1
|
Berselli A, Menziani MC, Muniz-Miranda F. Structure and Energetics of PET-Hydrolyzing Enzyme Complexes: A Systematic Comparison from Molecular Dynamics Simulations. J Chem Inf Model 2024; 64:8236-8257. [PMID: 39432831 DOI: 10.1021/acs.jcim.4c01369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
Discovered in 2016, the enzyme PETase, secreted by bacterial Ideonella Sakaiensis 201-F6, has an excellent hydrolytic activity toward poly(ethylene terephthalate) (PET) at room temperature, while it decreases at higher temperatures due to the low thermostability. Many variants have been engineered to overcome this limitation, which hinders industrial application. In this work, we systematically compare PETase wild-type (WT) and four mutants (DuraPETase, ThermoPETase, FastPETase, and HotPETase) using standard molecular dynamics (MD) simulations and unbinding free energy calculations. In particular, we analyze the enzymes' structural characteristics and binding to a tetrameric PET chain (PET4) under two temperature conditions: T1─300 K and T2─350 K. Our results indicate that (i) PET4 forms stable complexes with the five enzymes at room temperature (∼300 K) and (ii) most of the interactions are localized close to the active site of the protein, where the W185 and Y87 residues interact with the aromatic rings of the substrate. Specifically, (iii) the W185 side-chain explores different conformations in each variant (a phenomenon known in the literature as "W185 wobbling"). This suggests that the binding pocket retains structural plasticity and flexibility among the variants, facilitating substrate recognition and localization events at moderate temperatures. Moreover, (iv) PET4 establishes aromatic interactions with the catalytic H237 residue, stabilizing the catalytic triad composed of residues S160-H237-D206, and helping the system achieve an effective configuration for the hydrolysis reaction. Conversely, (v) the binding affinity decreases at a higher temperature (∼350 K), retaining moderate interactions only for HotPETase. Finally, (vi) MD simulations of complexes formed with poly(ethylene-2,5-furan dicarboxylate) (PEF) show no persistent interactions, suggesting that these enzymes are not yet optimized for binding this alternative semiaromatic plastic polymer. Our study offers valuable insights into the structural stability of these enzymes and the molecular determinants driving PET binding onto their surfaces, sheds light on the mechanistic steps that precede the onset of hydrolysis, and provides a foundation for future enzyme optimization.
Collapse
Affiliation(s)
- Alessandro Berselli
- Department of Chemical and Geological Sciences (DSCG), University of Modena and Reggio Emilia (UNIMORE), Via Campi 103, 41125 Modena, Italy
| | - Maria Cristina Menziani
- Department of Chemical and Geological Sciences (DSCG), University of Modena and Reggio Emilia (UNIMORE), Via Campi 103, 41125 Modena, Italy
| | - Francesco Muniz-Miranda
- Department of Chemical and Geological Sciences (DSCG), University of Modena and Reggio Emilia (UNIMORE), Via Campi 103, 41125 Modena, Italy
| |
Collapse
|
2
|
Yuce M, Ates B, Yasar NI, Sungur FA, Kurkcuoglu O. A computational workflow to determine drug candidates alternative to aminoglycosides targeting the decoding center of E. coli ribosome. J Mol Graph Model 2024; 131:108817. [PMID: 38976944 DOI: 10.1016/j.jmgm.2024.108817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/08/2024] [Accepted: 07/03/2024] [Indexed: 07/10/2024]
Abstract
The global antibiotic resistance problem necessitates fast and effective approaches to finding novel inhibitors to treat bacterial infections. In this study, we propose a computational workflow to identify plausible high-affinity compounds from FDA-approved, investigational, and experimental libraries for the decoding center on the small subunit 30S of the E. coli ribosome. The workflow basically consists of two molecular docking calculations on the intact 30S, followed by molecular dynamics (MD) simulations coupled with MM-GBSA calculations on a truncated ribosome structure. The parameters used in the molecular docking suits, Glide and AutoDock Vina, as well as in the MD simulations with Desmond were carefully adjusted to obtain expected interactions for the ligand-rRNA complexes. A filtering procedure was followed, considering a fingerprint based on aminoglycoside's binding site on the 30S to obtain seven hit compounds either with different clinical usages or aminoglycoside derivatives under investigation, suggested for in vitro studies. The detailed workflow developed in this study promises an effective and fast approach for the estimation of binding free energies of large protein-RNA and ligand complexes.
Collapse
Affiliation(s)
- Merve Yuce
- Istanbul Technical University, Department of Chemical Engineering, Istanbul, 34469, Turkey.
| | - Beril Ates
- Istanbul Technical University, Department of Chemical Engineering, Istanbul, 34469, Turkey.
| | - Nesrin Isil Yasar
- Istanbul Technical University, Computational Science and Engineering Division, Informatics Institute, Istanbul, 34469, Turkey.
| | - Fethiye Aylin Sungur
- Istanbul Technical University, Computational Science and Engineering Division, Informatics Institute, Istanbul, 34469, Turkey.
| | - Ozge Kurkcuoglu
- Istanbul Technical University, Department of Chemical Engineering, Istanbul, 34469, Turkey.
| |
Collapse
|
3
|
Simon JP, Dong S. In-silico screening of missense nsSNPs in Delta-opioid receptor protein and their restoring tendency on MCRT interaction; focusing on dynamic nature. Int J Biol Macromol 2024; 275:133710. [PMID: 38977046 DOI: 10.1016/j.ijbiomac.2024.133710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/30/2024] [Accepted: 07/05/2024] [Indexed: 07/10/2024]
Abstract
Delta-opioid receptor protein (OPRD1) is one of the potential targets for treating pain. The presently available opioid agonists are known to cause unnecessary side effects. To discover a novel opioid agonist, our research group has synthesized a chimeric peptide MCRT and proved its potential activity through in vivo analysis. Non-synonymous SNPs (nsSNPs) missense mutations affect the functionality and stability of proteins leading to diseases. The current research was focused on understanding the role of MCRT in restoring the binding tendency of OPRD1 nsSNPs missense mutations on dynamic nature in comparison with Deltorphin-II and morphiceptin. The deleterious effects of nsSNPs were analyzed using various bioinformatics tools for predicting structural, functional, and oncogenic influence. The shortlisted nine nsSNPs were predicted for allergic reactions, domain changes, post-translation modification, multiple sequence alignment, secondary structure, molecular dynamic simulation (MDS), and peptide docking influence. Further, the docked complex of three shortlisted deleterious nsSNPs was analyzed using an MDS study, and the highly deleterious shortlisted nsSNP A149T was further analyzed for higher trajectory analysis. MCRT restored the binding tendency influence caused by nsSNPs on the dynamics of stability, functionality, binding affinity, secondary structure, residues connection, motion, and folding of OPRD1 protein.
