[go: up one dir, main page]
More Web Proxy on the site http://driver.im/ Skip to main content
Elsevier - PMC Mpox Collection logoLink to Elsevier - PMC Mpox Collection
. 2023 Apr 22;161:106971. doi: 10.1016/j.compbiomed.2023.106971

Identification of core therapeutic targets for Monkeypox virus and repurposing potential of drugs against them: An in silico approach

Anshuman Sahu a,1, Mahendra Gaur b,a,1, Nimai Charan Mahanandia c, Enketeswara Subudhi d, Ranjit Prasad Swain e, Bharat Bhusan Subudhi a,
PMCID: PMC10122558  PMID: 37211001

Abstract

Monkeypox virus (mpox virus) outbreak has rapidly spread to 82 non-endemic countries. Although it primarily causes skin lesions, secondary complications and high mortality (1–10%) in vulnerable populations have made it an emerging threat. Since there is no specific vaccine/antiviral, it is desirable to repurpose existing drugs against mpox virus. With little knowledge about the lifecycle of mpox virus, identifying potential inhibitors is a challenge. Nevertheless, the available genomes of mpox virus in public databases represent a goldmine of untapped possibilities to identify druggable targets for the structure-based identification of inhibitors. Leveraging this resource, we combined genomics and subtractive proteomics to identify highly druggable core proteins of mpox virus. This was followed by virtual screening to identify inhibitors with affinities for multiple targets. 125 publicly available genomes of mpox virus were mined to identify 69 highly conserved proteins. These proteins were then curated manually. These curated proteins were funnelled through a subtractive proteomics pipeline to identify 4 highly druggable, non-host homologous targets namely; A20R, I7L, Top1B and VETFS. High-throughput virtual screening of 5893 highly curated approved/investigational drugs led to the identification of common as well as unique potential inhibitors with high binding affinities. The common inhibitors, i.e., batefenterol, burixafor and eluxadoline were further validated by molecular dynamics simulation to identify their best potential binding modes. The affinity of these inhibitors suggests their repurposing potential. This work can encourage further experimental validation for possible therapeutic management of mpox.

Keywords: Monkeypox, mpox virus, Genome mining, Core proteome, Subtractive proteomics, HTVS, Repurposing, Molecular dynamics simulation, MM/PBSA

Graphical abstract

Image 1

1. Introduction

In the backdrop of ongoing COVID-19 pandemic, the detection of the first case of Monkeypox (mpox) in Europe on 7th May 2022 set the alarm for potentially another pandemic [1]. Although this has not affected society with the same magnitude, the rapid spread to more than 80 non-endemic countries has prompted the World Health Organization (WHO) to declare this outbreak a “Public Health Emergency of International Concern” on 23rd July 2022 [2]. Mpox is a reemerging zoonotic disease common to the Central and Western parts of the African continent [3]. However, the current outbreak which is the most dispersed and largest known to date, has fuelled the fear of the next COVID-19 like a global pandemic in some circles of the international community.

Mpox virus, a double-stranded DNA virus belongs to the family Poxviridae, sub-family Chordopoxviridae, and genus orthopox virus that also includes the Variola virus which is responsible for smallpox [4]. Although its carriers are not limited to monkeys, its naming can be traced back to its first isolation from captive cynomolgus monkeys by a Danish lab in 1958 [5]. The first case of human mpox virus infection was documented in 1970 in the Democratic Republic of Congo, DRC (then Zaire) in an infant [6]. Mpox has since become endemic in DRC and has expanded to other Central and Western African countries [7]. The first human case and the subsequent outbreak of mpox outside Africa was documented in the USA in the year 2003 [[8], [9], [10], [11]]. The human-to-human transmission of mpox occurs primarily through skin lesions of infected humans or animals, respiratory droplets, body fluids and contaminated materials [7,12]. However, the transmission dynamics of the current outbreak in non-endemic countries remain cryptic as in some cases the detected cases do not have any associations with endemic regions [13]. Moreover, animal reservoirs are yet to be identified in these non-endemic regions [13]. Therefore, it has been suggested that the current epidemic in non-endemic regions might be a culmination of multiple mpox virus imports from endemic regions accompanied by silent, undocumented, cryptic inter-human transmissions [13].

Mpox virus has a double-layered cell membrane and a brick-shaped appearance. It exploits its double-stranded DNA to replicate inside the host's cytoplasm [14,15]. The genome of mpox virus is linear, approximately 197,000 bp long and includes hairpin termini. It consists of ≥198 non-overlapping open reading frames (ORFs). The central coding region is highly conserved and is flanked by variable regions with inverted terminal repeats on both sides [[16], [17], [18]]. Half of these ORFs are integral to viral replication and morphogenesis and are well conserved amongst other pox viruses [19]. The remaining half consists of “accessory” genes involved in immunomodulation, pathogenesis and host tropism. Many of them are yet to be functionally characterised [19]. As such these genes may be dispensable to the virus during replication [20]. As of now, pathogenesis and life cycle of mpox virus is very poorly understood. Nonetheless, limited in vitro studies suggest mpox virus can infect most mammalian cells [21,22]. The mature virion most likely attaches itself to host cell via glycosaminoglycans lining the host cell surface or modules of the extracellular matrix and external virion proteins [23,24]. Following attachment, the mature virion enters the host cell either by a low endosomal pathway or by merging with the plasma membrane in a pH-dependent manner to release the viral core into the cytoplasm [18,24]. Inside, the intracellular immature virion synthesises viral proteins to form an intracellular enveloped virion to eventually exit the host cell as an extracellular mature virion [15]. However, our knowledge of viral receptors that enables a virion to attach itself to a host cell remains elusive to this day [15].

The clinical presentation of mpox is somewhat similar to smallpox, albeit milder with three distinct phases [25]. The first phase (incubation phase) ranges from 7 to 14 days [25,26]. The second (prodrome) phase includes fever fluctuating between 38.5 °C and 40.5 °C, accompanied by muscle aches, headache, backache, chills and lymphadenopathy. This phase distinguishes it from smallpox and chickenpox. The third (rash) stage is characterised by macular rash that advances into papular, vesicular, and pustular stages resulting in crusts which eventually fall off [25]. The facial rashes can gradually progress across the body into the genitalia [27]. Complications include nutritional deterioration in patients with rashes in the oral cavity [15], permanent facial distortions upon the healing of facial lesions [28], loss of vision due to corneal infection [29], bacterial superinfections, and bronchopneumonia due to coinfection with influenza [30]. Rare serious complications include myocarditis, epiglottitis and sceptic shock resulting in mortality on account of the exaggerated immune response [31]. Although the infection is generally mild, immunocompromised individuals, children, pregnant women, elderly population and persons with comorbidity like HIV/AIDS may be prone to severe outcomes leading to death [13].

Two FDA approved vaccines initially developed as anecdotes against smallpox are currently being contemplated for effective immunisation against mpox. The first ACAM2000, is a next-generation vaccine similar to the discontinued Dryvax, with an efficacy of 85% [32]. MVA-BN (JYNNEOS in the U.S.) is the second next-generation vaccine developed with the modified Ankara strain (MVA) of the Vaccinia virus [33]. ACAM2000 is contraindicated in cases with atopic dermatitis, immunocompromised individuals, and pregnant women whereas the potency of MVA-BN remains to be validated in clinical trials [33,34]. Vaccinia immunoglobulin (VIG) has also been suggested from successful past experiences with smallpox [35]. Currently, tecovirimat (NCT00728689), cidofovir and brincidofovir (NCT01143181) are the only available antivirals against mpox. Their potency against orthopox virus induced diseases in animal studies has been well documented. However, scientific data of their effectiveness in individuals infected with the currently circulating strains of mpox virus WA clade-II remains speculative [15,36]. Moreover, the development of resistance against tecovirimat and cidofovir has also been documented in orthopox viruses by virtue of acquisition of drug-resistant mutations in F13L or E9L [37]. Thus, taken together, the current therapeutic arsenal against mpox is almost deserted.

Drug repurposing offers a tangible solution, considering the immediate necessity and prohibitive time/cost involved in new drug development. Structure-based virtual screening of drug libraries is viable strategy to find the repurposing potential of drugs. However, the selection of a target and the availability of quality structures are keys to the success of this approach. With little information about the targets of mpox virus and their structure, virtual screening against mpox virus is a challenge. Nonetheless, the available genomes in public repositories are a goldmine of untapped possibilities. However, judicious selection of target is necessary for successful repurposing. Subtractive proteomics approach has generally been used to prioritise bacterial targets [[38], [39], [40], [41], [42]]. This approach has rarely been used to select viral targets as the number protein targets are usually very limited. Since, no data is available on the protein targets of mpox virus and relatively much higher number proteins targets are involved, it is worthwhile to use subtractive proteomics approach to prioritise the drug target and find potential inhibitors for repurposing against mpox disease (Fig. 1 ).

Fig. 1.

Fig. 1

Computational framework of the genome-to-drug approach used in this study. The first phase involves the identification of core orthologs/proteins from the multiple genomes of mpox virus by adopting subtractive proteomics. The second phase involves docking-based high-throughput virtual screening of a non-redundant library against the identified core orthologs for the identification of potential drug candidates followed by molecular dynamics simulation of drug-target complexes.

2. Materials and methods

2.1. Global dataset of publicly available mpox virus genomes and quality assessment

All publicly available mpox virus genome assemblies were downloaded from the National Centre for Biotechnology Information (NCBI) on 3rd June 2022.

2.2. Annotation of mpox virus genomes

To avoid artificial differences resulting from different genome annotation pipelines, the genome assemblies downloaded from GenBank were reannotated with the Prokka v 1.14.6 [43] with kingdom = Virus, genus = orthopox virus and species = ‘Monkeypox virus’.

2.3. Identification and re-annotation of mpox virus core genome

Clustering orthologs in viruses can ease the screening for drug candidates [44,45]. Therefore, to obtain the core proteome of mpox virus, the proteomes from all the genomes were clustered into groups of orthologs using the OrthoFinder v2.5.4 [46,47] with default parameters. The OrthoFinder surmises homologous regions and determines the orthogroups (OG's) by using a combination of the BLAST search and the Markov Cluster Algorithm (MCA) [47]. For this study, we defined core orthologs as ORFs present in all the strains of mpox virus. Therefore, only the ORFs shared by all (100%) the strains of mpox virus were considered as core orthologs. Further, to improve the functional annotations of all core proteins, we searched the consensus sequence of these proteins against the NCBI, UniProt and KEGG databases.

2.4. Subtractive proteomics and identification of druggable core proteome of mpox virus

We devised a suitable subtractive proteomics strategy [48] to pinpoint the highly druggable core proteins of mpox virus. We did so by screening out undesirable proteins from the core proteomic dataset of mpox virus by applying a set of rationally chosen parameters in the order listed below.

2.4.1. Screening intracellular core proteins of mpox

Intracellular proteins can be easily isolated and overexpressed for experimental studies as compared to membranous proteins. Taking this into view, the core proteins of mpox virus were first sorted as globular proteins and membrane-associated proteins by assessing their sub-cellular localization using the DeepTMHMM server v2.0 [49], CCTOP [50], MEMSAT-SVM [51] and TOPCONS [52]. A consensus of all the predictors was considered to interpret their sub-cellular localization. The predicted transmembrane proteins were filtered out and the globular proteins were selected for our next downstream analysis.

2.4.2. Screening for enzymatic proteins

These globular core proteins were then sorted as either enzymes or non-enzymes by using the DEEPre [53], an enzyme function predictor that employs deep learning to predict the Enzyme Commission Number (EC number) of the input protein sequence, EMBL EBI’S Enzyme portal (https://www.ebi.ac.uk/enzymeportal/) and scientific literature.

2.4.3. Screening of non-host homologues

The absence of shared homology with human proteins is a key feature of a viable target structure to achieve selectivity and avoid toxicity. To achieve this, the pool of mpox virus enzymes was piped into NCBI-BLASTp with an e-value of 0.0001 against the entire set of human proteomes (taxid: 9606). The list of final enzymes based on this criterion was selected and screened further to assess their druggability.

2.4.4. Homology modeling, model refinement and model quality assessment

The evaluation of a modeled 3D protein structure with high precision is a core element of computational structure prediction [54]. The rapid emergence of revolutionary protein structure prediction methods accompanied by highly efficient structure evaluation tools has paved new avenues to construct high quality protein models for computational studies. In this study, the screened non-host homologous enzymes were modeled on the RoseTTAFold [55] server hosted by RosettaCommons (https://robetta.bakerlab.org) before the assessment of their druggability. The RoseTTAFold uses a three-track neural network to fold the amino-acid sequences of a protein into a 3D model. The Molprobity [56] was then used for the optimization of stereochemistry and quality evaluation of the modeled proteins. The optimized proteins were further qualitatively evaluated using 3 independent programs including the PROCHECK [57], VERIFY3D [58] and ERRAT [59].

2.4.5. Assessment of druggability

Druggability is defined as the ability of a protein to undergo high-affinity tethering with drug-like molecules [60]. This is a key feature for prioritizing and validating putative targets in pathogens [61]. Thus, the druggability of the final list of 3D-modeled non-host homologous enzymes was assessed using the CavityPlus [62], DoGSiteScorer [63] and DeepSite [64]. We set an average pKd ≥6.5, an average binding affinity of the associated pocket, for predicting druggable pockets without ligand using the CavityPlus [65]. The DoGSiteScorer identifies potential binding sites (called pockets). Proteins with pockets having druggability scores ≥0.8 were retained and the pocket with the highest druggability score was selected. The DeepSite [64] employs a machine learning algorithm based on deep convolutional neural networks (DCNNs) for predicting ligand-binding sites in proteins and provides cartesian coordinates of the centre of the identified binding pockets. A cut-off confidence score of 0.9 (90%) was used to predict the druggable pockets using the DeepSite. The consensus results of these three predictors were taken for predicting druggable enzymes.

2.5. Physicochemical characterization of druggable core proteins

Physicochemical properties of the screened druggable enzymes were further assessed as these features have a critical role in the identification of potential druggable targets in the early stages of drug development. Physicochemical properties like isoelectric point (pI), molecular weight, aliphatic index, instability index, extinction co-efficient and residues accessibility were computed with the ProtParam tools (https://www.expasy.org/resources/protparam).

2.6. Collection of FDA approved/investigational drugs

For high-throughput virtual screening, we prepared a non-redundant dataset of FDA approved/investigational drugs. The Probes & Drugs portal (P&D) is a collection of high-quality bioactive compounds including drugs and probes with their targets and experimental values collected from different sources and updated on a daily basis [66]. We downloaded the approved/investigational drugs from the Probes & Drugs portal (P&D) on 13th July 2022. This dataset included molecules approved for clinical use, clinical testing, veterinary use or neutraceutical applications.

2.7. In silico REOS and PAINS filtering

The SMILES of all the molecules were subjected to rapid elimination of swill (REOS) and pan-assay interferences (PAINS) filter using the RDKit [67], a python-based software (https://rdkit.org). Further, we implemented our in-house workflows using the Konstanz Information Miner (KNIME, v4.2.3; http://knime.org) followed by removal of redundancy.

2.8. Ligand preparation

All the non-redundant ligands were pre-processed using the OPLS_2005 force field in the LigPrep v5.3 module to obtain the accurate 3D energy minimized Lewis structures. Furthermore, EpiK v5.3 was used to generate the subsequent broad chemical, structurally diversified stereoisomers at pH 7.0 ± 2.0.

2.9. Virtual screening of FDA approved/investigational small molecules in the highly druggable binding sites

The Glide program and its virtual screening workflow were applied to identify hits against the highly druggable target proteins by incorporating three docking protocols; high throughput virtual screening (HTVS), Standard Precision (SP) mode and Extra Precision (XP) mode [68,69]. A grid box of size 12 Å was assigned around the centroid of the binding sites predicted for each target structure. The van der Waals radii (1.00 Å) of the receptor along with partial atomic charge (0.25) was kept unchanged. Default docking parameters were used, and no constraints were included. For screening curated FDA library against all the targets, we used the xglide module of the Schrodinger suite at every step of the glide docking protocol. All the docked poses of each drug were transferred from HTVS to SP to filter out the false-positive results. The SP docked poses were then piped to XP docking where the false positives were further eliminated based upon ligand-receptor shape complementarity. These XP docked poses were then ranked according to a more stringent scoring function i.e., XP glide docking score (XP GScore).

