scholarly journals Chikungunya Virus Inhibitor Study based on Molecular Docking Experiments

Author(s):  
A. Saraiva ◽  
Soares Jeferson ◽  
Castro Miranda ◽  
Jose Sousa ◽  
N. Ferreira ◽  
...  
2020 ◽  
Vol 21 (4) ◽  
pp. 325-340 ◽  
Author(s):  
Saeed Anwar ◽  
Jarin T. Mourosi ◽  
Md. Fahim Khan ◽  
Mohammad J. Hosen

Background: Chikungunya is an arthropod-borne viral disease characterized by abrupt onset of fever frequently accompanied by joint pain, which has been identified in over 60 countries in Africa, the Americas, Asia, and Europe. Methods: Regardless of the availability of molecular knowledge of this virus, no definite vaccine or other remedial agents have been developed yet. In the present study, a combination of B-cell and T-cell epitope predictions, followed by molecular docking simulation approach has been carried out to design a potential epitope-based peptide vaccine, which can trigger a critical immune response against the viral infections. Results: A total of 52 sequences of E1 glycoprotein from the previously reported isolates of Chikungunya outbreaks were retrieved and examined through in silico methods to identify a potential B-cell and T-cell epitope. From the two separate epitope prediction servers, five potential B-cell epitopes were selected, among them “NTQLSEAHVEKS” was found highly conserved across strains and manifests high antigenicity with surface accessibility, flexibility, and hydrophilicity. Similarly, two highly conserved, non-allergenic, non-cytotoxic putative T-cell epitopes having maximum population coverage were screened to bind with the HLA-C 12*03 molecule. Molecular docking simulation revealed potential T-cell based epitope “KTEFASAYR” as a vaccine candidate for this virus. Conclusion: A combination of these B-cell and T-cell epitope-based vaccine can open up a new skyline with broader therapeutic application against Chikungunya virus with further experimental and clinical investigation.


PeerJ ◽  
2016 ◽  
Vol 4 ◽  
pp. e2602 ◽  
Author(s):  
Adrian Oo ◽  
Pouya Hassandarvish ◽  
Sek Peng Chin ◽  
Vannajan Sanghiran Lee ◽  
Sazaly Abu Bakar ◽  
...  

BackgroundThe re-emerging,Aedes spp.transmitted Chikungunya virus (CHIKV) has recently caused large outbreaks in a wide geographical distribution of the world including countries in Europe and America. Though fatalities associated with this self-remitting disease were rarely reported, quality of patients’ lives have been severely diminished by polyarthralgia recurrence. Neither effective antiviral treatment nor vaccines are available for CHIKV. Our previous in vitro screening showed that hesperetin, a bioflavonoid exhibits inhibitory effect on the virus intracellular replication. Here, we present a study using the computational approach to identify possible target proteins for future mechanistic studies of hesperetin.Methods3D structures of CHIKV nsP2 (3TRK) and nsP3 (3GPG) were retrieved from Protein Data Bank (PDB), whereas nsP1, nsP4 and cellular factor SPK2 were modeled using Iterative Threading Assembly Refinement (I-TASSER) server based on respective amino acids sequence. We performed molecular docking on hesperetin against all four CHIKV non-structural proteins and SPK2. Proteins preparation and subsequent molecular docking were performed using Discovery Studio 2.5 and AutoDock Vina 1.5.6. The Lipinski’s values of the ligand were computed and compared with the available data from PubChem. Two non-structural proteins with crystal structures 3GPG and 3TRK in complexed with hesperetin, demonstrated favorable free energy of binding from the docking study, were further explored using molecular dynamics (MD) simulations.ResultsWe observed that hesperetin interacts with different types of proteins involving hydrogen bonds, pi-pi effects, pi-cation bonding and pi-sigma interactions with varying binding energies. Among all five tested proteins, our compound has the highest binding affinity with 3GPG at −8.5 kcal/mol. The ligand used in this study also matches the Lipinski’s rule of five in addition to exhibiting closely similar properties with that of in PubChem. The docking simulation was performed to obtain a first guess of the binding structure of hesperetin complex and subsequently analysed by MD simulations to assess the reliability of the docking results. Root mean square deviation (RMSD) of the simulated systems from MD simulations indicated that the hesperetin complex remains stable within the simulation timescale.DiscussionThe ligand’s tendencies of binding to the important proteins for CHIKV replication were consistent with our previous in vitro screening which showed its efficacy in blocking the virus intracellular replication. NsP3 serves as the highest potential target protein for the compound’s inhibitory effect, while it is interesting to highlight the possibility of interrupting CHIKV replication via interaction with host cellular factor. By complying the Lipinski’s rule of five, hesperetin exhibits drug-like properties which projects its potential as a therapeutic option for CHIKV infection.


