1
|
Daniels MH, Castro J, Lee YT, Gotur D, Knockenhauer KE, Grigoriu S, Lockbaum GJ, Cheong JE, Lu C, Brennan D, Buker SM, Liu J, Yao S, Sparling BA, Sickmier EA, Ribich S, Blakemore SJ, Silver SJ, Boriack-Sjodin PA, Duncan KW, Copeland RA. Discovery of ATX968: An Orally Available Allosteric Inhibitor of DHX9. J Med Chem 2025. [PMID: 40298172 DOI: 10.1021/acs.jmedchem.5c00252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
DHX9 is an RNA/DNA helicase integral in the maintenance of genome stability that has emerged as an attractive target for oncology drug discovery. Disclosed herein is the discovery and optimization of a series of DHX9 inhibitors. Compound 1 was identified as a partial inhibitor of DHX9 ATPase activity but a full inhibitor of unwinding activity. Binding of 1 to a pocket distinct from the ATP binding site was confirmed by X-ray crystallography, enabling structure-based drug optimization. During this optimization, a sulfur-halogen bond was identified that increased on-target residence time without impacting equilibrium binding affinity. Analysis shows that cell potency more closely correlates with residence time than with equilibrium measurements of binding affinity or biochemical potency. Further optimization of potency and ADME properties led to the identification of ATX968, a potent and selective DHX9 inhibitor that is efficacious in a tumor xenograft model of microsatellite instability-high (MSI-H) colorectal cancer.
Collapse
Affiliation(s)
- Matthew H Daniels
- Accent Therapeutics, Inc., 1050 Waltham Street, Lexington, Massachusetts 02421, United States
| | - Jennifer Castro
- Accent Therapeutics, Inc., 1050 Waltham Street, Lexington, Massachusetts 02421, United States
| | - Young-Tae Lee
- Accent Therapeutics, Inc., 1050 Waltham Street, Lexington, Massachusetts 02421, United States
| | - Deepali Gotur
- Accent Therapeutics, Inc., 1050 Waltham Street, Lexington, Massachusetts 02421, United States
| | - Kevin E Knockenhauer
- Accent Therapeutics, Inc., 1050 Waltham Street, Lexington, Massachusetts 02421, United States
| | - Simina Grigoriu
- Accent Therapeutics, Inc., 1050 Waltham Street, Lexington, Massachusetts 02421, United States
| | - Gordon J Lockbaum
- Accent Therapeutics, Inc., 1050 Waltham Street, Lexington, Massachusetts 02421, United States
| | - Jae Eun Cheong
- Accent Therapeutics, Inc., 1050 Waltham Street, Lexington, Massachusetts 02421, United States
| | - Chuang Lu
- Accent Therapeutics, Inc., 1050 Waltham Street, Lexington, Massachusetts 02421, United States
| | - David Brennan
- Accent Therapeutics, Inc., 1050 Waltham Street, Lexington, Massachusetts 02421, United States
| | - Shane M Buker
- Accent Therapeutics, Inc., 1050 Waltham Street, Lexington, Massachusetts 02421, United States
| | - Julie Liu
- Accent Therapeutics, Inc., 1050 Waltham Street, Lexington, Massachusetts 02421, United States
| | - Shihua Yao
- Accent Therapeutics, Inc., 1050 Waltham Street, Lexington, Massachusetts 02421, United States
| | - Brian A Sparling
- Accent Therapeutics, Inc., 1050 Waltham Street, Lexington, Massachusetts 02421, United States
| | - E Allen Sickmier
- Accent Therapeutics, Inc., 1050 Waltham Street, Lexington, Massachusetts 02421, United States
| | - Scott Ribich
- Accent Therapeutics, Inc., 1050 Waltham Street, Lexington, Massachusetts 02421, United States
| | - Steve J Blakemore
- Accent Therapeutics, Inc., 1050 Waltham Street, Lexington, Massachusetts 02421, United States
| | - Serena J Silver
- Accent Therapeutics, Inc., 1050 Waltham Street, Lexington, Massachusetts 02421, United States
| | - P Ann Boriack-Sjodin
- Accent Therapeutics, Inc., 1050 Waltham Street, Lexington, Massachusetts 02421, United States
| | - Kenneth W Duncan
- Accent Therapeutics, Inc., 1050 Waltham Street, Lexington, Massachusetts 02421, United States
| | - Robert A Copeland
- Accent Therapeutics, Inc., 1050 Waltham Street, Lexington, Massachusetts 02421, United States
| |
Collapse
|
2
|
Lockbaum GJ, Lynes MM, Shen SA, Liu J, Holt N, Nayak SP, Knockenhauer KE, Yao S, Sickmier EA, Raman A, Wu J, Case A, Shehaj L, Buker SM, Grigoriu S, Ribich S, Blakemore SJ, Sparling BA, Duncan KW, Copeland RA, Silver SJ, Boriack-Sjodin PA. Characterization of exoribonuclease XRN1 as a cancer target and identification of adenosine-3',5'-bisphosphate as a potent enzyme inhibitor. Commun Biol 2025; 8:589. [PMID: 40205031 PMCID: PMC11982291 DOI: 10.1038/s42003-025-08005-y] [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: 11/14/2024] [Accepted: 03/26/2025] [Indexed: 04/11/2025] Open
Abstract
XRN1 (5'-3' exoribonuclease 1) degrades RNA from the 5' → 3' direction and utilizes both single- and double-stranded RNA as substrates. XRN1 plays a critical role in mRNA turnover as well as regulating the cellular response to viral infection. XRN1 also protects the cell by preventing endogenous double-stranded RNA accumulation. XRN1 was identified as a putative vulnerability in a subset of cancer cell lines through analysis of publicly available CRISPR data. The role of XRN1 was explored using a set of non-small cell lung cancer cell lines with differential predicted XRN1 dependency to validate XRN1 as an oncology target. In predicted sensitive cell lines, XRN1 knockout reduced proliferation, increased apoptosis and activated the pPKR and MDA5 dsRNA sensing pathways. To facilitate drug discovery targeting XRN1, a suite of biochemical and biophysical assays was developed. These assays were used to characterize adenosine-3',5'-bisphosphate (pAp), a non-selective nuclease inhibitor, as a nanomolar inhibitor of XRN1. Additionally, the crystal structure of human XRN1 was solved with pAp bound, demonstrating distinct interactions for the compound in the XRN1 active site. These studies provide a strong foundation for the discovery of potent, selective inhibitors of XRN1 as a novel approach to cancer therapeutics.
