1
|
Alexandrov V, Vilenchik M, Kantidze O, Tsutskiridze N, Kharchilava D, Lhewa P, Shishkin A, Gankin Y, Kirpich A. Novel Efficient Multistage Lead Optimization Pipeline Experimentally Validated for DYRK1B Selective Inhibitors. J Med Chem 2022; 65:13784-13792. [PMID: 36239428 PMCID: PMC9619999 DOI: 10.1021/acs.jmedchem.2c00988] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
![]()
In addition to general challenges in drug discovery such as the
identification of lead compounds in time- and cost-effective ways,
specific challenges also exist. Particularly, it is necessary to develop
pharmacological inhibitors that effectively discriminate between closely
related molecular targets. DYRK1B kinase is considered a valuable
target for cancer-specific mono- or combination chemotherapy; however,
the inhibition of its closely related DYRK1A kinase is not beneficial.
Existing inhibitors target both kinases with essentially the same
efficiency, and the unavailability of the DYRK1B crystal structure
makes the discovery of DYRK1B-specific inhibitors even more challenging.
Here, we propose a novel multi-stage compound discovery pipeline aimed
at in silico identification of both potent and selective
small molecules from a large set of initial candidates. The method
uses structure-based docking and ligand-based quantitative structure–activity
relationship modeling. This approach allowed us to identify lead and
runner-up small-molecule compounds targeting DYRK1B with high efficiency
and specificity.
Collapse
Affiliation(s)
- Vadim Alexandrov
- Liquid Algo LLC, Hopewell Junction, New York12533, United States
| | - Maria Vilenchik
- Felicitex Therapeutics, Natick, Massachusetts01760, United States
| | - Omar Kantidze
- Quantori LLC, Cambridge, Massachusetts02142, United States
| | - Nika Tsutskiridze
- Quantori LLC, Cambridge, Massachusetts02142, United States.,Tbilisi State Medical University, Tbilisi0186, Georgia
| | - Daviti Kharchilava
- Quantori LLC, Cambridge, Massachusetts02142, United States.,Tbilisi State Medical University, Tbilisi0186, Georgia
| | - Pema Lhewa
- Department of Population Health Sciences, School of Public Health, Georgia State University, Atlanta, Georgia30303, United States
| | - Aleksandr Shishkin
- Department of Population Health Sciences, School of Public Health, Georgia State University, Atlanta, Georgia30303, United States
| | - Yuriy Gankin
- Quantori LLC, Cambridge, Massachusetts02142, United States
| | - Alexander Kirpich
- Department of Population Health Sciences, School of Public Health, Georgia State University, Atlanta, Georgia30303, United States
| |
Collapse
|
2
|
Deboever E, Fistrovich A, Hulme C, Dunckley T. The Omnipresence of DYRK1A in Human Diseases. Int J Mol Sci 2022; 23:ijms23169355. [PMID: 36012629 PMCID: PMC9408930 DOI: 10.3390/ijms23169355] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/11/2022] [Accepted: 08/17/2022] [Indexed: 01/13/2023] Open
Abstract
The increasing population will challenge healthcare, particularly because the worldwide population has never been older. Therapeutic solutions to age-related disease will be increasingly critical. Kinases are key regulators of human health and represent promising therapeutic targets for novel drug candidates. The dual-specificity tyrosine-regulated kinase (DYRKs) family is of particular interest and, among them, DYRK1A has been implicated ubiquitously in varied human diseases. Herein, we focus on the characteristics of DYRK1A, its regulation and functional role in different human diseases, which leads us to an overview of future research on this protein of promising therapeutic potential.