Collapse
Affiliation(s)
- Jerine Peter Simon
- Department of Animal and Biomedical Sciences, School of Life Sciences, Lanzhou University, 222 Tianshui South Road, Lanzhou 730000, China
| | - Shouliang Dong
- Department of Animal and Biomedical Sciences, School of Life Sciences, Lanzhou University, 222 Tianshui South Road, Lanzhou 730000, China,; Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, 222 Tianshui South Road, Lanzhou 730000, China.
| |
Collapse
|
4
|
Lim CP, Leow CH, Lim HT, Kok BH, Chuah C, Oliveira JIN, Jones M, Leow CY. Insights into structural vaccinology harnessed for universal coronavirus vaccine development. Clin Exp Vaccine Res 2024; 13:202-217. [PMID: 39144127 PMCID: PMC11319108 DOI: 10.7774/cevr.2024.13.3.202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 05/10/2024] [Accepted: 05/15/2024] [Indexed: 08/16/2024] Open
Abstract
Structural vaccinology is pivotal in expediting vaccine design through high-throughput screening of immunogenic antigens. Leveraging the structural and functional characteristics of antigens and immune cell receptors, this approach employs protein structural comparison to identify conserved patterns in key pathogenic components. Molecular modeling techniques, including homology modeling and molecular docking, analyze specific three-dimensional (3D) structures and protein interactions and offer valuable insights into the 3D interactions and binding affinity between vaccine candidates and target proteins. In this review, we delve into the utilization of various immunoinformatics and molecular modeling tools to streamline the development of broad-protective vaccines against coronavirus disease 2019 variants. Structural vaccinology significantly enhances our understanding of molecular interactions between hosts and pathogens. By accelerating the pace of developing effective and targeted vaccines, particularly against the rapidly mutating severe acute respiratory syndrome coronavirus 2 and other prevalent infectious diseases, this approach stands at the forefront of advancing immunization strategies. The combination of computational techniques and structural insights not only facilitates the identification of potential vaccine candidates but also contributes to the rational design of vaccines, fostering a more efficient and targeted approach to combatting infectious diseases.
Collapse
Affiliation(s)
- Chin Peng Lim
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Gelugor, Malaysia
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Gelugor, Malaysia
| | - Chiuan Herng Leow
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Gelugor, Malaysia
| | - Hui Ting Lim
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Gelugor, Malaysia
| | - Boon Hui Kok
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Gelugor, Malaysia
| | - Candy Chuah
- Faculty of Medicine, Asian Institute of Medical Science and Technology University, Bedong, Malaysia
| | - Jonas Ivan Nobre Oliveira
- Department of Biophysics and Pharmacology, Bioscience Center, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Malcolm Jones
- School of Veterinary Science, The University of Queensland, Gatton, Australia
| | - Chiuan Yee Leow
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Gelugor, Malaysia
| |
Collapse
|
5
|
Sahu D, Gupta C, Yennamalli RM, Sharma S, Roy S, Hasan S, Gupta P, Sharma VK, Kashyap S, Kumar S, Dwivedi VP, Zhao X, Panda AK, Das HR, Liu CJ. Novel peptide inhibitor of human tumor necrosis factor-α has antiarthritic activity. Sci Rep 2024; 14:12935. [PMID: 38839973 PMCID: PMC11153517 DOI: 10.1038/s41598-024-63790-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 06/03/2024] [Indexed: 06/07/2024] Open
Abstract
The inhibition of tumor necrosis factor (TNF)-α trimer formation renders it inactive for binding to its receptors, thus mitigating the vicious cycle of inflammation. We designed a peptide (PIYLGGVFQ) that simulates a sequence strand of human TNFα monomer using a series of in silico methods, such as active site finding (Acsite), protein-protein interaction (PPI), docking studies (GOLD and Flex-X) followed by molecular dynamics (MD) simulation studies. The MD studies confirmed the intermolecular interaction of the peptide with the TNFα. Fluorescence-activated cell sorting and fluorescence microscopy revealed that the peptide effectively inhibited the binding of TNF to the cell surface receptors. The cell culture assays showed that the peptide significantly inhibited the TNFα-mediated cell death. In addition, the nuclear translocation of the nuclear factor kappa B (NFκB) was significantly suppressed in the peptide-treated A549 cells, as observed in immunofluorescence and gel mobility-shift assays. Furthermore, the peptide protected against joint damage in the collagen-induced arthritis (CIA) mouse model, as revealed in the micro focal-CT scans. In conclusion, this TNFα antagonist would be helpful for the prevention and repair of inflammatory bone destruction and subsequent loss in the mouse model of CIA as well as human rheumatoid arthritis (RA) patients. This calls upon further clinical investigation to utilize its potential effect as an antiarthritic drug.
Collapse
Affiliation(s)
- Debasis Sahu
- Product Development Cell, National Institute of Immunology, New Delhi, India.
- Department of Orthopedics Surgery, New York University School of Medicine, New York, NY, USA.
- Science Habitat, Ubioquitos Inc, London, ON, Canada.
| | - Charu Gupta
- School of Biomedical Sciences, Galgotias University, Greater Noida, UP, India
- CSIR-Institute of Genomics and Integrative Biology, Delhi, India
| | - Ragothaman M Yennamalli
- Department of Bioinformatics, School of Chemical and Biotechnology, SASTRA Deemed to Be University, Thanjavur, Tamil Nadu, India
| | - Shikha Sharma
- Amity Institute of Forensic Sciences, Amity University, Noida, Uttar Pradesh, India
- Science Habitat, Ubioquitos Inc, London, ON, Canada
| | - Saugata Roy
- CSIR-Institute of Genomics and Integrative Biology, Delhi, India
| | - Sadaf Hasan
- Department of Orthopedics Surgery, New York University School of Medicine, New York, NY, USA
| | - Pawan Gupta
- Department of Pharmaceutical Chemistry, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, Maharashtra, India
| | - Vishnu Kumar Sharma
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research (NIPER), Mohali, Punjab, India
| | - Sujit Kashyap
- Division of Pediatric Rheumatology, University of California San Francisco, San Francisco, CA, USA
- Department of Genetics, University of Delhi, Delhi, India
| | - Santosh Kumar
- Immunobiology Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Ved Prakash Dwivedi
- Immunobiology Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Xiangli Zhao
- Department of Orthopedics Surgery, New York University School of Medicine, New York, NY, USA
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA
| | - Amulya Kumar Panda
- Product Development Cell, National Institute of Immunology, New Delhi, India
| | - Hasi Rani Das
- CSIR-Institute of Genomics and Integrative Biology, Delhi, India
| | - Chuan-Ju Liu
- Department of Orthopedics Surgery, New York University School of Medicine, New York, NY, USA
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
6
|
Yasmin S, Ansari MY, Pandey K, Dikhit MR. Identification of potential vaccine targets for elicitation of host immune cells against SARS-CoV-2 by reverse vaccinology approach. Int J Biol Macromol 2024; 265:130754. [PMID: 38508555 DOI: 10.1016/j.ijbiomac.2024.130754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 03/06/2024] [Accepted: 03/07/2024] [Indexed: 03/22/2024]
Abstract
The COVID-19 pandemic has emerged as a critical global health crisis, demanding urgent and effective strategies for containment. While some knowledge exists about epitope sequences recognized by human immune cells and their activation of CD8+ T cells within the HLA context, comprehensive information remains limited. This study employs reverse vaccinology to explore antigenic HLA-restricted T-cell epitopes capable of eliciting durable immunity. Screening reveals 187 consensus epitopes, with 23 offering broad population coverage worldwide, spanning over 5000 HLA alleles. Sequence alignment analysis highlights the genetic distinctiveness of these peptides from Homo sapiens and their intermediate to high TAP binding efficiency. Notably, these epitopes share 100 % sequence identity across strains from nine countries, indicating potential for a uniform protective immune response among diverse ethnic populations. Docking simulations further confirm their binding capacity with the HLA allele, validating them as promising targets for SARS-CoV-2 immune recognition. The anticipated epitopes are connected with suitable linkers and adjuvant, and then assessed for its translational efficacy within a bacterial expression vector through computational cloning. Through docking, it is observed that the chimeric vaccine construct forms lasting hydrogen bonds with Toll-like receptor (TLR4), while immune simulation illustrates an increased cytotoxic response aimed at CD8+ T cells. This comprehensive computational analysis suggests the chimeric vaccine construct's potential to provoke a robust immune response against SARS-CoV-2. By delineating these antigenic fragments, our study offers valuable insights into effective vaccine and immunotherapy development against COVID-19, contributing significantly to global efforts in combating this infectious threat.
