunya virus/growth & development , Chikungunya virus/isolation & purification , Humans , Mosquito Vectors/virology
7.
Emerg Microbes Infect ; 10(1): 305-316, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33539255

ABSTRACT

A chimeric Eilat/ Chikungunya virus (EILV/CHIKV) was previously reported to replicate only in mosquito cells but capable of inducing robust adaptive immunity in animals. Here, we initially selected C7/10 cells to optimize the production of the chimeric virus. A two-step procedure produced highly purified virus stocks, which was shown to not cause hypersensitive reactions in a mouse sensitization study. We further optimized the dose and characterized the kinetics of EILV/CHIKV-induced immunity. A single dose of 108 PFU was sufficient for induction of high levels of CHIKV-specific IgM and IgG antibodies, memory B cell and CD8+ T cell responses. Compared to the live-attenuated CHIKV vaccine 181/25, EILV/CHIKV induced similar levels of CHIKV-specific memory B cells, but higher CD8+ T cell responses at day 28. It also induced stronger CD8+, but lower CD4+ T cell responses than another live-attenuated CHIKV strain (CHIKV/IRES) at day 55 post-vaccination. Lastly, the purified EILV/CHIKV triggered antiviral cytokine responses and activation of antigen presenting cell (APC)s in vivo, but did not induce APCs alone upon in vitro exposure. Overall, our results demonstrate that the EILV/CHIKV vaccine candidate is safe, inexpensive to produce and a potent inducer of both innate and adaptive immunity in mice.


Subject(s)
Chikungunya Fever/prevention & control , Chikungunya virus/immunology , Viral Vaccines/immunology , Animals , Antibodies, Viral/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Culture Techniques , Cell Line , Chikungunya Fever/immunology , Chikungunya Fever/virology , Chikungunya virus/genetics , Chikungunya virus/growth & development , Culicidae , Female , Humans , Mice , Viral Vaccines/administration & dosage , Viral Vaccines/genetics
8.
Protein Pept Lett ; 28(5): 508-519, 2021.
Article in English | MEDLINE | ID: mdl-33121397

ABSTRACT

AIM: To identify and characterize peptide binders to truncated recombinant chikungunya virus envelope protein 2. BACKGROUND: Despite extensive research on the chikungunya virus (CHIKV), the specific antiviral treatment's unavailability has stressed the need for the urgent development of therapeutics. The Envelope protein 2 (E2) of CHIKV that displays putative receptor binding sites and specific epitopes for virus neutralizing antibodies is a critical target for the therapeutic intervention. OBJECTIVE: The study aims to identify the unique peptides that can bind to truncated E2 protein of CHIKV and further explore their properties as potential therapeutic candidate. METHODS: A stretch of CHIKV-E2 (rE2), which is prominently exposed on the surface of virion, was used as bait protein to identify peptide binders to the CHIKV-rE2 using a 12-mer phage display peptide library. Three rounds of biopanning yielded several peptide binders to CHIKV-rE2 and their binding affinities were compared by phage ELISA. Additionally, a fully flexible-blind docking simulation investigated the possible binding modes of the selected peptides. Furthermore, the selected peptides were characterized and their ADMET properties were explored in silico. RESULTS: Five peptides were identified as potential binders based on their robust reactivity to the bait protein. The selected peptides appeared to interact with the crucial residues that were notably exposed on the surface of E1-E2 trimeric structure. The explored in silico studies suggested their non-allergenicity, non-toxicity and likeliness to be antiviral. CONCLUSION: The potential binding peptides of CHIKV-rE2 protein were identified using phage display technology and characterized in silico. The selected peptides could be further used for the development of therapeutics against the CHIKV infection.>.


Subject(s)
Chikungunya virus/chemistry , Computer Simulation , Peptide Library , Viral Envelope Proteins/chemistry , Chikungunya virus/growth & development , Viral Envelope Proteins/genetics
9.
Nature ; 589(7843): 615-619, 2021 01.
Article in English | MEDLINE | ID: mdl-33328629