2.10. Molecular dynamics simulations: system preparation and data generation

To evaluate the binding stability of the selected common top hits at the predicted highly druggable active sites of the non-host homologous core proteins (enzymes), we employed the molecular dynamics (MD) simulations of the protein-ligand complexes in the water environment. Firstly, we prepared the conformational ensembles of all four apo proteins over the course of 400 ns MD simulation at a temperature of 300 K followed by simulation of the protein-ligand complexes over the course of 150 ns at the same temperature. The GROMACS suite v2021.1 [70] with charmm36-jul2021 force field was used to perform the MD simulation of apo proteins for the generation of ensembles and evaluation of the stability of bound ligands. The topology of proteins was generated by the in-built module of GROMACS and the topology of ligands was generated using the CHARMM General Force Field (CGenFF) web server (https://cgenff.umaryland.edu). Each system was solvated in a cubic periodic box with Tip3P charm-modified water model by keeping a 12 Å distance between the system and the water box. All the systems were neutralized by adding NaCl salt of concentration 0.15 M.

The structure of each system was minimized through a maximum of 50,000 steps using the steepest descent method of energy minimization. The equilibrations of each system were performed in two phases i.e. NVT (constant number of particles, volume and temperature) and NPT (constant number of particles, pressure and temperature) ensembles over the course of 5 ns each followed by MD production at contant temperature and pressure (1 bar and 300K). For ensemble simulation and MD production, each whole system was divided into two temperature-coupling groups of protein/protein-ligand and water-ions respectively. LINear Constraint Solver (LINCS) algorithm [71] was used to constraint all the bonds and isolated angles. Similarly, the velocity Verlet algorithm was used to integrate Newton's equations of motion. The temperature and pressure coupling on each ensemble was performed by V-rescale (modified Berendsen thermostat), Berendsen (NPT) and Parrinello-Rahman methods. However, no pressure coupling was used in NVT ensembles. For calculating the long-range (Electrostatic) interactions, the Particle Mesh Ewald (PME) method with a 1.2 nm cutoff was used. The output trajectories were recorderd every 10 ps for subsequent analysis.

2.11. Molecular dynamics simulations: stability, clustering, essential dynamics and binding free energy analysis

Following MD data productions, the trajectories of all the systems was compared by analysis of root mean square deviation (RMSD), root mean square fluctuation (RMSF), radius of gyrations (Rg), solvent accessible surface area (SASA) and amount of hydrogens bonds (HB) using the in-built module of the GROMACS. The analysis of positional changes of secondary structure elements (SSE) was accomplished by in-built module of biotite python library [72]. The dominant conformations of the trajectory data was inferenced by clustering Cα atoms RMSD of all the systems using TTClust [73]. The optimum cluster numbers was identified by elbow method. To understand the structural changes at Cα atoms level over 150 ns time of MD simulation, MODE-TASK tool were used the study the principal component analysis (PCA) using singular value decomposition (SVD) of the cartesian coordinates as well as internal coordinates [74,75]. The first two eigenvectors (principal component 1 and 2) of PCA was plotted using the ggplot2 R package [76]. To compare the collective motions of Cα atoms over the simulation time, the first essential principal component mode of PCA was represented as porcupine plots using Normal Mode Wizard (NMWiz), a ProDy python package based plugin in VMD [77,78]. The binding free energies and energies decomposition of key-interacting residues for all the complexes were calculated by AMBER's MMPBSA.py engine based gmx_MMPBSA tool [79,80] using molecular mechanics/Poisson-Boltzman surface area (MM/PBSA) model. The analysis module of the gmx_MMPBSA tool were used to infer and visualize the energies data. The average distance of bound ligands from the key-interacting residues, obtained from MM/PBSA analysis, were then calculated using the in-built gmx distance dismodule of the GROMACS.

3. Result

3.1. The meaning of the open reading frame (ORF) and core proteins in this study

A clarification is necessary on what we mean by an ORF. This is because there is a slight discrepancy in how different authors like to define the term “Open Reading Frame” or “ORF”. In the bioinformatics community it is a standard practice to identify an ORF such that the evidence of translation is not required, a definition that virological community may be less acquainted with. For the purpose of this study, we defined an ORF having start codon, stop codon and no internal stop codon as a “protein-coding gene” which translates into a functional protein that contributes to viral replication, transmission, immune evasion or overall fitness. However, we also acknowledge that translation itself may not be a criterion significant enough for an ORF to be a protein-coding gene. We acknowledge that an act of translation may have a function even if the peptide produced is non-functional as in the case of regulatory uORFs [81]. Therefore, each predicted ORF can be important even if it is annotated as a hypothetical protein.

Likewise, the term “core proteins” used here frequently has been used to describe those proteins that are highly conserved across all the strains of mpox virus. This clarification is necessary as some members of virology community may be more accustomed to the term “core protein” as the proteins associated directly with the nucleocapsid.

3.2. A compendium of 125 mpox virus genomes was developed and explored

All the genome assemblies of mpox virus available as of 3rd June 2022, were retrieved from the NCBI. Our dataset included 24 newly assembled mpox virus genomes from the latest mpox outbreak described in non-endemic countries. In total, our final dataset comprised of 125 complete genome assemblies of mpox virus. The mean genome size of mpox virus was 196318 bp (∼0.2 Mb). The highest number (275) of ORFs was identified in the strain mpox_FRA_2022_TLS67 (ON602722) whereas the strain mpox_Nig_2017_298464 (MG693724) harboured the least number of ORFs (193). The mean ORFs per genome was ∼213. Due to lack of pre-existing annotations, approximately half of the total ORFs could be successfully annotated whereas the other remaining half remained unannotated or hypothetical. The detailed metadata regarding the sample isolation information and genome statistics like genome size, GC content (%), gene count and completeness etc have been summarised in Supplementary Table S1.

3.3. Core proteome assessment identified sixty-nine core proteins from mpox virus genomes

Core proteins are major contributors to the replication and survival of a virus. Hence, developing novel therapeutics against such proteins could stop infectious pathways. Moreover, screening core proteins of an organism are advantageous as it helps to reduce the dependency on genome completeness [82]. In our core proteome assessment of 125 mpox virus strains, OrthoFinder assigned 26293 genes (99.4% of the total) to 294 ortho groups with 166 unassigned genes (0.6% of genes). Fifty percent of all genes were in orthogroups with 124 or more genes (G50 = 124) and were contained in the largest 101 ortho groups (O50 = 101). There were 69 ortho groups (core ortho groups) with all species present and 58 of these consisted entirely of single-copy genes. Two ortho groups were species-specific ortho groups with only 4 genes. The gene count in every orthogroup for each strain and strain-wise statistics is presented in Supplementary Tables S2 and S3. Every set of the 69 core proteins was then aligned to create a consensus protein sequence of each ortholog. Thus, in doing so, we created a dataset comprising the core proteome of all the 125 mpox virus genomes. Unfortunately, many predicted core proteins remained unannotated for the reasons mentioned previously. To overcome this problem, we reannotated every protein of our core proteomic dataset through a series of extensive manual curations. The detailed annotation of the core orthogroups dataset has been presented in Table 1 .

Table 1.

Table summarising the reannotation of the 69 core proteins identified in 125 genomes of mpox virus.

Core_Og Length (aa) Similar to Uniprot ID Protein name Gene % identity alignment length Mismatches E-Value Closest crystal structure (% Identity, % Coverage)
OG000 344 P42926 Serine proteinase inhibitor 2 (Serp-2) (Serpin-2) SPI-2 65.70 344 117 2.7E-149 1F0C_A (60.07, 89)
OG010 492 Q80DV6 DNA helicase; Transcript termination protein A18 A19R 97.16 493 13 0.0E+00 6JDE_A (21.76, 68)
OG011 1880 Q8QPZ7 Poxvirus B22R protein, putative transmembrane glycoprotein B22R 86.3 1829 18 0.0E+00
OG012 479 Q8QMZ9 Poly(A) polymerase catalytic subunit (EC 2.7.7.19) PAPL 99.17 479 4 0.0E+00 3ER8_C (98.75, 100)
OG013 1164 Q8V4V3 DNA-dependent RNA polymerase 132 kDa subunit, Rpo132 99.66 1164 4 0.0E+00 6RFL_B (98.97, 100)
OG014 167 P0DSW8 truncated interferon antagonist D8 (Host range protein 2) D8L 98.67 150 2 1.8E-105 5CYW_B (98, 89)
OG015 155 P0DSX4 Protein C6 C6L 92.21 154 12 3.0E-101
OG016 316 P17370 Protein C4 C4L 94.94 316 16 0.0E+00 8AG3_C (44.04, 99)
OG018 273 P23372 Protein E8 E8R 98.90 273 3 0.0E+00
OG019 665 P21093 R1L protein, Protein O1 O1L 97.30 666 17 0.0E+00
OG020 152 P33004 Protein J1 J1R 98.01 151 3 2.6E-104
OG024 206 P21039 Protein C5 C5L 94.82 193 10 7.8E-131 4HXI_A (23.81, 40)
OG025 43 P20639 Protein K3 K3L 95.24 42 2 1.6E-23 1LUZ_A (92.86, 97)
OG026 424 P20537 Phospholipase-D-like protein K4 K4L 98.11 424 8 0.0E+00 7E0M_A (24.28, 88)
OG027 214 Q6RZR2 Apoptosis regulator OPG045 OPG045 87.10 222 21 2.9E-134 2VTY_A (89.94, 78)
OG028 486 P21013 Kelch repeat protein F3 F3L 97.08 479 14 0.0E+00 6N3H_A (25.46, 47)
OG029 319 O57175 Ribonucleoside-diphosphate reductase small chain (EC 1.17.4.1) OPG048 99.06 319 3 0.0E+00 1H0N_A (80.94, 99)
OG030 64 P24360 Protein F8 F8L 96.92 65 1 2.4E-39
OG031 354 P21052 Protein F11 F11L 97.18 354 10 0.0E+00
OG032 49 P0DTM8 Protein OPG059 OPG059 95.8 48 2 4.4E-40
OG033 101 P68455 Phosphoprotein F17 TF17R 97.03 101 3 6.6E-70
OG034 153 P21605 RNA-binding protein E3 (p25) E3L 88.89 153 17 9.7E-96 1OYI_A (73.33, 29)
OG035 259 P21603 DNA-directed RNA polymerase 30 kDa polypeptide (EC 2.7.7.6) RPO30 97.68 259 6 0.0E+00 6RIC_S (97.68, 100)
OG036 166 P68447 Protein E7 E7R 93.98 166 10 2.3E-113
OG037 95 P21050 Probable FAD-linked sulfhydryl oxidase E10 (EC 1.8.3.2) E10R 98.95 95 1 2.6E-66
OG038 129 P68448 viral core protein E11 E11L 97.67 129 3 1.6E-91 6RFL_Q (97.67, 100)
OG039 269 P12923 ssDNA-binding phosphoprotein, Protein I3 I3L 98.51 269 4 0.0E+00
OG042 423 P20501 Core protease I7 I7L 99.05 423 4 0.0E+00
OG043 591 P21022 Metalloendopeptidase G1 G1L 98.64 590 8 0.0E+00
OG044 220 P68456 Late transcription elongation factor G2, Protein G2 G2R 98.64 220 3 5.5E-161
OG045 124 Q8V507 Glutaredoxin-2 EVM065 100.00 124 0 4.5E-91 2G2Q_A (97.56, 99)
OG046 340 P07611 Myristoylated protein G9, Protein F1 G9R 98.53 340 5 0.0E+00
OG047 250 P0DOT7 Protein L1; Virion membrane protein M25 L1R 99.20 250 2 0.0E+00 4U6H_E (98.91, 73)
OG048 251 P20981 core protein VP8 L4R 98.81 251 3 0.0E+00
OG049 129 P68608 DNA-dependent RNA polymerase 22 kDa subunit rpo22 Rpo22 100.00 129 0 2.7E-92 6RFL_E (100, 100)
OG051 171 P20495 Dual specificity protein phosphatase H1 H1L 98.83 171 2 1.0E-118 2RF6_A (97.66, 100)
OG052 189 P0DSY9 IMV membrane protein, Late protein H2 H2R 100.00 189 0 3.7E-145
OG053 324 P20497 Envelope protein H3 H3L 94.14 324 19 0.0E+00 5EJ0_A (93.67, 73)
OG054 314 P68697 DNA topoisomerase 1B Top1B 99.36 314 2 0.0E+00 2H7G_X (97.77, 100)
OG055 146 O57208 Late protein H7 H7R 97.95 146 3 1.7E-105 4W60_A (96.58, 100)
OG056 845 Q80DX6 mRNA capping enzyme catalytic 97 kDa subunit E1R 99.05 845 7 0.0E+00 4CKB_A (98.94, 100)
OG057 637 P04308 Early transcription factor 70 kDa small subunit VETFS 99.84 637 1 0.0E+00 7AMV_W (99.69, 100)
OG058 161 P04310 DNA-dependent RNA polymerase 18 kDa subunit rpo18 Rpo18 97.52 161 4 5.0E-117 6RFL_G (96.89, 100)
OG059 304 Q8V4Y0 Cell Surface Binding Protein, carbonic anhydrase homolog E8L 99.67 304 1 0.0E+00 4E9O_X (93.89, 86)
OG060 287 P20980 mRNA-capping enzyme 33 kDa small subunit D12L 98.96 287 3 0.0E+00 2VDW_B (98.61, 100)
OG061 551 P68440 Scaffold protein D13, Rifampicin resistance protein D13L 99.09 551 5 0.0E+00 6BED_A (99.09, 100)
OG062 150 P0DSV3 Viral late gene transcription factor 2 VLTF2 100.00 150 0 1.3E-110
OG063 644 P20643 Virion core protein 4b, p4b A3L 99.07 644 6 0.0E+00
OG064 281 P20983 39 kDa core protein A4L 95.37 281 13 5.5E-135
OG065 161 P68610 DNA-dependent RNA polymerase 19 kDa subunit Rpo19 97.56 164 1 8.1E-94 6RFL_F (97.56, 100)
OG066 891 P0DOL1 Virion core protein 4a precursor A10L 97.87 892 18 0.0E+00
OG067 318 P20988 Protein A11 A11R 99.37 318 2 0.0E+00
OG068 190 P0DOK9 25 kDa core protein A12L 98.42 190 15 5.7E-99
OG069 196 P0DOR5 Virion membrane protein A17 precursor, 23 kDa late protein A17L 97.45 196 4 1.3E-96
OG070 77 P20994 Zinc finger-like protein, Protein A19 A19L 97.40 77 2 3.5E-51 2DFY_C (42.86, 54)
OG071 426 P68709 DNA polymerase processivity factor component A20 A20R 97.18 426 12 0.0E+00 6ZXP_A (96.75, 28)
OG072 696 P24759 A-type inclusion protein A25 (ATI) A25 95.40 696 28 0.0E+00
OG073 146 Q8V4U9 Envelope protein A28 homolog (Protein A30) A30L 100.00 146 0 5.4E-106
OG074 305 Q6RZF4 DNA-directed RNA polymerase 35 kDa subunit (EC 2.7.7.6) RPO35 98.36 305 5 0.0E+00 6RFL_C (97.71, 100)
OG075 181 P68616 Protein A33 A33R 96.67 180 6 3.0E-130 4LQF_A (92.31, 50)
OG077 227 P68618 Protein A36 A36R 95.02 221 9 5.9E-134
OG078 213 P21064 Protein A41 A41L 95.24 210 8 6.5E-147 2VGA_A (97.48, 92)
OG079 346 P26670 3-beta-hydroxy-Delta(5)-steroid dehydrogenase (EC 1.1.1.145) SALF7L 98.84 346 4 0.0E+00 6JKG_A (27.02, 69)
OG080 125 Q8V4T3 Cu–Zn superoxide dismutase-like protein A46R 100.00 125 0 9.8E-85 1P1V_A (30.67, 86)
OG081 204 Q80DS7 Thymidylate kinase (EC 2.7.4.9) (dTMP kinase) TMK 98.53 204 3 3.9E-151 2V54_A (98.53, 100)
OG083 334 P21069 Protein A51 A51R 96.11 334 13 0.0E+00
OG085 176 P68443 Protein B6 B6R 88.14 177 16 6.9E-109
OG086 282 P21098 Pseudokinase B12 B12 97.17 283 7 0.0E+00 2LAV_A (32.88, 95)
OG089 190 P17365 Protein C13 (Protein B23R) C13L 95.79 190 8 3.5E-131