Collapse
Affiliation(s)
- Sabina Yasmin
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University (KKU), Abha 62529, Saudi Arabia
| | - Mohammad Yousuf Ansari
- Department of Pharmaceutical Chemistry, M.M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala 133207, India.
| | - Krishna Pandey
- Department of Clinical Medicine, ICMR-Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna 800007, India
| | - Manas Ranjan Dikhit
- Department of Bioinformatics, ICMR-Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna 800007, India.
| |
Collapse
|
7
|
Baammi S, El Allali A, Daoud R. Unleashing Nature's potential: a computational approach to discovering novel VEGFR-2 inhibitors from African natural compound using virtual screening, ADMET analysis, molecular dynamics, and MMPBSA calculations. Front Mol Biosci 2023; 10:1227643. [PMID: 37800126 PMCID: PMC10548200 DOI: 10.3389/fmolb.2023.1227643] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/07/2023] [Indexed: 10/07/2023] Open
Abstract
One of the characteristic features of cancer is angiogenesis, the process by which new, aberrant blood vessels are formed from pre-existing blood vessels. The process of angiogenesis begins when VEGF binds to its receptor, the VEGF receptor (VEGFR). The formation of new blood vessels provides nutrients that can promote the growth of cancer cells. When it comes to new blood vessel formation, VEGFR2 is a critical player. Therefore, inhibiting VEGFR2 is an effective way to target angiogenesis in cancer treatment. The aim of our research was to find new VEGFR-2 inhibitors by performing a virtual screening of 13313 from African natural compounds using different in silico techniques. Using molecular docking calculations and ADMET properties, we identified four compounds that exhibited a binding affinity ranging from -11.0 kcal/mol to -11.5 Kcal/mol when bound to VEGFR-2. These four compounds were further analyzed with 100 ns simulations to determine their stability and binding energy using the MM-PBSA method. After comparing the compounds with Regorafenib, a drug approved for anti-angiogenesis treatment, it was found that all the candidates (EANPDB 252, NANPDB 4577, and NANPDB 4580), with the exception of EANPDB 76, could target VEGFR-2 similarly effectively to Regorafenib. Therefore, we recommend three of these agents for anti-angiogenesis treatment because they are likely to deactivate VEGFR-2 and thus inhibit angiogenesis. However, it should be noted that the safety and suitability of these agents for clinical use needs further investigation, as the computer-assisted study did not include in vitro or in vivo experiments.
Collapse
Affiliation(s)
- Soukayna Baammi
- African Genome Center, Mohammed VI Polytechnic University, Ben Guerir, Morocco
| | - Achraf El Allali
- African Genome Center, Mohammed VI Polytechnic University, Ben Guerir, Morocco
| | - Rachid Daoud
- Chemical and Biochemical Sciences-Green Processing Engineering, Mohammed VI Polytechnic University, Ben Guerir, Morocco
| |
Collapse
|
8
|
Upadhyay A, Ekenna C. A New Tool to Study the Binding Behavior of Intrinsically Disordered Proteins. Int J Mol Sci 2023; 24:11785. [PMID: 37511544 PMCID: PMC10380747 DOI: 10.3390/ijms241411785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/07/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
Understanding the binding behavior and conformational dynamics of intrinsically disordered proteins (IDPs) is crucial for unraveling their regulatory roles in biological processes. However, their lack of stable 3D structures poses challenges for analysis. To address this, we propose an algorithm that explores IDP binding behavior with protein complexes by extracting topological and geometric features from the protein surface model. Our algorithm identifies a geometrically favorable binding pose for the IDP and plans a feasible trajectory to evaluate its transition to the docking position. We focus on IDPs from Homo sapiens and Mus-musculus, investigating their interaction with the Plasmodium falciparum (PF) pathogen associated with malaria-related deaths. We compare our algorithm with HawkDock and HDOCK docking tools for quantitative (computation time) and qualitative (binding affinity) measures. Our results indicated that our method outperformed the compared methods in computation performance and binding affinity in experimental conformations.
Collapse
Affiliation(s)
- Aakriti Upadhyay
- Department of Computer Science, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, USA
| | - Chinwe Ekenna
- Department of Computer Science, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, USA
| |
Collapse
|
9
|
Kamga Youmbi FI, Kengne Tchendji V, Tayou Djamegni C. P-FARFAR2: A multithreaded greedy approach to sampling low-energy RNA structures in Rosetta FARFAR2. Comput Biol Chem 2023; 104:107878. [PMID: 37167861 DOI: 10.1016/j.compbiolchem.2023.107878] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 04/23/2023] [Accepted: 05/01/2023] [Indexed: 05/13/2023]
Abstract
RNA (ribonucleic acid) structure prediction finds many applications in health science and drug discovery due to its importance in several life regulatory processes. But despite significant advances in the close field of protein prediction, RNA 3D structure still poses a tremendous challenge to predict, especially for large sequences. In this regard, the approach unfolded by Rosetta FARFAR2 (Fragment Assembly of RNA with Full-Atom Refinement, version 2) has shown promising results, but the algorithm is non-deterministic by nature. In this paper, we develop P-FARFAR2: a parallel enhancement of FARFAR2 that increases its ability to assemble low-energy structures via multithreaded exploration of random configurations in a greedy manner. This strategy, appearing in the literature under the term "parallel mechanism", is made viable through two measures: first, the synchronization window is coarsened to several Monte Carlo cycles; second, all but one of the threads are differentiated as auxiliary and set to perform a weakened version of the problem. Following empirical analysis on a diverse range of RNA structures, we report achieving statistical significance in lowering the energy levels of ensuing samples. And consequently, despite the moderate-to-weak correlation between energy levels and prediction accuracy, this achievement happens to propagate to accuracy measurements.