Collapse
Affiliation(s)
- Estelle Deboever
- ASU-Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- Correspondence: (E.D.); (T.D.)
| | - Alessandra Fistrovich
- Department of Chemistry and Biochemistry, College of Science, The University of Arizona, Tucson, AZ 85721, USA
- Division of Drug Discovery and Development, Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA
| | - Christopher Hulme
- Department of Chemistry and Biochemistry, College of Science, The University of Arizona, Tucson, AZ 85721, USA
- Division of Drug Discovery and Development, Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA
| | - Travis Dunckley
- ASU-Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- Correspondence: (E.D.); (T.D.)
| |
Collapse
|
3
|
da Costa RA, da Rocha JAP, Pinheiro AS, da Costa ADSS, da Rocha ECM, Silva RC, Gonçalves ADS, Santos CBR, Brasil DDSB. A Computational Approach Applied to the Study of Potential Allosteric Inhibitors Protease NS2B/NS3 from Dengue Virus. Molecules 2022; 27:molecules27134118. [PMID: 35807364 PMCID: PMC9268547 DOI: 10.3390/molecules27134118] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/01/2022] [Accepted: 06/02/2022] [Indexed: 12/10/2022] Open
Abstract
Dengue virus (DENV) is a danger to more than 400 million people in the world, and there is no specific treatment. Thus, there is an urgent need to develop an effective method to combat this pathology. NS2B/NS3 protease is an important biological target due it being necessary for viral replication and the fact that it promotes the spread of the infection. Thus, this study aimed to design DENV NS2B/NS3pro allosteric inhibitors from a matrix compound. The search was conducted using the Swiss Similarity tool. The compounds were subjected to molecular docking calculations, molecular dynamics simulations (MD) and free energy calculations. The molecular docking results showed that two compounds, ZINC000001680989 and ZINC000001679427, were promising and performed important hydrogen interactions with the Asn152, Leu149 and Ala164 residues, showing the same interactions obtained in the literature. In the MD, the results indicated that five residues, Lys74, Leu76, Asn152, Leu149 and Ala166, contribute to the stability of the ligand at the allosteric site for all of the simulated systems. Hydrophobic, electrostatic and van der Waals interactions had significant effects on binding affinity. Physicochemical properties, lipophilicity, water solubility, pharmacokinetics, druglikeness and medicinal chemistry were evaluated for four compounds that were more promising, showed negative indices for the potential penetration of the Blood Brain Barrier and expressed high human intestinal absorption, indicating a low risk of central nervous system depression or drowsiness as the the side effects. The compound ZINC000006694490 exhibited an alert with a plausible level of toxicity for the purine base chemical moiety, indicating hepatotoxicity and chromosome damage in vivo in mouse, rat and human organisms. All of the compounds selected in this study showed a synthetic accessibility (SA) score lower than 4, suggesting the ease of new syntheses. The results corroborate with other studies in the literature, and the computational approach used here can contribute to the discovery of new and potent anti-dengue agents.
Collapse
Affiliation(s)
- Renato A. da Costa
- Graduate Program in Science and Environment, Institute of Exact and Natural Sciences, Federal University of Pará (UFPA), Belém 66075-110, PA, Brazil; (A.S.P.); (A.d.S.S.d.C.); (D.d.S.B.B.)
- Federal Institute of Education, Science and Technology of Pará Campus Castanhal, Castanhal 68740-970, PA, Brazil
- Correspondence: ; Tel.: +55-91-985484622
| | - João A. P. da Rocha
- Federal Institute of Education, Science and Technology of Pará—Campus Bragança, Bragança 68600-000, PA, Brazil; (J.A.P.d.R.); (E.C.M.d.R.)
| | - Alan S. Pinheiro
- Graduate Program in Science and Environment, Institute of Exact and Natural Sciences, Federal University of Pará (UFPA), Belém 66075-110, PA, Brazil; (A.S.P.); (A.d.S.S.d.C.); (D.d.S.B.B.)
| | - Andréia do S. S. da Costa
- Graduate Program in Science and Environment, Institute of Exact and Natural Sciences, Federal University of Pará (UFPA), Belém 66075-110, PA, Brazil; (A.S.P.); (A.d.S.S.d.C.); (D.d.S.B.B.)
| | - Elaine C. M. da Rocha
- Federal Institute of Education, Science and Technology of Pará—Campus Bragança, Bragança 68600-000, PA, Brazil; (J.A.P.d.R.); (E.C.M.d.R.)