3.4. Subtractive proteomics successfully identifies four highly druggable, non-host homologous core proteins (enzymes)

The reannotated 69 core proteins of mpox virus were subjected to a set of multi-stage screenings to identify the druggable core proteins of mpox virus as shown in Table 2 . In the first stage, 56 globular proteins were identified based on their sub-cellular localization. These are most likely to be comprised of the proteins associated with the nucleocapsid, transcription factors and enzymes that may be critical to the virus lifecycle. In the second stage, we further screened out enzymes from the set of 56 globular core proteins. From the in silico assessment, we identified 23 core globular proteins which are most likely enzymes. Subsequently, in the third stage, the shared homology with human host proteins was assessed to limit the off-target issues. Eight enzymes namely, DNA-dependent RNA polymerase 132 kDa subunit (Rpo132), Phospholipase-D-like protein K4 (K4L), Ribonucleoside-diphosphate reductase small chain (OPG048), dual specificity protein phosphatase H1 (H1L), 3-beta-hydroxy-Delta(5)-steroid dehydrogenase (SALF7L), Cu–Zn superoxide dismutase-like protein (A46R), thymidylate kinase (TMK) and pseudokinase B12 (B12) shared 24.95%, 47.23%, 81.25%, 28.78%, 41.24%, 30%, 41.55% and 32.88% sequence identity with human proteins DNA-directed RNA polymerase II 140 kDa polypeptide (3J0K_B), 5′-3′ exonuclease PLD3 (Q8IV08), Human ribonucleotide reductase subunit R2 (D6W4Z6), Dual specificity protein phosphatase 22 (6LVQ_A), 3 beta-hydroxysteroid dehydrogenase type 7 (Q9H2F3), Superoxide dismutase [Cu–Zn] (3GTV_A), Thymidylate Kinase (1E98_A) and Vaccinia-related kinase 1 (2LAV_A) respectively. These proteins were subsequently discarded as they exceeded the permitted threshold (e-value = 0.0001). The 15 remaining non-host homologous enzymes were processed for druggability studies. 12 out of these 15 enzymes shared excellent coverages and sequence similarities with the solved crystal structures of proteins from the PDB database. On the contrary, DNA polymerase processivity factor component A20 (A20R) shared 96.75% similarity with 6ZXP_A at very low coverage (28%) whereas crystal structures even remotely similar to core cysteine protease (I7L) and metalloendopeptidase G1 (G1L) were not available (Table 1). In the absence of experimentally derived high-resolution structures, modeled proteins present suitable alternatives to examine structural perturbations in proteins. Therefore, to overcome this problem we applied the hybrid, a top performer of CASP-5 and highly accurate deep learning-based modeling method the RoseTTAFold to model the 3D structures of all the 15 non-host homologous enzymes. The RoseTTAFold [55] employs a 3-track neural network to fold a protein sequence into a model. The accuracy and validation assessment of all the predicted structures were assessed through the MolProbity web server [56]. Out of these 15, the 7 enzymes successfully modeled by the RoseTTAFold were subjected to druggability assessments for the time being. Of these 7 core non-host homologous enzymes, only 4 enzymes namely the I7L, DNA topoisomerase 1B (Top1B), early transcription factor 70 kDa small subunit (VETFS) and A20R complied with the thresholds of druggability prediction tools. The druggability scores (DScore), simple score (SScore), binding affinity (pKd), volume and surface area associated with the predicted highly druggable binding site of the four proteins have been summarised in Supplementary Table S4. The model quality statistics of these four druggable proteins are presented in Table 3 . Unfortunately, this study was impelled to abandon the druggability assessment of 8 out of these 15 enzymes (Group 5, Table 2). These 8 proteins will be assessed for their druggability in our future endeavors. Hence, by combining genomics and an extensive subtractive proteomic screening on the core proteomic dataset of mpox virus we finally identified 4 highly druggable, globular, non-host homologous core proteins (Table 2). The modeled proteins and their predicted druggable sites have been illustrated in Fig. 2 .

Table 2.

Table summarising the subtractive proteomics strategy and its outcomes. The sixty-nine core proteins were screened through four stages to pinpoint therapeutic targets with highly desirable features. Four proteins namely; I7L, Top1B, VETFS, and A20R were identified as highly druggable, globular, non-host homologous, enzymatic proteins and were subjected to virtual screening in this study. Eight proteins namely, A19R, PAPL, Rpo30, E10R, G1L, Rpo22, E1R and Rpo35 passed the first three stages of screening and remain under investigation for druggability assessment. Proteins that fit under a particular group of are shaded in blue

3.4.

3.4.

Table 3.

Physicochemical properties and model quality statistics of the four proteins of mpox virus identified through subtractive proteomics.

Features Core protease I7 DNA topoisomerase 1B Early transcription factor 70 kDa small subunit DNA polymerase processivity factor
Gene Name I7L TopIB VETFS A20R
Enzyme Commision (EC) 3.2.22.- 5.99.1.2 3.6.4.13 3.-
KEGG Orthology (KO) Identifier K21082
Closest Crystal Structure 2H7G (97.77%, 100%)) 7AMV (99.69%, 100%) 6ZXP (96.75%, 28%)
Length (Amino Acid) 423 aa 314 aa 637 aa 426 aa
Mol. Weight (Dalton) 49023.57 36665.56 73844.91 49147.14
pI 7.85 9.50 6.93 5.62
Instability Index 33.81 40.22 34.45 36.35
Aliphatic Index 83.14 88.03 97.85 92.77
GRAVY −0.22 −0.32 −0.17 −0.26
Predicted IDDT Score 0.64 0.87 0.70 0.80
Overall Quality Score 95.42 95.75 95.66 91.87
MolProbity Score 1.44 (96th percentile) 0.90 (100th percentile) 1.40 (97th percentile) 1.37 (98th percentile)
Clash Score 3.49 (97th percentile) 1.53 (99th percentile) 2.78 (98th percentile) 2.88 (98th percentile)
Poor Rotamers (%) 0 0 0 0
RamaPlot Most Favoured (%) 95.72 (403 aa) 100.00 (312 aa) 95.12 (604 aa) 95.80 (406 aa)
RamaPlot Allowed (%) 3.8 (16 aa) 0 3.48 (22 aa) 4 (17 aa)
RamaPlot Disallowed (%) 0.48 (2 aa) 0 1.42 (9 aa) 0.24 (1 aa)

Fig. 2.

Fig. 2

Homology models of four highly druggable core proteins of mpox virus. (A) Model of DNA polymerase processivity factor component A20 (A20R). (B) Model of Core cysteine proteinase (I7L). (C) Model of DNA topoisomerases type 1B (Top1B). (D) Model of Early transcription factor 70 kDa small subunit (VETFS). The figures of the models are represented as spectrum coloured cartoons. The predicted active site residues are styled as lacorice (stick) with purple blue colour. The active site residue information has been provided in Supplementary Table S4. The figures have been illustrated with Pymol v2.5.3.

3.5. The druggable proteins are soluble and highly stable over a wide range of temperatures

The physicochemical properties of the 4 highly druggable proteins were predicted (Table 3). Proteins with instability index above 40 are considered unstable whereas proteins with an instability index below 40 are considered stable. The instability value of Top1B was slightly above 40. Therefore, this enzyme could be slightly unstable. The instability values of the remaining 3 proteins were below 40 and reflect their highly stable nature. This prediction was also supported by the high aliphatic index of these proteins which suggests their stability over a wide range of temperatures. Similarly, the hydrophilic features and solubility of a protein are reflected in its grand averages of hydropathy (GRAVY) score. The GRAVY scores of all 4 enzymes were below 0 which reflects upon their globular and soluble nature.

3.6. A curated library of 5893 FDA approved/investigational drugs was prepared

For high-throughput virtual screening (HTVS), we collected ∼19 thousand FDA-approved/investigational drugs of approved, investigational and experimental categories from the Probes and Drugs Portal, a highly curated chemical space portal comprising data from ∼50 different sources [66]. Discontinued FDA-approved/investigational drugs were excluded from our selection. For filtering problematic functional groups and false positives, we implemented the REOS and PAINS filters on the collected library. The PAINS and REOS filter are both based on the RDKit substructure counter and compare the substructures present in the input database with a list of problematic functional groups. After filtering, ∼32% (5893) of drugs remained in the dataset and were subjected to the ligand preparation stage. Finally, a set of 12,048 stereoisomers corresponding to 5893 drugs were obtained following library preparation with the LigPrep module of the Schrodinger suite.

3.7. Burixafor, batefenterol and eluxadoline are the top hits common to the four highly druggable enzymes of mpox virus

We performed the multi-step (HTVS, SP and XP) docking of 12,048 stereoisomers corresponding to 5893 drugs against all the identified target proteins by using the Schrodinger suite's Glide module. Numerous binding modes of all the drugs across the 4 targets were predicted. The docking scores varied between −13.36 kcal/mol to −0.50 kcal/mol for core protease I7L, −10.74 kcal/mol to 2.19 kcal/mol for Top1B, −11.55 kcal/mol to 0.75 kcal/mol for VETFS and −12.30 kcal/mol to 0.35 kcal/mol for A20R as shown in Fig. 3 . Interestingly, the drug batefenterol (BAT) was among the top 30 hits for all the proteins namely; A20R, I7L, Top1B and VETFS whereas burixafor (BUR) and eluxadoline (ELU) were amongst the top 30 hits for I7L, Top1B and VETFS. The binding affinities of BAT with A20R, I7L, VETFS and Top1B ranged between −12.07 kcal/mol to −8.89 kcal/mol. The binding affinities of BUR with I7L, VETFS and Top1B ranged between −11.84 kcal/mol to −8.29 kcal/mol. Similarly, the binding affinities of ELU with I7L, VETFS and Top1B ranged between −12.73 kcal/mol to −8.14 kcal/mol respectively. Likewise, tobramycin (PD001728), dibekacin (PD001070) and GLPG-0187 (PD058153) were identified as hits common only to I7L and VETFS and were among their top 30 hits. The XP GScore, XP HBond energy and ligand efficiency of the top 30 hits for each target is presented in Supplementary Tables S5–S8. The primary indications of the top five hits against all the targets is summarised in Table 4 .

Fig. 3.

Fig. 3

Distribution of glide XP docking score (kcal/mol) against DNA polymerase processivity factor component A20 (A20R), Core cysteine proteinase (I7L), DNA topoisomerases type 1B (Top1B) and Early transcription factor 70 kDa small subunit (VETFS).

Table 4.

Mode of actions, application and 2D structure of shortlisted top five docked drugs with high binding affinity for each target. The mode of actions, applications was collected from DrugBank, Inxight Drugs portal and PubChem database.

Target Drug PDID Drug Status Primary Indications (1. Mode of Action; 2. Treatment) 2D Structure
I7L Eluxadoline PD008978 Approved
  • 1.

    An agonist of mixed mu-opioid receptor.

Image 1
  • 2.

    Used for the treatment of irritable bowel syndrome with diarrhea.

I7L, VETFS Burixafor PD058763 Investigational
  • 1.Inhibitor of CXC chemokine receptor 4 (CXCR4)

Image 2
  • 2.

    Used in trials studying treatment of Hodgkin's disease, Non-hodgkin's Lymphoma and Multiple Myeloma.

I7L Cefotiam PD010157 Approved
  • 1.Inhibits the bacterial cell wall biosynthesis.

Image 3
  • 2. Used for the treating a myriad of bacterial infections.

I7L Tobramycin PD001728 Approved
  • 1.Inhibits the synthesis of protein by binding to ribosome 30S subunit in bacterial cells.

Image 4
  • 2.

    Used for the treating a myriad of bacterial infections.

I7L Danegaptide PD058923 Investigational
  • 1.A selective 2nd generation gap junction modifier in adjacent cardiomyocytes.

Image 5
  • 2.

    Used in studying treatment of chronic atrial fibrillation (AF) and postoperative AF in large animal models.

Top1B Carbaphosphonate PD059915 Investigational
  • 1.3-dehydroquinate synthase inhibitor (Staphylococcus aureus)

Image 6
Top1B α-d-galacturonic acid PD006851 Investigational
  • 1.

    Backbone of pectin, cellular binders in the peel of many different fruits and vegetables.

Image 7
  • 2.

    Used as: antidiarrheal drug, food emulsifier, food stabilizer, food thickening agent and food gelling agent.

Top1B Adenosine phosphonoacetic acid PD059913 Investigational
  • 1.

    ndp kinase (human) inhibitor.

Image 8
Top1B Guanosine-2′,3′-O-methylidenephosphonate PD006419 Investigational
  • 1.

    catalyze the phosphorolytic breakdown of the N-glycosidic bond in the beta-(deoxy) ribonucleoside molecules

Image 9
Top1B Deferitazole PD058219 Investigational
  • 1.

    An iron cheater

Image 10
  • 2.

    Used in trials studying the treatment and basic science of Beta-thalassemia.

VETFS DB02785 PD059991 Investigational
  • 1.

    Gag-Pol polyprotein inhibitor

Image 11
  • 2.

    Used as an antiviral (antiHIV-1)

VETFS Radezolid PD012727 Investigational
  • 1.

    A 2nd generation's oxazolidinone antibiotic, inhibits the RRBP1.

Image 12
  • 2.

    Used in trials for studying the treatment of Abscess, Infectious Skin Diseases.

VETFS Rapastinel PD070040 Investigational
  • 1.

    NMDA receptor modulator with glycine-site partial agonist properties.

Image 13
  • 2.

    Used in trials studying the treatment of Major and Obsessive-Compulsive Disorder (OCD).

I7L, A20R Batefenterol PD058591 Investigational
  • 1.

    A bifunctional muscarinic (M2 and M3 receptors) antagonist and β2-agonist

Image 14
  • 2.

    Used in trials for studying the treatment of Chronic Obstructive Pulmonary Disorder (COPD).

A20R Nebivolol PD009358 Approved
  • 1.

    β-1 adrenergic receptor antagonist (beta blockers).

Image 15
  • 2.

    Used to treat high blood pressure either alone or in combination with other medications.

A20R Pimozide PD001879 Approved
  • 1.

    Dopamine type 2 receptors blocker

Image 16
  • 2.

    Used to control motor or verbal tics caused by Tourette's disorder.

A20R BAZ2-ICR PD046767 Investigational
  • 1.

    Epithelial sodium channel blocker

Image 17
  • 2.

    Used in trials for studying cystic fibrosis and chronic bronchitis.

A20R PF-610355 PD058896 Investigational
  • 1.

    A novel Ultra-Long-Acting β2-Adrenoreceptor agonist.

Image 18
  • 2.

    Used for studying the treatment of Asthma and Chronic Obstructive Pulmonary Disease.

3.8. RMSD, RMSF, Rg and SASA calculations suggests stability of the ligand-bound complexes

Two independent sets of MD simulations were undertaken in this study by using the GROMACS package. The first set involved the model refinement of four proteins A20R, I7L, Top1B and VETFS to reduce stearic clashes among their residues. This was achieved by simulating every protein for 400 ns. The trajectories were subjected to clustering using the TTClust. The centroid of the cluster with the least mean rmsd between the frames were selected for protein-ligand complex MD simulation. The summary of cluster analysis of the trajectories after 400 ns of simulation has been presented in Supplementary Fig. S1. Thereafter, to access the conformational stabilities of the 12 protein-ligand complexes during the 150 ns of MD production, various quality control parameters like RMSD, Rg, SASA, RMSF, H-Bonds, secondary structure, essential dynamics, MM/GBSA binding free energies and distance of ligand from active site were examined.