Collapse
Affiliation(s)
| | - Vianney Kengne Tchendji
- Department of Mathematics and Computer Science, University of Dschang, PO Box 67, Dschang, Cameroon.
| | - Clémentin Tayou Djamegni
- Department of Mathematics and Computer Science, University of Dschang, PO Box 67, Dschang, Cameroon; Department of Computer Engineering, University of Dschang, PO Box 134, Bandjoun, Cameroon.
| |
Collapse
|
10
|
Agarwal R, Smith JC. Speed vs Accuracy: Effect on Ligand Pose Accuracy of Varying Box Size and Exhaustiveness in AutoDock Vina. Mol Inform 2023; 42:e2200188. [PMID: 36262028 DOI: 10.1002/minf.202200188] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 10/19/2022] [Indexed: 11/11/2022]
Abstract
Structure-based virtual high-throughput screening involves docking chemical libraries to targets of interest. A parameter pertinent to the accuracy of the resulting pose is the root mean square deviation (RMSD) from a known crystallographic structure, i. e., the 'docking power'. Here, using a popular algorithm, Autodock Vina, as a model program, we evaluate the effects of varying two common docking parameters: the box size (the size of docking search space) and the exhaustiveness of the global search (the number of independent runs starting from random ligand conformations) on the RMSD from the PDBbind v2017 refined dataset of experimental protein-ligand complexes. Although it is clear that exhaustiveness is an important parameter, there is wide variation in the values used, with variation between 1 and >100. We, therefore, evaluated a combination of cubic boxes of different sizes and five exhaustiveness values (1, 8, 25, 50, 75, 100) within the range of those commonly adopted. The results show that the default exhaustiveness value of 8 performs well overall for most box sizes. In contrast, for all box sizes, but particularly for large boxes, an exhaustiveness value of 1 led to significantly higher median RMSD (mRMSD) values. The docking power was slightly improved with an exhaustiveness of 25, but the mRMSD changes little with values higher than 25. Therefore, although low exhaustiveness is computationally faster, the results are more likely to be far from reality, and, conversely, values >25 led to little improvement at the expense of computational resources. Overall, we recommend users to use at least the default exhaustiveness value of 8 for virtual screening calculations.
Collapse
Affiliation(s)
- Rupesh Agarwal
- UT/ORNL Center for Molecular Biophysics, Oak Ridge National Laboratory, Oak Ridge, TN 37831-6309, USA.,Department of Biochemistry and Cellular and Molecular Biology, University of Tennessee, Knoxville, 14311 Cumberland Avenue, Knoxville, TN 37996-1939, USA
| | - Jeremy C Smith
- UT/ORNL Center for Molecular Biophysics, Oak Ridge National Laboratory, Oak Ridge, TN 37831-6309, USA.,Department of Biochemistry and Cellular and Molecular Biology, University of Tennessee, Knoxville, 14311 Cumberland Avenue, Knoxville, TN 37996-1939, USA
| |
Collapse
|
11
|
Prakash R, Goodlett DW, Varghese S, Andrys J, Gbadamosi FA, Arriaza RH, Patel M, Tiwari PB, Borowski T, Chruszcz M, Shimizu LS, Upadhyay G. Development of fluorophore labeled or biotinylated anticancer small molecule NSC243928. Bioorg Med Chem 2023; 79:117171. [PMID: 36680947 PMCID: PMC9892358 DOI: 10.1016/j.bmc.2023.117171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 12/23/2022] [Accepted: 01/09/2023] [Indexed: 01/15/2023]
Abstract
Small molecule NSC243928 binds with LY6K, a potential target for the treatment of triple-negative breast cancer, and induces cancer cell death with an unclear mechanism. We have developed chemical tools to identify the molecular mechanisms of NSC243928-LY6K interaction. Herein, we report on the development and synthesis of biotinylated and fluorophore-tethered derivatives of NSC243928 guided by docking studies and molecular dynamics. Surface plasmon resonance assay indicates that these derivatives retained a direct binding with LY6K protein. Confocal analysis revealed that nitrobenzoxadiazole (NBD) fluorophore tagged NSC243928 is retained in LY6K expressing cancer cells. These novel modified compounds will be employed in future in vitro and in vivo studies to understand the molecular mechanisms of NSC243928 mediated cancer cell death. These studies will pave the path for developing novel targeted therapeutics and understanding any potential side-effects of these treatments for hard-to-treat cancers such as triple-negative breast cancer or other cancers with high expression of LY6K.
Collapse
Affiliation(s)
- Rahul Prakash
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC 29208, USA
| | - Dustin W Goodlett
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC 29208, USA
| | - Sheelu Varghese
- Henry M. Jackson Foundation, Bethesda, MD, USA; Department of Pathology, Uniformed Services University, Bethesda, MD, USA
| | - Justyna Andrys
- Jerzy Haber Institute of Catalysis and Surface Chemistry, Polish Academy of Science, Niezapominajek 8, Krakow 30-239, Poland
| | - Fahidat A Gbadamosi
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC 29208, USA
| | - Ricardo H Arriaza
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC 29208, USA
| | - Megha Patel
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC 29208, USA
| | - Purushottam B Tiwari
- Department of Oncology, Georgetown University Medical Center, Washington, DC, USA
| | - Tomasz Borowski
- Jerzy Haber Institute of Catalysis and Surface Chemistry, Polish Academy of Science, Niezapominajek 8, Krakow 30-239, Poland
| | - Maksymilian Chruszcz
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC 29208, USA
| | - Linda S Shimizu
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC 29208, USA
| | - Geeta Upadhyay
- John P. Murtha Cancer Center, Bethesda, MD, USA; Department of Pathology, Uniformed Services University, Bethesda, MD, USA; Department of Oncology, Georgetown University Medical Center, Washington, DC, USA.