Collapse
Affiliation(s)
| | | | | | - Julie Liu
- Accent Therapeutics, Lexington, MA, 02421, USA
| | | | | | | | - Shihua Yao
- Accent Therapeutics, Lexington, MA, 02421, USA
| | | | | | - Jie Wu
- Accent Therapeutics, Lexington, MA, 02421, USA
| | - April Case
- Accent Therapeutics, Lexington, MA, 02421, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
3
|
Viviani LG, Iijima TS, Piccirillo E, Rezende L, Alegria TGP, Netto LES, T.-do Amaral A, Miyamoto S. Identification of Novel Human 15-Lipoxygenase-2 (h15-LOX-2) Inhibitors Using a Virtual Screening Approach. J Med Chem 2025; 68:307-323. [PMID: 39700339 PMCID: PMC11726667 DOI: 10.1021/acs.jmedchem.4c01884] [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: 08/10/2024] [Revised: 12/05/2024] [Accepted: 12/09/2024] [Indexed: 12/21/2024]
Abstract
The human 15-lipoxygenase-2 (h15-LOX-2) catalyzes mainly the regio- and stereospecific oxygenation of arachidonate to its corresponding hydroperoxide (15(S)-HpETE). h15-LOX-2 is implicated in the biosynthesis of inflammatory lipid mediators and plays a role in the development of atherosclerotic plaques, but it is still underexploited as a drug target. Here, to search for novel h15-LOX-2 inhibitors, we used a virtual screening (VS) approach consisting of shape-based matching, two-dimensional (2D) structural "dissimilarity", docking, and visual inspection filters, which were applied to a "curated" ZINC database (∼8 × 106 compounds). The VS was experimentally validated, and six micromolar-range inhibitors were identified among 13 tested compounds (46.2%). The Ki values could be determined for two inhibitors, compounds 10 (Ki = 16.4 ± 8.1 μM) and 13 (Ki = 15.1 ± 7.6 μM), which showed a mixed-type mechanism of inhibition. Overall, the identified inhibitors fulfill drug-like criteria and are structurally novel compared with known h15-LOX-2 inhibitors.
Collapse
Affiliation(s)
- Lucas G. Viviani
- Department
of Biochemistry, Institute of Chemistry, University of São Paulo, Av. Prof. Lineu Prestes 748, 05508-000 São Paulo, Brazil
| | - Thais S. Iijima
- Department
of Biochemistry, Institute of Chemistry, University of São Paulo, Av. Prof. Lineu Prestes 748, 05508-000 São Paulo, Brazil
| | - Erika Piccirillo
- Center
for Medicinal Chemistry (CQMED), State University
of Campinas, Av. André
Tosello 550, 13083-886 Campinas, Brazil
| | - Leandro Rezende
- Department
of Biochemistry, Institute of Chemistry, University of São Paulo, Av. Prof. Lineu Prestes 748, 05508-000 São Paulo, Brazil
| | - Thiago G. P. Alegria
- Department
of Genetics and Evolutionary Biology, Institute of Biosciences, University of São Paulo, Rua do Matão, 277, 05508-090 São Paulo, Brazil
| | - Luis Eduardo S. Netto
- Department
of Genetics and Evolutionary Biology, Institute of Biosciences, University of São Paulo, Rua do Matão, 277, 05508-090 São Paulo, Brazil
| | - Antonia T.-do Amaral
- Department
of Fundamental Chemistry, Institute of Chemistry, University of São Paulo, Av. Prof. Lineu Prestes 748, 05508-000 São Paulo, Brazil
| | - Sayuri Miyamoto
- Department
of Biochemistry, Institute of Chemistry, University of São Paulo, Av. Prof. Lineu Prestes 748, 05508-000 São Paulo, Brazil
| |
Collapse
|
4
|
Maboko LM, Theron A, Panayides J, Cordier W, Fisher D, Steenkamp V. Evaluating Blood-Brain Barrier Permeability, Cytotoxicity, and Activity of Potential Acetylcholinesterase Inhibitors: In Vitro and In Silico Study. Pharmacol Res Perspect 2024; 12:e70043. [PMID: 39651604 PMCID: PMC11841676 DOI: 10.1002/prp2.70043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 11/08/2024] [Accepted: 11/14/2024] [Indexed: 12/11/2024] Open
Abstract
Acetylcholinesterase inhibitors (AChEIs) remain the first-line treatment for Alzheimer's disease. However, these drugs are largely symptomatic and often associated with adverse effects. This study aimed to evaluate novel pharmacophores for their in vitro AChEI activity, blood-brain barrier (BBB) permeability, and cytotoxic potential, hypothesizing that a combination of AChEIs could enhance symptom management while minimizing toxicity. A library of 1453 synthetic pharmacophores was assessed using in vitro and in silico methods to determine their feasibility as an inhibitor of the AChE enzyme. An in-house miniaturized Ellman's assay determined acellular AChEI activities, while pharmacokinetic properties were evaluated using the SwissADME web tool. The combinational effects of in silico BBB-permeable pharmacophores and donepezil were examined using a checkerboard AChEI assay. Cytotoxicity of active compounds and their synergistic combinations was assessed in SH-SY5Y neuroblastoma and bEnd.5 cells using the sulforhodamine B assay. Cellular AChEI activity of active in silico BBB-permeable predicted compounds was determined using an SH-SY5Y AChE-based assay. An in vitro BBB model was used to assess the effect of compounds on the integrity of the bEnd.5 monolayer. Out of the screened compounds, 12 demonstrated 60% AChEI activity at 5 μM, with compound A51 showing the lowest IC50 (0.20 μM). Five compounds were identified as BBB-permeable, with the donepezil-C53 combination at ¼IC50 exhibiting the strongest synergy (CI = 0.82). Compounds A136 and C129, either alone or with donepezil, showed cytotoxicity. Notably, compound C53, both alone and in combination with donepezil, demonstrated high AChEI activity and promising BBB permeability, warranting further investigation.