RMSD characterises a protein-ligand complex's conformational stability in its dynamic state during simulation. A low difference in RMSD indicates low and consistent fluctuation between the ensembles which suggests a protein-ligand complex is stable. To ensure the sampling method's reliability, the RMSDs of the proteins' alpha carbon atoms (C-α) were analysed by plotting them against the time scale of 150 ns from the starting structure (Fig. 4 ). None of the A20R ligand associated complexes could achieve equilibrium after 150 ns of simulation (Fig. 4A). The RMSD plot of the I7L-BAT complex (Fig. 4B) depicted an increasing trend for the first 45 ns. However, the ensembles that followed gradually traced a constant trajectory for the next 105 ns i.e., from 45 ns to 150 ns with consistent minor fluctuations within a permissible window of 3 Å to 4.5 Å and an average RMSD of 0.63 ± 0.26 Å. The trajectory of the I7L-BUR complex (Fig. 4B) followed a stable trend for the first 55 ns while fluctuating between a small window of 2 Å to 3 Å. The complex then encountered a minor fluctuation before finally settling into a relatively flat trajectory with an average RMSD of 0.9 ± 0.3 Å for the next 95 ns i.e., from 55 ns to 150 ns within a permissible window of 2.5 Å to 4.5 Å. Similarly, the trajectory of I7L-ELU complex (Fig. 4B) projected an increasing trend for the first 35 ns. Thereafter, the ensembles adopted a relatively flat trajectory with an average RMSD of 0.72 ± 0.28 Å for the last 115 ns i.e., from 35 ns to 150 ns within a small window of 3 Å to 5 Å. These results indicate the protein I7L has successfully achieved conformational stability with the three ligands. The ensembles of Top1B-BAT complex (Fig. 4C) traced out a relatively flat trajectory with consistent minor fluctuations for the first 85 ns within a confined permissible space between 2 Å to 4 Å. Minor blips within very narrow time frames of (25–30) ns and (45–48) ns were also encountered. This was followed by high fluctuations for the next 30 ns i.e., from (85–115) ns following which the complex again reverted back to its original 2 Å to 4 Å window for the last 35 ns. All the stable ensembles of Top1B-BAT complex that fluctuated within the 2 Å to 4 Å permissible window had an average RMSD of 1.16 ± 0.85 Å. Strangely, this pattern repeated itself again in Top1B-BUR and Top1B-ELU complexes respectively (Fig. 4C). The Top1B-BUR complex retained a consistent trajectory for the first 40 ns within an allowable window of 2 Å to 4 Å. This complex then navigated through a similar violent RMSD fluctuations for the next 55 ns i.e., from (40–95) ns before finally returning back to its initial 2 Å to 4 Å window for the last 55 ns with consistent minor fluctuations. All the stable ensembles of Top1B-BUR complex that fluctuated within the 2 Å to 4 Å permissible window had an average RMSD of 0.88 ± 0.59 Å. The ensembles of Top1B-ELU complex followed an increasing trend for the first 20 ns. Thereafter, the ensembles retained a stable trajectory throughout the entire 150 ns between a 2 Å to 4 Å window with an average RMSD of 1.17 ± 0.77 Å with the exception of two small time frames between 75 ns to 85 ns and 115 ns–120 ns respectively where major fluctuations in RMSD were clearly distinguishable (Fig. 4C). The ensembles of VETFS-BAT complex pursued an upward trajectory for the first 10 ns (Fig. 4D). Thereafter, the ensembles retained a stable trajectory for the next 140 ns i.e., from 10 ns to 150 ns with constant but minor fluctuations within an allowable window between 3 Å to 5 Å with an average RMSD of 0.57 ± 0.36 Å. The ensembles of VETFS-BUR complex traced an ascending step ladder trajectory for the first 70 ns before gradually settling into a stable trajectory for the remaining 80 ns i.e., from 70 ns to 150 ns (Fig. 4D). The stable ensembles during this time frame constantly fluctuated within a narrow and acceptable window between 4 Å to 6 Å with an average RMSD of 0.88 ± 0.39 Å. A rising trend was observed in the trajectory of VETFS-ELU complex for the first 40 ns (Fig. 4D). The trajectory thereafter declined which was followed by a steady rise in the next 45 ns before finally achieving stability for the last 55 ns i.e., from 85 ns to 150 ns. The stable ensembles during this time frame fluctuated within a 6 Å to 8 Å window with an average RMSD of 0.88 ± 0.44 Å. It is also interesting to note that the VETFS-BAT system achieved equilibrium within a very short duration in comparison to its BUR and ELU bound counterparts. This suggests, in comparison to VETFS-BAT complex, VETFS-BUR and VETFS-ELU complexes had to navigate through several conformational transitions to accommodate their respective ligands before finally achieving stable conformations. Overall, these results indicate that the enzyme VETFS has successfully acquired conformational stability with all the three ligands. Taken together, only nine complexes that showed conformational stability were considered for further examination. The density distribution of RMSDs of these nine stable complexes have also been provided in Supplementary Fig. S2.

Fig. 4.

Fig. 4

Dynamics of RMSD of the mpox virus ligand bound protein complexes during 150 ns of simulation. (A) RMSD of A20R ligand bound complexes. (B) RMSD of I7L ligand bound complexes. (C) RMSD of Top1B ligand bound complexes. (D) RMSD of VETFS ligand bound complexes. The colours red, blue and green represent the colours of the three ligands batefenterol, burixafor and eluxadoline respectively in complex with the respective proteins. The illustrations were created with Tableau.

Subsequently, the radius of gyration (Rg) of the 9 stable protein-ligand complexes were also assessed (Fig. 5 ). Rg examines a protein's compactness where a constant Rg suggests that the system has achieved a relatively constant shape and size during the entire simulation. The average radius of gyration of I7L-BAT, I7L-BUR and I7L-ELU complexes were 23.16 ± 0.19 Å, 22.74 ± 0.14 Å and 23.16 ± 0.24 Å respectively. These complexes successfully retained their compactness without undergoing any major structural re-arrangements during the entire 150 ns simulation as demonstrated by the consistency of their time dependent Rg plot (Fig. 5A). Similarly, the Rg of the Top1B-BUR and Top1B-ELU complexes also remained fairly consistent during the entire simulation with average Rg's of 25.51 ± 0.51 Å and 25.34 ± 0.52 Å respectively (Fig. 5B). The average Rg of Top1B-BAT complex was 24.71 ± 0.69 Å during the entire simulation with some minor fluctuations. The Rg of Top1B-BAT complex remained consistent for the first 75 ns following which it experienced a sharp decline and a subsequent upturn in the next 55 ns i.e., between 75 ns and 130 ns before retracing its original trajectory in the last 20 ns (Fig. 5B). Overall, the consistent Rg of the three Top1B-ligand bound complexes suggest they have retained their structural integrity and are relatively compact after 150 ns of simulation. The VETFS-BAT complex also remained compact with an average Rg of 30.05 ± 0.27 Å (Fig. 5C). This was evident from its time-evolution Rg plot which remained fairly consistent during the entire simulation. The Rg of VETFS-BUR and VETFS-ELU complexes remained steady for the first 70 ns (Fig. 5C). Thereafter, the Rg of VETFS-BUR declined linearly for the next 30 ns before retaining its steadiness for the last 50 ns. On the contrary, the Rg of VETFS-ELU complex witnessed a linear increment for the next 10 ns before becoming relatively constant for the last 70 ns. This suggests that the presence of BUR in the active site of VETFS has enhanced its compactness. It also suggests that BUR is probably better adapted to VETFS in comparison to ELU. The Rg of VETFS-BUR and VETFS-ELU complex was 29.59 ± 0.32 Å and 30.64 ± 0.39 Å respectively. Taken together trends of Rg of all the nine complexes are in congruence with their corresponding RMSD trends that reflect the relative stabilities and compactness of the 9 ligand bound enzyme complexes.

Fig. 5.

Fig. 5

Dynamics of radius of gyration (Rg) and solvent accessible surface area (SASA) of the mpox virus ligand bound protein complexes during 150 ns of simulation. (A) Rg of I7L ligand bound complexes. (B) Rg of Top1B ligand bound complexes. (C) Rg of VETFS ligand bound complexes. (D) SASA of I7L ligand bound complexes. (E) SASA of Top1B ligand bound complexes. (F) SASA of VETFS ligand bound complexes. The colours red, blue and green represent the colours of the three ligands batefenterol, burixafor and eluxadoline respectively in complex with the three proteins. The illustrations were created with Tableau.

SASA is another frequently studied quality control parameter that is examined to scrutinize a protein-ligand complex's conformational stability. The solvent environment around the protein plays a crucial role in retaining a protein's folding and governs the protein–ligand interaction processes, orientation, and stability. The time evolution SASA plots clearly indicate that all the nine complexes are relatively well equilibrated during the entire 150 ns simulation (Fig. 5D, E and 5F). The SASA values varied within a narrow range between 233.4 ± 4.43 nm2 - 242.11 ± 6.67 nm2 for I7L-ligand complexes, 191.76 ± 3.32 nm2 - 192.50 ± 3.04 nm2 for Top1B-ligand complexes and 339.96 ± 4.35 nm2–348.41 ± 5.27 nm2 for VETFS-ligand bound complexes respectively. These evidences are in agreement with their corresponding RMSDs and suggest that all the proteins have successfully attained conformational stabilities with their respective ligands without undergoing any major alterations in their available surface area.

Root Mean Square Fluctuation (RMSF) was computed to examine the positional fluctuation of each amino acid residue around its mean position. The Cα atoms of all the I7L complexes shared very similar RMSF trends (Fig. 6 A). This case was also similar for the Cα atoms of Top1B (Fig. 6B) and VETFS (Fig. 6C) ligand bound complexes. The Cα atoms associated with the loops experienced high fluctuations whereas the Cα atoms associated with α-helix and β-sheets were comparatively rigid in all the complexes. High fluctuations were were also associated with the C terminal residues in all the nine complexes. Amongst the I7L complexes, the highest average atomic fluctuation of 1.83 ± 0.97 nm was recorded in the residues of the ELU bound complex. Similarly, amongst the Top1B and VETFS complexes, highest average atomic fluctuations of 3.19 ± 1.6 nm and 2.71 ± 1.27 nm was recorded in the residues of BUR and ELU bound complexes respectively. From Fig. 6B it is quite evident that high atomic fluctuations of all the Top1B complexes are mostly restricted to the residues from 1 to 70 which encompass the protein's N-terminal domain. The fluctuations in the residues that encompass the ligand bound C-terminal DNA binding domain (76–314) are relative restricted which hints at the conformational rigidity of this domain. This suggests that the C-terminal DNA binding domain of Top1B has been successfully stabilized by the three ligands and the wild fluctuations in Top1B's RMSD trajectories are most probably related to the flexibility of its N-terminal domain.

Fig. 6.

Fig. 6

Dynamics of RMSF and intermolecular hydrogen bonds of the mpox virus ligand bound protein complexes during 150 ns of simulation. (A) RMSF of I7L ligand bound complexes. (B) RMSF of Top1B ligand bound complexes. (C) RMSF of VETFS ligand bound complexes. (D) H-Bonds of I7L ligand bound complexes. (E) H-Bonds of Top1B ligand bound complexes. (F) H-Bonds of VETFS ligand bound complexes. The three colours red, blue and green represent the colours of the three ligands batefenterol, burixafor and eluxadoline respectively in complex with the three proteins. The illustrations were created with Tableau.

The molecular dialogues between a ligand and the flexible active site amino acid residues are largely driven by H-bond interactions. We further examined the time evolution plots of H-bonds involved in the molecular interaction of the three ligands BAT, BUR and ELU with each of the three proteins namely, I7L, Top1B and VETFS. The nine protein-ligand complexes depicted differential intermolecular H-bonding patterns during the 150 ns of MD run (Fig. 6D, E and 6F). The equilibrated complexes of protein I7L formed an average of 2.31 ± 1.36, 2.1 ± 1.19 and 1.5 ± 1.03 H-bonds per frame with the ligands BAT, BUR and ELU respectively. The equilibrated complexes of Top1B formed an average of 1.28 ± 1.03, 1.79 ± 1.17 and 1.55 ± 1.06 H-bonds per frame with the ligands BAT, BUR and ELU respectively. Similarly, the fully equilibrated complexes of VETFS formed an average of 1.14 ± 0.86, 1.705 ± 1.22 and 1.59 ± 0.65 H-bonds per frame with the ligands BAT, BUR and ELU respectively. H-bond interactions govern the correct orientation and stability of a ligand in a protein's active site which subsequently drives the protein's overall compactness and structural conformity. Therefore, the compactness and structural rigidity of I7L, Top1B and VETFS as inferred from their stable time evolution RMSD, Rg and SASA plots are most probably driven by the these H-bond interactions.

3.9. The stabilities of the protein-ligand complexes are largely driven by switching between secondary structural elements

Changes in secondary structure as a function of time were also mapped for every complex using the Biolite, a Python-based molecular biology suite. Helices and β-sheets are stable amongst the secondary structure elements whereas coils, loops and turns are highly flexible. Constant switchings between coils, bends and turns were clearly visible in all the complexes during their simulations. These switchings probably contribute in parts to the molecular motions of all the complexes. The integrity of all the major α-helices and β-sheets remained intact in all the complexes. These α-helices and β-sheets are most likely responsible for the global conformational rigidity of the proteins. Analysis of the time evolution map of the secondary structure elements resulted in some interesting observations. The flexible turn between the residues 40 and 50 in the I7L-ELU complex gradually transformed into a more rigid α-helix after 10 ns and remained constant for the remaining duration of the simulation (Supplementary Fig. S3). This correlates well with its RMSD plot where the stability of the complex was achieved immediately after 10 ns. This observation suggests that ELU mediated stabilization of the enzyme I7L was most likely driven by the metamorphosis of a turn between the residues 40 and 50 into an α-helix. The turn at the residues between 70 and 75 of Top1B-BUR complex was replaced by a more flexible coil between 50 ns and 95 ns which probably explains the corresponding sharp RMSD and RMSF fluctuations (Supplementary Fig. S4). The residues between 420 and 430 in VETFS-BUR complex continued to switch between a turn and α-helix for the first 85 ns before smoothly transitioning into an α-helical structure for the remaineder of the simulation (Supplementary Fig. S5). This correlates well with the RMSD trajectory of this complex and suggests that ELU driven stabilization of VETFS is regulated by the formation of an α-helix. Overall, these results suggest that the transition of unstable protein-ligand complexes into their equilibrated states and vice versa are largely driven by switching between secondary structural elements.

3.10. The equilibrated ensembles of the protein-ligand complexes are grouped into many clusters

The ensembles of I7L, Top1B and VETFS ligand bound complexes generated after 150 ns of the simulation were clustered with the TTClust to identify the clusters with stable ensembles and the corresponding centroid structures that represent these clusters. The TTClust analyses the trajectories to generate graphical imagery like distribution within the clusters, the relative distance between clusters and hierarchical cluster dendrograms. Fig. 7 and Supplementary Fig. S6 depict the results of the cluster analysis of I7L complexes during the 150 ns of MD simulation. The equilibrated ensembles of I7L-BAT complex was subgrouped into 3 clusters (C2, C3, C4), whereas those of I7L-BUR and I7L-ELU complex were further subgrouped into 2 clusters (C2, C3) each. The equilibrated ensembles of Top1B-BAT, Top1B-BUR and Top1B-ELU complexes were also subgrouped into 2 clusters (C1, C2) each (Fig. 8 and Supplementary Fig. S6). Similarly, the equilibrated ensembles of VETFS-BAT, VETFS-BUR and VETFS-ELU complexes were subgrouped into 3 (C1, C2, C3), 2 (C3, C4) and 1 (C3) clusters respectively (Fig. 9 and Supplementary Fig. S6). The distribution of these equilibrated ensembles into different clusters suggests that they probably sample small conformational subspaces with minor structural differences. The protein-ligand interactions experienced by the centroid frames of these stable sub-clusters are summarised in Table 5 .

Fig. 7.

Fig. 7

Cluster analysis of the I7L ligand bound complexes. (A) I7L-BAT. (B) I7L-BUR. (C) I7L-ELU. The linear plot depicting the distribution of conformations (frames) over 150 ns of simulatuion. Close view of initial frame, centroid frame of the most populated cluster and 3D alignment of initial frame with centroid frame are presented below each linear plot from left to right. The total number of clusters and frames per cluster has been detailed in Supplementary Fig. S6. The colours in the time evolution linear plots represent the cluster to which a frame belongs at a given point of time. The cartoons of initial (far left) and centroid frames are illustrated as spectrum. Close view of aligned frames (far right) where the initial and centroid frames are illustrated as limegreen and violetpurple coloured cartoons respectively. The bound ligands with initial and centroid frames are illustrated in red and orange colours respectively.

Fig. 8.

Fig. 8

Cluster analysis of the Top1B ligand bound complexes. (A) Top1B-BAT. (B) Top1B-BUR. (C) Top1B-ELU. The linear plot depicting the distribution of conformations (frames) over 150 ns of simulatuion. Close view of initial frame, centroid frame of the most populated cluster and 3D alignment of initial frame with centroid frame are presented below each linear plot from left to right. The total number of clusters and frames per cluster has been detailed in Supplementary Fig. S6. The colours in the time evolution linear plots represent the cluster to which a frame belongs at a given point of time. The representation of colouring scheme are same as described in Fig. 7.