| |
Collapse
|
12
|
Miah MM, Tabassum N, Afroj Zinnia M, Islam ABMMK. Drug and Anti-Viral Peptide Design to Inhibit the Monkeypox Virus by Restricting A36R Protein. Bioinform Biol Insights 2022; 16:11779322221141164. [PMID: 36570327 PMCID: PMC9772960 DOI: 10.1177/11779322221141164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 11/06/2022] [Indexed: 12/24/2022] Open
Abstract
Most recently, monkeypox virus (MPXV) has emanated as a global public health threat. Unavailability of effective medicament against MPXV escalates demand for new therapeutic agent. In this study, in silico strategies were conducted to identify novel drug against the A36R protein of MPXV. The A36R protein of MPXV is responsible for the viral migration, adhesion, and vesicle trafficking to the host cell. To block the A36R protein, 4893 potential antiviral peptides (AVPs) were retrieved from DRAMP and SATPdb databases. Finally, 57 sequences were screened based on peptide filtering criteria, which were then modeled. Likewise, 31 monkeypox virus A36R protein sequences were collected from NCBI protein database to find consensus sequence and to predict 3D protein model. The refined and validated models of the A36R protein and AVP peptides were used to predict receptor-ligand interactions using DINC 2 server. Three peptides that showed best interactions were SATPdb10193, SATPdb21850, and SATPdb26811 with binding energies -6.10, -6.10, and -6.30 kcal/mol, respectively. Small molecules from drug databases were also used to perform virtual screening against the A36R protein. Among databases, Enamine-HTSC showed strong affinity with docking scores ranging from -8.8 to 9.8 kcal/mol. Interaction of target protein A36R with the top 3 peptides and the most probable drug (Z55287118) examined by molecular dynamic (MD) simulation. Trajectory analyses (RMSD, RMSF, SASA, and Rg) confirmed the stable nature of protein-ligand and protein-peptide complexes. This work suggests that identified top AVPs and small molecules might interfere with the function of the A36R protein of MPXV.
Collapse
Affiliation(s)
| | - Nuzhat Tabassum
- Department of Pharmacy, East West University, Dhaka, Bangladesh
| | | | - Abul Bashar Mir Md. Khademul Islam
- Department of Genetic Engineering & Biotechnology, University of Dhaka, Dhaka, Bangladesh,Abul Bashar Mir Md. Khademul Islam, Department of Genetic Engineering and Biotechnology, University of Dhaka, Nilkhet Rd, Dhaka 1000, Bangladesh.
| |
Collapse
|
13
|
Vij S, Thakur R, Rishi P. Reverse engineering approach: a step towards a new era of vaccinology with special reference to Salmonella. Expert Rev Vaccines 2022; 21:1763-1785. [PMID: 36408592 DOI: 10.1080/14760584.2022.2148661] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Salmonella is responsible for causing enteric fever, septicemia, and gastroenteritis in humans. Due to high disease burden and emergence of multi- and extensively drug-resistant Salmonella strains, it is becoming difficult to treat the infection with existing battery of antibiotics as we are not able to discover newer antibiotics at the same pace at which the pathogens are acquiring resistance. Though vaccines against Salmonella are available commercially, they have limited efficacy. Advancements in genome sequencing technologies and immunoinformatics approaches have solved the problem significantly by giving rise to a new era of vaccine designing, i.e. 'Reverse engineering.' Reverse engineering/vaccinology has expedited the vaccine identification process. Using this approach, multiple potential proteins/epitopes can be identified and constructed as a single entity to tackle enteric fever. AREAS COVERED This review provides details of reverse engineering approach and discusses various protein and epitope-based vaccine candidates identified using this approach against typhoidal Salmonella. EXPERT OPINION Reverse engineering approach holds great promise for developing strategies to tackle the pathogen(s) by overcoming the limitations posed by existing vaccines. Progressive advancements in the arena of reverse vaccinology, structural biology, and systems biology combined with an improved understanding of host-pathogen interactions are essential components to design new-generation vaccines.
Collapse
Affiliation(s)
- Shania Vij
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Reena Thakur
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Praveen Rishi
- Department of Microbiology, Panjab University, Chandigarh, India
| |
Collapse
|
14
|
Albumin/Thiacalix[4]arene Nanoparticles as Potential Therapeutic Systems: Role of the Macrocycle for Stabilization of Monomeric Protein and Self-Assembly with Ciprofloxacin. Int J Mol Sci 2022; 23:ijms231710040. [PMID: 36077448 PMCID: PMC9455997 DOI: 10.3390/ijms231710040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/23/2022] [Accepted: 08/30/2022] [Indexed: 11/17/2022] Open
Abstract
The therapeutic application of serum albumin is determined by the relative content of the monomeric form compared to dimers, tetramers, hexamers, etc. In this paper, we propose and develop an approach to synthesize the cone stereoisomer of p-tert-butylthiacalix[4]arene with sulfobetaine fragments stabilization of monomeric bovine serum albumin and preventing aggregation. Spectral methods (UV-vis, CD, fluorescent spectroscopy, and dynamic light scattering) established the influence of the synthesized compounds on the content of monomeric and aggregated forms of BSA even without the formation of stable thiacalixarene/protein associates. The effect of thiacalixarenes on the efficiency of protein binding with the antibiotic ciprofloxacin was shown by fluorescence spectroscopy. The binding constant increases in the presence of the macrocycles, likely due to the stabilization of monomeric forms of BSA. Our study clearly shows the potential of this macrocycle design as a platform for the development of the fundamentally new approaches for preventing aggregation.
Collapse
|
15
|
Li C, Sun J, Li LW, Wu X, Palade V. An Effective Swarm Intelligence Optimization Algorithm for Flexible Ligand Docking. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2022; 19:2672-2684. [PMID: 34375285 DOI: 10.1109/tcbb.2021.3103777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
In general, flexible ligand docking is used for docking simulations under the premise that the position of the binding site is already known, and meanwhile it can also be used without prior knowledge of the binding site. However, most of the optimization search algorithms used in popular docking software are far from being ideal in the first case, and they can hardly be directly utilized for the latter case due to the relatively large search area. In order to design an algorithm that can flexibly adapt to different sizes of the search area, we propose an effective swarm intelligence optimization algorithm in this paper, called diversity-controlled Lamarckian quantum particle swarm optimization (DCL-QPSO). The highlights of the algorithm are a diversity-controlled strategy and a modified local search method. Integrated with the docking environment of Autodock, the DCL-QPSO is compared with Autodock Vina, Glide and other two Autodock-based search algorithms for flexible ligand docking. Experimental results revealed that the proposed algorithm has a performance comparable to those of Autodock Vina and Glide for dockings within a certain area around the binding sites, and is a more effective solver than all the compared methods for dockings without prior knowledge of the binding sites.