| | - Rai. C. Silva
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto 14040-901, SP, Brazil;
| | - Arlan da S. Gonçalves
- Federal Institute of Education, Science and Technology of Espírito Santo, Vila Velha 29106-010, ES, Brazil;
| | - Cleydson B. R. Santos
- Laboratory of Modeling and Computational Chemistry, Department of Biological and Health Sciences, Federal University of Amapá, Macapá 68902-280, AP, Brazil;
| | - Davi do S. B. Brasil
- Graduate Program in Science and Environment, Institute of Exact and Natural Sciences, Federal University of Pará (UFPA), Belém 66075-110, PA, Brazil; (A.S.P.); (A.d.S.S.d.C.); (D.d.S.B.B.)
| |
Collapse
|
4
|
Shukla R, Kumar A, Kelvin DJ, Singh TR. Disruption of DYRK1A-induced hyperphosphorylation of amyloid-beta and tau protein in Alzheimer's disease: An integrative molecular modeling approach. Front Mol Biosci 2022; 9:1078987. [PMID: 36741918 PMCID: PMC9892649 DOI: 10.3389/fmolb.2022.1078987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 12/20/2022] [Indexed: 01/20/2023] Open
Abstract
Alzheimer's disease (AD) is a neurological disorder caused by the abnormal accumulation of hyperphosphorylated proteins. Dual-specificity tyrosine phosphorylation-regulated kinase 1A (DYRK1A) is a dual phosphorylation enzyme which phosphorylates the amyloid-β (Aβ) and neurofibrillary tangles (NFTs). A high throughput virtual screening approach was applied to screen a library of 98,071 compounds against DYRK1A using different programs including AutoDock Vina, Smina, and idock. Based on the binding affinities, we selected 330 compounds for absorption, distribution, metabolism, excretion, and toxicity (ADMET) analysis. Various pharmacokinetics parameters were predicted using the admetSAR server, and based on the pharmacokinetics results, 14 compounds were selected for cross-docking analysis using AutoDock. Cross-docking analysis revealed four compounds, namely, ZINC3843365 (-11.07 kcal/mol-1), ZINC2123081 (-10.93 kcal/mol-1), ZINC5220992 (-10.63 kcal/mol-1), and ZINC68569602 (-10.35 kcal/mol-1), which had the highest negative affinity scores compared to the 10 other molecules analyzed. Density functional theory (DFT) analysis was conducted for all the four top-ranked compounds. The molecular interaction stability of these four compounds with DYRK1A has been evaluated using molecular dynamics (MD) simulations on 100 nanoseconds followed by principal component analysis (PCA) and binding free energy calculations. The Gibbs free energy landscape analysis suggested the metastable state and folding pattern of selected docking complexes. Based on the present study outcome, we propose four antagonists, viz., ZINC3843365, ZINC2123081, ZINC5220992, and ZINC68569602 as potential inhibitors against DYRK1A and to reduce the amyloid-β and neurofibrillary tangle burden. These screened molecules can be further investigated using a number of in vitro and in vivo experiments.
Collapse
Affiliation(s)
- Rohit Shukla
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology (JUIT), Waknaghat, Himachal Pradesh, India
- Centre for Excellence in Healthcare Technologies and Informatics (CEHTI), Jaypee University of Information Technology (JUIT), Waknaghat, Himachal Pradesh, India
| | - Anuj Kumar
- Laboratory of Immunity, Shantou University Medical College, Shantou, China
- Department of Microbiology and Immunology, Canadian Centre for Vaccinology CCfV, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - David J. Kelvin
- Laboratory of Immunity, Shantou University Medical College, Shantou, China
- Department of Microbiology and Immunology, Canadian Centre for Vaccinology CCfV, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
- *Correspondence: David J. Kelvin, ; Tiratha Raj Singh,
| | - Tiratha Raj Singh
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology (JUIT), Waknaghat, Himachal Pradesh, India
- Centre for Excellence in Healthcare Technologies and Informatics (CEHTI), Jaypee University of Information Technology (JUIT), Waknaghat, Himachal Pradesh, India
- *Correspondence: David J. Kelvin, ; Tiratha Raj Singh,
| |
Collapse
|