1
|
Agoni C, Fernández-Díaz R, Timmons PB, Adelfio A, Gómez H, Shields DC. Molecular Modelling in Bioactive Peptide Discovery and Characterisation. Biomolecules 2025; 15:524. [PMID: 40305228 PMCID: PMC12025251 DOI: 10.3390/biom15040524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 03/12/2025] [Accepted: 04/01/2025] [Indexed: 05/02/2025] Open
Abstract
Molecular modelling is a vital tool in the discovery and characterisation of bioactive peptides, providing insights into their structural properties and interactions with biological targets. Many models predicting bioactive peptide function or structure rely on their intrinsic properties, including the influence of amino acid composition, sequence, and chain length, which impact stability, folding, aggregation, and target interaction. Homology modelling predicts peptide structures based on known templates. Peptide-protein interactions can be explored using molecular docking techniques, but there are challenges related to the inherent flexibility of peptides, which can be addressed by more computationally intensive approaches that consider their movement over time, called molecular dynamics (MD). Virtual screening of many peptides, usually against a single target, enables rapid identification of potential bioactive peptides from large libraries, typically using docking approaches. The integration of artificial intelligence (AI) has transformed peptide discovery by leveraging large amounts of data. AlphaFold is a general protein structure prediction tool based on deep learning that has greatly improved the predictions of peptide conformations and interactions, in addition to providing estimates of model accuracy at each residue which greatly guide interpretation. Peptide function and structure prediction are being further enhanced using Protein Language Models (PLMs), which are large deep-learning-derived statistical models that learn computer representations useful to identify fundamental patterns of proteins. Recent methodological developments are discussed in the context of canonical peptides, as well as those with modifications and cyclisations. In designing potential peptide therapeutics, the main outstanding challenge for these methods is the incorporation of diverse non-canonical amino acids and cyclisations.
Collapse
Affiliation(s)
- Clement Agoni
- School of Medicine, University College Dublin, D04 C1P1 Dublin, Ireland;
- Conway Institute of Biomolecular and Biomedical Science, University College Dublin, D04 C1P Dublin, Ireland
- Discipline of Pharmaceutical Sciences, School of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa
| | - Raúl Fernández-Díaz
- School of Medicine, University College Dublin, D04 C1P1 Dublin, Ireland;
- IBM Research, D15 HN66 Dublin, Ireland
| | | | - Alessandro Adelfio
- Nuritas Ltd., Joshua Dawson House, D02 RY95 Dublin, Ireland; (P.B.T.); (A.A.); (H.G.)
| | - Hansel Gómez
- Nuritas Ltd., Joshua Dawson House, D02 RY95 Dublin, Ireland; (P.B.T.); (A.A.); (H.G.)
| | - Denis C. Shields
- School of Medicine, University College Dublin, D04 C1P1 Dublin, Ireland;
- Conway Institute of Biomolecular and Biomedical Science, University College Dublin, D04 C1P Dublin, Ireland
| |
Collapse
|
2
|
Basciu A, Athar M, Kurt H, Neville C, Malloci G, Muredda FC, Bosin A, Ruggerone P, Bonvin AMJJ, Vargiu AV. Toward the Prediction of Binding Events in Very Flexible, Allosteric, Multidomain Proteins. J Chem Inf Model 2025; 65:2052-2065. [PMID: 39907634 PMCID: PMC11863385 DOI: 10.1021/acs.jcim.4c01810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 01/08/2025] [Accepted: 01/13/2025] [Indexed: 02/06/2025]
Abstract
Knowledge of the structures formed by proteins and small molecules is key to understand the molecular principles of chemotherapy and for designing new and more effective drugs. During the early stage of a drug discovery program, it is customary to predict ligand-protein complexes in silico, particularly when screening large compound databases. While virtual screening based on molecular docking is widely used for this purpose, it generally fails in mimicking binding events associated with large conformational changes in the protein, particularly when the latter involve multiple domains. In this work, we describe a new methodology to generate bound-like conformations of very flexible and allosteric proteins bearing multiple binding sites by exploiting only information on the unbound structure and the putative binding sites. The protocol is validated on the paradigm enzyme adenylate kinase, for which we generated a significant fraction of bound-like structures. A fraction of these conformations, employed in ensemble-docking calculations, allowed to find native-like poses of substrates and inhibitors (binding to the active form of the enzyme), as well as catalytically incompetent analogs (binding the inactive form). Our protocol provides a general framework for the generation of bound-like conformations of challenging drug targets that are suitable to host different ligands, demonstrating high sensitivity to the fine chemical details that regulate protein's activity. We foresee applications in virtual screening, in the prediction of the impact of amino acid mutations on structure and dynamics, and in protein engineering.
Collapse
Affiliation(s)
- Andrea Basciu
- Physics
Department, University of Cagliari, Cittadella
Universitaria, Monserrato
(CA) I-09042, Italy
| | - Mohd Athar
- Physics
Department, University of Cagliari, Cittadella
Universitaria, Monserrato
(CA) I-09042, Italy
| | - Han Kurt
- Physics
Department, University of Cagliari, Cittadella
Universitaria, Monserrato
(CA) I-09042, Italy
| | - Christine Neville
- Institute
for Computational Molecular Science, Temple
University, 1925 N. 12th Street, Philadelphia, Pennsylvania 19122, United States
- Department
of Biology, Temple University, 1900 North 12th Street, Philadelphia, Pennsylvania 19122, United States
| | - Giuliano Malloci
- Physics
Department, University of Cagliari, Cittadella
Universitaria, Monserrato
(CA) I-09042, Italy
| | - Fabrizio C. Muredda
- Physics
Department, University of Cagliari, Cittadella
Universitaria, Monserrato
(CA) I-09042, Italy
| | - Andrea Bosin
- Physics
Department, University of Cagliari, Cittadella
Universitaria, Monserrato
(CA) I-09042, Italy
| | - Paolo Ruggerone
- Physics
Department, University of Cagliari, Cittadella
Universitaria, Monserrato
(CA) I-09042, Italy
| | - Alexandre M. J. J. Bonvin
- Bijvoet
Centre for Biomolecular Research, Faculty of Science - Chemistry, Utrecht University, Padualaan 8, Utrecht 3584 CH, The Netherlands
| | - Attilio V. Vargiu
- Physics
Department, University of Cagliari, Cittadella
Universitaria, Monserrato
(CA) I-09042, Italy
| |
Collapse
|
3
|
Alvarado YJ, González-Paz L, Paz JL, Loroño-González MA, Santiago Contreras J, Lossada C, Vivas A, Marrero-Ponce Y, Martinez-Rios F, Rodriguez-Lugo P, Balladores Y, Vera-Villalobos J. Biological Implications of the Intrinsic Deformability of Human Acetylcholinesterase Induced by Diverse Compounds: A Computational Study. BIOLOGY 2024; 13:1065. [PMID: 39765732 PMCID: PMC11672903 DOI: 10.3390/biology13121065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/26/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025]
Abstract
The enzyme acetylcholinesterase (AChE) plays a crucial role in the termination of nerve impulses by hydrolyzing the neurotransmitter acetylcholine (ACh). The inhibition of AChE has emerged as a promising therapeutic approach for the management of neurological disorders such as Lewy body dementia and Alzheimer's disease. The potential of various compounds as AChE inhibitors was investigated. In this study, we evaluated the impact of natural compounds of interest on the intrinsic deformability of human AChE using computational biophysical analysis. Our approach incorporates classical dynamics, elastic networks (ENM and NMA), statistical potentials (CUPSAT and SWOTein), energy frustration (Frustratometer), and volumetric cavity analyses (MOLE and PockDrug). The results revealed that cyanidin induced significant changes in the flexibility and rigidity of AChE, especially in the distribution and volume of internal cavities, compared to model inhibitors such as TZ2PA6, and through a distinct biophysical-molecular mechanism from the other inhibitors considered. These findings suggest that cyanidin could offer potential mechanistic pathways for future research and applications in the development of new treatments for neurodegenerative diseases.
Collapse
Affiliation(s)
- Ysaías J. Alvarado
- Laboratorio de Química Biofísica Experimental y Teórica (LQBET), Instituto Venezolano de Investigaciones Científicas (IVIC), Centro de Biomedicina Molecular (CBM), Maracaibo 4001, Zulia, República Bolivariana de Venezuela; (Y.J.A.); (P.R.-L.)
| | - Lenin González-Paz
- Laboratorio de Modelado, Dinamica y Bioquímica Subcelular (LMDBS), Instituto Venezolano de Investigaciones Científicas (IVIC), Centro de Biomedicina Molecular (CBM), Maracaibo 4001, Zulia, República Bolivariana de Venezuela; (C.L.); (A.V.)
| | - José L. Paz
- Departamento Académico de Química Inorgánica, Facultad de Química e Ingeniería Química, Universidad Nacional Mayor de San Marcos, Lima 15081, Peru
| | - Marcos A. Loroño-González
- Departamento Académico de Fisicoquímica, Facultad de Química e Ingeniería Química, Universidad Nacional Mayor de San Marcos, Lima 15081, Peru;
| | - Julio Santiago Contreras
- Departamento Académico de Química Orgánica, Facultad de Química e Ingeniería Química, Universidad Nacional Mayor de San Marcos, Lima 15081, Peru;
| | - Carla Lossada
- Laboratorio de Modelado, Dinamica y Bioquímica Subcelular (LMDBS), Instituto Venezolano de Investigaciones Científicas (IVIC), Centro de Biomedicina Molecular (CBM), Maracaibo 4001, Zulia, República Bolivariana de Venezuela; (C.L.); (A.V.)
| | - Alejandro Vivas
- Laboratorio de Modelado, Dinamica y Bioquímica Subcelular (LMDBS), Instituto Venezolano de Investigaciones Científicas (IVIC), Centro de Biomedicina Molecular (CBM), Maracaibo 4001, Zulia, República Bolivariana de Venezuela; (C.L.); (A.V.)
| | - Yovani Marrero-Ponce
- Facultad de Ingeniería, Universidad Panamericana, Augusto Rodin 498, Insurgentes Mixcoac, Benito Juárez, Ciudad de México 03920, México or (Y.M.-P.); (F.M.-R.)
- Grupo de Medicina Molecular y Traslacional (MeM&T), Colegio de Ciencias de la Salud (COCSA), Universidad San Francisco de Quito (USFQ), Escuela de Medicina, Edificio de Especialidades Médicas, Diego de Robles y vía interoceánica, Quito 170157, Ecuador
| | - Felix Martinez-Rios
- Facultad de Ingeniería, Universidad Panamericana, Augusto Rodin 498, Insurgentes Mixcoac, Benito Juárez, Ciudad de México 03920, México or (Y.M.-P.); (F.M.-R.)
| | - Patricia Rodriguez-Lugo
- Laboratorio de Química Biofísica Experimental y Teórica (LQBET), Instituto Venezolano de Investigaciones Científicas (IVIC), Centro de Biomedicina Molecular (CBM), Maracaibo 4001, Zulia, República Bolivariana de Venezuela; (Y.J.A.); (P.R.-L.)
| | - Yanpiero Balladores
- Laboratorio de Física de la Materia Condensada, Instituto Venezolano de Investigaciones Científicas (IVIC), Apartado 20632, Caracas, República Bolivariana de Venezuela;
| | - Joan Vera-Villalobos
- Laboratorio de Análisis Químico Instrumental (LAQUINS), Facultad de Ciencias Naturales y Matemáticas, Departamento de Química y Ciencias Ambientales, Escuela Superior Politécnica del Litoral, Guayaquil ECO90211, Ecuador;
| |
Collapse
|
4
|
Herlah B, Goričan T, Benedik NS, Grdadolnik SG, Sosič I, Perdih A. Simulation- and AI-directed optimization of 4,6-substituted 1,3,5-triazin-2(1 H)-ones as inhibitors of human DNA topoisomerase IIα. Comput Struct Biotechnol J 2024; 23:2995-3018. [PMID: 39135887 PMCID: PMC11318567 DOI: 10.1016/j.csbj.2024.06.037] [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: 05/03/2024] [Revised: 06/29/2024] [Accepted: 06/30/2024] [Indexed: 08/15/2024] Open
Abstract
The 4,6-substituted-1,3,5-triazin-2(1H)-ones are promising inhibitors of human DNA topoisomerase IIα. To further develop this chemical class targeting the enzyme´s ATP binding site, the triazin-2(1H)-one substitution position 6 was optimized. Inspired by binding of preclinical substituted 9H-purine derivative, bicyclic substituents were incorporated at position 6 and the utility of this modification was validated by a combination of molecular simulations, dynamic pharmacophores, and free energy calculations. Considering also predictions of Deepfrag, a software developed for structure-based lead optimization based on deep learning, compounds with both bicyclic and monocyclic substitutions were synthesized and investigated for their inhibitory activity. The SAR data showed that the bicyclic substituted compounds exhibited good inhibition of topo IIα, comparable to their mono-substituted counterparts. Further evaluation on a panel of human protein kinases showed selectivity for the inhibition of topo IIα. Mechanistic studies indicated that the compounds acted predominantly as catalytic inhibitors, with some exhibiting topo IIα poison effects at higher concentrations. Integration of STD NMR experiments and molecular simulations, provided insights into the binding model and highlighted the importance of the Asn120 interaction and hydrophobic interactions with substituents at positions 4 and 6. In addition, NCI-60 screening demonstrated cytotoxicity of the compounds with bicyclic substituents and identified sensitive human cancer cell lines, underlining the translational relevance of our findings for further preclinical development of this class of compounds. The study highlights the synergy between simulation and AI-based approaches in efficiently guiding molecular design for drug optimization, which has implications for further preclinical development of this class of compounds.