Collapse
|
16
|
Conev A, Devaurs D, Rigo MM, Antunes DA, Kavraki LE. 3pHLA-score improves structure-based peptide-HLA binding affinity prediction. Sci Rep 2022; 12:10749. [PMID: 35750701 PMCID: PMC9232595 DOI: 10.1038/s41598-022-14526-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 06/08/2022] [Indexed: 12/30/2022] Open
Abstract
Binding of peptides to Human Leukocyte Antigen (HLA) receptors is a prerequisite for triggering immune response. Estimating peptide-HLA (pHLA) binding is crucial for peptide vaccine target identification and epitope discovery pipelines. Computational methods for binding affinity prediction can accelerate these pipelines. Currently, most of those computational methods rely exclusively on sequence-based data, which leads to inherent limitations. Recent studies have shown that structure-based data can address some of these limitations. In this work we propose a novel machine learning (ML) structure-based protocol to predict binding affinity of peptides to HLA receptors. For that, we engineer the input features for ML models by decoupling energy contributions at different residue positions in peptides, which leads to our novel per-peptide-position protocol. Using Rosetta's ref2015 scoring function as a baseline we use this protocol to develop 3pHLA-score. Our per-peptide-position protocol outperforms the standard training protocol and leads to an increase from 0.82 to 0.99 of the area under the precision-recall curve. 3pHLA-score outperforms widely used scoring functions (AutoDock4, Vina, Dope, Vinardo, FoldX, GradDock) in a structural virtual screening task. Overall, this work brings structure-based methods one step closer to epitope discovery pipelines and could help advance the development of cancer and viral vaccines.
Collapse
Affiliation(s)
- Anja Conev
- grid.21940.3e0000 0004 1936 8278Department of Computer Science, Rice University, Houston, 77005 USA
| | - Didier Devaurs
- grid.4305.20000 0004 1936 7988MRC Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU UK
| | - Mauricio Menegatti Rigo
- grid.21940.3e0000 0004 1936 8278Department of Computer Science, Rice University, Houston, 77005 USA
| | | | - Lydia E. Kavraki
- grid.21940.3e0000 0004 1936 8278Department of Computer Science, Rice University, Houston, 77005 USA
| |
Collapse
|
17
|
Liu Z, Yang Y, Li D, Lv X, Chen X, Dai Q. Prediction of the RNA Tertiary Structure Based on a Random Sampling Strategy and Parallel Mechanism. Front Genet 2022; 12:813604. [PMID: 35069706 PMCID: PMC8769045 DOI: 10.3389/fgene.2021.813604] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 11/19/2021] [Indexed: 12/14/2022] Open
Abstract
Background: Macromolecule structure prediction remains a fundamental challenge of bioinformatics. Over the past several decades, the Rosetta framework has provided solutions to diverse challenges in computational biology. However, it is challenging to model RNA tertiary structures effectively when the de novo modeling of RNA involves solving a well-defined small puzzle. Methods: In this study, we introduce a stepwise Monte Carlo parallelization (SMCP) algorithm for RNA tertiary structure prediction. Millions of conformations were randomly searched using the Monte Carlo algorithm and stepwise ansatz hypothesis, and SMCP uses a parallel mechanism for efficient sampling. Moreover, to achieve better prediction accuracy and completeness, we judged and processed the modeling results. Results: A benchmark of nine single-stranded RNA loops drawn from riboswitches establishes the general ability of the algorithm to model RNA with high accuracy and integrity, including six motifs that cannot be solved by knowledge mining-based modeling algorithms. Experimental results show that the modeling accuracy of the SMCP algorithm is up to 0.14 Å, and the modeling integrity on this benchmark is extremely high. Conclusion: SMCP is an ab initio modeling algorithm that substantially outperforms previous algorithms in the Rosetta framework, especially in improving the accuracy and completeness of the model. It is expected that the work will provide new research ideas for macromolecular structure prediction in the future. In addition, this work will provide theoretical basis for the development of the biomedical field.
Collapse
Affiliation(s)
- Zhendong Liu
- School of Computer Science and Technology, Shandong Jianzhu University, Jinan, China
| | - Yurong Yang
- School of Computer Science and Technology, Shandong Jianzhu University, Jinan, China
| | - Dongyan Li
- School of Computer Science and Technology, Shandong Jianzhu University, Jinan, China
| | - Xinrong Lv
- School of Computer Science and Technology, Shandong Jianzhu University, Jinan, China
| | - Xi Chen
- School of Computer Science and Technology, Shandong Jianzhu University, Jinan, China
| | - Qionghai Dai
- Department of Automation, Tsinghua University, Beijing, China
| |
Collapse
|
18
|
Hall-Swan S, Devaurs D, Rigo MM, Antunes DA, Kavraki LE, Zanatta G. DINC-COVID: A webserver for ensemble docking with flexible SARS-CoV-2 proteins. Comput Biol Med 2021; 139:104943. [PMID: 34717233 PMCID: PMC8518241 DOI: 10.1016/j.compbiomed.2021.104943] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/27/2021] [Accepted: 10/11/2021] [Indexed: 12/16/2022]
Abstract
An unprecedented research effort has been undertaken in response to the ongoing COVID-19 pandemic. This has included the determination of hundreds of crystallographic structures of SARS-CoV-2 proteins, and numerous virtual screening projects searching large compound libraries for potential drug inhibitors. Unfortunately, these initiatives have had very limited success in producing effective inhibitors against SARS-CoV-2 proteins. A reason might be an often overlooked factor in these computational efforts: receptor flexibility. To address this issue we have implemented a computational tool for ensemble docking with SARS-CoV-2 proteins. We have extracted representative ensembles of protein conformations from the Protein Data Bank and from in silico molecular dynamics simulations. Twelve pre-computed ensembles of SARS-CoV-2 protein conformations have now been made available for ensemble docking via a user-friendly webserver called DINC-COVID (dinc-covid.kavrakilab.org). We have validated DINC-COVID using data on tested inhibitors of two SARS-CoV-2 proteins, obtaining good correlations between docking-derived binding energies and experimentally-determined binding affinities. Some of the best results have been obtained on a dataset of large ligands resolved via room temperature crystallography, and therefore capturing alternative receptor conformations. In addition, we have shown that the ensembles available in DINC-COVID capture different ranges of receptor flexibility, and that this diversity is useful in finding alternative binding modes of ligands. Overall, our work highlights the importance of accounting for receptor flexibility in docking studies, and provides a platform for the identification of new inhibitors against SARS-CoV-2 proteins.
Collapse
Affiliation(s)
- Sarah Hall-Swan
- Department of Computer Science, Rice University, Houston, 77005, Texas, United States
| | - Didier Devaurs
- MRC Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU, United Kingdom
| | - Mauricio M. Rigo
- Department of Computer Science, Rice University, Houston, 77005, Texas, United States
| | - Dinler A. Antunes
- Department of Computer Science, Rice University, Houston, 77005, Texas, United States,Department of Biology and Biochemistry, University of Houston, Houston, 77005, Texas, United States,Corresponding author. Department of Computer Science, Rice University, Houston, 77005, Texas, United States
| | - Lydia E. Kavraki
- Department of Computer Science, Rice University, Houston, 77005, Texas, United States,Corresponding author
| | - Geancarlo Zanatta
- Department of Physics, Federal University of Ceará, Fortaleza, CE, Brazil,Corresponding author
| |
Collapse
|
19
|
Svetlov D, Artsimovitch I. Reductionism Ad Absurdum: The Misadventures of Structural Biology in the Time of Coronavirus. ACS Infect Dis 2021; 7:2948-2952. [PMID: 34613689 PMCID: PMC8507565 DOI: 10.1021/acsinfecdis.1c00492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Indexed: 01/18/2023]
Abstract
The tragic consequences of the COVID-19 pandemic have led to admirable responses by the global scientific community, including a profound acceleration in the pace of research and exchange of findings. However, this has had considerable costs of its own, as erroneous conclusions have propagated faster than researchers have been able to detect and correct them. We illustrate the specific misunderstandings that have resulted from reductionist approaches to the study of SARS-CoV-2 RNA-dependent RNA polymerase (RdRp), which are but one instance of a regrettably growing trend in structural biology. Far from merely being cautionary tales about the conduct of scientific research, these errors have had significant practical impact, by hampering a correct understanding of RdRp structure and mechanism, its inhibition by nucleoside analogues such as remdesivir, and the discovery and characterization of such analogues. After correcting these misunderstandings, we close with several recommendations for a broader correction of the course of scientific research.