Collapse
Affiliation(s)
- L. M. Maboko
- Department of Pharmacology, School of Medicine, Faculty of Health SciencesUniversity of PretoriaPretoriaSouth Africa
| | - A. Theron
- Future Production: Chemicals, Council for Scientific and Industrial ResearchPretoriaSouth Africa
| | - J.‐L. Panayides
- Future Production: Chemicals, Council for Scientific and Industrial ResearchPretoriaSouth Africa
| | - W. Cordier
- Department of Pharmacology, School of Medicine, Faculty of Health SciencesUniversity of PretoriaPretoriaSouth Africa
| | - D. Fisher
- Department of Medical BioSciences, Faculty of Natural Sciences, Neurobiology Research GroupUniversity of Western CapeCape TownSouth Africa
| | - V. Steenkamp
- Department of Pharmacology, School of Medicine, Faculty of Health SciencesUniversity of PretoriaPretoriaSouth Africa
| |
Collapse
|
5
|
Visvanathan R, Le DT, Dhital S, Rali T, Davis RA, Williamson G. Inhibition of Human Salivary and Pancreatic α-Amylase by Resveratrol Oligomers. J Med Chem 2024; 67:18753-18763. [PMID: 39501642 PMCID: PMC11571111 DOI: 10.1021/acs.jmedchem.4c01042] [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: 05/02/2024] [Revised: 09/16/2024] [Accepted: 09/18/2024] [Indexed: 11/15/2024]
Abstract
A key strategy to mitigate postprandial hyperglycemia involves inhibiting α-amylases, which commence the starch digestion process in the gut. This study examined the inhibitory effects of resveratrol and stilbenoid tetramers, vaticanol B, (-)-hopeaphenol, and vatalbinoside A on human salivary and pancreatic α-amylases experimentally and through molecular docking studies. Vaticanol B demonstrated the most potent inhibition with IC50 values of 5.3 ± 0.3 μM for salivary and 6.1 ± 0.5 μM for pancreatic α-amylase (compared to acarbose with IC50 values of 1.2 ± 0.1 μM and 0.5 ± 0.0 μM, respectively). Kinetic analysis suggested a competitive inhibition mode for vaticanol B. Resveratrol and vatalbinoside A were poor inhibitors of human α-amylases, while (-)-hopeaphenol exhibited moderate inhibition. Molecular docking supported the inhibition data, and several aspects of the structural configurations explained the stronger inhibition exerted by vaticanol B. Overall, vaticanol B shows promise as a natural alternative to acarbose for inhibiting α-amylase.