Collapse
Affiliation(s)
- Barbara Herlah
- National Institute of Chemistry, Hajdrihova 19, SI 1000 Ljubljana, Slovenia
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, SI 1000 Ljubljana, Slovenia
| | - Tjaša Goričan
- National Institute of Chemistry, Hajdrihova 19, SI 1000 Ljubljana, Slovenia
| | - Nika Strašek Benedik
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, SI 1000 Ljubljana, Slovenia
| | | | - Izidor Sosič
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, SI 1000 Ljubljana, Slovenia
| | - Andrej Perdih
- National Institute of Chemistry, Hajdrihova 19, SI 1000 Ljubljana, Slovenia
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, SI 1000 Ljubljana, Slovenia
| |
Collapse
|
5
|
Basciu A, Athar M, Kurt H, Neville C, Malloci G, Muredda FC, Bosin A, Ruggerone P, Bonvin AMJJ, Vargiu AV. Predicting binding events in very flexible, allosteric, multi-domain proteins. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.02.597018. [PMID: 38895346 PMCID: PMC11185556 DOI: 10.1101/2024.06.02.597018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Knowledge of the structures formed by proteins and small molecules is key to understand the molecular principles of chemotherapy and for designing new and more effective drugs. During the early stage of a drug discovery program, it is customary to predict ligand-protein complexes in silico, particularly when screening large compound databases. While virtual screening based on molecular docking is widely used for this purpose, it generally fails in mimicking binding events associated with large conformational changes in the protein, particularly when the latter involve multiple domains. In this work, we describe a new methodology to generate bound-like conformations of very flexible and allosteric proteins bearing multiple binding sites by exploiting only information on the unbound structure and the putative binding sites. The protocol is validated on the paradigm enzyme adenylate kinase, for which we generated a significant fraction of bound-like structures. A fraction of these conformations, employed in ensemble-docking calculations, allowed to find native-like poses of substrates and inhibitors (binding to the active form of the enzyme), as well as catalytically incompetent analogs (binding the inactive form). Our protocol provides a general framework for the generation of bound-like conformations of challenging drug targets that are suitable to host different ligands, demonstrating high sensitivity to the fine chemical details that regulate protein's activity. We foresee applications in virtual screening, in the prediction of the impact of amino acid mutations on structure and dynamics, and in protein engineering.
Collapse
Affiliation(s)
- Andrea Basciu
- Physics Department, University of Cagliari, Cittadella Universitaria, I-09042 Monserrato (CA), Italy
| | - Mohd Athar
- Physics Department, University of Cagliari, Cittadella Universitaria, I-09042 Monserrato (CA), Italy
| | - Han Kurt
- Physics Department, University of Cagliari, Cittadella Universitaria, I-09042 Monserrato (CA), Italy
| | - Christine Neville
- Institute for Computational Molecular Science, Temple University, 1925 N. 12th Street Philadelphia, PA 19122, U.S.A
- Department of Biology, Temple University, 1900 North 12th Street, Philadelphia, PA 19122, U.S.A
| | - Giuliano Malloci
- Physics Department, University of Cagliari, Cittadella Universitaria, I-09042 Monserrato (CA), Italy
| | - Fabrizio C. Muredda
- Physics Department, University of Cagliari, Cittadella Universitaria, I-09042 Monserrato (CA), Italy
| | - Andrea Bosin
- Physics Department, University of Cagliari, Cittadella Universitaria, I-09042 Monserrato (CA), Italy
| | - Paolo Ruggerone
- Physics Department, University of Cagliari, Cittadella Universitaria, I-09042 Monserrato (CA), Italy
| | - Alexandre M. J. J. Bonvin
- Bijvoet Centre for Biomolecular Research, Faculty of Science - Chemistry, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Attilio V. Vargiu
- Physics Department, University of Cagliari, Cittadella Universitaria, I-09042 Monserrato (CA), Italy
| |
Collapse
|
6
|
Pala D, Clark DE. Caught between a ROCK and a hard place: current challenges in structure-based drug design. Drug Discov Today 2024; 29:104106. [PMID: 39029868 DOI: 10.1016/j.drudis.2024.104106] [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: 04/11/2024] [Revised: 06/27/2024] [Accepted: 07/13/2024] [Indexed: 07/21/2024]
Abstract
The discipline of structure-based drug design (SBDD) is several decades old and it is tempting to think that the proliferation of experimental structures for many drug targets might make computer-aided drug design (CADD) straightforward. However, this is far from true. In this review, we illustrate some of the challenges that CADD scientists face every day in their work, even now. We use Rho-associated protein kinase (ROCK), and public domain structures and data, as an example to illustrate some of the challenges we have experienced during our project targeting this protein. We hope that this will help to prevent unrealistic expectations of what CADD can accomplish and to educate non-CADD scientists regarding the challenges still facing their CADD colleagues.
Collapse
Affiliation(s)
- Daniele Pala
- Medicinal Chemistry and Drug Design Technologies Department, Chiesi Farmaceutici S.p.A, Research Center, Largo Belloli 11/a, 43122 Parma, Italy
| | - David E Clark
- Charles River, 6-9 Spire Green Centre, Flex Meadow, Harlow CM19 5TR, UK.
| |
Collapse
|
7
|
Wang Z, Zhou F, Wang Z, Hu Q, Li YQ, Wang S, Wei Y, Zheng L, Li W, Peng X. Fully Flexible Molecular Alignment Enables Accurate Ligand Structure Modeling. J Chem Inf Model 2024; 64:6205-6215. [PMID: 39074901 DOI: 10.1021/acs.jcim.4c00669] [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: 07/31/2024]
Abstract
Accurate protein-ligand binding poses are the prerequisites of structure-based binding affinity prediction and provide the structural basis for in-depth lead optimization in small molecule drug design. However, it is challenging to provide reasonable predictions of binding poses for different molecules due to the complexity and diversity of the chemical space of small molecules. Similarity-based molecular alignment techniques can effectively narrow the search range, as structurally similar molecules are likely to have similar binding modes, with higher similarity usually correlated to higher success rates. However, molecular similarity is not consistently high because molecules often require changes to achieve specific purposes, leading to reduced alignment precision. To address this issue, we propose a new alignment method─Z-align. This method uses topological structural information as a criterion for evaluating similarity, reducing the reliance on molecular fingerprint similarity. Our method has achieved success rates significantly higher than those of other methods at moderate levels of similarity. Additionally, our approach can comprehensively and flexibly optimize bond lengths and angles of molecules, maintaining a high accuracy even when dealing with larger molecules. Consequently, our proposed solution helps in achieving more accurate binding poses in protein-ligand docking problems, facilitating the development of small molecule drugs. Z-align is freely available as a web server at https://cloud.zelixir.com/zalign/home.
Collapse
Affiliation(s)
- Zhihao Wang
- School of Physics, Shandong University, Jinan, 250100, China
| | - Fan Zhou
- Shanghai Zelixir Biotech, Shanghai, 200030, China
| | - Zechen Wang
- School of Physics, Shandong University, Jinan, 250100, China
| | - Qiuyue Hu
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yong-Qiang Li
- School of Physics, Shandong University, Jinan, 250100, China
| | - Sheng Wang
- Shanghai Zelixir Biotech, Shanghai, 200030, China
| | - Yanjie Wei
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Liangzhen Zheng
- Shanghai Zelixir Biotech, Shanghai, 200030, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Weifeng Li
- School of Physics, Shandong University, Jinan, 250100, China
| | - Xiangda Peng
- Shanghai Zelixir Biotech, Shanghai, 200030, China
| |
Collapse
|
8
|
Patnaik SK, Ayyamperumal S, Jade D, Palathoti N, Akey KS, Jupudi S, Harrison MA, Ponnambalam S, Mj N, Mjn C. Virtual high throughput screening of natural peptides against ErbB1 and ErbB2 to identify potential inhibitors for cancer chemotherapy. J Biomol Struct Dyn 2024; 42:5551-5574. [PMID: 37387589 DOI: 10.1080/07391102.2023.2226744] [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: 10/13/2022] [Accepted: 06/13/2023] [Indexed: 07/01/2023]
Abstract
Human epidermal growth factor receptors (EGFR), namely ErbB1/HER1, ErbB2/HER2/neu, ErbB3/HER3, and ErbB4/HER4, the trans-membrane family of tyrosine kinase receptors, are overexpressed in many types of cancers. These receptors play an important role in cell proliferation, differentiation, invasion, metastasis and angiogenesis including unregulated activation of cancer cells. Overexpression of ErbB1 and ErbB2 that occurs in several types of cancers is associated with poor prognosis leading to resistance to ErbB1-directed therapies. In this connection, promising strategy to overcome the disadvantages of the existing chemotherapeutic drugs is the use of short peptides as anticancer agents. In the present study, we have performed virtual high throughput screening of natural peptides against ErbB1 and ErbB2 to identify potential dual inhibitors and identified five inhibitors based on their binding affinities, ADMET analysis, MD simulation studies and calculation of free energy of binding. These natural peptides could be further exploited for developing drugs for treating cancer.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Sunil Kumar Patnaik
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, Tamil Nadu, India
| | - Selvaraj Ayyamperumal
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, Tamil Nadu, India
| | - Dhananjay Jade
- School of Biomedical Sciences, University of Leeds, Leeds, UK
| | - Nagarjuna Palathoti
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, Tamil Nadu, India
| | - Krishna Swaroop Akey
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, Tamil Nadu, India
| | - Srikanth Jupudi
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, Tamil Nadu, India
| | | | | | - Nanjan Mj
- JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, Tamil Nadu, India
| | - Chandrasekar Mjn
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, Tamil Nadu, India
- School of Life Sciences, JSS Academy of Higher Education & Research(Ooty Campus), Ooty, Tamil Nadu, India
| |
Collapse
|
9
|
Herlah B, Janežič M, Ogris I, Grdadolnik SG, Kološa K, Žabkar S, Žegura B, Perdih A. Nature-inspired substituted 3-(imidazol-2-yl) morpholines targeting human topoisomerase IIα: Dynophore-derived discovery. Biomed Pharmacother 2024; 175:116676. [PMID: 38772152 DOI: 10.1016/j.biopha.2024.116676] [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: 03/12/2024] [Revised: 04/22/2024] [Accepted: 04/29/2024] [Indexed: 05/23/2024] Open
Abstract
The molecular nanomachine, human DNA topoisomerase IIα, plays a crucial role in replication, transcription, and recombination by catalyzing topological changes in the DNA, rendering it an optimal target for cancer chemotherapy. Current clinical topoisomerase II poisons often cause secondary tumors as side effects due to the accumulation of double-strand breaks in the DNA, spurring the development of catalytic inhibitors. Here, we used a dynamic pharmacophore approach to develop catalytic inhibitors targeting the ATP binding site of human DNA topoisomerase IIα. Our screening of a library of nature-inspired compounds led to the discovery of a class of 3-(imidazol-2-yl) morpholines as potent catalytic inhibitors that bind to the ATPase domain. Further experimental and computational studies identified hit compound 17, which exhibited selectivity against the human DNA topoisomerase IIα versus human protein kinases, cytotoxicity against several human cancer cells, and did not induce DNA double-strand breaks, making it distinct from clinical topoisomerase II poisons. This study integrates an innovative natural product-inspired chemistry and successful implementation of a molecular design strategy that incorporates a dynamic component of ligand-target molecular recognition, with comprehensive experimental characterization leading to hit compounds with potential impact on the development of more efficient chemotherapies.