Collapse
Affiliation(s)
- Dmitri Svetlov
- Svetlov Scientific
Software, Pasadena, California 91106, United States
| | - Irina Artsimovitch
- Department of Microbiology and The Center for RNA
Biology, The Ohio State University, Columbus, Ohio 43210,
United States
| |
Collapse
|
20
|
Sun L, Fu T, Zhao D, Fan H, Zhong S. Divide-and-link peptide docking: a fragment-based peptide docking protocol. Phys Chem Chem Phys 2021; 23:22647-22660. [PMID: 34596658 DOI: 10.1039/d1cp02098f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Protein-peptide interactions are crucial for various important cellular regulations, and are also a basis for understanding protein-protein interactions, protein folding and peptide drug design. Due to the limited structural data obtained using experimental methods, it is necessary to predict protein-peptide interaction modes using computational methods. In the present work, we designed a fragment-based docking protocol, Divide-and-Link Peptide Docking (DLPepDock), to predict protein-peptide binding modes. This protocol contains the following steps: dividing the peptide into fragments and separately docking the fragments using a third-party small molecular docking tool, linking the docked fragmental poses to form the whole peptide conformations via fragmental coordinate transformation using our in-house program, removing unreasonable poses according to several geometrical filters, extracting representative conformations after clustering for further minimization using the steepest descent and conjugation gradient methods based on a full-atom molecular force field and finally scoring using the MM/PBSA binding energy calculation implemented in Amber. When tested on the LEADS-PEP benchmark data set of 26 diverse complexes with peptides of 6-12 residues, FlexPepDock ab initio and AutoDock CrankPep achieved superior results. DLPepDock performed better than the other 15 docking protocols implemented in nine docking programs (HPepDock, DockThor, rDock, Glide, LeDock, AutoDock, AutoDock Vina, Surflex, and GOLD). The Linux scripts to call the third-party tools and run all the calculations.
Collapse
Affiliation(s)
- Lu Sun
- School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, 116024, P. R. China.
| | - Tingting Fu
- School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, 116024, P. R. China. .,School of Tropical Medicine and Laboratory Medicine, Hainan Medical University, Haikou, Hainan, 570102, P. R. China
| | - Dan Zhao
- School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, 116024, P. R. China.
| | - Hongjun Fan
- State Key Laboratory of Molecular Reaction Dynamics, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Liaoning, 116023, P. R. China
| | - Shijun Zhong
- School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, 116024, P. R. China.
| |
Collapse
|
21
|
Nazarova A, Khannanov A, Boldyrev A, Yakimova L, Stoikov I. Self-Assembling Systems Based on Pillar[5]arenes and Surfactants for Encapsulation of Diagnostic Dye DAPI. Int J Mol Sci 2021; 22:6038. [PMID: 34204914 PMCID: PMC8199762 DOI: 10.3390/ijms22116038] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 05/31/2021] [Accepted: 05/31/2021] [Indexed: 12/12/2022] Open
Abstract
In this paper, we report the development of the novel self-assembling systems based on oppositely charged Pillar[5]arenes and surfactants for encapsulation of diagnostic dye DAPI. For this purpose, the aggregation behavior of synthesized macrocycles and surfactants in the presence of Pillar[5]arenes functionalized by carboxy and ammonium terminal groups was studied. It has been demonstrated that by varying the molar ratio in Pillar[5]arene-surfactant systems, it is possible to obtain various types of supramolecular systems: host-guest complexes at equimolar ratio of Pillar[5]arene-surfactant and interpolyelectrolyte complexes (IPECs) are self-assembled materials formed in aqueous medium by two oppositely charged polyelectrolytes (macrocycle and surfactant micelles). It has been suggested that interaction of Pillar[5]arenes with surfactants is predominantly driven by cooperative electrostatic interactions. Synthesized stoichiometric and non-stoichiometric IPECs specifically interact with DAPI. UV-vis, luminescent spectroscopy and molecular docking data show the structural feature of dye-loaded IPEC and key role of the electrostatic, π-π-stacking, cation-π interactions in their formation. Such a strategy for the design of supramolecular Pillar[5]arene-surfactant systems will lead to a synergistic interaction of the two components and will allow specific interaction with the third component (drug or fluorescent tag), which will certainly be in demand in pharmaceuticals and biomedical diagnostics.
Collapse
Affiliation(s)
| | | | | | - Luidmila Yakimova
- A.M. Butlerov’ Chemistry Institute of Kazan Federal University, 18 Kremlyovskaya Str., 420008 Kazan, Russia; (A.N.); (A.K.); (A.B.)
| | - Ivan Stoikov
- A.M. Butlerov’ Chemistry Institute of Kazan Federal University, 18 Kremlyovskaya Str., 420008 Kazan, Russia; (A.N.); (A.K.); (A.B.)
| |
Collapse
|
22
|
Li C, Sun J, Palade V. MSLDOCK: Multi-Swarm Optimization for Flexible Ligand Docking and Virtual Screening. J Chem Inf Model 2021; 61:1500-1515. [PMID: 33657798 DOI: 10.1021/acs.jcim.0c01358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Autodock and its various variants are widely utilized docking approaches, which adopt optimization methods as search algorithms for flexible ligand docking and virtual screening. However, many of them have their limitations, such as poor accuracy for dockings with highly flexible ligands and low docking efficiency. In this paper, a multi-swarm optimization algorithm integrated with Autodock environment is proposed to design a high-performance and high-efficiency docking program, namely, MSLDOCK. The search algorithm is a combination of the random drift particle swarm optimization with a novel multi-swarm strategy and the Solis and Wets local search method with a modified implementation. Due to the algorithm's structure, MSLDOCK also has a multithread mode. The experimental results reveal that MSLDOCK outperforms other two Autodock-based approaches in many aspects, such as self-docking, cross-docking, and virtual screening accuracies as well as docking efficiency. Moreover, compared with three non-Autodock-based docking programs, MSLDOCK can be a reliable choice for self-docking and virtual screening, especially for dealing with highly flexible ligand docking problems. The source code of MSLDOCK can be downloaded for free from https://github.com/lcmeteor/MSLDOCK.