Fig. 9.

Fig. 9

Cluster analysis of the VETFS ligand bound complexes. (A) VETFS-BAT. (B) VETFS-BUR. (C) VETFS-ELU. The linear plot depicting the distribution of conformations (frames) over 150 ns of simulatuion. Close view of initial frame, centroid frame of the most populated cluster and 3D alignment of initial frame with centroid frame are presented below each linear plot from left to right. The total number of clusters and frames per cluster has been detailed in Supplementary Fig. S6. The colours in the time evolution linear plots represent the cluster to which a frame belongs at a given point of time. The representation of colouring scheme are same as described in Fig. 7.

Table 5.

Protein-ligand interactions of the representative centroid complex of each cluster that falls within the stable region (RMSD) of the trajectories in every complex.

Target Ligand Cluster Hydrogen Bond
Hydrophobic Interaction
Halogen Bonds
Salt Bridge
Residue Sidechain Distance (H-A, Å) Distance (D-A, Å) Residue Distance (Å) Residue Sidechain Distance (Å) Residue Distance (Å)
I7L
Batefenterol
C2
GLU108 FALSE 2.09 2.97 LEU109 3.70
ASN295 TRUE 3.51 3.88 TYR238 3.63
LYS243 3.78




ILE298
3.73





C3
SER078 FALSE 2.52 3.46 LEU077 3.87
SER078 TRUE 2.41 3.21 ARG099 3.73 GLU108 TRUE 2.87
SER078 TRUE 2.96 3.81 PRO264 3.66
ARG099 TRUE 3.40 4.10 PHE267 3.64
TYR100 TRUE 2.47 3.35 PHE275 3.70
GLU108 FALSE 1.79 2.76 PHE278 3.83
ILE298 3.77




ILE316
3.62





C4
HIS076 FALSE 2.89 3.41 ARG099 3.81 GLU108 TRUE 2.62
SER078 FALSE 3.25 3.69 LEU109 3.53
SER078 TRUE 2.37 3.21 ASN262 3.97
SER078 TRUE 3.13 4.06 PHE267 3.89
ARG099 TRUE 3.02 3.44 PHE275 3.89
ARG099 TRUE 3.08 4.06 ILE316 3.76
ASN262
TRUE
3.37
4.04







Burixafor
C2
THR111 TRUE 2.74 3.55 VAL047 3.55 ASP297 4.61
SER200 FALSE 3.03 4.01 PRO064 3.91
LYS206 TRUE 2.65 3.38
LYS243 TRUE 3.23 3.76
ASN293 FALSE 2.13 2.96
THR294 TRUE 3.03 3.94
ASN295
TRUE
1.81
2.77







C3
THR111 TRUE 2.86 3.59 TYR046 3.49
THR111 TRUE 2.99 3.59 LYS050 3.55
LYS243 TRUE 3.63 4.08 TYR051 3.58
VAL060 3.97




PRO064
3.84





Eluxadoline
C2
THR111 TRUE 1.99 2.89 LYS243 5.00
SER240
FALSE
2.20
3.18







C3
SER240 FALSE 2.04 3.02 TYR046 3.81 LYS243 4.37
HIS241 FALSE 3.16 3.48 ILE113 3.64
TYR238 3.76
LEU239 3.70




LEU239
3.83





Top1B
Batefenterol
C1
ARG223 TRUE 3.42 3.87 LEU122 3.94
ARG223 FALSE 3.06 3.85 LEU122 3.67
HIS265 TRUE 3.02 3.48 PHE131 3.40
TYR274 FALSE 1.78 2.76 PHE131 3.90
LEU146 3.90
GLU205 3.66
VAL208 3.78
VAL208 3.79
ARG223 3.32




VAL227
3.96





C2
MET126 FALSE 2.30 3.26 LEU122 3.84
ILE129 FALSE 2.03 2.90 ARG130 3.88
ARG130 TRUE 3.17 3.76 PHE131 3.58
ARG130 TRUE 3.18 3.76 VAL208 3.47
PHE131 FALSE 2.46 3.39 ILE212 3.63
GLY132 FALSE 2.33 3.09 LEU222 3.53
TYR136 TRUE 3.07 4.00
TYR136 TRUE 3.40 4.00
GLU261
FALSE
2.75
3.24







Burixafor
C1
PHE128 FALSE 3.18 3.85 PHE128 3.57
ARG130 TRUE 2.88 3.44 PHE128 3.85
HIS172 TRUE 2.31 3.06 ILE129 3.79
PHE164 3.29
HIS172 3.97
PHE174 3.99




TYR233
3.74





C2
MET126 FALSE 2.02 2.97 ILE129 3.92
ARG130 FALSE 2.75 3.39 PHE164 3.62
GLU173 FALSE 2.02 2.94 VAL175 3.60
ARG223 TRUE 1.80 2.75
ARG223
TRUE
1.89
2.80







Eluxadoline
C1
ARG130 FALSE 3.06 3.56 HIS172 3.87
PHE131 FALSE 2.02 3.02
ARG223 TRUE 3.17 4.03
THR230 TRUE 1.86 2.84
VAL262
FALSE
1.86
2.73







C2
ARG130 FALSE 2.81 3.30
PHE131 FALSE 1.71 2.69 ARG223 4.80
ARG223 TRUE 2.22 3.01
VAL262 FALSE 2.22 2.90
GLY264
FALSE
2.49
3.15







C5
ARG130 FALSE 2.24 2.97 ARG130 3.80
PHE131 FALSE 2.22 3.20 PHE164 3.88
VAL262 FALSE 1.93 2.91 HIS172 3.72
VAL263
FALSE
2.80
3.79







VETFS Batefenterol
C1
ARG262 TRUE 1.65 2.65 PHE271 3.47 ASP443 5.20
ASP272 FALSE 2.38 3.36 TYR276 3.90
TYR276 3.92




THR444
3.78





C2
TYR258 TRUE 1.97 2.88 ASP272 3.72 ASP443 5.07
ARG262 TRUE 3.14 3.84 MET275 3.78
ARG262 TRUE 2.38 3.30 TYR477 3.61
ASP272
FALSE
2.10
3.06







C3
ARG262 TRUE 1.86 2.82 PHE271 3.60 LYS273 TRUE 3.59 ASP443 5.25
ASP272 FALSE 2.10 3.09 TYR276 3.53
TYR276 3.62
ASP443 3.91




TYR477
3.54





Burixafor
C3
ASN170 TRUE 2.93 3.63 PHE445 3.88
ASN497
FALSE
3.12
4.05







C4
SER166 FALSE 1.82 2.79
GLU494 TRUE 2.69 3.52 PHE445 3.88
GLU494 TRUE 2.62 3.52
ASN497
FALSE
2.25
3.09







Eluxadoline C3 ARG262 TRUE 1.95 2.88 PHE271 3.83
ARG262 TRUE 3.34 3.93 PHE271 3.68
MET275 3.67
TYR477 3.70

3.11. PCA suggests the motions of the protein ligand complexes are relatively constricted

Protein function is controlled by shifting between various conformations. This ability to flexibly switch between multiple conformations is regulated by a set of their collective motions. These motions are critical to different biological processes and have a key contribution in the transmission of biological signals. A fair amount of flexibility as well as rigidity is necessary, especially of the binding site residues for any protein to be functional. A tighter ligand interaction would essentially restrict a protein's motion, thereby preventing it from sampling the critical conformations necessary for its functionality. Therefore, essential dynamics (ED) analysis was attempted to better understand the collective motions of all the nine ligand bound protein complexes in the 2D conformational phase space during the 150 ns simulation. As a multi-variate statistical method, ED is reliant upon diagonal covariance matrix of a protein's Cα atom to trace its global motion through Eigen vectors (EVs) popularly called principal components (PCs) and eigenvalues [83]. The EVs describe the global direction of motion of the atoms and the associated eigenvalues depict the atomic contribution of motion in MD trajectories of a ligand bound system which is essentially controlled by a proteins' secondary structure. The collective dynamic motions of the proteins captured through the projections of EVs (PC1 and PC2) are shown in Fig. 10 . The PCA statistics and the percentage of variance covered by the first three eigenvectors of I7L, Top1B and VETFS complexes are summarised in Supplementary Table S9. As visible from the figure, the first two PCs accounted for the bulk of all the internal motions. This indicates that the vectors mapped within this conformational subspace elucidate the essential subspace of the system. The trace values calculated from the covariance matrices of I7L-BAT, I7L-BUR, I7L-ELU, Top1B-BAT, Top1B-BUR, Top1B -ELU, VETFS-BAT, VETFS-BUR and VETFS-ELU complexes were 17.09 nm2, 16.99 nm2, 18.22 nm2, 30.52 nm2, 40.01 nm2, 23.69 nm2, 44.98 nm2, 42.12 nm2 and 57.12 nm2 respectively. The trace values of Top1B-BUR, VETFS-BAT, VETFS-BUR and VETFS-ELU complexes were relatively higher compared to the other five complexes. These results are consistent with higher occupation of the conformational subspace by these complexes which is clearly visible in the form of dispersed spectral dots and further hints at greater flexibility of these proteins. Therefore, as a consequence of higher fluctuations these complexes had to sample a much wider conformational space most probably to accommodate the ligands before accomplishing the ensembles in their dynamically equilibrated states. It is interesting to note that the molecular motions of the 3 Top1B complexes are relatively constricted (as supported by their low phase space occupancy). This further strengthen the idea that ensembles of all the Top1B complexes have achieved as state of dynamic equilibrium with their respective ligands. Overall, ED analysis suggests the collective molecular motions of all the 9 complexes are restricted to a small and localised conformational space. These results are consistent with their corresponding RMSD, SASA and Rg plots which further strengthen the notion that the three proteins namely, I7L, Top1B and VETFS have been stabilized by the ligands BAT, BUR and ELU.

Fig. 10.

Fig. 10

Principal component analysis revealing the dynamics of motion for all nine mapped ligand-bound complexes. Projection of motion in phase space along the PC1 and PC2 is drawn as scatter plot. (A) PCA plot of I7L-BAT complex. (B) PCA plot of I7L-BUR complex. (C) PCA plot of I7L-ELU complex. (D) (A) PCA plot of Top1B-BAT complex. (E) PCA plot of Top1B-BUR complex. (F) PCA plot of Top1B-ELU complex. (G) PCA plot of VETFS-BAT complex. (H) PCA plot of VETFS-BUR complex. (I) PCA plot of VETFS-ELU complex. The coloured dots in every PCA plot correspond to the sub clusters formed by each protein in complex with its respective ligand during 150 ns of simulation. The explained variance (%) of each principal component are presented in brackets.

3.12. Porcupine plot assessment suggest stability of Top1B's ligand bound DNA binding domain

Porcupine plots were developed to gain quantitative insights into the molecular motion of the 9 protein-ligand complexes. To picture the molecular motions, a cone was drawn along the C-alpha atom of each residue in the direction traced by its extreme projection on PC1 (Fig. 11 ) where the length of the cone signifies the amplitude of motion of the Cα atom. It shows that a vast majority of all the fluctuations are closely associated with the regions representing the loops/coils in all the protein ligand complexes. These results are completely in tune with their corresponding RMSF plots. Similarly, cones were also observed on the residues neighbouring the ligands in all the complexes. These fluctuations have most likely occurred to accommodate the ligands in the active site cavity. Interestingly, large protruding cones were clearly visible in the N-terminal domains of all the Top1B complexes which was not visible in their ligand bound C-terminal DNA binding domain. This clearly suggests the large fluctuations in Top1B complexes are particularly associated with the N-terminal domain. This is perfectly in sync with their RMSF plots where large fluctuations were specifically confined to the residues of the N-terminal domain. Combined together, the rmsf plots and porcupine plots of the Top1B complexes evidently explain the large fluctuations in RMSDs of all the Top1B complexes. These correlations beyond doubt clarify that the target C-terminal DNA binding domain of Top1B has successfully established an equilibrium with the ligands BAT, BUR and ELU.

Fig. 11.

Fig. 11

Porcupine plot displaying the motion of mpox virus proteins I7L, Top1B and VETFS with the ligands BAT, BUR and ELU obtained from PC1 during the 150 ns of simulation. The colours pink, blue, yellow and white represent α-helix, 3–10 helix, β-sheet and loops/coils of the proteins respectively. The ligands bound to the proteins are depicted in green colour. The fluctuations in Cα atoms above 2 Å are presented in the form of red coloured cones where the length of the cones represent amplitude of the Cα atoms from their mean positions. The illustrations were created with VMD.

3.13. MM/PBSA assessment suggests stable binding of the ligands BAT, BUR and ELU with the three proteins I7L, Top1B and VETFS

The assessment of RMSD trajectories suggest all the nine protein-ligand systems stabilized in the last 50 ns. Therefore, MM/PBSA was used to estimate the binding free energy and understand the associated molecular interactions between the proteins I7L, Top1B, VETFS and the ligands BAT, BUR and ELU for the last 50 ns of MD run. Supplementary Fig. S7 suggests the trajectories of binding free energy of all the nine protein-ligand complexes remained almost constant in the last 50 ns. Fig. 12 suggests that BAT, BUR and ELU bind favourably with all the three proteins. The three proteins, I7L, Top1B, VETFS, had highest binding affinity (ΔGmmpbsa) of −29.63 ± 7.53 kcal/mol, -32.72 ± 5.17 kcal/mol and -24.96 ± 4.48 kcal/mol respectively with BAT suggesting BAT might be the most suitable ligand for their inhibition. The decomposition of the average binding free energy of all the nine complexes into energy terms suggests the binding of the ligands BAT, BUR and ELU in the active site of majority of the proteins is stabilized by Van der Waal's (vdW) energies (Fig. 12). However, another critical energy term average electrostatic (ΔEele) seems to be the major contributor towards the stabilization of Top1B-ELU (ΔEele = −59.94 ± 11.95 kcal/mol) and VETFS-ELU (ΔEele = −42.85 ± 10.55 kcal/mol) complex respectively.

Fig. 12.

Fig. 12

The binding free energy terms derived from MM/PBSA calculations relative to the binding of BAT, BUR and ELU with the three proteins I7L, Top1B and VETFS. The colour coding schemes for different energy terms are shown at the top of the figure.

We further employed per residue binding free energy decomposition to determine the molecular interactions of every ligand with the binding pocket residues of the proteins BAT, BUR and ELU (Fig. 13 ). Per residue decomposition analysis suggests the active site residues energetically favour the stable binding of BAT with protein I7L. The free energy decomposition analysis plot suggests the active site residues Arg99, Ile298, Ile316, Leu109, Phe256, Pro110 and Pro264 favour the stable binding of BAT with I7L. The residues Met67, Pro64, Pro110, Pro114, Ser112, Thr111, Tyr46, Tyr238, Val47 favour the binding of BUR to the protein I7L. Similarly, the active site residues Asp258, Leu239, Pro264, Ser112 and Tyr238 favour the binding of ELU with I7L. The residues Pro 110, Ser112 and Pro264 of I7L protein were common to two of the three ligands. This suggests these residues might be important to I7L. The residues Met126 and Phe131 favoured the binding of BAT to the active site of Top1B. The residues Arg223, Gly264, Ile129, Phe127, Phe128, Phe174, Tyr233, Val 262, and Val 263 favoured the stable binding of BUR in the active site of Top1B whereas the pocket residues Arg130, Ile129, Phe131, Val 262 and Val 263 contributed towards the favourable binding of ELU in Top1B's active site. The residues Ile129, Phe 131, Val262 and Val263 were common to two of the three ligands bound to Top1B. Therefore, these residues might be important to Top1B. The residues Lys273, Met 275, Tyr276 and Tyr477 favoured the binding of BAT in the active site of protein VETFS whereas the active site residues Arg262, Met275, Phe271, Tyr276 guided the favourable binding of ELU with VETFS. Residues favouring the binding of BUR with VETFS could not be identified. The residues Met275 and Tyr276 were common to both the ligands. As such, these residues might be critical to VETFS.

Fig. 13.

Fig. 13

The per residue decomposition plots (MM/PBSA) representing the binding free energy contributed by the residues of the active sites of the three proteins I7L, Top1B and VETFS which energetically contribute towards the stabilization of the three ligands BAT, BUR and ELU.