Collapse
Affiliation(s)
- Barbara Herlah
- National Institute of Chemistry, Hajdrihova 19, Ljubljana SI 1000, Slovenia; University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, Ljubljana SI 1000, Slovenia
| | - Matej Janežič
- National Institute of Chemistry, Hajdrihova 19, Ljubljana SI 1000, Slovenia
| | - Iza Ogris
- National Institute of Chemistry, Hajdrihova 19, Ljubljana SI 1000, Slovenia; University of Ljubljana, Faculty of Medicine, Vrazov trg 2, Ljubljana SI 1000, Slovenia
| | | | - Katja Kološa
- National institute of Biology, Department of Genetic Toxicology and Cancer Biology, Večna pot 121, Ljubljana SI 1000, Slovenia
| | - Sonja Žabkar
- National institute of Biology, Department of Genetic Toxicology and Cancer Biology, Večna pot 121, Ljubljana SI 1000, Slovenia
| | - Bojana Žegura
- National institute of Biology, Department of Genetic Toxicology and Cancer Biology, Večna pot 121, Ljubljana SI 1000, Slovenia
| | - Andrej Perdih
- National Institute of Chemistry, Hajdrihova 19, Ljubljana SI 1000, Slovenia; University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, Ljubljana SI 1000, Slovenia.
| |
Collapse
|
10
|
Wang M, Fa S, Zhang G, Yu J, Zhang Q. Sequentially Controlled Recognition of Different Proteins Using Programmable Protein Imprinted Nanospheres. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2304957. [PMID: 37518853 DOI: 10.1002/smll.202304957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/20/2023] [Indexed: 08/01/2023]
Abstract
Although protein imprinted materials with multiple templates are developed to selectively separate different proteins, it is difficult to achieve the programmed adsorption and separation of different proteins using one material, because the available protein imprinted materials are constructed through irreversible crosslinking and their structures are unprogrammable and non-reconstructive. Herein, a novel nanosphere (MS@PTL-g-PNIPAM) is designed, which not only is temperature and pH responsive but also can dynamically reversibly crosslink/de-crosslink under ultraviolet light of different wavelengths. With the help of the dynamically reversible photo-crosslinking, the nanospheres can be repeatedly programmed into protein imprinted nanospheres toward different target proteins. Moreover, the prepared imprinted nanospheres can easily achieve the controlled rebinding and release of target proteins, benefiting from the introduced temperature- and pH-responsive moieties. As a consequence, this study realizes the specific separation of different target proteins from protein mixture and the real bovine blood sequentially by programming one material. It is resource saving, time saving, recyclable, and it will provide convenience for protein imprinted materials to use in the blood purification, drug delivery, and virus detection.
Collapse
Affiliation(s)
- Mingqi Wang
- Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| | - Shixin Fa
- Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| | - Guoxian Zhang
- Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| | - Jiate Yu
- Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| | - Qiuyu Zhang
- Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| |
Collapse
|
11
|
Kwon S, Seok C. CSAlign and CSAlign-Dock: Structure alignment of ligands considering full flexibility and application to protein-ligand docking. Comput Struct Biotechnol J 2022; 21:1-10. [PMID: 36514334 PMCID: PMC9719078 DOI: 10.1016/j.csbj.2022.11.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 11/22/2022] [Accepted: 11/23/2022] [Indexed: 11/27/2022] Open
Abstract
Structure prediction of protein-ligand complexes, called protein-ligand docking, is a critical computational technique that can be used to understand the underlying principle behind the protein functions at the atomic level and to design new molecules regulating the functions. Protein-ligand docking methods have been employed in structure-based drug discovery for hit discovery and lead optimization. One of the important technical challenges in protein-ligand docking is to account for protein conformational changes induced by ligand binding. A small change such as a single side-chain rotation upon ligand binding can hinder accurate docking. Here we report an increase in docking performance achieved by structure alignment to known complex structures. First, a fully flexible compound-to-compound alignment method CSAlign is developed by global optimization of a shape score. Next, the alignment method is combined with a docking algorithm to dock a new ligand to a target protein when a reference protein-ligand complex structure is available. This alignment-based docking method, called CSAlign-Dock, showed superior performance to ab initio docking methods in cross-docking benchmark tests. Both CSAlign and CSAlign-Dock are freely available as a web server at https://galaxy.seoklab.org/csalign.
Collapse
Affiliation(s)
- Sohee Kwon
- Department of Chemistry, Seoul National University, Seoul 08826, South Korea
- Galux Inc, Seoul 08738, South Korea
| | - Chaok Seok
- Department of Chemistry, Seoul National University, Seoul 08826, South Korea
- Galux Inc, Seoul 08738, South Korea
| |
Collapse
|
12
|
Modeling receptor flexibility in the structure-based design of KRAS G12C inhibitors. J Comput Aided Mol Des 2022; 36:591-604. [PMID: 35930206 PMCID: PMC9512760 DOI: 10.1007/s10822-022-00467-0] [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: 05/05/2022] [Accepted: 07/15/2022] [Indexed: 11/15/2022]
Abstract
KRAS has long been referred to as an ‘undruggable’ target due to its high affinity for its cognate ligands (GDP and GTP) and its lack of readily exploited allosteric binding pockets. Recent progress in the development of covalent inhibitors of KRASG12C has revealed that occupancy of an allosteric binding site located between the α3-helix and switch-II loop of KRASG12C—sometimes referred to as the ‘switch-II pocket’—holds great potential in the design of direct inhibitors of KRASG12C. In studying diverse switch-II pocket binders during the development of sotorasib (AMG 510), the first FDA-approved inhibitor of KRASG12C, we found the dramatic conformational flexibility of the switch-II pocket posing significant challenges toward the structure-based design of inhibitors. Here, we present our computational approaches for dealing with receptor flexibility in the prediction of ligand binding pose and binding affinity. For binding pose prediction, we modified the covalent docking program CovDock to allow for protein conformational mobility. This new docking approach, termed as FlexCovDock, improves success rates from 55 to 89% for binding pose prediction on a dataset of 10 cross-docking cases and has been prospectively validated across diverse ligand chemotypes. For binding affinity prediction, we found standard free energy perturbation (FEP) methods could not adequately handle the significant conformational change of the switch-II loop. We developed a new computational strategy to accelerate conformational transitions through the use of targeted protein mutations. Using this methodology, the mean unsigned error (MUE) of binding affinity prediction were reduced from 1.44 to 0.89 kcal/mol on a set of 14 compounds. These approaches were of significant use in facilitating the structure-based design of KRASG12C inhibitors and are anticipated to be of further use in the design of covalent (and noncovalent) inhibitors of other conformationally labile protein targets.
Collapse
|
13
|
Simončič M, Lukšič M, Druchok M. Machine learning assessment of the binding region as a tool for more efficient computational receptor-ligand docking. J Mol Liq 2022; 353:118759. [PMID: 35273421 PMCID: PMC8903148 DOI: 10.1016/j.molliq.2022.118759] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
We present a combined computational approach to protein-ligand binding, which consists of two steps: (1) a deep neural network is used to locate a binding region on a target protein, and (2) molecular docking of a ligand is performed within the specified region to obtain the best pose using Autodock Vina. Our in-house designed neural network was trained using the PepBDB dataset. Although the training dataset consisted of protein-peptide complexes, we show that the approach is not limited to peptides, but also works remarkably well for a large class of non-peptide ligands. The results are compared with those in which the binding region (first step) was provided by Accluster. In cases where no prior experimental data on the binding region are available, our deep neural network provides a fast and effective alternative to classical software for its localization. Our code is available at https://github.com/mksmd/NNforDocking.
Collapse
Affiliation(s)
- Matjaž Simončič
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, SI-1000 Ljubljana, Slovenia
| | - Miha Lukšič
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, SI-1000 Ljubljana, Slovenia
| | - Maksym Druchok
- Institute for Condensed Matter Physics, 1 Svientsitskii Str., UA-79011 Lviv, Ukraine
- SoftServe Inc., 2d Sadova Str., UA-79021 Lviv, Ukraine
| |
Collapse
|
14
|
Design, Synthesis and Evaluation of Fused Bicyclo[2.2.2]octene as a Potential Core Scaffold for the Non-Covalent Inhibitors of SARS-CoV-2 3CLpro Main Protease. Pharmaceuticals (Basel) 2022; 15:ph15050539. [PMID: 35631364 PMCID: PMC9145702 DOI: 10.3390/ph15050539] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 04/22/2022] [Accepted: 04/24/2022] [Indexed: 12/22/2022] Open
Abstract
The emergence of SARS-CoV-2, responsible for the global COVID-19 pandemic, requires the rapid development of novel antiviral drugs that would contribute to an effective treatment alongside vaccines. Drug repurposing and development of new molecules targeting numerous viral targets have already led to promising drug candidates. To this end, versatile molecular scaffolds with high functionalization capabilities play a key role. Starting with the clinically used conformationally flexible HIV-1 protease inhibitors that inhibit replication of SARS-CoV-2 and bind major protease 3CLpro, we designed and synthesized a series of rigid bicyclo[2.2.2]octenes fused to N-substituted succinimides to test whether this core scaffold could support the development of non-covalent 3CLpro inhibitors. Inhibition assays confirmed that some compounds can inhibit the SARS-CoV-2 main protease; the most promising compound 11a inhibited 3CLpro in micromolar range (IC50 = 102.2 μM). Molecular simulations of the target-ligand complex in conjunction with dynophore analyses and endpoint free energy calculations provide additional insight and first recommendations for future optimization. The fused bicyclo[2.2.2]octenes can be used as a new potential starting point in the development of non-covalent SARS-CoV-2 3CLpro protease inhibitors and the study also substantiates the potential of this versatile scaffold for the development of biologically active molecules.
Collapse
|
15
|
Djokovic N, Ruzic D, Rahnasto-Rilla M, Srdic-Rajic T, Lahtela-Kakkonen M, Nikolic K. Expanding the Accessible Chemical Space of SIRT2 Inhibitors through Exploration of Binding Pocket Dynamics. J Chem Inf Model 2022; 62:2571-2585. [PMID: 35467856 DOI: 10.1021/acs.jcim.2c00241] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Considerations of binding pocket dynamics are one of the crucial aspects of the rational design of binders. Identification of alternative conformational states or cryptic subpockets could lead to the discovery of completely novel groups of the ligands. However, experimental characterization of pocket dynamics, besides being expensive, may not be able to elucidate all of the conformational states relevant for drug discovery projects. In this study, we propose the protocol for computational simulations of sirtuin 2 (SIRT2) binding pocket dynamics and its integration into the structure-based virtual screening (SBVS) pipeline. Initially, unbiased molecular dynamics simulations of SIRT2:inhibitor complexes were performed using optimized force field parameters of SIRT2 inhibitors. Time-lagged independent component analysis (tICA) was used to design pocket-related collective variables (CVs) for enhanced sampling of SIRT2 pocket dynamics. Metadynamics simulations in the tICA eigenvector space revealed alternative conformational states of the SIRT2 binding pocket and the existence of a cryptic subpocket. Newly identified SIRT2 conformational states outperformed experimentally resolved states in retrospective SBVS validation. After performing prospective SBVS, compounds from the under-represented portions of the SIRT2 inhibitor chemical space were selected for in vitro evaluation. Two compounds, NDJ18 and NDJ85, were identified as potent and selective SIRT2 inhibitors, which validated the in silico protocol and opened up the possibility for generalization and broadening of its application. The anticancer effects of the most potent compound NDJ18 were examined on the triple-negative breast cancer cell line. Results indicated that NDJ18 represents a promising structure suitable for further evaluation.
Collapse
Affiliation(s)
- Nemanja Djokovic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia
| | - Dusan Ruzic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia
| | - Minna Rahnasto-Rilla
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, 70210 Kuopio, Finland
| | - Tatjana Srdic-Rajic
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia
| | | | - Katarina Nikolic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia
| |
Collapse
|
16
|
Trisciuzzi D, Siragusa L, Baroni M, Autiero I, Nicolotti O, Cruciani G. Getting Insights into Structural and Energetic Properties of Reciprocal Peptide-Protein Interactions. J Chem Inf Model 2022; 62:1113-1125. [PMID: 35148095 DOI: 10.1021/acs.jcim.1c01343] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Peptide-protein interactions play a key role for many cellular and metabolic processes involved in the onset of largely spread diseases such as cancer and neurodegenerative pathologies. Despite the progress in the structural characterization of peptide-protein interfaces, the in-depth knowledge of the molecular details behind their interactions is still a daunting task. Here, we present the first comprehensive in silico morphological and energetic study of peptide binding sites by focusing on both peptide and protein standpoints. Starting from the PixelDB database, a nonredundant benchmark collection of high-quality 3D crystallographic structures of peptide-protein complexes, a classification analysis of the most representative categories based on the nature of each cocrystallized peptide has been carried out. Several interpretable geometrical and energetic descriptors have been computed both from peptide and target protein sides in the attempt to unveil physicochemical and structural causative correlations. Finally, we investigated the most frequent peptide-protein residue pairs at the binding interface and made extensive energetic analyses, based on GRID MIFs, with the aim to study the peptide affinity-enhancing interactions to be further exploited in rational drug design strategies.