ABSTRACT

Positive-sense single-stranded RNA viruses, such as coronaviruses, flaviviruses and alphaviruses, carry out transcription and replication inside virus-induced membranous organelles within host cells1-7. The remodelling of the host-cell membranes for the formation of these organelles is coupled to the membrane association of viral replication complexes and to RNA synthesis. These viral niches allow for the concentration of metabolites and proteins for the synthesis of viral RNA, and prevent the detection of this RNA by the cellular innate immune system8. Here we present the cryo-electron microscopy structure of non-structural protein 1 (nsP1) of the alphavirus chikungunya virus, which is responsible for RNA capping and membrane binding of the viral replication machinery. The structure shows the enzyme in its active form, assembled in a monotopic membrane-associated dodecameric ring. The structure reveals the structural basis of the coupling between membrane binding, oligomerization and allosteric activation of the capping enzyme. The stoichiometry-with 12 active sites in a single complex-redefines viral replication complexes as RNA synthesis reactors. The ring shape of the complex implies it has a role in controlling access to the viral organelle and ensuring the exit of properly capped viral RNA. Our results provide high-resolution information about the membrane association of the replication machinery of positive-sense single-stranded RNA viruses, and open up avenues for the further characterization of viral replication on cell membranes and the generation of antiviral agents.


Subject(s)
Cell Membrane/metabolism , Chikungunya virus/growth & development , Chikungunya virus/ultrastructure , Cryoelectron Microscopy , RNA Caps/metabolism , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism , Virus Replication , Animals , Catalytic Domain , Cell Line , Cell Membrane/chemistry , Chikungunya virus/chemistry , Chikungunya virus/genetics , Models, Molecular , RNA Caps/chemistry , RNA, Viral/biosynthesis , RNA, Viral/chemistry , RNA, Viral/metabolism , Viral Nonstructural Proteins/ultrastructure
10.
J Cell Mol Med ; 24(12): 6988-6999, 2020 06.
Article in English | MEDLINE | ID: mdl-32374474

ABSTRACT

Outbreaks of infections with viruses like Sars-CoV-2, Ebola virus and Zika virus lead to major global health and economic problems because of limited treatment options. Therefore, new antiviral drug candidates are urgently needed. The promising new antiviral drug candidate silvestrol effectively inhibited replication of Corona-, Ebola-, Zika-, Picorna-, Hepatis E and Chikungunya viruses. Besides a direct impact on pathogens, modulation of the host immune system provides an additional facet to antiviral drug development because suitable immune modulation can boost innate defence mechanisms against the pathogens. In the present study, silvestrol down-regulated several pro- and anti-inflammatory cytokines (IL-6, IL-8, IL-10, CCL2, CCL18) and increased TNF-α during differentiation and activation of M1-macrophages, suggesting that the effects of silvestrol might cancel each other out. However, silvestrol amplified the anti-inflammatory potential of M2-macrophages by increasing expression of anti-inflammatory surface markers CD206, TREM2 and reducing release of pro-inflammatory IL-8 and CCL2. The differentiation of dendritic cells in the presence of silvestrol is characterized by down-regulation of several surface markers and cytokines indicating that differentiation is impaired by silvestrol. In conclusion, silvestrol influences the inflammatory status of immune cells depending on the cell type and activation status.


Subject(s)
Antiviral Agents/pharmacology , Betacoronavirus/drug effects , Cytokines/genetics , Dendritic Cells/drug effects , Immunologic Factors/pharmacology , Macrophages/drug effects , Triterpenes/pharmacology , Betacoronavirus/growth & development , Betacoronavirus/immunology , Cell Differentiation/drug effects , Chikungunya virus/drug effects , Chikungunya virus/growth & development , Chikungunya virus/immunology , Cytokines/classification , Cytokines/immunology , Dendritic Cells/immunology , Dendritic Cells/virology , Ebolavirus/drug effects , Ebolavirus/growth & development , Ebolavirus/immunology , Gene Expression Profiling , Gene Expression Regulation/drug effects , Hepatitis E virus/drug effects , Hepatitis E virus/growth & development , Hepatitis E virus/immunology , Humans , Immunity, Innate/drug effects , Macrophages/immunology , Macrophages/virology , Organ Specificity , Picornaviridae/drug effects , Picornaviridae/growth & development , Picornaviridae/immunology , Primary Cell Culture , SARS-CoV-2 , Signal Transduction , Zika Virus/drug effects , Zika Virus/growth & development , Zika Virus/immunology
11.
J Biol Chem ; 295(23): 7941-7957, 2020 06 05.
Article in English | MEDLINE | ID: mdl-32341071