All the ligand bound complexes maintained a favourable distance below 10 Å between the centre of mass of the interacting active site residues and centre of mass of their associated ligands with the exception of VETFS-BUR complex in the last 50 ns of simulation (Supplementary Fig. S8). This suggests stable binding modes of these ligands with the active site residues of the three proteins. The distance between the centre of mass of the ligand BUR and the centre of mass of the active site pocket residues of VETFS was far greater than 10 Å. This suggests BUR has been displaced from the active site of VETFS and therefore is no longer bound to it. Therefore, taken together only eight complexes namely, I7L-BAT, I7L-BUR, I7L-ELU, Top1B-BAT, Top1B-BUR, Top1B-ELU, VETFS-BAT and VETFS-ELU have successfully attained a state of dynamic equilibrium. These findings suggest the ligands BAT and ELU have the potential to function as inhibitors of all the three enzymes whereas BUR can potentially inhibit the enzymes I7L and Top1B.

4. Discussion

In this study, 125 genomes of mpox virus were collected from the public repository. Mining of these genomes led to the identification of four highly druggable non-host homologous enzymes namely, I7L, Top1B, VETFS and the A20R. Further, screening of a non-redundant library of 5893 compounds yielded BAT, BUR and ELU as potential inhibitors with an affinity for multiple targets. Interestingly, these molecules were amongst our top 30 hits for every target. Following MD simulation, the interactions of these ligands with three targets namely I7L, Top1B, VETFS were demonstrated to be stable. We fully acknowledge that our findings currently lack in vivo and in vitro antiviral experimental validations. However, we wanted to make our findings available to the scientific community at the earliest with the hope that our findings can be suitably exploited by others in the fight against the global mpox epidemic.

The presently ongoing mpox outbreak is unique as it is the most dispersed and the largest to be ever documented in non-endemic countries. Its rapid rise and dissemination across the globe are a cause of great concern. However, the only probable saving grace is the fact that the current strains under circulation are closely related to the less infectious strains of clade II [84,85]. Nevertheless, a global outbreak of its more nefarious and highly virulent cousin from clade I (with case fatality rate >10%) [86] can never be ruled out in the future. Currently, there are no specific antivirals against mpox. Therefore, it is highly desirable to quickly identify potential therapeutics against mpox. Identifying ‘drug repurposing’ potential through in silico analysis is an approach to fasten antiviral development. However, the availability of valid, druggable targets is key to the success of this strategy. Unfortunately, no information is available about the druggable targets of mpox virus.

Genes that are conserved across diverse phylogenetic lineages are most likely to be essential and their subsequent protein products are likely to have a critical role in a pathogen's lifecycle and virulence [87,88]. In the current study, 69 highly conserved proteins were identified in all genomes of mpox virus. The ease of access to the therapeutic targets under investigation is paramount for an experimental biologist to rapidly investigate and verify the outcomes of in silico studies. Globular proteins provide a significant advantage over membrane-associated proteins in this regard. Thus, a pool of 56 globular proteins was selected for further downstream investigations.

Since the identified possible core transcription factors associated with mpox virus remain functionally uncharacterised, targeting them without prior scientific evidence might not yield desirable results. Hence, the possibility of transcription factors as therapeutic targets was ruled out by subtractive proteomic approach from the list of identified globular proteins for the time being. On the contrary, the identified core enzymes shared sequence similarities and structural identities with the enzymes of its nearest neighbour, the vaccinia virus, whose functionalities are fairly well characterised. Therefore, only the 23 enzymes from the pool of 56 globular proteins were pursued further to identify therapeutic targets.

An ideal drug target in a pathogen must always be non-homologous to the host's proteome to minimise cross-reactivity and related side effects [89,90]. However, as clearly visible from our study, 8 enzymes shared significant sequence homology with the critical human proteins. Therefore, these enzymes were eliminated from this study. The remaining 15 non-host homologous enzymes were prioritised for further examination.

To be considered a therapeutic target, a protein should be critically involved in the signalling or metabolic pathway of a virus. Secondly, the biological function of this protein must be tuneable by binding of the drug candidates with high affinity. These combined features were together termed “druggability” by Hopkins and Groom [91]. Therefore, druggability is a critical step in the selection, categorisation, and validation of suitable drug targets in the early stages of drug discovery. Unfortunately, we were impelled to temporarily halt our pursuit of assessing the druggability of 8 enzymes because of some unavoidable circumstances mentioned previously. Accordingly, the 7 remaining host non-homologous enzymes were subjected to druggability assessment using CavityPlus [62], DoGSiteScorer [63] and DeepSite [64]. The outcomes were interesting and, in a manner, similar to Cheng et al. (2007); only 4 key enzymes of mpox virus were predicted as “druggable”. The remaining 3 enzymes namely, DNA-dependent RNA polymerase 18 kDa subunit (Rpo18), mRNA-capping enzyme 33 kDa small subunit (D12L) and DNA-dependent RNA polymerase 19 kDa subunit (Rpo19) were predicted as “undruggable”. Taken together, 4 highly druggable, host non-homologous enzymes of mpox virus were identified. Their physicochemical properties supported their stability and suitability for further analysis.

The I7L gene product i.e., core cysteine protease was identified as one of the four therapeutic targets. This is the main candidate protein for viral core protein proteinase (vCPP) activity in the viruses which is essential for the production of other non-structural proteins. Like other virus core proteases, the mpox virus I7L protein also contains putative catalytic residues (His241 and Cys328) in its highly druggable binding site [92]. Top1B DNA topoisomerase relaxes DNA supercoils by iteratively cleaving and rejoining one strand of the DNA duplex through a covalent DNA-(3′-phosphotyrosyl)-enzyme intermediate [93]. This makes it a critical chokepoint of the mpox virus replication machinery and an ideal therapeutic target. VETFS was identified as the third target. This protein is expressed late in the infectious cycle of orthopox virus and regulates the cascade mechanism of gene transcription in the virus. VETFS is recruited by RNA polymerase associated protein (RAP94, 94 kDa) to form the early transcription complex including core RNA polymerase and binds to early gene promoters [94]. Inhibition of this can potentially prevent the assembly of the functional early transcription complex necessary for the successful expression of genes pertaining to the regulation of its lifecycle, virulence, and immunomodulation. Previous studies [95,96] suggest A20R in vaccinia virus as a processivity factor on the essentially distributive viral DNA polymerase along with D4R, D5R and H5R proteins [95]. Thus, targeting these core enzymes can choke the replication, translation and assembly of the mpox virus into the mature virion particles. Therefore, these 4 proteins can be considered potential therapeutic targets for mpox. However, the actual expression of these targets must be verified, and their cellular functions must be characterised to validate them.

With drug repurposing as the objective, a highly curated library of 5893 FDA approved/investigational drugs was screened through HTVS against these targets. We employed the Schrodinger suite's Glide module for this purpose. The Glide module follows a three-stage screening protocol namely, HTVS, SP and XP where the sampling and scoring gradually become more extensive and stringent [69,97]. Moreover, the XGlide module allowed us to screen the library against multiple target proteins simultaneously. A list of the top 30 hits corresponding to 0.5% of the drugs contained in the FDA approved/investigational library was also compiled for every target protein. BAT, BUR and ELU were the common top-ranked hits against every protein target as visible from the list of top 30 hits. Targeting multiple non-structural proteins is expected to enhance antiviral potency [98,99]. It is also expected to overcome resistance due to point mutations in targets. Since some of these targets are also conserved in other strains/viruses, multi-targeting can lead to a broad spectrum of action [100]. BAT is an approved drug for treating pulmonary diseases [101,102]. BUR is used for treating multiple myeloma, Hodgkin's disease, and non-Hodgkin's lymphoma [103]. ELU is primarily prescribed for bowel syndrome with diarrhea [104]. Although the primary indication of these drugs do not overlap with viral pathology, any overlap of the gene involved in these indications with that of viral disease cannot be ruled out at this stage.

Molecular dynamics (MD) simulation mimics the flexible behaviour of biomolecules, conformational changes in proteins and protein-ligand interactions to paint a realistic picture with atomic resolution with reference to time. Therefore, to gain further insights into the structural dynamics and stabilities of BAT, BUR and ELU with all the enzymes namely, A20R, I7L, Top1B and VETFS, MD simulations were executed for 150 ns for every complex.

A set of quality control parameters like RMSD, RMSF, Rg, SASA and H-Bond were applied on all the twelve protein-ligand complexes to infer their stability. Unfortunately, none of the ligands could stabilize the enzyme A20R. Although the RMSD trajectory of A20R-BUR appeared to be stable for the first 90 ns, it continued its upward ascension for the remaining time duration. Hence, it was not possible to draw a reliable conclusion about the stability of A20R-BUR complex. On the contrary, RMSD trajectories of the remaining 9 complexes suggested that BAT, BUR and ELU successfully stabilized the enzymes I7L, Top1B and VETFS. Their RMSD trajectories were also coherent with their Rg and SASA trajectories. Such findings suggest the stable ligand bound complexes have successfully retained their compactness throughout the simulations. Prominent transformations in the secondary structures of some stable ligand bound proteins were also identified after examining their time dependent secondary structure evolution graph. These transformations in the secondary structural elements might be key drivers of ligand dependent stabilization of these complexes. Therefore, we believe a better understanding of these structural changes may be fundamental in designing antiviral therapeutics against these proteins of mpox virus.

The complexes of the enzyme Top1B presented a very interesting case in itself. The RMSDs of the Top1B ligand bound complexes fluctuated irregularly while transitioning between their stable conformations. However, upon further examining their Rg, SASA, H-Bond and PCA plots, a more cleaner picture of their relative stability was obtained. The assessment of RMSF plots and porcupine plots further vindicated the stabilities of these complexes. Top1B is a multidomain protein comprising of a N-terminal DNA binding domain and a C-terminal domain which is separated by a hinge comprising of three residues Gly73, Lys71 and Met75. Our assessments suggest this hinge has added to the flexibility of Top1B's overall structure by granting a higher degree of flexibility to the N-terminal domain whereas the desired target DNA binding C-terminal domain was relatively stable upon ligand binding. Therefore, in future drug discovery endeavours researchers are expected to be mindful of studying the two domains of Top1B enzyme independently to avoid false negative predictions. Similarly, the RMSD trajectories and all the quality control parameters clearly indicated towards the stable binding of the VETFS-BUR complex initially. However, further assessment of the distance between the centre of mass of the ligand BUR and the interacting active site residues of VETFS suggests that the ligand has moved away from the active site pocket and is therefore no longer bound to it. This somewhat explains the increased compactness (Rg) of VETFS-BUR complex which was wrongly interpreted for a stable protein-ligand complex earlier.

Clinically, BAT does not show any general or cardiovascular adverse events when inhaled even after 6 continuous weeks of use for chronic obstructive pulmonary disease [105]. Burixafor is also well tolerated in clinical applications against myolema and lumphoma [103]. Besides, ELU has been demonstrated to be safe in a 12 week long treatment against irritable bowel syndrome with diarrhea [106]. Taken together BAT, BUR and ELU show potential for repurposing against mpox. However, it requires further experimental, pre-clinical as well as clinical validations. Nonetheless, this is an attempt to leverage multi-omics mining strategies to identify therapeutic targets of mpox virus and their potential inhibitors. As newly sequenced genomes of mpox virus gradually get deposited in the public repositories, our findings can easily be reauthenticated by incorporating newer genomes. This can encourage research for repurposing drugs against mpox.

5. Conclusion

This study demonstrated the integration of genomics and subtractive proteomics to identify therapeutic targets for mpox virus. Although this requires further validation, our in silico analysis and prior literature indicate their druggability and relevance as therapeutic targets. This can encourage structure-based analysis for finding antivirals for mpox. The virtual screening followed by MD simulation suggested batefenterol, burixafor and eluxadoline as potential inhibitors of multiple mpox virus targets. The clinical safety of these drugs also supports their suitability for repurposing. This can encourage experimental validations to repurpose these as antivirals for the therapeutic management of mpox virus infection.

Author contributions

Conceptualization: AS, BBS; Formal Analysis: AS, MG; Funding Acquisition: BBS; Investigation: AS, MG; Methodology: AS, MG, NCM, BBS; Project administration: BBS; Resources: BBS; Supervision: BBS; Visualization: AS, MG; Writing – original draft: AS, MG; Writing – review & editing: AS, MG, NCM, ES, RPS, BBS.

Declaration of competing interest

The authors declare that they have no known competing financial interests or personal relationships that can influence the work reported in this paper.

Acknowledgements

We thank Dean, School of Pharmaceutical Sciences, Siksha ‘O’ Anusandhan (Deemed to be University), Bhubaneswar, Odisha, India for the facilities. MG is supported by a fellowship funded from by Department of Biotechnology (DBT), Ministry of Science and Technology, New Delhi, India (Grant Id: BT/INF/22/SP45078/2022). The authors would like to acknowledge the Indian Council of Medical Research (ICMR), Ministry of Health & Family Welfare, New Delhi, India (Grant ID: AMR/DHR/GIA/4/ECD-II-2020) for providing high-performance computational resources for this study. We acknowledge the Bioinformatics Resources and Applications Facility (BRAF), C-DAC, Pune, India for providing timely access to their high-performance computing clusters, which allowed us to perform complex simulations and data analysis for this study.

Footnotes

Appendix A

Supplementary data to this article can be found online at https://doi.org/10.1016/j.compbiomed.2023.106971.

Appendix A. Supplementary data

The following are the Supplementary data to this article:

Multimedia component 1
mmc1.docx (6.1MB, docx)
Multimedia component 2
mmc2.xlsx (31.5KB, xlsx)
Multimedia component 3
mmc3.xlsx (20.1KB, xlsx)
Multimedia component 4
mmc4.xlsx (119.2KB, xlsx)