Collapse
Affiliation(s)
- Daniela Trisciuzzi
- Department of Pharmacy, Pharmaceutical Sciences, Università degli Studi di Bari "Aldo Moro", 70125 Bari, Italy.,Molecular Horizon s.r.l., Via Montelino, 30, 06084 Bettona (PG), Italy
| | - Lydia Siragusa
- Molecular Horizon s.r.l., Via Montelino, 30, 06084 Bettona (PG), Italy.,Molecular Discovery Ltd., Kinetic Business Centre, Theobald Street, Elstree, Borehamwood, Hertfordshire WD6 4PJ, United Kingdom
| | - Massimo Baroni
- Molecular Discovery Ltd., Kinetic Business Centre, Theobald Street, Elstree, Borehamwood, Hertfordshire WD6 4PJ, United Kingdom
| | - Ida Autiero
- Molecular Horizon s.r.l., Via Montelino, 30, 06084 Bettona (PG), Italy.,National Research Council, Institute of Biostructures and Bioimaging, 80138 Naples, Italy
| | - Orazio Nicolotti
- Department of Pharmacy, Pharmaceutical Sciences, Università degli Studi di Bari "Aldo Moro", 70125 Bari, Italy
| | - Gabriele Cruciani
- Department of Chemistry, Biology and Biotechnology, Università degli Studi di Perugia, via Elce di Sotto, 8, 06123 Perugia (PG), Italy
| |
Collapse
|
17
|
Basciu A, Callea L, Motta S, Bonvin AM, Bonati L, Vargiu AV. No dance, no partner! A tale of receptor flexibility in docking and virtual screening. VIRTUAL SCREENING AND DRUG DOCKING 2022. [DOI: 10.1016/bs.armc.2022.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
18
|
Adelusi TI, Oyedele AQK, Boyenle ID, Ogunlana AT, Adeyemi RO, Ukachi CD, Idris MO, Olaoba OT, Adedotun IO, Kolawole OE, Xiaoxing Y, Abdul-Hammed M. Molecular modeling in drug discovery. INFORMATICS IN MEDICINE UNLOCKED 2022. [DOI: 10.1016/j.imu.2022.100880] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
|
19
|
Janežič M, Valjavec K, Loboda KB, Herlah B, Ogris I, Kozorog M, Podobnik M, Grdadolnik SG, Wolber G, Perdih A. Dynophore-Based Approach in Virtual Screening: A Case of Human DNA Topoisomerase IIα. Int J Mol Sci 2021; 22:ijms222413474. [PMID: 34948269 PMCID: PMC8703789 DOI: 10.3390/ijms222413474] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/23/2021] [Accepted: 12/10/2021] [Indexed: 12/04/2022] Open
Abstract
In this study, we utilized human DNA topoisomerase IIα as a model target to outline a dynophore-based approach to catalytic inhibitor design. Based on MD simulations of a known catalytic inhibitor and the native ATP ligand analog, AMP-PNP, we derived a joint dynophore model that supplements the static structure-based-pharmacophore information with a dynamic component. Subsequently, derived pharmacophore models were employed in a virtual screening campaign of a library of natural compounds. Experimental evaluation identified flavonoid compounds with promising topoisomerase IIα catalytic inhibition and binding studies confirmed interaction with the ATPase domain. We constructed a binding model through docking and extensively investigated it with molecular dynamics MD simulations, essential dynamics, and MM-GBSA free energy calculations, thus reconnecting the new results to the initial dynophore-based screening model. We not only demonstrate a new design strategy that incorporates a dynamic component of molecular recognition, but also highlight new derivates in the established flavonoid class of topoisomerase II inhibitors.
Collapse
Affiliation(s)
- Matej Janežič
- National Institute of Chemistry, Hajdrihova 19, SI-1000 Ljubljana, Slovenia; (M.J.); (K.V.); (K.B.L.); (B.H.); (I.O.); (M.K.); (M.P.); (S.G.G.)
- Laboratory for Structural Bioinformatics, RIKEN Center for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
| | - Katja Valjavec
- National Institute of Chemistry, Hajdrihova 19, SI-1000 Ljubljana, Slovenia; (M.J.); (K.V.); (K.B.L.); (B.H.); (I.O.); (M.K.); (M.P.); (S.G.G.)
| | - Kaja Bergant Loboda
- National Institute of Chemistry, Hajdrihova 19, SI-1000 Ljubljana, Slovenia; (M.J.); (K.V.); (K.B.L.); (B.H.); (I.O.); (M.K.); (M.P.); (S.G.G.)
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, SI-1000 Ljubljana, Slovenia
| | - Barbara Herlah
- National Institute of Chemistry, Hajdrihova 19, SI-1000 Ljubljana, Slovenia; (M.J.); (K.V.); (K.B.L.); (B.H.); (I.O.); (M.K.); (M.P.); (S.G.G.)
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, SI-1000 Ljubljana, Slovenia
| | - Iza Ogris
- National Institute of Chemistry, Hajdrihova 19, SI-1000 Ljubljana, Slovenia; (M.J.); (K.V.); (K.B.L.); (B.H.); (I.O.); (M.K.); (M.P.); (S.G.G.)
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, SI-1000 Ljubljana, Slovenia
| | - Mirijam Kozorog
- National Institute of Chemistry, Hajdrihova 19, SI-1000 Ljubljana, Slovenia; (M.J.); (K.V.); (K.B.L.); (B.H.); (I.O.); (M.K.); (M.P.); (S.G.G.)
| | - Marjetka Podobnik
- National Institute of Chemistry, Hajdrihova 19, SI-1000 Ljubljana, Slovenia; (M.J.); (K.V.); (K.B.L.); (B.H.); (I.O.); (M.K.); (M.P.); (S.G.G.)
| | - Simona Golič Grdadolnik
- National Institute of Chemistry, Hajdrihova 19, SI-1000 Ljubljana, Slovenia; (M.J.); (K.V.); (K.B.L.); (B.H.); (I.O.); (M.K.); (M.P.); (S.G.G.)
| | - Gerhard Wolber
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Straße 2-4, 14195 Berlin, Germany;
| | - Andrej Perdih
- National Institute of Chemistry, Hajdrihova 19, SI-1000 Ljubljana, Slovenia; (M.J.); (K.V.); (K.B.L.); (B.H.); (I.O.); (M.K.); (M.P.); (S.G.G.)
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, SI-1000 Ljubljana, Slovenia
- Correspondence: ; Tel.: +386-1-4760-376
| |
Collapse
|
20
|
Redhair M, Atkins WM. Analytical and functional aspects of protein-ligand interactions: Beyond induced fit and conformational selection. Arch Biochem Biophys 2021; 714:109064. [PMID: 34715072 DOI: 10.1016/j.abb.2021.109064] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/18/2021] [Accepted: 10/20/2021] [Indexed: 10/20/2022]
Abstract
Ligand-dependent changes in protein conformation are foundational to biology. Historical mechanistic models for substrate-specific proteins are induced fit (IF) and conformational selection (CS), which invoke a change in protein conformation after ligand binds or before ligand binds, respectively. These mechanisms have important, but rarely discussed, functional relevance because IF vs. CS can differentially affect a protein's substrate specificity or promiscuity, and its regulatory properties. The modern view of proteins as conformational ensembles in both ligand free and bound states, together with the realization that most proteins exhibit some substrate promiscuity, demands a deeper interpretation of the historical models and provides an opportunity to improve mechanistic analyses. Here we describe alternative analytical strategies for distinguishing the historical models, including the more complex expanded versions of IF and CS. Functional implications of the different models are described. We provide an alternative perspective based on protein ensembles interacting with ligand ensembles that clarifies how a single protein can 'apparently' exploit different mechanisms for different ligands. Mechanistic information about protein ensembles can be optimized when they are probed with multiple ligands.
Collapse
Affiliation(s)
- Michelle Redhair
- Department of Medicinal Chemistry, Box 375610, University of Washington, Seattle, WA, 98177, USA
| | - William M Atkins
- Department of Medicinal Chemistry, Box 375610, University of Washington, Seattle, WA, 98177, USA.
| |
Collapse
|
21
|
Chheda PR, Cooling GT, Dean SF, Propp J, Hobbs KF, Spies MA. Decrypting a Cryptic Allosteric Pocket in H. pylori Glutamate Racemase. Commun Chem 2021; 4:172. [PMID: 35673630 PMCID: PMC9169614 DOI: 10.1038/s42004-021-00605-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 11/08/2021] [Indexed: 01/27/2023] Open
Abstract
One of our greatest challenges in drug design is targeting cryptic allosteric pockets in enzyme targets. Drug leads that do bind to these cryptic pockets are often discovered during HTS campaigns, and the mechanisms of action are rarely understood. Nevertheless, it is often the case that the allosteric pocket provides the best option for drug development against a given target. In the current studies we present a successful way forward in rationally exploiting the cryptic allosteric pocket of H. pylori glutamate racemase, an essential enzyme in this pathogen's life cycle. A wide range of computational and experimental methods are employed in a workflow leading to the discovery of a series of natural product allosteric inhibitors which occupy the allosteric pocket of this essential racemase. The confluence of these studies reveals a fascinating source of the allosteric inhibition, which centers on the abolition of essential monomer-monomer coupled motion networks.
Collapse
Affiliation(s)
- Pratik Rajesh Chheda
- Division of Medicinal and Natural Products Chemistry, Department of Pharmaceutical Sciences and Experimental Therapeutics, The University of Iowa, Iowa City, IA 52242 USA
| | - Grant T. Cooling
- Division of Medicinal and Natural Products Chemistry, Department of Pharmaceutical Sciences and Experimental Therapeutics, The University of Iowa, Iowa City, IA 52242 USA
| | - Sondra F. Dean
- Division of Medicinal and Natural Products Chemistry, Department of Pharmaceutical Sciences and Experimental Therapeutics, The University of Iowa, Iowa City, IA 52242 USA
| | - Jonah Propp
- Division of Medicinal and Natural Products Chemistry, Department of Pharmaceutical Sciences and Experimental Therapeutics, The University of Iowa, Iowa City, IA 52242 USA
| | - Kathryn F. Hobbs
- Department of Biochemistry, Carver College of Medicine, The University of Iowa, Iowa City, IA 52242 USA
| | - M. Ashley Spies
- Division of Medicinal and Natural Products Chemistry, Department of Pharmaceutical Sciences and Experimental Therapeutics, The University of Iowa, Iowa City, IA 52242 USA
- Department of Biochemistry, Carver College of Medicine, The University of Iowa, Iowa City, IA 52242 USA
| |
Collapse
|
22
|
Bhunia SS, Saxena AK. Efficiency of Homology Modeling Assisted Molecular Docking in G-protein Coupled Receptors. Curr Top Med Chem 2021; 21:269-294. [PMID: 32901584 DOI: 10.2174/1568026620666200908165250] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/20/2020] [Accepted: 09/01/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Molecular docking is in regular practice to assess ligand affinity on a target protein crystal structure. In the absence of protein crystal structure, the homology modeling or comparative modeling is the best alternative to elucidate the relationship details between a ligand and protein at the molecular level. The development of accurate homology modeling (HM) and its integration with molecular docking (MD) is essential for successful, rational drug discovery. OBJECTIVE The G-protein coupled receptors (GPCRs) are attractive therapeutic targets due to their immense role in human pharmacology. The GPCRs are membrane-bound proteins with the complex constitution, and the understanding of their activation and inactivation mechanisms is quite challenging. Over the past decade, there has been a rapid expansion in the number of solved G-protein-coupled receptor (GPCR) crystal structures; however, the majority of the GPCR structures remain unsolved. In this context, HM guided MD has been widely used for structure-based drug design (SBDD) of GPCRs. METHODS The focus of this review is on the recent (i) developments on HM supported GPCR drug discovery in the absence of GPCR crystal structures and (ii) application of HM in understanding the ligand interactions at the binding site, virtual screening, determining receptor subtype selectivity and receptor behaviour in comparison with GPCR crystal structures. RESULTS The HM in GPCRs has been extremely challenging due to the scarcity in template structures. In such a scenario, it is difficult to get accurate HM that can facilitate understanding of the ligand-receptor interactions. This problem has been alleviated to some extent by developing refined HM based on incorporating active /inactive ligand information and inducing protein flexibility. In some cases, HM proteins were found to outscore crystal structures. CONCLUSION The developments in HM have been highly operative to gain insights about the ligand interaction at the binding site and receptor functioning at the molecular level. Thus, HM guided molecular docking may be useful for rational drug discovery for the GPCRs mediated diseases.