ABSTRACT

Chikungunya fever is a re-emerging zoonotic disease caused by chikungunya virus (CHIKV), a member of the Alphavirus genus in the Togaviridae family. Only a few studies have reported on the host factors required for intracellular CHIKV trafficking. Here, we conducted an imaging-based siRNA screen to identify human host factors for intracellular trafficking that are involved in CHIKV infection, examined their interactions with CHIKV proteins, and investigated the contributions of these proteins to CHIKV infection. The results of the siRNA screen revealed that host endosomal sorting complexes required for transport (ESCRT) proteins are recruited during CHIKV infection. Co-immunoprecipitation analyses revealed that both structural and nonstructural CHIKV proteins interact with hepatocyte growth factor-regulated tyrosine kinase substrate (HGS), a component of the ESCRT-0 complex. We also observed that HGS co-localizes with the E2 protein of CHIKV and with dsRNA, a marker of the replicated CHIKV genome. Results from gene knockdown analyses indicated that, along with other ESCRT factors, HGS facilitates both genome replication and post-translational steps during CHIKV infection. Moreover, we show that ESCRT factors are also required for infections with other alphaviruses. We conclude that during CHIKV infection, several ESCRT factors are recruited via HGS and are involved in viral genome replication and post-translational processing of viral proteins.


Subject(s)
Chikungunya Fever/metabolism , Chikungunya Fever/virology , Chikungunya virus/growth & development , Chikungunya virus/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Virus Replication , Chikungunya virus/genetics , HEK293 Cells , Humans , Virus Replication/genetics
12.
Molecules ; 25(6)2020 Mar 11.
Article in English | MEDLINE | ID: mdl-32168734

ABSTRACT

Nucleoside analogs are widely used for the treatment of viral diseases (Hepatitis B/C, herpes and human immunodeficiency virus, HIV) and various malignancies. ALS-8176, a prodrug of the 4'-chloromethyl-2'-deoxy-2'-fluoro nucleoside ALS-8112, was evaluated in hospitalized infants for the treatment of respiratory syncytial virus (RSV), but was abandoned for unclear reasons. Based on the structure of ALS-8112, a series of novel 4'-modified-2'-deoxy-2'-fluoro nucleosides were synthesized. Newly prepared compounds were evaluated against RSV, but also against a panel of RNA viruses, including Dengue, West Nile, Chikungunya, and Zika viruses. Unfortunately, none of the compounds showed marked antiviral activity against these viruses.


Subject(s)
Antiviral Agents/chemical synthesis , Deoxycytidine/analogs & derivatives , Deoxyribonucleosides/chemical synthesis , Prodrugs/chemical synthesis , Animals , Antiviral Agents/pharmacology , Cell Line, Tumor , Chikungunya virus/drug effects , Chikungunya virus/growth & development , Cricetulus , Dengue Virus/drug effects , Dengue Virus/growth & development , Deoxycytidine/chemical synthesis , Deoxycytidine/pharmacology , Deoxyribonucleosides/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/virology , Hepatocytes/drug effects , Hepatocytes/virology , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/virology , Microbial Sensitivity Tests , Primary Cell Culture , Prodrugs/pharmacology , Respiratory Syncytial Virus, Human/drug effects , Respiratory Syncytial Virus, Human/growth & development , T-Lymphocytes/drug effects , T-Lymphocytes/virology , Treatment Failure , Virus Replication/drug effects , West Nile virus/drug effects , West Nile virus/growth & development , Zika Virus/drug effects , Zika Virus/growth & development
13.
J Gen Virol ; 100(11): 1541-1553, 2019 11.
Article in English | MEDLINE | ID: mdl-31613205

ABSTRACT

Chikungunya virus (CHIKV) has caused large-scale epidemics of fever, rash and arthritis since 2004. This unprecedented re-emergence has been associated with mutations in genes encoding structural envelope proteins, providing increased fitness in the secondary vector Aedes albopictus. In the 2008-2013 CHIKV outbreaks across Southeast Asia, an R82S mutation in non-structural protein 4 (nsP4) emerged early in Malaysia or Singapore and quickly became predominant. To determine whether this nsP4-R82S mutation provides a selective advantage in host cells, which may have contributed to the epidemic, the fitness of infectious clone-derived CHIKV with wild-type nsP4-82R and mutant nsP4-82S were compared in Ae. albopictus and human cell lines. Viral infectivity, dissemination and transmission in Ae. albopictus were not affected by the mutation when the two variants were tested separately. In competition, the nsP4-82R variant showed an advantage over nsP4-82S in dissemination to the salivary glands, but only in late infection (10 days). In human rhabdomyosarcoma (RD) and embryonic kidney (HEK-293T) cell lines coinfected at a 1 : 1 ratio, wild-type nsP4-82R virus was rapidly outcompeted by nsP4-82S virus as early as one passage (3 days). In conclusion, the nsP4-R82S mutation provides a greater selective advantage in human cells than in Ae. albopictus, which may explain its apparent natural selection during CHIKV spread in Southeast Asia. This is an unusual example of a naturally occurring mutation in a non-structural protein, which may have facilitated epidemic transmission of CHIKV.