References

  • 1.World Health Organization (16 May 2022), Monkeypox - United Kingdom of Great Britain and Northern Ireland 2022. https://www.who.int/emergencies/disease-outbreak-news/item/2022-DON381
  • 2.World Health Organization, WHO Director-General’s statement at the press conference following IHR Emergency Committee regarding the multi-country outbreak of monkeypox - 23 July 2022 2022. https://www.who.int/director-general/speeches/detail/who-director-general-s-statement-on-the-press-conference-following-IHR-emergency-committee-regarding-the-multi-country-outbreak-of-monkeypox-23-july-2022
  • 3.Kozlov M. Monkeypox goes global: why scientists are on alert. Nature. 2022;606:15–16. doi: 10.1038/d41586-022-01421-8. [DOI] [PubMed] [Google Scholar]
  • 4.(ICTV) ICoToV. Virus Taxonomy Release. 2020. https://talk.ictvonline.org/taxonomy
  • 5.von Magnus P., Andersen E.K., Petersen K.B., Birch-Andersen A. A pox-like disease in cynomolgus monkeys, acta pathol. Microbiol. Scand. 1959;46:156–176. doi: 10.1111/j.1699-0463.1959.tb00328.x. [DOI] [Google Scholar]
  • 6.Breman J.G. Kalisa-ruti, M. V steniowski, E. Zanotto, A.I. Gromyko, I. Arita, human monkeypox, 1970-79. Bull. World Health Organ. 1980;58:165–182. http://www.ncbi.nlm.nih.gov/pubmed/6249508 [PMC free article] [PubMed] [Google Scholar]
  • 7.Bunge E.M., Hoet B., Chen L., Lienert F., Weidenthaler H., Baer L.R., Steffen R. The changing epidemiology of human monkeypox—a potential threat? A systematic review. PLoS Negl. Trop. Dis. 2022;16 doi: 10.1371/journal.pntd.0010141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Centers for Disease Control and Prevention (CDC) Multistate outbreak of monkeypox–Illinois, Indiana, and Wisconsin, 2003., MMWR. Morb. Mortal. Wkly. Rep. 2003;52:537–540. http://www.ncbi.nlm.nih.gov/pubmed/12803191 [PubMed] [Google Scholar]
  • 9.Centers for Disease Control and Prevention (CDC) Update: multistate outbreak of monkeypox–Illinois, Indiana, Kansas, Missouri, Ohio, and Wisconsin, 2003., MMWR. Morb. Mortal. Wkly. Rep. 2003;52:561–564. http://www.ncbi.nlm.nih.gov/pubmed/12816106 [PubMed] [Google Scholar]
  • 10.Centers for Disease Control and Prevention (CDC) Update: multistate outbreak of monkeypox–Illinois, Indiana, Kansas, Missouri, Ohio, and Wisconsin, 2003., MMWR. Morb. Mortal. Wkly. Rep. 2003;52:642–646. http://www.ncbi.nlm.nih.gov/pubmed/12855947 [PubMed] [Google Scholar]
  • 11.Reed K.D., Melski J.W., Graham M.B., Regnery R.L., Sotir M.J., V Wegner M., Kazmierczak J.J., Stratman E.J., Li Y., Fairley J.A., Swain G.R., Olson V.A., Sargent E.K., Kehl S.C., Frace M.A., Kline R., Foldy S.L., Davis J.P., Damon I.K. The detection of monkeypox in humans in the Western Hemisphere., N. Engl. J. Med. 2004;350:342–350. doi: 10.1056/NEJMoa032299. [DOI] [PubMed] [Google Scholar]
  • 12.European Centre for Disease Prevention and Control Risk assessment: monkeypox multi-country outbreak. 2022. https://www.ecdc.europa.eu/en/publications-data/risk-assessment-monkeypox-multi-country-outbreak
  • 13.Alakunle E.F., Okeke M.I. Monkeypox virus: a neglected zoonotic pathogen spreads globally. Nat. Rev. Microbiol. 2022 doi: 10.1038/s41579-022-00776-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Damon I.K. Poxviruses. Manual Clin. Microbiol. 2006;2:1631–1640. n.d. [Google Scholar]
  • 15.ramazan azim okyay Another epidemic in the shadow of Covid 19 pandemic: a review of monkeypox. Eurasian J. Med. Oncol. 2022 doi: 10.14744/ejmo.2022.2022. [DOI] [Google Scholar]
  • 16.Shchelkunov S.N., Totmenin A.V., Babkin I.V., Safronov P.F., Ryazankina O.I., Petrov N.A., Gutorov V.V., Uvarova E.A., Mikheev M.V., Sisler J.R., Esposito J.J., Jahrling P.B., Moss B., Sandakhchiev L.S. Human monkeypox and smallpox viruses: genomic comparison. FEBS Lett. 2001;509:66–70. doi: 10.1016/S0014-5793(01)03144-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Kugelman J.R., Johnston S.C., Mulembakani P.M., Kisalu N., Lee M.S., Koroleva G., McCarthy S.E., Gestole M.C., Wolfe N.D., Fair J.N., Schneider B.S., Wright L.L., Huggins J., Whitehouse C.A., Wemakoy E.O., Muyembe-Tamfum J.J., Hensley L.E., Palacios G.F., Rimoin A.W. Genomic variability of monkeypox virus among humans, Democratic Republic of the Congo. Emerg. Infect. Dis. 2014;20:232–239. doi: 10.3201/eid2002.130118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Kmiec D., Kirchhoff F. Monkeypox: a new threat? Int. J. Mol. Sci. 2022;23 doi: 10.3390/ijms23147866. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Seet B.T., Johnston J.B., Brunetti C.R., Barrett J.W., Everett H., Cameron C., Sypula J., Nazarian S.H., Lucas A., McFadden G. Poxviruses and immune evasion. Annu. Rev. Immunol. 2003;21:377–423. doi: 10.1146/annurev.immunol.21.120601.141049. [DOI] [PubMed] [Google Scholar]
  • 20.Senkevich T.G., Yutin N., Wolf Y.I., Koonin E.V., Moss B. Ancient gene capture and recent gene loss shape the evolution of orthopoxvirus-host interaction genes. mBio. 2021;12 doi: 10.1128/mBio.01495-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Marennikova S.S., Seluhina E.M., Mal’ceva N.N., Cimiskjan K.L., Macevic G.R. Isolation and properties of the causal agent of a new variola-like disease (monkeypox) in man. Bull. World Health Organ. 1972;46:599–611. [PMC free article] [PubMed] [Google Scholar]
  • 22.McFadden G. Poxvirus tropism. Nat. Rev. Microbiol. 2005;3:201–213. doi: 10.1038/nrmicro1099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Realegeno S., Puschnik A.S., Kumar A., Goldsmith C., Burgado J., Sambhara S., Olson V.A., Carroll D., Damon I., Hirata T., Kinoshita T., Carette J.E., Satheshkumar P.S. Monkeypox virus host factor screen using haploid cells identifies essential role of GARP complex in extracellular virus formation. J. Virol. 2017;91 doi: 10.1128/jvi.00011-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Moss B. Membrane fusion during poxvirus entry. Semin. Cell Dev. Biol. 2016;60:89–96. doi: 10.1016/j.semcdb.2016.07.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Wilson M.E., Hughes J.M., McCollum A.M., Damon I.K. Human monkeypox. Clin. Infect. Dis. 2014;58:260–267. doi: 10.1093/cid/cit703. [DOI] [PubMed] [Google Scholar]
  • 26.Yinka-Ogunleye A., Aruna O., Dalhat M., Ogoina D., McCollum A., Disu Y., Mamadu I., Akinpelu A., Ahmad A., Burga J., Ndoreraho A., Nkunzimana E., Manneh L., Mohammed A., Adeoye O., Tom-Aba D., Silenou B., Ipadeola O., Saleh M., Adeyemo A., Nwadiutor I., Aworabhi N., Uke P., John D., Wakama P., Reynolds M., Mauldin M.R., Doty J., Wilkins K., Musa J., Khalakdina A., Adedeji A., Mba N., Ojo O., Krause G., Ihekweazu C., Mandra A., Davidson W., Olson V., Li Y., Radford K., Zhao H., Townsend M., Burgado J., Satheshkumar P.S. Outbreak of human monkeypox in Nigeria in 2017–18: a clinical and epidemiological report. Lancet Infect. Dis. 2019;19:872–879. doi: 10.1016/S1473-3099(19)30294-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Simpson K., Heymann D., Brown C.S., Edmunds W.J., Elsgaard J., Fine P., Hochrein H., Hoff N.A., Green A., Ihekweazu C., Jones T.C., Lule S., Maclennan J., McCollum A., Mühlemann B., Nightingale E., Ogoina D., Ogunleye A., Petersen B., Powell J., Quantick O., Rimoin A.W., Ulaeato D., Wapling A. Human monkeypox – after 40 years, an unintended consequence of smallpox eradication. Vaccine. 2020;38:5077–5081. doi: 10.1016/j.vaccine.2020.04.062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Chakraborty C., Bhattacharya M., Nandi S.S., Mohapatra R.K., Dhama K., Agoramoorthy G. Appearance and re-appearance of zoonotic disease during the pandemic period: long-term monitoring and analysis of zoonosis is crucial to confirm the animal origin of SARS-CoV-2 and monkeypox virus. Vet. Q. 2022;42:119–124. doi: 10.1080/01652176.2022.2086718. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Reynolds M.G., McCollum A.M., Nguete B., Lushima R.S., Petersen B.W. Improving the care and treatment of monkeypox patients in low-resource settings: applying evidence from contemporary biomedical and smallpox biodefense research. Viruses. 2017;9 doi: 10.3390/v9120380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Sklenovská N., Van Ranst M. Emergence of monkeypox as the most important orthopoxvirus infection in humans, front. Publ. Health. 2018;6 doi: 10.3389/fpubh.2018.00241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Thornhill J.P., Barkati S., Walmsley S., Rockstroh J., Antinori A., Harrison L.B., Palich R., Nori A., Reeves I., Habibi M.S., Apea V., Boesecke C., Vandekerckhove L., Yakubovsky M., Sendagorta E., Blanco J.L., Florence E., Moschese D., Maltez F.M., Goorhuis A., Pourcher V., Migaud P., Noe S., Pintado C., Maggi F., Hansen A.-B.E., Hoffmann C., Lezama J.I., Mussini C., Cattelan A., Makofane K., Tan D., Nozza S., Nemeth J., Klein M.B., Orkin C.M. SHARE-Net clinical group, monkeypox virus infection in humans across 16 countries - april-june 2022. N. Engl. J. Med. 2022 doi: 10.1056/NEJMoa2207323. [DOI] [PubMed] [Google Scholar]
  • 32.Fine P.E.M., Jezek Z., Grab B., Dixon H. The transmission potential of monkeypox virus in human populations, Int. J. Epidemiol. 1988;17:643–650. doi: 10.1093/ije/17.3.643. [DOI] [PubMed] [Google Scholar]
  • 33.Rao A.K., Petersen B.W., Whitehill F., Razeq J.H., Isaacs S.N., Merchlinsky M.J., Campos-Outcalt D., Morgan R.L., Damon I., Sánchez P.J., Bell B.P. Use of JYNNEOS (smallpox and monkeypox vaccine, live, nonreplicating) for preexposure vaccination of persons at risk for occupational exposure to orthopoxviruses: recommendations of the advisory committee on immunization practices — United States, 2022, MMWR. Morb. Mortal. Wkly. Rep. 2022;71:734–742. doi: 10.15585/mmwr.mm7122e1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Xiang Y., White A. Monkeypox Virus Emerges from The Shadow of Its More Infamous Cousin: Family Biology Matters. Emerg. Microbes Infect. 2022:1–14. doi: 10.1080/22221751.2022.2095309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Xiao Y., Isaacs S.N. Therapeutic vaccines and antibodies for treatment of orthopoxvirus infections. Viruses. 2010;2:2381–2403. doi: 10.3390/v2102381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Yang G., Pevear D.C., Davies M.H., Collett M.S., Bailey T., Rippen S., Barone L., Burns C., Rhodes G., Tohan S., Huggins J.W., Baker R.O., Buller R.L.M., Touchette E., Waller K., Schriewer J., Neyts J., DeClercq E., Jones K., Hruby D., Jordan R. An orally bioavailable antipoxvirus compound (ST-246) inhibits extracellular virus formation and protects mice from lethal orthopoxvirus challenge. J. Virol. 2005;79:13139–13149. doi: 10.1128/jvi.79.20.13139-13149.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Andrei G., Fiten P., Krečmerová M., Opdenakker G., Topalis D., Snoeck R. Poxviruses bearing DNA polymerase mutations show complex patterns of cross-resistance. Biomedicines. 2022;580(10) doi: 10.3390/biomedicines10030580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Karim M., Nazrul Islam M., Jewel G.M.N.A. Silico identification of potential drug targets by subtractive genome analysis of Enterococcus faecium DO. bioRxiv. 2020 doi: 10.1101/2020.02.14.948232. [DOI] [Google Scholar]
  • 39.Solanki V., Tiwari M., Tiwari V. Subtractive proteomic analysis of antigenic extracellular proteins and design a multi-epitope vaccine against Staphylococcus aureus. Microbiol. Immunol. 2021;65:302–316. doi: 10.1111/1348-0421.12870. [DOI] [PubMed] [Google Scholar]
  • 40.Ahmad S., Navid A., Akhtar A.S., Azam S.S., Wadood A., Pérez-Sánchez H., Genomics Subtractive, Docking Molecular, Dynamics Molecular. Simulation revealed LpxC as a potential drug target against multi-drug resistant Klebsiella pneumoniae, interdiscip. Sci. Comput. Life Sci. 2019;11:508–526. doi: 10.1007/s12539-018-0299-y. [DOI] [PubMed] [Google Scholar]
  • 41.Solanki V., Tiwari V. Subtractive proteomics to identify novel drug targets and reverse vaccinology for the development of chimeric vaccine against Acinetobacter baumannii. Sci. Rep. 2018;8 doi: 10.1038/s41598-018-26689-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Uddin R., Jamil F. Prioritization of potential drug targets against P. aeruginosa by core proteomic analysis using computational subtractive genomics and Protein-Protein interaction network. Comput. Biol. Chem. 2018;74:115–122. doi: 10.1016/j.compbiolchem.2018.02.017. [DOI] [PubMed] [Google Scholar]
  • 43.Seemann T. Prokka: rapid prokaryotic genome annotation. Bioinformatics. 2014;30:2068–2069. doi: 10.1093/bioinformatics/btu153. [DOI] [PubMed] [Google Scholar]
  • 44.Kristensen D.M., Waller A.S., Yamada T., Bork P., Mushegian A.R., Koonin E.V., Gene Clusters Orthologous, Signature Taxon. Genes for viruses of prokaryotes. J. Bacteriol. 2013;195:941–950. doi: 10.1128/JB.01801-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Nogueira W.G., Jaiswal A.K., Tiwari S., Ramos R.T.J., Ghosh P., Barh D., Azevedo V., Soares S.C. Computational identification of putative common genomic drug and vaccine targets in Mycoplasma genitalium. Genomics. 2021;113:2730–2743. doi: 10.1016/j.ygeno.2021.06.011. [DOI] [PubMed] [Google Scholar]
  • 46.Emms D.M., Kelly S. OrthoFinder: phylogenetic orthology inference for comparative genomics. Genome Biol. 2019;238(20) doi: 10.1186/s13059-019-1832-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Emms D.M., Kelly S. OrthoFinder: solving fundamental biases in whole genome comparisons dramatically improves orthogroup inference accuracy. Genome Biol. 2015;16 doi: 10.1186/s13059-015-0721-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Acebrón-García-de-Eulate M., Blundell T.L., Vedithi S.C. Strategies for drug target identification in Mycobacterium leprae, Drug Discov. Today Off. 2021;26:1569–1573. doi: 10.1016/j.drudis.2021.03.026. [DOI] [PubMed] [Google Scholar]
  • 49.Hallgren J., Tsirigos K.D., Damgaard Pedersen M., Juan J., Armenteros A., Marcatili P., Nielsen H., Krogh A., Winther O. DeepTMHMM predicts alpha and beta transmembrane proteins using deep neural networks. bioRxiv. 2022 doi: 10.1101/2022.04.08.487609. 2022.04.08.487609. [DOI] [Google Scholar]
  • 50.Dobson L., Reményi I., Tusnády G.E. CCTOP: a Consensus Constrained TOPology prediction web server. Nucleic Acids Res. 2015;43:W408–W412. doi: 10.1093/nar/gkv451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Nugent T., Jones D.T. Detecting pore-lining regions in transmembrane protein sequences. BMC Bioinformatics. 2012;169(13) doi: 10.1186/1471-2105-13-169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Tsirigos K.D., Peters C., Shu N., Käll L., Elofsson A. The TOPCONS web server for consensus prediction of membrane protein topology and signal peptides, Nucleic Acids Res. 2015;43:W401–W407. doi: 10.1093/nar/gkv485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Li Y., Wang S., Umarov R., Xie B., Fan M., Li L., Gao X. DEEPre: sequence-based enzyme EC number prediction by deep learning. Bioinformatics. 2018;34:760–769. doi: 10.1093/bioinformatics/btx680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Tahir ul Qamar M., Mirza M.U., Song J.-M., Rao M.J., Zhu X., Chen L.-L. Probing the structural basis of Citrus phytochrome B using computational modelling and molecular dynamics simulation approaches. J. Mol. Liq. 2021;340 doi: 10.1016/j.molliq.2021.116895. [DOI] [Google Scholar]