2018 ◽  
Vol 16 (1) ◽  
Author(s):  
Muhammad Tahir ul Qamar ◽  
Amna Bari ◽  
Muhammad Muzammal Adeel ◽  
Arooma Maryam ◽  
Usman Ali Ashfaq ◽  
...  

2021 ◽  
Vol 18 (1) ◽  
Author(s):  
Poonam Patil ◽  
Megha Agrawal ◽  
Shahdab Almelkar ◽  
Manish Kumar Jeengar ◽  
Ashwini More ◽  
...  

Abstract Background Chikungunya virus (CHIKV), a serious health problem in several tropical countries, is the causative agent of chikungunya fever. Approved antiviral therapies or vaccines for the treatment or prevention of CHIKV infections are not available. As diverse natural phenolic compounds have been shown to possess antiviral activities, we explored the antiviral activity of α-Mangostin, a xanthanoid, against CHIKV infection. Methods The in vitro prophylactic and therapeutic effects of α-Mangostin on CHIKV replication in Vero E6 cells were investigated by administering it under pre, post and cotreatment conditions. The antiviral activity was determined by foci forming unit assay, quantitative RT-PCR and cell-based immune-fluorescence assay. The molecular mechanism of inhibitory action was further proposed using in silico molecular docking studies. Results In vitro studies revealed that 8 µM α-Mangostin completely inhibited CHIKV infectivity under the cotreatment condition. CHIKV replication was also inhibited in virus-infected mice. This is the first in vivo study which clearly showed that α-Mangostin is effective in vivo by significantly reducing virus replication in serum and muscles. Molecular docking indicated that α-Mangostin can efficiently interact with the E2–E1 heterodimeric glycoprotein and the ADP-ribose binding cavity of the nsP3 macrodomain. Conclusions The findings suggest that α-Mangostin can inhibit CHIKV infection and replication through possible interaction with multiple CHIKV target proteins and might act as a prophylactic/therapeutic agent against CHIKV.


Author(s):  
Kristina Kovacikova ◽  
Marina Gorostiola González ◽  
Rhian Jones ◽  
Juan Reguera ◽  
Alba Gigante ◽  
...  

Chikungunya virus (CHIKV) nonstructural protein 1 (nsP1) harbours the methyltransferase (MTase) and guanylyltransferase (GTase) activities needed for viral RNA capping and represents a promising antiviral drug target. We compared the antiviral efficacy of nsP1 inhibitors belonging to the MADTP, CHVB and FHNA series [6’-fluoro-homoneplanocin A (FHNA), its 3’-keto form and 6'-β-Fluoro-homoaristeromycin]. Cell-based phenotypic cross-resistance assays revealed that the CHVB and MADTP series shared a similar mode of action that differed from that of the FHNA series. In biochemical assays with purified Semliki Forest virus and CHIKV nsP1, CHVB compounds strongly inhibited MTase and GTase activities, while MADTP-372 had a moderate inhibitory effect. FHNA did not directly inhibit enzymatic activity of CHIKV nsP1. The first of its kind molecular docking studies with the cryo-EM structure of CHIKV nsP1, which is assembled into a dodecameric ring, revealed that the MADTP and CHVB series bind at the SAM-binding site in the capping domain, where they would function as (non)competitive inhibitors. The FHNA series was predicted to bind at the secondary binding pocket in the Ring-Aperture Membrane-Binding and Oligomerization domain, potentially interfering with membrane binding and oligomerization of nsP1. Our cell-based and enzymatic assays, in combination with molecular docking and mapping of compound-resistance mutations to the nsP1 structure allowed us to group nsP1 inhibitors into functionally distinct classes. This study identified druggable pockets in the nsP1 dodecameric structure and provides a basis for rational design, optimization and combination of inhibitors of this unique and promising antiviral drug target.


Sign in / Sign up

Export Citation Format

Share Document