Subject(s)
Chikungunya virus/growth & development , Genetic Fitness , Mutation, Missense , Viral Nonstructural Proteins/genetics , Virulence Factors/genetics , Aedes , Animals , Cell Line , Chikungunya virus/genetics , Humans , Mutant Proteins/genetics , Selection, Genetic
14.
Nature ; 574(7777): 259-263, 2019 10.
Article in English | MEDLINE | ID: mdl-31554973

ABSTRACT

Chikungunya virus (CHIKV) is a re-emerging alphavirus that is transmitted to humans by mosquito bites and causes musculoskeletal and joint pain1,2. Despite intensive investigations, the human cellular factors that are critical for CHIKV infection remain unknown, hampering the understanding of viral pathogenesis and the development of anti-CHIKV therapies. Here we identified the four-and-a-half LIM domain protein 1 (FHL1)3 as a host factor that is required for CHIKV permissiveness and pathogenesis in humans and mice. Ablation of FHL1 expression results in the inhibition of infection by several CHIKV strains and o'nyong-nyong virus, but not by other alphaviruses and flaviviruses. Conversely, expression of FHL1 promotes CHIKV infection in cells that do not normally express it. FHL1 interacts directly with the hypervariable domain of the nsP3 protein of CHIKV and is essential for the replication of viral RNA. FHL1 is highly expressed in CHIKV-target cells and is particularly abundant in muscles3,4. Dermal fibroblasts and muscle cells derived from patients with Emery-Dreifuss muscular dystrophy that lack functional FHL15 are resistant to CHIKV infection. Furthermore,  CHIKV infection  is undetectable in Fhl1-knockout mice. Overall, this study shows that FHL1 is a key factor expressed by the host that enables CHIKV infection and identifies the interaction between nsP3 and FHL1 as a promising target for the development of anti-CHIKV therapies.


Subject(s)
Chikungunya Fever/virology , Chikungunya virus/pathogenicity , Host-Derived Cellular Factors/metabolism , Host-Pathogen Interactions , Intracellular Signaling Peptides and Proteins/metabolism , LIM Domain Proteins/metabolism , Muscle Proteins/metabolism , Animals , Cells, Cultured , Chikungunya Fever/drug therapy , Chikungunya virus/drug effects , Chikungunya virus/genetics , Chikungunya virus/growth & development , Female , Fibroblasts/virology , HEK293 Cells , Host-Derived Cellular Factors/genetics , Humans , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , LIM Domain Proteins/deficiency , LIM Domain Proteins/genetics , Male , Mice , Muscle Proteins/deficiency , Muscle Proteins/genetics , Myoblasts/virology , O'nyong-nyong Virus/growth & development , O'nyong-nyong Virus/pathogenicity , Protein Binding , RNA, Viral/biosynthesis , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Virus Replication
15.
Indian J Med Res ; 149(6): 771-777, 2019 06.
Article in English | MEDLINE | ID: mdl-31496530

ABSTRACT

Background & objectives: Chikungunya virus (CHIKV), a mosquito-borne arthritogenic virus causes infections ranging from febrile illness to debilitating polyarthralgia in humans. Re-emergence of the virus has affected millions of people in Africa and Asia since 2004. During the outbreak, a new lineage of the virus has evolved as an adaptation for enhanced replication and transmission by Aedes albopictus mosquito. A study was designed to compare the susceptibility of four vertebrate cell lines, namely Vero E6 (African green monkey kidney), BHK-21 (Baby hamster kidney), RD (human rhabdomyosarcoma), A-549 (human alveolar basal epithelial cell) and C6/36 (Ae. albopictus) to Asian genotype and two lineages of East, Central and South African (E1:A226 and E1:A226V) of CHIKV. Methods: One-step growth kinetics of different CHIKV strains was carried out in the above five cell lines to determine the growth kinetics and virus yield. Virus titre was determined by 50 per cent tissue culture infectious dose assay and titres were calculated by the Reed and Muench formula. Growth and virus yield of the three strains in Ae. aegypti mosquitoes was studied by intrathoracic inoculation and virus titration in Vero E6 cell line. Results: Virus titration showed Vero E6, C6/36 and BHK-21 cell lines are high virus yielding with all the three lineages while RD and A-549 yielded low virus titres. C6/36 cell line was the most sensitive and yielded the maximum titre. Ae. aegypti mosquitoes, when inoculated with high titre virus, yielded an almost equal growth with the three strains while rapid growth of E1:A226V and Asian strain was observed with 1 log virus. Interpretation & conclusions: C6/36 cell line was found to be the most sensitive and high yielding for CHIKV irrespective of lineages while Vero E6 and BHK-21 cell lines yielded high titres and may find application for vaccine/diagnostic development. Infection of Ae. aegypti mosquitoes with the three CHIKV strains gave almost identical pattern of growth.