  • 55.Baek M., DiMaio F., Anishchenko I., Dauparas J., Ovchinnikov S., Lee G.R., Wang J., Cong Q., Kinch L.N., Dustin Schaeffer R., Millán C., Park H., Adams C., Glassman C.R., DeGiovanni A., Pereira J.H., Rodrigues A.V., Van Dijk A.A., Ebrecht A.C., Opperman D.J., Sagmeister T., Buhlheller C., Pavkov-Keller T., Rathinaswamy M.K., Dalwadi U., Yip C.K., Burke J.E., Christopher Garcia K., Grishin N.V., Adams P.D., Read R.J., Baker D. Accurate prediction of protein structures and interactions using a three-track neural network. Science. 2021;373:871–876. doi: 10.1126/SCIENCE.ABJ8754/SUPPL_FILE/ABJ8754_MDAR_REPRODUCIBILITY_CHECKLIST.PDF. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Williams C.J., Headd J.J., Moriarty N.W., Prisant M.G., Videau L.L., Deis L.N., Verma V., Keedy D.A., Hintze B.J., Chen V.B., Jain S., Lewis S.M., Arendall W.B., Snoeyink J., Adams P.D., Lovell S.C., Richardson J.S., Richardson D.C. MolProbity: more and better reference data for improved all-atom structure validation. Protein Sci. 2018;27:293–315. doi: 10.1002/pro.3330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Laskowski R.A., MacArthur M.W., Moss D.S., Thornton J.M. PROCHECK: a program to check the stereochemical quality of protein structures. J. Appl. Crystallogr. 1993;26:283–291. doi: 10.1107/s0021889892009944. [DOI] [Google Scholar]
  • 58.Eisenberg D., Lüthy R., Bowie J.U. VERIFY3D: assessment of protein models with three-dimensional profiles. Methods Enzymol. 1997;97:77022–77028. doi: 10.1016/S0076-6879. [DOI] [PubMed] [Google Scholar]
  • 59.Colovos C., Yeates T.O. Verification of protein structures: patterns of nonbonded atomic interactions. Protein Sci. 1993;2:1511–1519. doi: 10.1002/PRO.5560020916. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Liu T., Altman R. Identifying druggable targets by protein microenvironments matching: application to transcription factors, CPT pharmacometrics syst. Pharmacol. 2014;3:93. doi: 10.1038/psp.2013.66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Jamal S.B., Hassan S.S., Tiwari S., Viana M.V., De Jesus Benevides L., Ullah A., Turjanski A.G., Barh D., Ghosh P., Costa D.A., Silva A., Röttger R., Baumbach J., Azevedo V.A.C. An integrative in-silico approach for therapeutic target identification in the human pathogen Corynebacterium diphtheriae. PLoS One. 2017;12 doi: 10.1371/journal.pone.0186401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Xu Y., Wang S., Hu Q., Gao S., Ma X., Zhang W., Shen Y., Chen F., Lai L., Pei J. CavityPlus: a web server for protein cavity detection with pharmacophore modelling, allosteric site identification and covalent ligand binding ability prediction, Nucleic Acids Res. 2018;46:W374–W379. doi: 10.1093/nar/gky380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Volkamer A., Kuhn D., Rippmann F., Rarey M. Dogsitescorer: a web server for automatic binding site prediction, analysis and druggability assessment, Bioinformatics. 2012;28:2074–2075. doi: 10.1093/bioinformatics/bts310. [DOI] [PubMed] [Google Scholar]
  • 64.Jiménez J., Doerr S., Martínez-Rosell G., Rose A.S., De Fabritiis G. DeepSite: protein-binding site predictor using 3D-convolutional neural networks. Bioinformatics. 2017;33:3036–3042. doi: 10.1093/bioinformatics/btx350. [DOI] [PubMed] [Google Scholar]
  • 65.Zhang J., Gan Y., Li H., Yin J., He X., Lin L., Xu S., Fang Z., wook Kim B., Gao L., Ding L., Zhang E., Ma X., Li J., Li L., Xu Y., Horne D., Xu R., Yu H., Gu Y., Huang W. Inhibition of the CDK2 and Cyclin A complex leads to autophagic degradation of CDK2 in cancer cells. Nat. Commun. 2022;13 doi: 10.1038/s41467-022-30264-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Skuta C., Popr M., Muller T., Jindrich J., Kahle M., Sedlak D., Svozil D., Bartunek P. Probes & Drugs portal: an interactive, open data resource for chemical biology. Nat. Methods. 2017;14:759–760. doi: 10.1038/nmeth.4365. [DOI] [PubMed] [Google Scholar]
  • 67.RDKit Open-Source cheminformatics software (RRID: SCR_014274), (n.d. http://www.rdkit.org
  • 68.Repasky M.P., Shelley M., Friesner R.A. Flexible ligand docking with glide. Curr. Protoc. Bioinforma. 2007 doi: 10.1002/0471250953.bi0812s18. [DOI] [PubMed] [Google Scholar]
  • 69.Friesner R.A., Murphy R.B., Repasky M.P., Frye L.L., Greenwood J.R., Halgren T.A., Sanschagrin P.C., Mainz D.T. Extra precision glide: docking and scoring incorporating a model of hydrophobic enclosure for protein-ligand complexes. J. Med. Chem. 2006;49:6177–6196. doi: 10.1021/jm051256o. [DOI] [PubMed] [Google Scholar]
  • 70.Abraham M.J., Murtola T., Schulz R., Páll S., Smith J.C., Hess B., Lindah E. Gromacs: high performance molecular simulations through multi-level parallelism from laptops to supercomputers. SoftwareX. 1–2. 2015:19–25. doi: 10.1016/j.softx.2015.06.001. [DOI] [Google Scholar]
  • 71.Hess B., Bekker H., Berendsen H.J.C., Fraaije J.G.E.M. LINCS: a linear constraint solver for molecular simulations, J. Comput. Chem. 1997;18:1463–1472. doi: 10.1002/(SICI)1096-987X(199709)18:12<1463::AID-JCC4>3.0.CO;2-H. [DOI] [Google Scholar]
  • 72.Kunzmann P., Hamacher K. Biotite: a unifying open source computational biology framework in Python. BMC Bioinformatics. 2018;346(19) doi: 10.1186/s12859-018-2367-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Tubiana T., Carvaillo J.-C., Boulard Y., Bressanelli S. TTClust: a versatile molecular simulation trajectory clustering program with graphical summaries, J. Chem. Inf. Model. 2018;58:2178–2182. doi: 10.1021/acs.jcim.8b00512. [DOI] [PubMed] [Google Scholar]
  • 74.David C.C., Jacobs D.J. Principal component analysis: a method for determining the essential dynamics of proteins. Methods Mol. Biol. 2014 doi: 10.1007/978-1-62703-658-0_11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Ross C., Nizami B., Glenister M., Amamuddy O.S., Atilgan A.R., Atilgan C., Bishop Ö.T. MODE-TASK: large-scale protein motion tools. Bioinformatics. 2018 doi: 10.1093/bioinformatics/bty427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Wickham H. 2016. ggplot2: Elegant Graphics for Data Analysis.https://ggplot2.tidyverse.org [Google Scholar]
  • 77.William H., Andrew D., Klaus S. VMD: visual molecular dynamics. J. Mol. Graph. 1996;14:33–38. doi: 10.1016/0263-7855(96)00018-5. [DOI] [PubMed] [Google Scholar]
  • 78.Zhang S., Krieger J.M., Zhang Y., Kaya C., Kaynak B., Mikulska-Ruminska K., Doruker P., Li H., Bahar I. ProDy 2.0: increased scale and scope after 10 years of protein dynamics modelling with Python. Bioinformatics. 2021;37:3657–3659. doi: 10.1093/bioinformatics/btab187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Valdés-Tresanco M.S., Valdés-Tresanco M.E., Valiente P.A., Moreno E. gmx_MMPBSA: a new tool to perform end-state free energy calculations with GROMACS, J. Chem. Theor. Comput. 2021;17:6281–6291. doi: 10.1021/acs.jctc.1c00645. [DOI] [PubMed] [Google Scholar]
  • 80.Miller B.R., McGee T.D., Swails J.M., Homeyer N., Gohlke H., Roitberg A.E. MMPBSA.py : an efficient program for end-state free energy calculations. J. Chem. Theor. Comput. 2012;8:3314–3321. doi: 10.1021/ct300418h. [DOI] [PubMed] [Google Scholar]
  • 81.Upstream ORFs are prevalent translational repressors in vertebrates | the EMBO Journal, (n.d. https://www.embopress.org/doi/full/10.15252/embj.201592759 [DOI] [PMC free article] [PubMed]
  • 82.Vilela Rodrigues T.C., Jaiswal A.K., de Sarom A., de Castro Oliveira L., Freire Oliveira C.J., Ghosh P., Tiwari S., Miranda F.M., de Jesus Benevides L., Ariston de Carvalho Azevedo V., de Castro Soares S. Reverse vaccinology and subtractive genomics reveal new therapeutic targets against Mycoplasma pneumoniae : a causative agent of pneumonia. R. Soc. Open Sci. 2019;6 doi: 10.1098/rsos.190907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Dehury B., Behera S.K., Mahapatra N. Structural dynamics of Casein Kinase I (CKI) from malarial parasite Plasmodium falciparum (Isolate 3D7): insights from theoretical modelling and molecular simulations. J. Mol. Graph. Model. 2017;71:154–166. doi: 10.1016/j.jmgm.2016.11.012. [DOI] [PubMed] [Google Scholar]
  • 84.Isidro J., Borges V., Pinto M., Sobral D., Santos J.D., Nunes A., Mixão V., Ferreira R., Santos D., Duarte S., Vieira L., Borrego M.J., Núncio S., de Carvalho I.L., Pelerito A., Cordeiro R., Gomes J.P. Phylogenomic characterization and signs of microevolution in the 2022 multi-country outbreak of monkeypox virus. Nat. Med. 2022;28:1569–1572. doi: 10.1038/s41591-022-01907-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Happi C., Adetifa I., Mbala P., Njouom R., Nakoune E., Happi A., Ndodo N., Ayansola O., Mboowa G., Bedford T., Neher R.A., Roemer C., Hodcroft E., Tegally H., O'Toole A., Rambaut A., Pybus O., Kraemer M.U.G., Wilkinson E., Isidro J., Borges V., Pinto M., Gomes J.P., Freitas L., Resende P.C., Lee R.T.C., Maurer-Stroh S., Baxter C., Lessells R., Ogwell A.E., Kebede Y., Tessema S.K., de Oliveira T. Urgent need for a non-discriminatory and non-stigmatizing nomenclature for monkeypox virus. PLoS Biol. 2022;20 doi: 10.1371/journal.pbio.3001769. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Kozlov M. Monkeypox goes global: why scientists are on alert. Nature. 2022;606:15–16. doi: 10.1038/d41586-022-01421-8. [DOI] [PubMed] [Google Scholar]
  • 87.Mushegian AR K.E. Proc Natl Acad Sci USA. 1996. A minimal gene set for cellular life derived by comparison of complete bacterial genomes. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Elhefnawi M., Alaidi O., Mohamed N., Kamar M., El-Azab I., Zada S., Siam R. Identification of novel conserved functional motifs across most Influenza A viral strains. Virol. J. 2011;8 doi: 10.1186/1743-422X-8-44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Yan F., Gao F. A systematic strategy for the investigation of vaccines and drugs targeting bacteria. Comput. Struct. Biotechnol. J. 2020;18:1525–1538. doi: 10.1016/j.csbj.2020.06.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Shiragannavar S.S., Shettar A.K., Madagi S.B., Sarawad S. Subtractive genomics approach in identifying polysacharide biosynthesis protein as novel drug target against Eubacterium nodatum. Asian J. Pharm. Pharmacol. 2019;5:382–392. doi: 10.31024/ajpp.2019.5.2.24. [DOI] [Google Scholar]
  • 91.Hopkins A.L., Groom C.R. The druggable genome, Nat. Rev. Drug Discov. 2002;1:727–730. doi: 10.1038/nrd892. [DOI] [PubMed] [Google Scholar]
  • 92.Byrd C.M., Bolken T.C., Hruby D.E. The vaccinia virus I7L gene product is the core protein proteinase, J. Virol. 2002;76:8973–8976. doi: 10.1128/JVI.76.17.8973-8976.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Reed B., Yakovleva L., Shuman S., Ghose R. Characterization of DNA binding by the isolated N-terminal domain of vaccinia virus DNA topoisomerase IB. Biochemistry. 2017;56:3307–3317. doi: 10.1021/acs.biochem.7b00042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Gershon P.D., Moss B. Early transcription factor subunits are encoded by vaccinia virus late genes. Proc. Natl. Acad. Sci. 1990;87:4401–4405. doi: 10.1073/pnas.87.11.4401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Ishii K., Moss B. Role of vaccinia virus A20R protein in DNA replication: construction and characterization of temperature-sensitive mutants. J. Virol. 2001;75:1656–1663. doi: 10.1128/JVI.75.4.1656-1663.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Klemperer N., McDonald W., Boyle K., Unger B., Traktman P. The A20R protein is a stoichiometric component of the processive form of vaccinia virus DNA polymerase, J. Virol. 2001;75:12298–12307. doi: 10.1128/JVI.75.24.12298-12307.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Clark A.J., Tiwary P., Borrelli K., Feng S., Miller E.B., Abel R., Friesner R.A., Berne B.J. Prediction of protein-ligand binding poses via a combination of induced fit docking and metadynamics simulations. J. Chem. Theor. Comput. 2016;12:2990–2998. doi: 10.1021/acs.jctc.6b00201. [DOI] [PubMed] [Google Scholar]
  • 98.Levitzki A., Klein S. My journey from tyrosine phosphorylation inhibitors to targeted immune therapy as strategies to combat cancer. Proc. Natl. Acad. Sci. 2019;116:11579–11586. doi: 10.1073/pnas.1816012116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Ismail H.M., Barton V., Phanchana M., Charoensutthivarakul S., Wong M.H.L., Hemingway J., Biagini G.A., O'Neill P.M., Ward S.A. Artemisinin activity-based probes identify multiple molecular targets within the asexual stage of the malaria parasites Plasmodium falciparum 3D7. Proc. Natl. Acad. Sci. U. S. A. 2016;113:2080–2085. doi: 10.1073/pnas.1600459113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Tassini S., Sun L., Lanko K., Crespan E., Langron E., Falchi F., Kissova M., Armijos-Rivera J.I., Delang L., Mirabelli C., Neyts J., Pieroni M., Cavalli A., Costantino G., Maga G., Vergani P., Leyssen P., Radi M. Discovery of multitarget agents active as broad-spectrum antivirals and correctors of cystic fibrosis transmembrane conductance regulator for associated pulmonary diseases. J. Med. Chem. 2017;60:1400–1416. doi: 10.1021/acs.jmedchem.6b01521. [DOI] [PubMed] [Google Scholar]
  • 101.Ambery C., Riddell K., Daley-Yates P. Open-label, randomized, 6-way crossover, single-dose study to determine the pharmacokinetics of batefenterol (GSK961081) and fluticasone furoate when administered alone or in combination. Clin. Pharmacol. Drug Dev. 2016;5:399–407. doi: 10.1002/cpdd.274. [DOI] [PubMed] [Google Scholar]
  • 102.Hughes A.D., Jones L.H. Dual-pharmacology muscarinic antagonist and β 2 agonist molecules for the treatment of chronic obstructive pulmonary disease. Future Med. Chem. 2011;3:1585–1605. doi: 10.4155/fmc.11.106. [DOI] [PubMed] [Google Scholar]
  • 103.Setia G., Hagog N., Jalilizeinali B., Funkhouser S., Pierzchanowski L., Lan F., Gabig T.G., Kiner-Strachan B., Kelleher K., Hsu M.-C., Chang L.-W., Schuster M.W., A Phase Open-label pilot study to evaluate the hematopoietic stem cell mobilization of TG-0054 combined with G-CSF in 12 patients with multiple myeloma, non-hodgkin lymphoma or Hodgkin lymphoma - an interim analysis. Blood. 2015;126 doi: 10.1182/blood.V126.23.515.515. 515–515. [DOI] [Google Scholar]
  • 104.Lembo A.J., Lacy B.E., Zuckerman M.J., Schey R., Dove L.S., Andrae D.A., Davenport J.M., McIntyre G., Lopez R., Turner L., Covington P.S. Eluxadoline for irritable bowel syndrome with diarrhea, N. Engl. J. Med. 2016;374:242–253. doi: 10.1056/NEJMoa1505180. [DOI] [PubMed] [Google Scholar]
  • 105.Crim C., Gotfried M., Spangenthal S., Watkins M., Emmett A., Crawford C., Baidoo C., Castro-Santamaria R. A randomized, controlled, repeat-dose study of batefenterol/fluticasone furoate compared with placebo in the treatment of COPD. BMC Pulm. Med. 2020;119(20) doi: 10.1186/s12890-020-1153-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Brenner D.M., Sayuk G.S., Gutman C.R., Jo E., Elmes S.J.R., Liu L.W.C., Cash B.D. Efficacy and safety of eluxadoline in patients with irritable bowel syndrome with diarrhea who report inadequate symptom control with loperamide: RELIEF phase 4 study. Am. J. Gastroenterol. 2019;114:1502–1511. doi: 10.14309/ajg.0000000000000327. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Multimedia component 1
mmc1.docx (6.1MB, docx)
Multimedia component 2
mmc2.xlsx (31.5KB, xlsx)
Multimedia component 3
mmc3.xlsx (20.1KB, xlsx)
Multimedia component 4
mmc4.xlsx (119.2KB, xlsx)

Articles from Computers in Biology and Medicine are provided here courtesy of Elsevier

RESOURCES