Collapse
Affiliation(s)
- Rizliya Visvanathan
- Molecular
Nutrition Group, Department of Nutrition, Dietetics and Food, Monash University, Victorian Heart Institute, Victoria
Heart Hospital, 631 Blackburn
Road, Clayton, VIC 3168, Australia
| | - Dang Truong Le
- Molecular
Nutrition Group, Department of Nutrition, Dietetics and Food, Monash University, Victorian Heart Institute, Victoria
Heart Hospital, 631 Blackburn
Road, Clayton, VIC 3168, Australia
- Bioresource
Processing Research Institute of Australia (BioPRIA), Department of
Chemical and Biological Engineering, Monash
University, Clayton, VIC 3800, Australia
| | - Sushil Dhital
- Bioresource
Processing Research Institute of Australia (BioPRIA), Department of
Chemical and Biological Engineering, Monash
University, Clayton, VIC 3800, Australia
| | - Topul Rali
- School
of Natural and Physical Sciences, The University
of Papua New Guinea, Port Moresby, Papua New Guinea
| | - Rohan A. Davis
- Institute
for Biomedicine and Glycomics, Griffith
University, Brisbane, QLD 4111, Australia
| | - Gary Williamson
- Molecular
Nutrition Group, Department of Nutrition, Dietetics and Food, Monash University, Victorian Heart Institute, Victoria
Heart Hospital, 631 Blackburn
Road, Clayton, VIC 3168, Australia
| |
Collapse
|
6
|
García Molina P, Saura-Sanmartin A, Berna J, Teruel JA, Muñoz Muñoz JL, Rodríguez López JN, García Cánovas F, García Molina F. Considerations about the inhibition of monophenolase and diphenolase activities of tyrosinase. Characterization of the inhibitor concentration which generates 50 % of inhibition, type and inhibition constants. A review. Int J Biol Macromol 2024; 267:131513. [PMID: 38608979 DOI: 10.1016/j.ijbiomac.2024.131513] [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: 12/31/2023] [Revised: 04/08/2024] [Accepted: 04/09/2024] [Indexed: 04/14/2024]
Abstract
Tyrosinase is a copper oxidase enzyme which catalyzes the first two steps in the melanogenesis pathway, L-tyrosine to L-dopa conversion and, then, to o-dopaquinone and dopachrome. Hypopigmentation and, above all, hyperpigmentation issues can be originated depending on their activity. This enzyme also promotes the browning of fruits and vegetables. Therefore, control of their activity by regulators is research topic of great relevance. In this work, we consider the use of inhibitors of monophenolase and diphenolase activities of the enzyme in order to accomplish such control. An experimental design and data analysis which allow the accurate calculation of the degree of inhibition of monophenolase activity (iM) and diphenolase activity (iD) are proposed. The IC50 values (amount of inhibitor that causes 50 % inhibition at a fixed substrate concentration) can be calculated for the two activities and from the values of IC50M (monophenolase) and IC50D(diphenolase). Additionally, the strength and type of inhibition can be deduced from these values. The data analysis from these IC50D values allows to obtain the values of [Formula: see text] or [Formula: see text] , or and [Formula: see text] from the values of IC50M. In all cases, the values of the different must satisfy their relationship with IC50M and IC50D.
Collapse
Affiliation(s)
- Pablo García Molina
- GENZ-Group of Research on Enzymology, Department of Biochemistry and Molecular Biology-A, Regional Campus of International Excellence "Campus Mare Nostrum", University of Murcia, Espinardo, Murcia, Spain
| | - Adrian Saura-Sanmartin
- Department of Organic Chemistry, Faculty of Chemistry, University of Murcia, E-30100 Espinardo, Murcia, Spain.
| | - Jose Berna
- Department of Organic Chemistry, Faculty of Chemistry, University of Murcia, E-30100 Espinardo, Murcia, Spain
| | - Jose Antonio Teruel
- Department of Biochemistry and Molecular Biology-A, Regional Campus of International Excellence "Campus Mare Nostrum", University of Murcia, Espinardo, Murcia, Spain
| | - Jose Luis Muñoz Muñoz
- Microbial Enzymology Lab, Department of Applied Sciences, Ellison Building A, University of Northumbria, Newcastle Upon Tyne, UK
| | - Jose Neptuno Rodríguez López
- GENZ-Group of Research on Enzymology, Department of Biochemistry and Molecular Biology-A, Regional Campus of International Excellence "Campus Mare Nostrum", University of Murcia, Espinardo, Murcia, Spain
| | - Francisco García Cánovas
- GENZ-Group of Research on Enzymology, Department of Biochemistry and Molecular Biology-A, Regional Campus of International Excellence "Campus Mare Nostrum", University of Murcia, Espinardo, Murcia, Spain
| | - Francisco García Molina
- Department of Anatomía Patológica, Hospital General Universitario Reina Sofía, Av. Intendente Jorge Palacios, 1, 30003 Murcia, Spain.
| |
Collapse
|
7
|
Gising J, Honarnejad S, Bras M, Baillie GL, McElroy SP, Jones PS, Morrison A, Beveridge J, Hallberg M, Larhed M. The Discovery of New Inhibitors of Insulin-Regulated Aminopeptidase by a High-Throughput Screening of 400,000 Drug-like Compounds. Int J Mol Sci 2024; 25:4084. [PMID: 38612894 PMCID: PMC11012289 DOI: 10.3390/ijms25074084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 03/25/2024] [Accepted: 04/02/2024] [Indexed: 04/14/2024] Open
Abstract
With the ambition to identify novel chemical starting points that can be further optimized into small drug-like inhibitors of insulin-regulated aminopeptidase (IRAP) and serve as potential future cognitive enhancers in the clinic, we conducted an ultra-high-throughput screening campaign of a chemically diverse compound library of approximately 400,000 drug-like small molecules. Three biochemical and one biophysical assays were developed to enable large-scale screening and hit triaging. The screening funnel, designed to be compatible with high-density microplates, was established with two enzyme inhibition assays employing either fluorescent or absorbance readouts. As IRAP is a zinc-dependent enzyme, the remaining active compounds were further evaluated in the primary assay, albeit with the addition of zinc ions. Rescreening with zinc confirmed the inhibitory activity for most compounds, emphasizing a zinc-independent mechanism of action. Additionally, target engagement was confirmed using a complementary biophysical thermal shift assay where compounds causing positive/negative thermal shifts were considered genuine binders. Triaging based on biochemical activity, target engagement, and drug-likeness resulted in the selection of 50 qualified hits, of which the IC50 of 32 compounds was below 3.5 µM. Despite hydroxamic acid dominance, diverse chemotypes with biochemical activity and target engagement were discovered, including non-hydroxamic acid compounds. The most potent compound (QHL1) was resynthesized with a confirmed inhibitory IC50 of 320 nM. Amongst these compounds, 20 new compound structure classes were identified, providing many new starting points for the development of unique IRAP inhibitors. Detailed characterization and optimization of lead compounds, considering both hydroxamic acids and other diverse structures, are in progress for further exploration.