Subject(s)
Aedes/virology , Chikungunya Fever/virology , Chikungunya virus/growth & development , Culicidae/virology , A549 Cells/virology , Africa/epidemiology , Animals , Asia/epidemiology , Chikungunya Fever/epidemiology , Chikungunya Fever/genetics , Chikungunya virus/genetics , Chikungunya virus/pathogenicity , Chlorocebus aethiops , Disease Outbreaks , Genotype , Humans , Mosquito Vectors/genetics , Mosquito Vectors/growth & development , Saliva/virology , Vero Cells/virology
16.
PLoS Negl Trop Dis ; 13(8): e0007610, 2019 08.
Article in English | MEDLINE | ID: mdl-31449523

ABSTRACT

Chikungunya virus (CHIKV) is a re-emerging arbovirus known to cause chronic myalgia and arthralgia with high morbidity. CHIKV is now considered endemic in many countries across Asia and Africa. In this study, the susceptibility of various human, mammalian and mosquito cell lines to CHIKV infection was evaluated. CHIKV infection was found to be cell-type dependent and virus strain-specific. Furthermore, SJCRH30 (human rhabdomyosarcoma cell line) was showed to be highly permissive to CHIKV infection, with maximum production of infectious virions observed at 12 h.p.i. Pre-infection treatment of SJCRH30 with various inhibitors of endocytosis, including monodansylcadaverine (receptor-mediated endocytic inhibitor), dynasore (clathrin-mediated endocytic inhibitor), as well as filipin (caveolin-mediated endocytosis inhibitor), resulted in minimal inhibition of CHIKV infection. In contrast, dose-dependent inhibition of CHIKV infection was observed with the treatment of macropinocytosis inhibitor, 5-(N-ethyl-N-isopropyl)amiloride (EIPA). Furthermore, siRNA-mediated knockdown of sortin nexin 9 (SNX9) a protein involved in macropinosome formation, also resulted in a significant dose-dependent reduction in viral titre. By performing a virus entry assay, CHIKV particles were also observed to colocalize with FITC-dextran, a macropinosome marker. This study shows for the first time, that the infectious entry of CHIKV into human muscle cells is mediated by macropinocytosis. Together, the data from this study may pave the way for the development of specific inhibitors that target the entry process of CHIKV into cells.


Subject(s)
Chikungunya Fever/virology , Chikungunya virus/physiology , Muscles/virology , Pinocytosis/physiology , Virus Internalization , Amiloride/analogs & derivatives , Amiloride/pharmacology , Animals , Cadaverine/analogs & derivatives , Cadaverine/pharmacology , Cell Line , Cell Survival , Chikungunya virus/growth & development , Clathrin/antagonists & inhibitors , Endocytosis/drug effects , Filipin/pharmacology , Gene Knockdown Techniques , Humans , Hydrazones/pharmacology , Kinetics , Pinocytosis/drug effects , Pinocytosis/genetics , RNA, Small Interfering , Rhabdomyosarcoma , Sorting Nexins/genetics , Viral Load , Viral Plaque Assay
17.
Article in English | MEDLINE | ID: mdl-31307983

ABSTRACT

The mosquito-borne chikungunya virus (CHIKV) causes acute pain and joint inflammation, and in recent years the virus has caused large epidemics in previously CHIKV-free geographic areas. To advance the understanding of host factors that antagonize CHIKV, we show that synthetic agonist of liver X receptor (LXR-623) inhibits CHIKV replication by upregulating the cholesterol exporter ABCA1 and that endogenous and pharmacological activation of interferon signaling pathway partners with LXR-623 to generate a superior antiviral state.