Collapse
Affiliation(s)
- Chao Li
- Department of Computer Science and Technology, Jiangnan University, No.1800, Lihu Avenue, Wuxi, Jiangsu 214122, PR China
| | - Jun Sun
- Department of Computer Science and Technology, Jiangnan University, No.1800, Lihu Avenue, Wuxi, Jiangsu 214122, PR China
| | - Vasile Palade
- Faculty of Engineering and Computing, Coventry University, Priory Street, Coventry CV1 5FB, U.K
| |
Collapse
|
23
|
Hall-Swan S, Antunes DA, Devaurs D, Rigo MM, Kavraki LE, Zanatta G. DINC-COVID: A webserver for ensemble docking with flexible SARS-CoV-2 proteins. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021. [PMID: 33501448 DOI: 10.1101/2021.01.21.427315] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Motivation Recent efforts to computationally identify inhibitors for SARS-CoV-2 proteins have largely ignored the issue of receptor flexibility. We have implemented a computational tool for ensemble docking with the SARS-CoV-2 proteins, including the main protease (Mpro), papain-like protease (PLpro) and RNA-dependent RNA polymerase (RdRp). Results Ensembles of other SARS-CoV-2 proteins are being prepared and made available through a user-friendly docking interface. Plausible binding modes between conformations of a selected ensemble and an uploaded ligand are generated by DINC, our parallelized meta-docking tool. Binding modes are scored with three scoring functions, and account for the flexibility of both the ligand and receptor. Additional details on our methods are provided in the supplementary material. Availability dinc-covid.kavrakilab.org. Supplementary information Details on methods for ensemble generation and docking are provided as supplementary data online. Contact geancarlo.zanatta@ufc.br , kavraki@rice.edu.
Collapse
|
24
|
Antunes DA, Abella JR, Hall-Swan S, Devaurs D, Conev A, Moll M, Lizée G, Kavraki LE. HLA-Arena: A Customizable Environment for the Structural Modeling and Analysis of Peptide-HLA Complexes for Cancer Immunotherapy. JCO Clin Cancer Inform 2020; 4:623-636. [PMID: 32667823 PMCID: PMC7397777 DOI: 10.1200/cci.19.00123] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2020] [Indexed: 02/07/2023] Open
Abstract
PURPOSE HLA protein receptors play a key role in cellular immunity. They bind intracellular peptides and display them for recognition by T-cell lymphocytes. Because T-cell activation is partially driven by structural features of these peptide-HLA complexes, their structural modeling and analysis are becoming central components of cancer immunotherapy projects. Unfortunately, this kind of analysis is limited by the small number of experimentally determined structures of peptide-HLA complexes. Overcoming this limitation requires developing novel computational methods to model and analyze peptide-HLA structures. METHODS Here we describe a new platform for the structural modeling and analysis of peptide-HLA complexes, called HLA-Arena, which we have implemented using Jupyter Notebook and Docker. It is a customizable environment that facilitates the use of computational tools, such as APE-Gen and DINC, which we have previously applied to peptide-HLA complexes. By integrating other commonly used tools, such as MODELLER and MHCflurry, this environment includes support for diverse tasks in structural modeling, analysis, and visualization. RESULTS To illustrate the capabilities of HLA-Arena, we describe 3 example workflows applied to peptide-HLA complexes. Leveraging the strengths of our tools, DINC and APE-Gen, the first 2 workflows show how to perform geometry prediction for peptide-HLA complexes and structure-based binding prediction, respectively. The third workflow presents an example of large-scale virtual screening of peptides for multiple HLA alleles. CONCLUSION These workflows illustrate the potential benefits of HLA-Arena for the structural modeling and analysis of peptide-HLA complexes. Because HLA-Arena can easily be integrated within larger computational pipelines, we expect its potential impact to vastly increase. For instance, it could be used to conduct structural analyses for personalized cancer immunotherapy, neoantigen discovery, or vaccine development.
Collapse
Affiliation(s)
| | | | - Sarah Hall-Swan
- Department of Computer Science, Rice University, Houston, TX
| | | | - Anja Conev
- Department of Computer Science, Rice University, Houston, TX
| | - Mark Moll
- Department of Computer Science, Rice University, Houston, TX
| | - Gregory Lizée
- Department of Melanoma Medical Oncology–Research, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | |
Collapse
|
25
|
Santos KB, Guedes IA, Karl ALM, Dardenne LE. Highly Flexible Ligand Docking: Benchmarking of the DockThor Program on the LEADS-PEP Protein-Peptide Data Set. J Chem Inf Model 2020; 60:667-683. [PMID: 31922754 DOI: 10.1021/acs.jcim.9b00905] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Protein-peptide interactions play a crucial role in many cellular and biological functions, which justify the increasing interest in the development of peptide-based drugs. However, predicting experimental binding modes and affinities in protein-peptide docking remains a great challenge for most docking programs due to some particularities of this class of ligands, such as the high degree of flexibility. In this paper, we present the performance of the DockThor program on the LEADS-PEP data set, a benchmarking set composed of 53 diverse protein-peptide complexes with peptides ranging from 3 to 12 residues and with up to 51 rotatable bonds. The DockThor performance for pose prediction on redocking studies was compared with some state-of-the-art docking programs that were also evaluated on the LEADS-PEP data set, AutoDock, AutoDock Vina, Surflex, GOLD, Glide, rDock, and DINC, as well as with the task-specific docking protocol HPepDock. Our results indicate that DockThor could dock 40% of the cases with an overall backbone RMSD below 2.5 Å when the top-scored docking pose was considered, exhibiting similar results to Glide and outperforming other protein-ligand docking programs, whereas rDock and HPepDock achieved superior results. Assessing the docking poses closest to the crystal structure (i.e., best-RMSD pose), DockThor achieved a success rate of 60% in pose prediction. Due to the great overall performance of handling peptidic compounds, the DockThor program can be considered as suitable for docking highly flexible and challenging ligands, with up to 40 rotatable bonds. DockThor is freely available as a virtual screening Web server at https://www.dockthor.lncc.br/ .
Collapse
Affiliation(s)
- Karina B Santos
- National Laboratory for Scientific Computing - LNCC , Petrópolis , Rio de Janeiro 25651-075 , Brazil
| | - Isabella A Guedes
- National Laboratory for Scientific Computing - LNCC , Petrópolis , Rio de Janeiro 25651-075 , Brazil
| | - Ana L M Karl
- National Laboratory for Scientific Computing - LNCC , Petrópolis , Rio de Janeiro 25651-075 , Brazil
| | - Laurent E Dardenne
- National Laboratory for Scientific Computing - LNCC , Petrópolis , Rio de Janeiro 25651-075 , Brazil
| |
Collapse
|