Collapse
Affiliation(s)
- Johan Gising
- The Beijer Laboratory, Science for Life Laboratory, Department of Medicinal Chemistry, Biomedical Centre, Uppsala University, P.O. Box 574, SE-751 23 Uppsala, Sweden; (J.B.); (M.L.)
| | - Saman Honarnejad
- Pivot Park Screening Centre, Kloosterstraat 9, 5349 AB Oss, The Netherlands; (S.H.); (M.B.)
| | - Maaike Bras
- Pivot Park Screening Centre, Kloosterstraat 9, 5349 AB Oss, The Netherlands; (S.H.); (M.B.)
| | - Gemma L. Baillie
- BioAscent Discovery Ltd., Bo‘Ness Road, Newhouse, Motherwell ML1 5UH, UK; (G.L.B.); (S.P.M.); (P.S.J.); (A.M.)
| | - Stuart P. McElroy
- BioAscent Discovery Ltd., Bo‘Ness Road, Newhouse, Motherwell ML1 5UH, UK; (G.L.B.); (S.P.M.); (P.S.J.); (A.M.)
| | - Philip S. Jones
- BioAscent Discovery Ltd., Bo‘Ness Road, Newhouse, Motherwell ML1 5UH, UK; (G.L.B.); (S.P.M.); (P.S.J.); (A.M.)
| | - Angus Morrison
- BioAscent Discovery Ltd., Bo‘Ness Road, Newhouse, Motherwell ML1 5UH, UK; (G.L.B.); (S.P.M.); (P.S.J.); (A.M.)
| | - Julia Beveridge
- The Beijer Laboratory, Science for Life Laboratory, Department of Medicinal Chemistry, Biomedical Centre, Uppsala University, P.O. Box 574, SE-751 23 Uppsala, Sweden; (J.B.); (M.L.)
| | - Mathias Hallberg
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Neuropharmacology and Addiction Research, Biomedical Centre, Uppsala University, P.O. Box 591, SE-751 24 Uppsala, Sweden;
| | - Mats Larhed
- The Beijer Laboratory, Science for Life Laboratory, Department of Medicinal Chemistry, Biomedical Centre, Uppsala University, P.O. Box 574, SE-751 23 Uppsala, Sweden; (J.B.); (M.L.)
| |
Collapse
|
8
|
Cui X, Huang Z, Deng S, Zhang Y, Li G, Wang L, Deng Y, Wu C. Benzofuran Derivatives from Cortex Mori Radicis and Their Cholinesterase-Inhibitory Activity. Molecules 2024; 29:315. [PMID: 38257228 PMCID: PMC10820097 DOI: 10.3390/molecules29020315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 12/26/2023] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
The phytochemical investigation of Cortex Mori Radicis led to the isolation and identification of a new prenylated benzofuranone (1) and four ring-opening derivatives (2-5) named albaphenol A-E, as well as nigranol A (6), together with ten 2-arylbenzofuran derivatives (7-16). The characterization of the structures of the new compounds and the structural revision of nigranol A (6) were conducted using the comprehensive analysis of spectroscopic data (1D/2D NMR, HRESIMS, CD, and XRD). Compounds 1-16 were tested for their inhibitory effects on acetylcholinesterase (AChE) and butyrylcholinesterase (BChE). Compounds 1 and 4 showed weak BChE-inhibitory activity (IC50 45.5 and 61.0 μM); six 2-arylbenzofuran derivatives showed more-potent BChE-inhibitory activity (IC50 2.5-32.8 μM) than the positive control galantamine (IC50 35.3 μM), while being inactive or weakly inhibitory toward AChE. Cathafuran C (14) exhibited the most potent and selective inhibitory activity against BChE in a competitive manner, with a Ki value of 1.7 μM. The structure-activity relationships of the benzofuran-type stilbenes were discussed. Furthermore, molecular docking and dynamic simulations were performed to clarify the interactions of the inhibitor-enzyme complex.
Collapse
Affiliation(s)
- Xiang Cui
- College of Life Sciences and Agronomy, Zhoukou Normal University, Zhoukou 466001, China; (X.C.)
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zehong Huang
- College of Life Sciences and Agronomy, Zhoukou Normal University, Zhoukou 466001, China; (X.C.)
| | - Shanshan Deng
- College of Life Sciences and Agronomy, Zhoukou Normal University, Zhoukou 466001, China; (X.C.)
| | - Yunxia Zhang
- College of Life Sciences and Agronomy, Zhoukou Normal University, Zhoukou 466001, China; (X.C.)
| | - Guoyin Li
- College of Life Sciences and Agronomy, Zhoukou Normal University, Zhoukou 466001, China; (X.C.)
| | - Lining Wang
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yanru Deng
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Changjing Wu
- College of Life Sciences and Agronomy, Zhoukou Normal University, Zhoukou 466001, China; (X.C.)