Collapse
Affiliation(s)
- Shome S Bhunia
- Global Institute of Pharmaceutical Education and Research, Kashipur, Uttarakhand, India
| | - Anil K Saxena
- Division of Medicinal and Process Chemistry, CSIR-CDRI, Lucknow 226031, India
| |
Collapse
|
23
|
Callea L, Bonati L, Motta S. Metadynamics-Based Approaches for Modeling the Hypoxia-Inducible Factor 2α Ligand Binding Process. J Chem Theory Comput 2021; 17:3841-3851. [PMID: 34082524 PMCID: PMC8280741 DOI: 10.1021/acs.jctc.1c00114] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
![]()
Several methods based
on enhanced-sampling molecular dynamics have
been proposed for studying ligand binding processes. Here, we developed
a protocol that combines the advantages of steered molecular dynamics
(SMD) and metadynamics. While SMD is proposed for investigating possible
unbinding pathways of the ligand and identifying the preferred one,
metadynamics, with the path collective variable (PCV) formalism, is
suggested to explore the binding processes along the pathway defined
on the basis of SMD, by using only two CVs. We applied our approach
to the study of binding of two known ligands to the hypoxia-inducible
factor 2α, where the buried binding cavity makes simulation
of the process a challenging task. Our approach allowed identification
of the preferred entrance pathway for each ligand, highlighted the
features of the bound and intermediate states in the free-energy surface,
and provided a binding affinity scale in agreement with experimental
data. Therefore, it seems to be a suitable tool for elucidating ligand
binding processes of similar complex systems.
Collapse
Affiliation(s)
- Lara Callea
- Department of Earth and Environmental Sciences, University of Milano-Bicocca, Piazza della Scienza 1, 20126 Milan, Italy
| | - Laura Bonati
- Department of Earth and Environmental Sciences, University of Milano-Bicocca, Piazza della Scienza 1, 20126 Milan, Italy
| | - Stefano Motta
- Department of Earth and Environmental Sciences, University of Milano-Bicocca, Piazza della Scienza 1, 20126 Milan, Italy
| |
Collapse
|
24
|
Current and Future Challenges in Modern Drug Discovery. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2021; 2114:1-17. [PMID: 32016883 DOI: 10.1007/978-1-0716-0282-9_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Drug discovery is an expensive, time-consuming, and risky business. To avoid late-stage failure, learnings from past projects and the development of new approaches are crucial. New modalities and emerging new target spaces allow the exploration of unprecedented indications or to address so far undrugable targets. Late-stage attrition is usually attributed to the lack of efficacy or to compound-related safety issues. Efficacy has been shown to be related to a strong genetic link to human disease, a better understanding of the target biology, and the availability of biomarkers to bridge from animals to humans. Compound safety can be improved by ligand optimization, which is becoming increasingly demanding for difficult targets. Therefore, new strategies include the design of allosteric ligands, covalent binders, and other modalities. Design methods currently heavily rely on artificial intelligence and advanced computational methods such as free energy calculations and quantum chemistry. Especially for quantum chemical methods, a more detailed overview is given in this chapter.
Collapse
|
25
|
Fenwick RB, Oyen D, van den Bedem H, Dyson HJ, Wright PE. Modeling of Hidden Structures Using Sparse Chemical Shift Data from NMR Relaxation Dispersion. Biophys J 2020; 120:296-305. [PMID: 33301748 DOI: 10.1016/j.bpj.2020.11.2267] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/30/2020] [Accepted: 11/11/2020] [Indexed: 12/24/2022] Open
Abstract
NMR relaxation dispersion measurements report on conformational changes occurring on the μs-ms timescale. Chemical shift information derived from relaxation dispersion can be used to generate structural models of weakly populated alternative conformational states. Current methods to obtain such models rely on determining the signs of chemical shift changes between the conformational states, which are difficult to obtain in many situations. Here, we use a "sample and select" method to generate relevant structural models of alternative conformations of the C-terminal-associated region of Escherichia coli dihydrofolate reductase (DHFR), using only unsigned chemical shift changes for backbone amides and carbonyls (1H, 15N, and 13C'). We find that CS-Rosetta sampling with unsigned chemical shift changes generates a diversity of structures that are sufficient to characterize a minor conformational state of the C-terminal region of DHFR. The excited state differs from the ground state by a change in secondary structure, consistent with previous predictions from chemical shift hypersurfaces and validated by the x-ray structure of a partially humanized mutant of E. coli DHFR (N23PP/G51PEKN). The results demonstrate that the combination of fragment modeling with sparse chemical shift data can determine the structure of an alternative conformation of DHFR sampled on the μs-ms timescale. Such methods will be useful for characterizing alternative states, which can potentially be used for in silico drug screening, as well as contributing to understanding the role of minor states in biology and molecular evolution.
Collapse
Affiliation(s)
- R Bryn Fenwick
- Department of Integrative Structural and Computational Biology and Skaggs Institute of Chemical Biology, The Scripps Research Institute, La Jolla, California.
| | - David Oyen
- Department of Integrative Structural and Computational Biology and Skaggs Institute of Chemical Biology, The Scripps Research Institute, La Jolla, California
| | - Henry van den Bedem
- SLAC National Accelerator Laboratory, Stanford University, Menlo Park, California, and Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California
| | - H Jane Dyson
- Department of Integrative Structural and Computational Biology and Skaggs Institute of Chemical Biology, The Scripps Research Institute, La Jolla, California
| | - Peter E Wright
- Department of Integrative Structural and Computational Biology and Skaggs Institute of Chemical Biology, The Scripps Research Institute, La Jolla, California.
| |
Collapse
|
26
|
Saikia S, Bordoloi M. Molecular Docking: Challenges, Advances and its Use in Drug Discovery Perspective. Curr Drug Targets 2020; 20:501-521. [PMID: 30360733 DOI: 10.2174/1389450119666181022153016] [Citation(s) in RCA: 271] [Impact Index Per Article: 54.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 06/08/2018] [Accepted: 08/28/2018] [Indexed: 01/21/2023]
Abstract
Molecular docking is a process through which small molecules are docked into the macromolecular structures for scoring its complementary values at the binding sites. It is a vibrant research area with dynamic utility in structure-based drug-designing, lead optimization, biochemical pathway and for drug designing being the most attractive tools. Two pillars for a successful docking experiment are correct pose and affinity prediction. Each program has its own advantages and drawbacks with respect to their docking accuracy, ranking accuracy and time consumption so a general conclusion cannot be drawn. Moreover, users don't always consider sufficient diversity in their test sets which results in certain programs to outperform others. In this review, the prime focus has been laid on the challenges of docking and troubleshooters in existing programs, underlying algorithmic background of docking, preferences regarding the use of docking programs for best results illustrated with examples, comparison of performance for existing tools and algorithms, state of art in docking, recent trends of diseases and current drug industries, evidence from clinical trials and post-marketing surveillance are discussed. These aspects of the molecular drug designing paradigm are quite controversial and challenging and this review would be an asset to the bioinformatics and drug designing communities.
Collapse
Affiliation(s)
- Surovi Saikia
- Natural Products Chemistry Group, CSIR North East Institute of Science & Technology, Jorhat-785006, Assam, India
| | - Manobjyoti Bordoloi
- Natural Products Chemistry Group, CSIR North East Institute of Science & Technology, Jorhat-785006, Assam, India
| |
Collapse
|
27
|
Wang Z, Sun H, Shen C, Hu X, Gao J, Li D, Cao D, Hou T. Combined strategies in structure-based virtual screening. Phys Chem Chem Phys 2020; 22:3149-3159. [PMID: 31995074 DOI: 10.1039/c9cp06303j] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The identification and optimization of lead compounds are inalienable components in drug design and discovery pipelines. As a powerful computational approach for the identification of hits with novel structural scaffolds, structure-based virtual screening (SBVS) has exhibited a remarkably increasing influence in the early stages of drug discovery. During the past decade, a variety of techniques and algorithms have been proposed and tested with different purposes in the scope of SBVS. Although SBVS has been a common and proven technology, it still shows some challenges and problems that are needed to be addressed, where the negative influence regardless of protein flexibility and the inaccurate prediction of binding affinity are the two major challenges. Here, focusing on these difficulties, we summarize a series of combined strategies or workflows developed by our group and others. Furthermore, several representative successful applications from recent publications are also discussed to demonstrate the effectiveness of the combined SBVS strategies in drug discovery campaigns.
Collapse
Affiliation(s)
- Zhe Wang
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, P. R. China.
| | - Huiyong Sun
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, P. R. China.
| | - Chao Shen
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, P. R. China.
| | - Xueping Hu
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, P. R. China.
| | - Junbo Gao
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, P. R. China.
| | - Dan Li
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, P. R. China.
| | - Dongsheng Cao
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410004, Hunan, P. R. China.
| | - Tingjun Hou
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, P. R. China.
| |
Collapse
|
28
|
Echeverría E, Velez Rueda AJ, Cabrera M, Juritz E, Burghi V, Fabián L, Davio C, Lorenzano Menna P, Fernández NC. Identification of inhibitors of the RGS homology domain of GRK2 by docking-based virtual screening. Life Sci 2019; 239:116872. [DOI: 10.1016/j.lfs.2019.116872] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 09/10/2019] [Accepted: 09/11/2019] [Indexed: 01/25/2023]
|
29
|
Molecular Docking Analysis of 120 Potential HPV Therapeutic Epitopes Using a New Analytical Method. Int J Pept Res Ther 2019. [DOI: 10.1007/s10989-019-09985-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
30
|
Coupling enhanced sampling of the apo-receptor with template-based ligand conformers selection: performance in pose prediction in the D3R Grand Challenge 4. J Comput Aided Mol Des 2019; 34:149-162. [DOI: 10.1007/s10822-019-00244-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 10/30/2019] [Indexed: 12/20/2022]
|
31
|
Molecular basis for the different interactions of congeneric substrates with the polyspecific transporter AcrB. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2019; 1861:1397-1408. [DOI: 10.1016/j.bbamem.2019.05.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 12/20/2018] [Accepted: 01/06/2019] [Indexed: 12/20/2022]
|
32
|
Lee ACL, Harris JL, Khanna KK, Hong JH. A Comprehensive Review on Current Advances in Peptide Drug Development and Design. Int J Mol Sci 2019; 20:ijms20102383. [PMID: 31091705 PMCID: PMC6566176 DOI: 10.3390/ijms20102383] [Citation(s) in RCA: 413] [Impact Index Per Article: 68.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 05/09/2019] [Accepted: 05/10/2019] [Indexed: 11/16/2022] Open
Abstract
Protein-protein interactions (PPIs) execute many fundamental cellular functions and have served as prime drug targets over the last two decades. Interfering intracellular PPIs with small molecules has been extremely difficult for larger or flat binding sites, as antibodies cannot cross the cell membrane to reach such target sites. In recent years, peptides smaller size and balance of conformational rigidity and flexibility have made them promising candidates for targeting challenging binding interfaces with satisfactory binding affinity and specificity. Deciphering and characterizing peptide-protein recognition mechanisms is thus central for the invention of peptide-based strategies to interfere with endogenous protein interactions, or improvement of the binding affinity and specificity of existing approaches. Importantly, a variety of computation-aided rational designs for peptide therapeutics have been developed, which aim to deliver comprehensive docking for peptide-protein interaction interfaces. Over 60 peptides have been approved and administrated globally in clinics. Despite this, advances in various docking models are only on the merge of making their contribution to peptide drug development. In this review, we provide (i) a holistic overview of peptide drug development and the fundamental technologies utilized to date, and (ii) an updated review on key developments of computational modeling of peptide-protein interactions (PepPIs) with an aim to assist experimental biologists exploit suitable docking methods to advance peptide interfering strategies against PPIs.
Collapse
Affiliation(s)
- Andy Chi-Lung Lee
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia.
- Radiation Biology Research Center, Institute for Radiological Research, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 333, Taiwan.
- Department of Radiation Oncology, Chang Gung Memorial Hospital, Linkou 333, Taiwan.
| | | | - Kum Kum Khanna
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia.
| | - Ji-Hong Hong
- Radiation Biology Research Center, Institute for Radiological Research, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 333, Taiwan.
- Department of Radiation Oncology, Chang Gung Memorial Hospital, Linkou 333, Taiwan.
| |
Collapse
|
33
|
Ding F, Peng W. Probing the local conformational flexibility in receptor recognition: mechanistic insight from an atomic-scale investigation. RSC Adv 2019; 9:13968-13980. [PMID: 35519308 PMCID: PMC9064033 DOI: 10.1039/c9ra01906e] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 04/28/2019] [Indexed: 12/13/2022] Open
Abstract
Inherent protein conformational flexibility is important for biomolecular recognition, but this critical property is often neglected in several studies. This event can lead to large deviations in the research results. In the current contribution, we disclose the effects of the local conformational flexibility on receptor recognition by using an atomic-scale computational method. The results indicated that both static and dynamic reaction modes have noticeable differences, and these originated from the structural features of the protein molecules. Dynamic interaction results displayed that the structural stability and conformational flexibility of the proteins had a significant influence on the recognition processes. This point related closely to the characteristics of the flexible loop regions where bixin located within the protein structures. The energy decomposition analyses and circular dichroism results validated the rationality of the recognition studies. More importantly, the conformational and energy changes of some residues around the bixin binding domain were found to be vital to biological reactions. These microscopic findings clarified the nature of the phenomenon that the local conformational flexibility could intervene in receptor recognition. Obviously, this report may provide biophysical evidence for the exploration of the structure-function relationships of the biological receptors in the human body.