Subject(s)
ATP Binding Cassette Transporter 1/genetics , Antiviral Agents/pharmacology , Chikungunya virus/drug effects , Fibroblasts/drug effects , Host-Pathogen Interactions/drug effects , Indazoles/pharmacology , Liver X Receptors/genetics , ATP Binding Cassette Transporter 1/antagonists & inhibitors , ATP Binding Cassette Transporter 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 1/metabolism , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Chikungunya virus/growth & development , Chikungunya virus/metabolism , Cholesterol/metabolism , Cholesterol/pharmacology , Fibroblasts/metabolism , Fibroblasts/virology , Gene Expression Regulation , Humans , Interferons/genetics , Interferons/metabolism , Liver X Receptors/antagonists & inhibitors , Liver X Receptors/metabolism , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Virus Replication/drug effects
18.
Viruses ; 11(6)2019 06 25.
Article in English | MEDLINE | ID: mdl-31242674

ABSTRACT

Chikungunya (CHIK) is a febrile arboviral illness caused by chikungunya virus (CHIKV) and has been identified in more than 60 countries across the globe. A major public health concern, the infection occurs as an acute febrile phase and a chronic arthralgic phase. The disease manifests differently in different age groups that can range from asymptomatic infection in the younger age group to a prolonged chronic phase in the elderly population. The present study was undertaken to evaluate strain-specific pathogenesis of ECSA genotype of CHIKV strains derived from clinical isolates in adult C57BL/6J mice model. The strain that was pathogenic and developed distinct acute and post-acute phase of CHIK infection was further evaluated for dose-dependent pathogenesis. Upon arriving on the optimal dose to induce clinical symptoms in the mice, the disease progression was evaluated across the acute and the post-acute phase of infection for a period of 15 days post-infection in two age groups of mice, namely eight weeks old and 20 weeks old mice groups. Biochemical, hematological, and virology attributes were measured and correlated to morbidity and linked neurotropism and limb thickness in the two age groups. Our results show that CHIKV exhibit strain-specific pathogenesis in C57BL/6J mice. Distinct dissimilarities were observed between the two age groups in terms of pathogenesis, viral clearance and host response to CHIKV infection.


Subject(s)
Chikungunya Fever/pathology , Chikungunya Fever/virology , Chikungunya virus/growth & development , Chikungunya virus/pathogenicity , Disease Models, Animal , Viral Tropism , Age Factors , Animals , Mice, Inbred C57BL
19.
PLoS Negl Trop Dis ; 13(6): e0007429, 2019 06.
Article in English | MEDLINE | ID: mdl-31166953

ABSTRACT

BACKGROUND: RNA interference is among the most important mechanisms that serve to restrict virus replication within mosquitoes, where microRNAs (miRNAs) are important in regulating viral replication and cellular functions. These miRNAs function by binding to complementary sequences mostly in the untranslated regions of the target. Chikungunya virus (CHIKV) genome consists of two open reading frames flanked by 5' and 3' untranslated regions on the two sides. A recent study from our laboratory has shown that Aedes miRNAs are regulated during CHIKV infection. The present study was undertaken to further understand the role of these miRNAs in CHIKV replication. METHODS/FINDINGS: We observe that miR-2944b-5p binds to the 3' untranslated region of CHIKV and the binding is abated when the binding sites are abolished. Loss-of-function studies of miR-2944b-5p using antagomirs, both in vitro and in vivo, reveal an increase in CHIKV viral replication, thereby directly implying a role of miR-2944b-5p in CHIKV replication. We further showed that the mitochondrial membrane potential of the mosquito cells is maintained by this miRNA during CHIKV replication, and cellular factor vps-13 plays a contributing role. CONCLUSIONS: Our study has opened new avenues to understand vector-virus interactions and provides novel insights into CHIKV replication in Aedes aegypti. Furthermore, our study has shown miR-2944b-5p to be playing role, where one of its target vps-13 also contributes, in maintaining mitochondrial membrane potential in Aedes aegypti.


Subject(s)
3' Untranslated Regions , Aedes/virology , Chikungunya virus/growth & development , Insect Proteins/metabolism , MicroRNAs/metabolism , Virus Replication , Animals , Female , Membrane Potentials , Mice, Inbred BALB C , Mitochondrial Membranes/physiology , Nucleic Acid Hybridization , RNA Interference
20.
Multimedia | Multimedia Resources | ID: multimedia-2575
SELECTION OF CITATIONS
SEARCH DETAIL
...