- Field Observation and Research Station of Green Agriculture in Dancheng County, Zhoukou 466001, China
| |
Collapse
|
9
|
Gotur D, Case A, Liu J, Sickmier EA, Holt N, Knockenhauer KE, Yao S, Lee YT, Copeland RA, Buker SM, Boriack-Sjodin PA. Development of assays to support identification and characterization of modulators of DExH-box helicase DHX9. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2023; 28:376-384. [PMID: 37625785 DOI: 10.1016/j.slasd.2023.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/02/2023] [Accepted: 08/22/2023] [Indexed: 08/27/2023]
Abstract
DHX9 is a DExH-box RNA helicase that utilizes hydrolysis of all four nucleotide triphosphates (NTPs) to power cycles of 3' to 5' directional movement to resolve and/or unwind double stranded RNA, DNA, and RNA/DNA hybrids, R-loops, triplex-DNA and G-quadraplexes. DHX9 activity is important for both viral amplification and maintaining genomic stability in cancer cells; therefore, it is a therapeutic target of interest for drug discovery efforts. Biochemical assays measuring ATP hydrolysis and oligonucleotide unwinding for DHX9 have been developed and characterized, and these assays can support high-throughput compound screening efforts under balanced conditions. Assay development efforts revealed DHX9 can use double stranded RNA with 18-mer poly(U) 3' overhangs and as well as significantly shorter overhangs at the 5' or 3' end as substrates. The enzymatic assays are augmented by a robust SPR assay for compound validation. A mechanism-derived inhibitor, GTPγS, was characterized as part of the validation of these assays and a crystal structure of GDP bound to cat DHX9 has been solved. In addition to enabling drug discovery efforts for DHX9, these assays may be extrapolated to other RNA helicases providing a valuable toolkit for this important target class.
Collapse
Affiliation(s)
- Deepali Gotur
- Accent Therapeutics, 1050 Waltham Street, Lexington, MA 02421, USA
| | - April Case
- Accent Therapeutics, 1050 Waltham Street, Lexington, MA 02421, USA
| | - Julie Liu
- Accent Therapeutics, 1050 Waltham Street, Lexington, MA 02421, USA
| | - E Allen Sickmier
- Accent Therapeutics, 1050 Waltham Street, Lexington, MA 02421, USA
| | - Nicholas Holt
- Accent Therapeutics, 1050 Waltham Street, Lexington, MA 02421, USA
| | | | - Shihua Yao
- Accent Therapeutics, 1050 Waltham Street, Lexington, MA 02421, USA
| | - Young-Tae Lee
- Accent Therapeutics, 1050 Waltham Street, Lexington, MA 02421, USA
| | | | - Shane M Buker
- Accent Therapeutics, 1050 Waltham Street, Lexington, MA 02421, USA
| | | |
Collapse
|
10
|
Suwanhom P, Nualnoi T, Khongkow P, Tipmanee V, Lomlim L. Novel Lawsone-Quinoxaline Hybrids as New Dual Binding Site Acetylcholinesterase Inhibitors. ACS OMEGA 2023; 8:32498-32511. [PMID: 37720764 PMCID: PMC10500570 DOI: 10.1021/acsomega.3c02683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 08/15/2023] [Indexed: 09/19/2023]
Abstract
A new family of lawsone-quinoxaline hybrids was designed, synthesized, and evaluated as dual binding site cholinesterase inhibitors (ChEIs). In vitro tests revealed that compound 6d was the most potent AChEI (IC50 = 20 nM) and BChEI (IC50 = 220 nM). The compound 6d did not show cytotoxicity against the SH-SY5Y neuronal cells (GI50 > 100 μM). In silico and enzyme kinetic experiments demonstrated that compound 6d bound to both the catalytic anionic site and the peripheral anionic site of HuAChE. The lawsone-quinoxaline hybrids exhibited potential for further development of potent acetylcholinesterase inhibitors for the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Paptawan Suwanhom
- Department
of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
- Phytomedicine
and Pharmaceutical Biotechnology Excellent Center (PPBEC), Faculty
of Pharmaceutical Sciences, Prince of Songkla
University, Hat Yai, Songkhla 90110, Thailand
| | - Teerapat Nualnoi
- Department
of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
| | - Pasarat Khongkow
- Department
of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
| | - Varomyalin Tipmanee
- Department
of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
| | - Luelak Lomlim
- Department
of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
- Phytomedicine
and Pharmaceutical Biotechnology Excellent Center (PPBEC), Faculty
of Pharmaceutical Sciences, Prince of Songkla
University, Hat Yai, Songkhla 90110, Thailand
| |
Collapse
|
11
|
Brooke H, Ghoshray M, Ibrahim A, Lloyd MD. Steady-state kinetic analysis of reversible enzyme inhibitors: A case study on calf intestine alkaline phosphatase. Methods Enzymol 2023; 690:39-84. [PMID: 37858536 DOI: 10.1016/bs.mie.2023.06.015] [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] [Indexed: 10/21/2023]
Abstract
Enzymes are important drug targets and inhibition of enzymatic activity is an important therapeutic strategy. Enzyme assays measuring catalytic activity are utilized in both the discovery and development of new drugs. Colorimetric assays based on the release of 4-nitrophenol from substrates are commonly used. 4-Nitrophenol is only partly ionized to 4-nitrophenolate under typical assay conditions (pH 7-9) leading to under-estimation of product formation rates due to the much lower extinction coefficient of 4-nitrophenol compared to 4-nitrophenolate. Determination of 4-nitrophenol pKa values based on absorbance at 405 nm as a function of experimental pH values is reported, allowing for calculation of a corrected extinction coefficient at the assay pH. Characterization of inhibitor properties using steady-state enzyme kinetics is demonstrated using calf intestine alkaline phosphatase and 4-nitrophenyl phosphate as substrate at pH ∼8.2. The following kinetic parameters were determined: Km= 40±3 µM; Vmax= 72.8±1.2 µmolmin-1mg protein-1; kcat= 9.70±0.16 s-1; kcat/Km= 2.44±0.16 × 105 M-1s-1 (mean± SEM, N = 4). Sodium orthovanadate and EDTA were used as model inhibitors and the following pIC50 values were measured using dose-response curves: 6.61±0.08 and 3.07±0.03 (mean±SEM, N = 4). Rapid dilution experiments determined that inhibition was reversible for sodium orthovanadate and irreversible for EDTA. A Ki value for orthovanadate of 51±8 nM (mean±SEM, N = 3) was determined. Finally, data analysis and statistical design of experiments are discussed.