Collapse
Affiliation(s)
- Fei Ding
- School of Environmental Science and Engineering, Chang'an University Xi'an 710064 China
- Key Laboratory of Subsurface Hydrology and Ecological Effect in Arid Region of Ministry of Education, Chang'an University No. 126 Yanta Road, Yanta District Xi'an 710064 China
| | - Wei Peng
- College of Chemistry and Chemical Engineering, Xiamen University Xiamen 361005 China +86-29-87092367 +86-29-87092367
- Department of Chemistry, China Agricultural University Beijing 100193 China
| |
Collapse
|
34
|
Basciu A, Malloci G, Pietrucci F, Bonvin AMJJ, Vargiu AV. Holo-like and Druggable Protein Conformations from Enhanced Sampling of Binding Pocket Volume and Shape. J Chem Inf Model 2019; 59:1515-1528. [PMID: 30883122 DOI: 10.1021/acs.jcim.8b00730] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Understanding molecular recognition of small molecules by proteins in atomistic detail is key for drug design. Molecular docking is a widely used computational method to mimic ligand-protein association in silico. However, predicting conformational changes occurring in proteins upon ligand binding is still a major challenge. Ensemble docking approaches address this issue by considering a set of different conformations of the protein obtained either experimentally or from computer simulations, e.g., molecular dynamics. However, holo structures prone to host (the correct) ligands are generally poorly sampled by standard molecular dynamics simulations of the apo protein. In order to address this limitation, we introduce a computational approach based on metadynamics simulations called ensemble docking with enhanced sampling of pocket shape (EDES) that allows holo-like conformations of proteins to be generated by exploiting only their apo structures. This is achieved by defining a set of collective variables that effectively sample different shapes of the binding site, ultimately mimicking the steric effect due to the ligand. We assessed the method on three challenging proteins undergoing different extents of conformational changes upon ligand binding. In all cases our protocol generates a significant fraction of structures featuring a low RMSD from the experimental holo geometry. Moreover, ensemble docking calculations using those conformations yielded in all cases native-like poses among the top-ranked ones.
Collapse
Affiliation(s)
- Andrea Basciu
- Dipartimento di Fisica , Università di Cagliari, Cittadella Universitaria , I- 09042 Monserrato (CA) , Italy
| | - Giuliano Malloci
- Dipartimento di Fisica , Università di Cagliari, Cittadella Universitaria , I- 09042 Monserrato (CA) , Italy
| | - Fabio Pietrucci
- Sorbonne Université , Muséum National d'Histoire Naturelle, UMR CNRS 7590, IRD, Institut de Minéralogie, de Physique des Matériaux et de Cosmochimie, IMPMC , F-75005 Paris , France
| | - Alexandre M J J Bonvin
- Bijvoet Center for Biomolecular Research, Faculty of Science - Chemistry , Utrecht University , Padualaan 8 , 3584 CH Utrecht , The Netherlands
| | - Attilio V Vargiu
- Dipartimento di Fisica , Università di Cagliari, Cittadella Universitaria , I- 09042 Monserrato (CA) , Italy.,Bijvoet Center for Biomolecular Research, Faculty of Science - Chemistry , Utrecht University , Padualaan 8 , 3584 CH Utrecht , The Netherlands
| |
Collapse
|
35
|
Abstract
Drugs modulate disease states through their actions on targets in the body. Determining these targets aids the focused development of new treatments, and helps to better characterize those already employed. One means of accomplishing this is through the deployment of in silico methodologies, harnessing computational analytical and predictive power to produce educated hypotheses for experimental verification. Here, we provide an overview of the current state of the art, describe some of the well-established methods in detail, and reflect on how they, and emerging technologies promoting the incorporation of complex and heterogeneous data-sets, can be employed to improve our understanding of (poly)pharmacology.
Collapse
Affiliation(s)
- Ryan Byrne
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH), Zurich, Switzerland
| | - Gisbert Schneider
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH), Zurich, Switzerland.
| |
Collapse
|
36
|
Guedes IA, Pereira FSS, Dardenne LE. Empirical Scoring Functions for Structure-Based Virtual Screening: Applications, Critical Aspects, and Challenges. Front Pharmacol 2018; 9:1089. [PMID: 30319422 PMCID: PMC6165880 DOI: 10.3389/fphar.2018.01089] [Citation(s) in RCA: 160] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Accepted: 09/07/2018] [Indexed: 12/19/2022] Open
Abstract
Structure-based virtual screening (VS) is a widely used approach that employs the knowledge of the three-dimensional structure of the target of interest in the design of new lead compounds from large-scale molecular docking experiments. Through the prediction of the binding mode and affinity of a small molecule within the binding site of the target of interest, it is possible to understand important properties related to the binding process. Empirical scoring functions are widely used for pose and affinity prediction. Although pose prediction is performed with satisfactory accuracy, the correct prediction of binding affinity is still a challenging task and crucial for the success of structure-based VS experiments. There are several efforts in distinct fronts to develop even more sophisticated and accurate models for filtering and ranking large libraries of compounds. This paper will cover some recent successful applications and methodological advances, including strategies to explore the ligand entropy and solvent effects, training with sophisticated machine-learning techniques, and the use of quantum mechanics. Particular emphasis will be given to the discussion of critical aspects and further directions for the development of more accurate empirical scoring functions.
Collapse
Affiliation(s)
- Isabella A Guedes
- Grupo de Modelagem Molecular em Sistemas Biológicos, Laboratório Nacional de Computação Científica, Petrópolis, Brazil
| | - Felipe S S Pereira
- Grupo de Modelagem Molecular em Sistemas Biológicos, Laboratório Nacional de Computação Científica, Petrópolis, Brazil
| | - Laurent E Dardenne
- Grupo de Modelagem Molecular em Sistemas Biológicos, Laboratório Nacional de Computação Científica, Petrópolis, Brazil
| |
Collapse
|
37
|
Pinzi L, Caporuscio F, Rastelli G. Selection of protein conformations for structure-based polypharmacology studies. Drug Discov Today 2018; 23:1889-1896. [PMID: 30099123 DOI: 10.1016/j.drudis.2018.08.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 08/03/2018] [Accepted: 08/06/2018] [Indexed: 11/29/2022]
Abstract
Several drugs exert their therapeutic effect through the modulation of multiple targets. Structure-based approaches hold great promise for identifying compounds with the desired polypharmacological profiles. These methods use knowledge of the protein binding sites to identify stereoelectronically complementary ligands. The selection of the most suitable protein conformations to be used in the design process is vital, especially for multitarget drug design in which the same ligand has to be accommodated in multiple binding pockets. Herein, we focus on currently available techniques for the selection of the most suitable protein conformations for multitarget drug design, compare the potential advantages and limitations of each method, and comment on how their combination could help in polypharmacology drug design.
Collapse
Affiliation(s)
- Luca Pinzi
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103, 41125, Modena, Italy
| | - Fabiana Caporuscio
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103, 41125, Modena, Italy
| | - Giulio Rastelli
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103, 41125, Modena, Italy.
| |
Collapse
|
38
|
De Paris R, Vahl Quevedo C, Ruiz DD, Gargano F, de Souza ON. A selective method for optimizing ensemble docking-based experiments on an InhA Fully-Flexible receptor model. BMC Bioinformatics 2018; 19:235. [PMID: 29929475 PMCID: PMC6013854 DOI: 10.1186/s12859-018-2222-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 05/29/2018] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND In the rational drug design process, an ensemble of conformations obtained from a molecular dynamics simulation plays a crucial role in docking experiments. Some studies have found that Fully-Flexible Receptor (FFR) models predict realistic binding energy accurately and improve scoring to enhance selectiveness. At the same time, methods have been proposed to reduce the high computational costs involved in considering the explicit flexibility of proteins in receptor-ligand docking. This study introduces a novel method to optimize ensemble docking-based experiments by reducing the size of an InhA FFR model at docking runtime and scaling docking workflow invocations on cloud virtual machines. RESULTS First, in order to find the most affordable cost-benefit pool of virtual machines, we evaluated the performance of the docking workflow invocations in different configurations of Azure instances. Second, we validated the gains obtained by the proposed method based on the quality of the Reduced Fully-Flexible Receptor (RFFR) models produced using AutoDock4.2. The analyses show that the proposed method reduced the model size by approximately 50% while covering at least 86% of the best docking results from the 74 ligands tested. Third, we tested our novel method using AutoDock Vina, a different docking software, and showed the positive accuracy achieved in the resulting RFFR models. Finally, our results demonstrated that the method proposed optimized ensemble docking experiments and is applicable to different docking software. In addition, it detected new binding modes, which would be unreachable if employing only the rigid structure used to generate the InhA FFR model. CONCLUSIONS Our results showed that the selective method is a valuable strategy for optimizing ensemble docking-based experiments using different docking software. The RFFR models produced by discarding non-promising snapshots from the original model are accurately shaped for a larger number of ligands, and the elapsed time spent in the ensemble docking experiments are considerably reduced.
Collapse
Affiliation(s)
- Renata De Paris
- Business Intelligence and Machine Learning Research Group—GPIN, School of Technology, PUCRS, Av. Ipiranga, 6681, Building 32, Room 628, Porto Alegre, RS, Brazil
| | - Christian Vahl Quevedo
- Business Intelligence and Machine Learning Research Group—GPIN, School of Technology, PUCRS, Av. Ipiranga, 6681, Building 32, Room 628, Porto Alegre, RS, Brazil
| | - Duncan D. Ruiz
- Business Intelligence and Machine Learning Research Group—GPIN, School of Technology, PUCRS, Av. Ipiranga, 6681, Building 32, Room 628, Porto Alegre, RS, Brazil
| | - Furia Gargano
- Bioinformatics and Biossystems Modeling and Simulation Lab—LABIO, School of Technology, PUCRS, Av. Ipiranga, 6681, Building 32, Room 602, Porto Alegre, RS, Brazil
| | - Osmar Norberto de Souza
- Bioinformatics and Biossystems Modeling and Simulation Lab—LABIO, School of Technology, PUCRS, Av. Ipiranga, 6681, Building 32, Room 602, Porto Alegre, RS, Brazil
| |
Collapse
|
39
|
Ciemny M, Kurcinski M, Kamel K, Kolinski A, Alam N, Schueler-Furman O, Kmiecik S. Protein-peptide docking: opportunities and challenges. Drug Discov Today 2018; 23:1530-1537. [PMID: 29733895 DOI: 10.1016/j.drudis.2018.05.006] [Citation(s) in RCA: 167] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 03/20/2018] [Accepted: 05/02/2018] [Indexed: 12/31/2022]
Abstract
Peptides have recently attracted much attention as promising drug candidates. Rational design of peptide-derived therapeutics usually requires structural characterization of the underlying protein-peptide interaction. Given that experimental characterization can be difficult, reliable computational tools are needed. In recent years, a variety of approaches have been developed for 'protein-peptide docking', that is, predicting the structure of the protein-peptide complex, starting from the protein structure and the peptide sequence, including variable degrees of information about the peptide binding site and/or conformation. In this review, we provide an overview of protein-peptide docking methods and outline their capabilities, limitations, and applications in structure-based drug design. Key challenges are also briefly discussed, such as modeling of large-scale conformational changes upon binding, scoring of predicted models, and optimal inclusion of varied types of experimental data and theoretical predictions into an integrative modeling process.