Collapse
Affiliation(s)
- Henry Brooke
- Department of Life Sciences, University of Bath, Claverton Down, Bath, United Kingdom
| | - Meghna Ghoshray
- Department of Life Sciences, University of Bath, Claverton Down, Bath, United Kingdom
| | - Archad Ibrahim
- Department of Life Sciences, University of Bath, Claverton Down, Bath, United Kingdom
| | - Matthew D Lloyd
- Department of Life Sciences, University of Bath, Claverton Down, Bath, United Kingdom.
| |
Collapse
|
12
|
Harman MAJ, Stanway SJ, Scott H, Demydchuk Y, Bezerra GA, Pellegrino S, Chen L, Brear P, Lulla A, Hyvönen M, Beswick PJ, Skynner MJ. Structure-Guided Chemical Optimization of Bicyclic Peptide ( Bicycle) Inhibitors of Angiotensin-Converting Enzyme 2. J Med Chem 2023. [PMID: 37433017 DOI: 10.1021/acs.jmedchem.3c00710] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2023]
Abstract
Angiotensin-converting enzyme 2 (ACE2) is a metalloprotease that cleaves angiotensin II, a peptide substrate involved in the regulation of hypertension. Here, we identified a series of constrained bicyclic peptides, Bicycle, inhibitors of human ACE2 by panning highly diverse bacteriophage display libraries. These were used to generate X-ray crystal structures which were used to inform the design of additional Bicycles with increased affinity and inhibition of ACE2 enzymatic activity. This novel structural class of ACE2 inhibitors is among the most potent ACE2 inhibitors yet described in vitro, representing a valuable tool to further probe ACE2 function and for potential therapeutic utility.
Collapse
Affiliation(s)
- Maximilian A J Harman
- BicycleTx Ltd., Portway Building Blocks A and B, Granta Park, Great Abington, Cambridge CB21 6GS, U.K
| | - Steven J Stanway
- BicycleTx Ltd., Portway Building Blocks A and B, Granta Park, Great Abington, Cambridge CB21 6GS, U.K
| | - Heather Scott
- BicycleTx Ltd., Portway Building Blocks A and B, Granta Park, Great Abington, Cambridge CB21 6GS, U.K
| | - Yuliya Demydchuk
- BicycleTx Ltd., Portway Building Blocks A and B, Granta Park, Great Abington, Cambridge CB21 6GS, U.K
| | - Gustavo Arruda Bezerra
- BicycleTx Ltd., Portway Building Blocks A and B, Granta Park, Great Abington, Cambridge CB21 6GS, U.K
| | - Simone Pellegrino
- BicycleTx Ltd., Portway Building Blocks A and B, Granta Park, Great Abington, Cambridge CB21 6GS, U.K
| | - Liuhong Chen
- BicycleTx Ltd., Portway Building Blocks A and B, Granta Park, Great Abington, Cambridge CB21 6GS, U.K
| | - Paul Brear
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1GA, U.K
| | - Aleksei Lulla
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1GA, U.K
| | - Marko Hyvönen
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1GA, U.K
| | - Paul J Beswick
- BicycleTx Ltd., Portway Building Blocks A and B, Granta Park, Great Abington, Cambridge CB21 6GS, U.K
| | - Michael J Skynner
- BicycleTx Ltd., Portway Building Blocks A and B, Granta Park, Great Abington, Cambridge CB21 6GS, U.K
| |
Collapse
|
13
|
Matvieieva N, Bessarabov V, Khainakova O, Duplij V, Bohdanovych T, Ratushnyak Y, Kuzmina G, Lisovyi V, Zderko N, Kobylinska N. Cichorium intybus L. “hairy” roots as a rich source of antioxidants and anti-inflammatory compounds. Heliyon 2023; 9:e14516. [PMID: 37101499 PMCID: PMC10123141 DOI: 10.1016/j.heliyon.2023.e14516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 03/02/2023] [Accepted: 03/08/2023] [Indexed: 03/19/2023] Open
Abstract
The present study aimed to determine the bioactive profile of various extracts of Cichorium intybus L. "hairy" roots. In particular, the total content of flavonoids as well as the reducing power, antioxidant and anti-inflammatory activity of the aqueous and ethanolic (70%) extracts were evaluated. The total content of flavonoids the ethanolic extract of the dry "hairy" root reached up to 121.3 mg (RE)/g, which was twofold greater than in the aqueous one. A total of 33 diverse polyphenols were identified by the LC-HRMS method. The experimental results showed a high amount of gallic (6.103 ± 0.008 mg/g) and caffeic (7.001 ± 0.068 mg/g) acids. In the "hairy" roots, the presence of rutin, apigenin, kaempferol, quercetin, and its derivatives was found in concentrations of 0.201±0.003 - 6.710±0.052 mg/g. The broad spectrum of pharmacological activities (antioxidant, anti-inflammatory, antimutagenic, anticarcinogenic, etc.) of the key flavonoids identified in the chicory "hairy" root extract was predicted by the General Unrestricted Structure-Activity Relationships algorithm based on in the substances detected in the extract. The evaluation of the antioxidant activity showed that the EC50 values of the ethanol and the aqueous extracts were 0.174 and 0.346 mg, respectively. Thus, the higher ability of the ethanol extract to scavenge the DPPH radical was observed. The calculated Michaelis and inhibition constants indicated that the ethanolic extract of C. intybus "hairy" roots is an efficient inhibitor of soybean 15-Lipoxygenase activity (IC50 = 84.13 ± 7.22 μM) in a mixed mechanism. Therefore, the obtained extracts could be the basis of herbal pharmaceuticals for the therapy of human diseases accompanied by oxidative stress and inflammation, including the pandemic coronavirus disease COVID-19.