Collapse
Affiliation(s)
- Maciej Ciemny
- Biological and Chemical Research Center, Faculty of Chemistry, University of Warsaw, Warsaw, Poland; Faculty of Physics, University of Warsaw, Warsaw, Poland
| | - Mateusz Kurcinski
- Biological and Chemical Research Center, Faculty of Chemistry, University of Warsaw, Warsaw, Poland
| | - Karol Kamel
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| | - Andrzej Kolinski
- Biological and Chemical Research Center, Faculty of Chemistry, University of Warsaw, Warsaw, Poland
| | - Nawsad Alam
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - Ora Schueler-Furman
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - Sebastian Kmiecik
- Biological and Chemical Research Center, Faculty of Chemistry, University of Warsaw, Warsaw, Poland.
| |
Collapse
|
40
|
Iglesias J, Saen‐oon S, Soliva R, Guallar V. Computational structure‐based drug design: Predicting target flexibility. WILEY INTERDISCIPLINARY REVIEWS-COMPUTATIONAL MOLECULAR SCIENCE 2018. [DOI: 10.1002/wcms.1367] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
| | | | | | - Victor Guallar
- Life Science DepartmentBarcelonaSpain
- ICREA, Passeig Lluís Companys 23BarcelonaSpain
| |
Collapse
|
41
|
Bertazzo M, Bernetti M, Recanatini M, Masetti M, Cavalli A. Fully Flexible Docking via Reaction-Coordinate-Independent Molecular Dynamics Simulations. J Chem Inf Model 2018; 58:490-500. [PMID: 29378136 DOI: 10.1021/acs.jcim.7b00674] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Predicting the geometry of protein-ligand binding complexes is of primary importance for structure-based drug discovery. Molecular dynamics (MD) is emerging as a reliable computational tool for use in conjunction with, or an alternative to, docking methods. However, simulating the protein-ligand binding process often requires very expensive simulations. This drastically limits the practical application of MD-based approaches. Here, we propose a general framework to accelerate the generation of putative protein-ligand binding modes using potential-scaled MD simulations. The proposed dynamical protocol has been applied to two pharmaceutically relevant systems (GSK-3β and the N-terminal domain of HSP90α). Our approach is fully independent of any predefined reaction coordinate (or collective variable). It identified the correct binding mode of several ligands and can thus save valuable computational time in dynamic docking simulations.
Collapse
Affiliation(s)
- Martina Bertazzo
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-Università di Bologna , Via Belmeloro 6, 40126, Bologna, Italy.,CompuNet, Istituto Italiano di Tecnologia , Via Morego 30, 16163, Genova, Italy
| | - Mattia Bernetti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-Università di Bologna , Via Belmeloro 6, 40126, Bologna, Italy.,CompuNet, Istituto Italiano di Tecnologia , Via Morego 30, 16163, Genova, Italy
| | - Maurizio Recanatini
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-Università di Bologna , Via Belmeloro 6, 40126, Bologna, Italy
| | - Matteo Masetti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-Università di Bologna , Via Belmeloro 6, 40126, Bologna, Italy
| | - Andrea Cavalli
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-Università di Bologna , Via Belmeloro 6, 40126, Bologna, Italy.,CompuNet, Istituto Italiano di Tecnologia , Via Morego 30, 16163, Genova, Italy
| |
Collapse
|
42
|
Vistoli G, Pedretti A, Mazzolari A, Testa B. Approaching Pharmacological Space: Events and Components. Methods Mol Biol 2018; 1800:245-274. [PMID: 29934897 DOI: 10.1007/978-1-4939-7899-1_12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
With a view to introducing the concept of pharmacological space and its potential applications in investigating and predicting the toxic mechanisms of xenobiotics, this opening chapter describes the logical relations between conformational behavior, physicochemical properties and binding spaces, which are seen as the three key elements composing the pharmacological space. While the concept of conformational space is routinely used to encode molecular flexibility, the concepts of property spaces and, particularly, of binding spaces are more innovative. Indeed, their descriptors can find fruitful applications (a) in describing the dynamic adaptability a given ligand experiences when inserted into a specific environment, and (b) in parameterizing the flexibility a ligand retains when bound to a biological target. Overall, these descriptors can conveniently account for the often disregarded entropic factors and as such they prove successful when inserted in ligand- or structure-based predictive models. Notably, and although binding space parameters can clearly be derived from MD simulations, the chapter will illustrate how docking calculations, despite their static nature, are able to evaluate ligand's flexibility by analyzing several poses for each ligand. Such an approach, which represents the founding core of the binding space concept, can find various applications in which the related descriptors show an impressive enhancing effect on the statistical performances of the resulting predictive models.
Collapse
Affiliation(s)
- Giulio Vistoli
- Dipartimento di Scienze Farmaceutiche Università degli Studi di Milano, Milan, Italy.
| | - Alessandro Pedretti
- Dipartimento di Scienze Farmaceutiche Università degli Studi di Milano, Milan, Italy
| | - Angelica Mazzolari
- Dipartimento di Scienze Farmaceutiche Università degli Studi di Milano, Milan, Italy
| | | |
Collapse
|
43
|
Noresson AL, Aurelius O, Öberg CT, Engström O, Sundin AP, Håkansson M, Stenström O, Akke M, Logan DT, Leffler H, Nilsson UJ. Designing interactions by control of protein-ligand complex conformation: tuning arginine-arene interaction geometry for enhanced electrostatic protein-ligand interactions. Chem Sci 2017; 9:1014-1021. [PMID: 29675148 PMCID: PMC5883865 DOI: 10.1039/c7sc04749e] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 12/01/2017] [Indexed: 01/13/2023] Open
Abstract
3-Benzamido-2-O-sulfo-galactosides can be designed to control protein conformation into forming entropically favourable galectin-3-arginine salt bridges with ligand sulfates.
We investigated galectin-3 binding to 3-benzamido-2-O-sulfo-galactoside and -thiodigalactoside ligands using a combination of site-specific mutagenesis, X-ray crystallography, computational approaches, and binding thermodynamics measurements. The results reveal a conformational variability in a surface-exposed arginine (R144) side chain in response to different aromatic C3-substituents of bound galactoside-based ligands. Fluorinated C3-benzamido substituents induced a shift in the side-chain conformation of R144 to allow for an entropically favored electrostatic interaction between its guanidine group and the 2-O-sulfate of the ligand. By contrast, binding of ligands with non-fluorinated substituents did not trigger a conformational change of R144. Hence, a sulfate–arginine electrostatic interaction can be tuned by the choice of ligand C3-benzamido structures to favor specific interaction modes and geometries. These results have important general implications for ligand design, as the proper choice of arginine–aromatic interacting partners opens up for ligand-controlled protein conformation that in turn may be systematically exploited in ligand design.
Collapse
Affiliation(s)
- A-L Noresson
- Centre for Analysis and Synthesis , Department of Chemistry , Lund University , Box 124 , SE-221 00 Lund , Sweden .
| | - O Aurelius
- Section for Biochemistry and Structural Biology , Center for Molecular Protein Science , Department of Chemistry , Lund University , Box 124 , SE-221 00 Lund , Sweden
| | - C T Öberg
- Centre for Analysis and Synthesis , Department of Chemistry , Lund University , Box 124 , SE-221 00 Lund , Sweden .
| | - O Engström
- Centre for Analysis and Synthesis , Department of Chemistry , Lund University , Box 124 , SE-221 00 Lund , Sweden .
| | - A P Sundin
- Centre for Analysis and Synthesis , Department of Chemistry , Lund University , Box 124 , SE-221 00 Lund , Sweden .
| | - M Håkansson
- SARomics Biostructures AB , Medicon Village , SE-223 81 Lund , Sweden
| | - O Stenström
- Biophysical Chemistry , Center for Molecular Protein Science , Department of Chemistry , Lund University , Box 124 , SE-221 00 Lund , Sweden
| | - M Akke
- Biophysical Chemistry , Center for Molecular Protein Science , Department of Chemistry , Lund University , Box 124 , SE-221 00 Lund , Sweden
| | - D T Logan
- Section for Biochemistry and Structural Biology , Center for Molecular Protein Science , Department of Chemistry , Lund University , Box 124 , SE-221 00 Lund , Sweden.,SARomics Biostructures AB , Medicon Village , SE-223 81 Lund , Sweden
| | - H Leffler
- Department of Laboratory Medicine , Section MIG , Lund University , Sölvegatan 23, SE-223 62 , Lund , Sweden
| | - U J Nilsson
- Centre for Analysis and Synthesis , Department of Chemistry , Lund University , Box 124 , SE-221 00 Lund , Sweden .
| |
Collapse
|
44
|
Gioia D, Bertazzo M, Recanatini M, Masetti M, Cavalli A. Dynamic Docking: A Paradigm Shift in Computational Drug Discovery. Molecules 2017; 22:2029. [PMID: 29165360 PMCID: PMC6150405 DOI: 10.3390/molecules22112029] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 11/18/2017] [Accepted: 11/19/2017] [Indexed: 12/18/2022] Open
Abstract
Molecular docking is the methodology of choice for studying in silico protein-ligand binding and for prioritizing compounds to discover new lead candidates. Traditional docking simulations suffer from major limitations, mostly related to the static or semi-flexible treatment of ligands and targets. They also neglect solvation and entropic effects, which strongly limits their predictive power. During the last decade, methods based on full atomistic molecular dynamics (MD) have emerged as a valid alternative for simulating macromolecular complexes. In principle, compared to traditional docking, MD allows the full exploration of drug-target recognition and binding from both the mechanistic and energetic points of view (dynamic docking). Binding and unbinding kinetic constants can also be determined. While dynamic docking is still too computationally expensive to be routinely used in fast-paced drug discovery programs, the advent of faster computing architectures and advanced simulation methodologies are changing this scenario. It is feasible that dynamic docking will replace static docking approaches in the near future, leading to a major paradigm shift in in silico drug discovery. Against this background, we review the key achievements that have paved the way for this progress.
Collapse
Affiliation(s)
- Dario Gioia
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-Universita' di Bologna, via Belmeloro 6, I-40126 Bologna, Italy.
| | - Martina Bertazzo
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-Universita' di Bologna, via Belmeloro 6, I-40126 Bologna, Italy.
- Computational Sciences, Istituto Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy.
| | - Maurizio Recanatini
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-Universita' di Bologna, via Belmeloro 6, I-40126 Bologna, Italy.
| | - Matteo Masetti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-Universita' di Bologna, via Belmeloro 6, I-40126 Bologna, Italy.
| | - Andrea Cavalli
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-Universita' di Bologna, via Belmeloro 6, I-40126 Bologna, Italy.
- Computational Sciences, Istituto Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy.
| |
Collapse
|
45
|
Sahlgren C, Meinander A, Zhang H, Cheng F, Preis M, Xu C, Salminen TA, Toivola D, Abankwa D, Rosling A, Karaman DŞ, Salo-Ahen OMH, Österbacka R, Eriksson JE, Willför S, Petre I, Peltonen J, Leino R, Johnson M, Rosenholm J, Sandler N. Tailored Approaches in Drug Development and Diagnostics: From Molecular Design to Biological Model Systems. Adv Healthc Mater 2017; 6. [PMID: 28892296 DOI: 10.1002/adhm.201700258] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Revised: 05/04/2017] [Indexed: 12/13/2022]
Abstract
Approaches to increase the efficiency in developing drugs and diagnostics tools, including new drug delivery and diagnostic technologies, are needed for improved diagnosis and treatment of major diseases and health problems such as cancer, inflammatory diseases, chronic wounds, and antibiotic resistance. Development within several areas of research ranging from computational sciences, material sciences, bioengineering to biomedical sciences and bioimaging is needed to realize innovative drug development and diagnostic (DDD) approaches. Here, an overview of recent progresses within key areas that can provide customizable solutions to improve processes and the approaches taken within DDD is provided. Due to the broadness of the area, unfortunately all relevant aspects such as pharmacokinetics of bioactive molecules and delivery systems cannot be covered. Tailored approaches within (i) bioinformatics and computer-aided drug design, (ii) nanotechnology, (iii) novel materials and technologies for drug delivery and diagnostic systems, and (iv) disease models to predict safety and efficacy of medicines under development are focused on. Current developments and challenges ahead are discussed. The broad scope reflects the multidisciplinary nature of the field of DDD and aims to highlight the convergence of biological, pharmaceutical, and medical disciplines needed to meet the societal challenges of the 21st century.
Collapse
Affiliation(s)
- Cecilia Sahlgren
- Faculty of Science and Engineering; Cell Biology; Åbo Akademi University; FI-20520 Turku Finland
- Turku Centre for Biotechnology; Åbo Akademi University and University of Turku; FI-20520 Turku Finland
- Department of Biomedical Engineering; Technical University of Eindhoven; 5613 DR Eindhoven Netherlands
| | - Annika Meinander
- Faculty of Science and Engineering; Cell Biology; Åbo Akademi University; FI-20520 Turku Finland
| | - Hongbo Zhang
- Faculty of Science and Engineering; Pharmaceutical Sciences Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| | - Fang Cheng
- Faculty of Science and Engineering; Cell Biology; Åbo Akademi University; FI-20520 Turku Finland
| | - Maren Preis
- Faculty of Science and Engineering; Pharmaceutical Sciences Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| | - Chunlin Xu
- Faculty of Science and Engineering; Natural Materials Technology; Åbo Akademi University; FI-20500 Turku Finland
| | - Tiina A. Salminen
- Faculty of Science and Engineering; Structural Bioinformatics Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| | - Diana Toivola
- Faculty of Science and Engineering; Cell Biology; Åbo Akademi University; FI-20520 Turku Finland
- Turku Center for Disease Modeling; University of Turku; FI-20520 Turku Finland
| | - Daniel Abankwa
- Department of Biomedical Engineering; Technical University of Eindhoven; 5613 DR Eindhoven Netherlands
| | - Ari Rosling
- Faculty of Science and Engineering; Polymer Technologies; Åbo Akademi University; FI-20500 Turku Finland
| | - Didem Şen Karaman
- Faculty of Science and Engineering; Pharmaceutical Sciences Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| | - Outi M. H. Salo-Ahen
- Faculty of Science and Engineering; Pharmaceutical Sciences Laboratory; Åbo Akademi University; FI-20520 Turku Finland
- Faculty of Science and Engineering; Structural Bioinformatics Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| | - Ronald Österbacka
- Faculty of Science and Engineering; Physics; Åbo Akademi University; FI-20500 Turku Finland
| | - John E. Eriksson
- Faculty of Science and Engineering; Cell Biology; Åbo Akademi University; FI-20520 Turku Finland
- Turku Centre for Biotechnology; Åbo Akademi University and University of Turku; FI-20520 Turku Finland
| | - Stefan Willför
- Faculty of Science and Engineering; Natural Materials Technology; Åbo Akademi University; FI-20500 Turku Finland
| | - Ion Petre
- Faculty of Science and Engineering; Computer Science; Åbo Akademi University; FI-20500 Turku Finland
| | - Jouko Peltonen
- Faculty of Science and Engineering; Physical Chemistry; Åbo Akademi University; FI-20500 Turku Finland
| | - Reko Leino
- Faculty of Science and Engineering; Organic Chemistry; Johan Gadolin Process Chemistry Centre; Åbo Akademi University; FI-20500 Turku Finland
| | - Mark Johnson
- Faculty of Science and Engineering; Structural Bioinformatics Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| | - Jessica Rosenholm
- Faculty of Science and Engineering; Pharmaceutical Sciences Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| | - Niklas Sandler
- Faculty of Science and Engineering; Pharmaceutical Sciences Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| |
Collapse
|
46
|
Sahlgren C, Meinander A, Zhang H, Cheng F, Preis M, Xu C, Salminen TA, Toivola D, Abankwa D, Rosling A, Karaman DŞ, Salo-Ahen OMH, Österbacka R, Eriksson JE, Willför S, Petre I, Peltonen J, Leino R, Johnson M, Rosenholm J, Sandler N. Tailored Approaches in Drug Development and Diagnostics: From Molecular Design to Biological Model Systems. Adv Healthc Mater 2017. [DOI: 10.1002/adhm.201700258 10.1002/adhm.201700258] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2023]
Affiliation(s)
- Cecilia Sahlgren
- Faculty of Science and Engineering; Cell Biology; Åbo Akademi University; FI-20520 Turku Finland
- Turku Centre for Biotechnology; Åbo Akademi University and University of Turku; FI-20520 Turku Finland
- Department of Biomedical Engineering; Technical University of Eindhoven; 5613 DR Eindhoven Netherlands
| | - Annika Meinander
- Faculty of Science and Engineering; Cell Biology; Åbo Akademi University; FI-20520 Turku Finland
| | - Hongbo Zhang
- Faculty of Science and Engineering; Pharmaceutical Sciences Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| | - Fang Cheng
- Faculty of Science and Engineering; Cell Biology; Åbo Akademi University; FI-20520 Turku Finland
| | - Maren Preis
- Faculty of Science and Engineering; Pharmaceutical Sciences Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| | - Chunlin Xu
- Faculty of Science and Engineering; Natural Materials Technology; Åbo Akademi University; FI-20500 Turku Finland
| | - Tiina A. Salminen
- Faculty of Science and Engineering; Structural Bioinformatics Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| | - Diana Toivola
- Faculty of Science and Engineering; Cell Biology; Åbo Akademi University; FI-20520 Turku Finland
- Turku Center for Disease Modeling; University of Turku; FI-20520 Turku Finland
| | - Daniel Abankwa
- Department of Biomedical Engineering; Technical University of Eindhoven; 5613 DR Eindhoven Netherlands
| | - Ari Rosling
- Faculty of Science and Engineering; Polymer Technologies; Åbo Akademi University; FI-20500 Turku Finland
| | - Didem Şen Karaman
- Faculty of Science and Engineering; Pharmaceutical Sciences Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| | - Outi M. H. Salo-Ahen
- Faculty of Science and Engineering; Pharmaceutical Sciences Laboratory; Åbo Akademi University; FI-20520 Turku Finland
- Faculty of Science and Engineering; Structural Bioinformatics Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| | - Ronald Österbacka
- Faculty of Science and Engineering; Physics; Åbo Akademi University; FI-20500 Turku Finland
| | - John E. Eriksson
- Faculty of Science and Engineering; Cell Biology; Åbo Akademi University; FI-20520 Turku Finland
- Turku Centre for Biotechnology; Åbo Akademi University and University of Turku; FI-20520 Turku Finland
| | - Stefan Willför
- Faculty of Science and Engineering; Natural Materials Technology; Åbo Akademi University; FI-20500 Turku Finland
| | - Ion Petre
- Faculty of Science and Engineering; Computer Science; Åbo Akademi University; FI-20500 Turku Finland
| | - Jouko Peltonen
- Faculty of Science and Engineering; Physical Chemistry; Åbo Akademi University; FI-20500 Turku Finland
| | - Reko Leino
- Faculty of Science and Engineering; Organic Chemistry; Johan Gadolin Process Chemistry Centre; Åbo Akademi University; FI-20500 Turku Finland
| | - Mark Johnson
- Faculty of Science and Engineering; Structural Bioinformatics Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| | - Jessica Rosenholm
- Faculty of Science and Engineering; Pharmaceutical Sciences Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| | - Niklas Sandler
- Faculty of Science and Engineering; Pharmaceutical Sciences Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| |
Collapse
|
47
|
Motta S, Bonati L. Modeling Binding with Large Conformational Changes: Key Points in Ensemble-Docking Approaches. J Chem Inf Model 2017; 57:1563-1578. [DOI: 10.1021/acs.jcim.7b00125] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Stefano Motta
- Department of Earth and Environmental
Sciences, University of Milano-Bicocca, Piazza della Scienza 1, 20126 Milan, Italy
| | - Laura Bonati
- Department of Earth and Environmental
Sciences, University of Milano-Bicocca, Piazza della Scienza 1, 20126 Milan, Italy
| |
Collapse
|
48
|
Sulimov AV, Zheltkov DA, Oferkin IV, Kutov DC, Katkova EV, Tyrtyshnikov EE, Sulimov VB. Evaluation of the novel algorithm of flexible ligand docking with moveable target-protein atoms. Comput Struct Biotechnol J 2017; 15:275-285. [PMID: 28377797 PMCID: PMC5367798 DOI: 10.1016/j.csbj.2017.02.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 02/28/2017] [Indexed: 11/28/2022] Open
Abstract
We present the novel docking algorithm based on the Tensor Train decomposition and the TT-Cross global optimization. The algorithm is applied to the docking problem with flexible ligand and moveable protein atoms. The energy of the protein-ligand complex is calculated in the frame of the MMFF94 force field in vacuum. The grid of precalculated energy potentials of probe ligand atoms in the field of the target protein atoms is not used. The energy of the protein-ligand complex for any given configuration is computed directly with the MMFF94 force field without any fitting parameters. The conformation space of the system coordinates is formed by translations and rotations of the ligand as a whole, by the ligand torsions and also by Cartesian coordinates of the selected target protein atoms. Mobility of protein and ligand atoms is taken into account in the docking process simultaneously and equally. The algorithm is realized in the novel parallel docking SOL-P program and results of its performance for a set of 30 protein-ligand complexes are presented. Dependence of the docking positioning accuracy is investigated as a function of parameters of the docking algorithm and the number of protein moveable atoms. It is shown that mobility of the protein atoms improves docking positioning accuracy. The SOL-P program is able to perform docking of a flexible ligand into the active site of the target protein with several dozens of protein moveable atoms: the native crystallized ligand pose is correctly found as the global energy minimum in the search space with 157 dimensions using 4700 CPU ∗ h at the Lomonosov supercomputer.
Collapse
Affiliation(s)
- Alexey V Sulimov
- Dimonta, Ltd, Nagornaya Street 15, Bldg. 8, Moscow 117186, Russia; Research Computer Center, Moscow State University, Leninskie Gory 1, Bldg. 4, Moscow 119992, Russia
| | - Dmitry A Zheltkov
- Faculty of Computational Mathematics and Cybernetics of Lomonosov Moscow State University, Leninskie Gory 1, Bldg. 52, Moscow 119992, Russia
| | - Igor V Oferkin
- Dimonta, Ltd, Nagornaya Street 15, Bldg. 8, Moscow 117186, Russia
| | - Danil C Kutov
- Dimonta, Ltd, Nagornaya Street 15, Bldg. 8, Moscow 117186, Russia; Research Computer Center, Moscow State University, Leninskie Gory 1, Bldg. 4, Moscow 119992, Russia
| | - Ekaterina V Katkova
- Dimonta, Ltd, Nagornaya Street 15, Bldg. 8, Moscow 117186, Russia; Research Computer Center, Moscow State University, Leninskie Gory 1, Bldg. 4, Moscow 119992, Russia
| | - Eugene E Tyrtyshnikov
- Faculty of Computational Mathematics and Cybernetics of Lomonosov Moscow State University, Leninskie Gory 1, Bldg. 52, Moscow 119992, Russia; Institute of Numerical Mathematics of Russian Academy of Sciences, Gubkin Street 8, Moscow, 119333, Russia
| | - Vladimir B Sulimov
- Dimonta, Ltd, Nagornaya Street 15, Bldg. 8, Moscow 117186, Russia; Research Computer Center, Moscow State University, Leninskie Gory 1, Bldg. 4, Moscow 119992, Russia
| |
Collapse
|
49
|
Bernetti M, Cavalli A, Mollica L. Protein-ligand (un)binding kinetics as a new paradigm for drug discovery at the crossroad between experiments and modelling. MEDCHEMCOMM 2017; 8:534-550. [PMID: 30108770 PMCID: PMC6072069 DOI: 10.1039/c6md00581k] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 01/25/2017] [Indexed: 12/14/2022]
Abstract
In the last three decades, protein and nucleic acid structure determination and comprehension of the mechanisms, leading to their physiological and pathological functions, have become a cornerstone of biomedical sciences. A deep understanding of the principles governing the fates of cells and tissue at the molecular level has been gained over the years, offering a solid basis for the rational design of drugs aimed at the pharmacological treatment of numerous diseases. Historically, affinity indicators (i.e. Kd and IC50/EC50) have been assumed to be valid indicators of the in vivo efficacy of a drug. However, recent studies pointed out that the kinetics of the drug-receptor binding process could be as important or even more important than affinity in determining the drug efficacy. This eventually led to a growing interest in the characterisation and prediction of the rate constants of protein-ligand association and dissociation. For instance, a drug with a longer residence time can kinetically select a given receptor over another, even if the affinity for both receptors is comparable, thus increasing its therapeutic index. Therefore, understanding the molecular features underlying binding and unbinding processes is of central interest towards the rational control of drug binding kinetics. In this review, we report the theoretical framework behind protein-ligand association and highlight the latest advances in the experimental and computational approaches exploited to investigate the binding kinetics.
Collapse
Affiliation(s)
- M Bernetti
- Department of Pharmacy and Biotechnology , University of Bologna , via Belmeloro 6 , 40126 Bologna , Italy
- CompuNet , Istituto Italiano di Tecnologia , via Morego 30 , 16163 Genova , Italy .
| | - A Cavalli
- Department of Pharmacy and Biotechnology , University of Bologna , via Belmeloro 6 , 40126 Bologna , Italy
- CompuNet , Istituto Italiano di Tecnologia , via Morego 30 , 16163 Genova , Italy .
| | - L Mollica
- CompuNet , Istituto Italiano di Tecnologia , via Morego 30 , 16163 Genova , Italy .
| |
Collapse
|
50
|
Peng W, Ding F. Enantioselective recognition of an isomeric ligand by a biomolecule: mechanistic insights into static and dynamic enantiomeric behavior and structural flexibility. MOLECULAR BIOSYSTEMS 2017; 13:2226-2234. [DOI: 10.1039/c7mb00378a] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Enantioselective biorecognition is a vital trigger that results in remarkable enantiomeric differences in the biochemical behavior of chiral substances.
Collapse
Affiliation(s)
- Wei Peng
- Laboratory for Computational Biochemistry & Molecular Design
- Department of Phytomedicine
- Qingdao Agricultural University
- Qingdao 266109
- China
| | - Fei Ding
- Laboratory for Computational Biochemistry & Molecular Design
- Department of Phytomedicine
- Qingdao Agricultural University
- Qingdao 266109
- China
| |
Collapse
|