Collapse
Affiliation(s)
- Nadiia Matvieieva
- Institute of Cell Biology and Genetic Engineering, National Academy of Sciences of Ukraine, 148 Zabolotnogo Str., Kyiv, 03143, Ukraine
| | - Volodymyr Bessarabov
- Kyiv National University of Technologies and Design, 2 Nemyrovycha-Danchenko Str., Kyiv, 01011, Ukraine
| | - Olena Khainakova
- University of Oviedo, 8 Julián Claveria Av., Oviedo, 33006, Spain
| | - Volodymyr Duplij
- Institute of Cell Biology and Genetic Engineering, National Academy of Sciences of Ukraine, 148 Zabolotnogo Str., Kyiv, 03143, Ukraine
| | - Taisa Bohdanovych
- Institute of Cell Biology and Genetic Engineering, National Academy of Sciences of Ukraine, 148 Zabolotnogo Str., Kyiv, 03143, Ukraine
| | - Yakiv Ratushnyak
- Institute of Cell Biology and Genetic Engineering, National Academy of Sciences of Ukraine, 148 Zabolotnogo Str., Kyiv, 03143, Ukraine
| | - Galina Kuzmina
- Kyiv National University of Technologies and Design, 2 Nemyrovycha-Danchenko Str., Kyiv, 01011, Ukraine
| | - Vadym Lisovyi
- Kyiv National University of Technologies and Design, 2 Nemyrovycha-Danchenko Str., Kyiv, 01011, Ukraine
| | - Nazar Zderko
- Kyiv National University of Technologies and Design, 2 Nemyrovycha-Danchenko Str., Kyiv, 01011, Ukraine
| | - Natalia Kobylinska
- Dumansky Institute of Colloid and Water Chemistry, National Academy of Sciences of Ukraine, 42 akad. Vernadskoho Blvd., Kyiv, 03142, Ukraine
- Corresponding author.
| |
Collapse
|
14
|
Thamm S, Willwacher MK, Aspnes GE, Bretschneider T, Brown NF, Buschbom-Helmke S, Fox T, Gargano EM, Grabowski D, Hoenke C, Matera D, Mueck K, Peters S, Reindl S, Riether D, Schmid M, Tautermann CS, Teitelbaum AM, Trünkle C, Veser T, Winter M, Wortmann L. Discovery of a Novel Potent and Selective HSD17B13 Inhibitor, BI-3231, a Well-Characterized Chemical Probe Available for Open Science. J Med Chem 2023; 66:2832-2850. [PMID: 36727857 PMCID: PMC9969402 DOI: 10.1021/acs.jmedchem.2c01884] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Genome-wide association studies in patients revealed HSD17B13 as a potential new target for the treatment of nonalcoholic steatohepatitis (NASH) and other liver diseases. However, the physiological function and the disease-relevant substrate of HSD17B13 remain unknown. In addition, no suitable chemical probe for HSD17B13 has been published yet. Herein, we report the identification of the novel potent and selective HSD17B13 inhibitor BI-3231. Through high-throughput screening (HTS), using estradiol as substrate, compound 1 was identified and selected for subsequent optimization resulting in compound 45 (BI-3231). In addition to the characterization of compound 45 for its functional, physicochemical, and drug metabolism and pharmacokinetic (DMPK) properties, NAD+ dependency was investigated. To support Open Science, the chemical HSD17B13 probe BI-3231 will be available to the scientific community for free via the opnMe platform, and thus can help to elucidate the pharmacology of HSD17B13.
Collapse
Affiliation(s)
- Sven Thamm
- Boehringer
Ingelheim Pharma GmbH & Co. KG, 88397 Biberach an der Riß, Germany,
| | | | - Gary E. Aspnes
- Boehringer
Ingelheim Pharma GmbH & Co. KG, 88397 Biberach an der Riß, Germany
| | - Tom Bretschneider
- Boehringer
Ingelheim Pharma GmbH & Co. KG, 88397 Biberach an der Riß, Germany
| | - Nicholas F. Brown
- Boehringer
Ingelheim Pharmaceuticals, Inc., 900 Ridgebury Road, PO Box 368, Ridgefield, Connecticut 06877-0368, United States
| | | | - Thomas Fox
- Boehringer
Ingelheim Pharma GmbH & Co. KG, 88397 Biberach an der Riß, Germany
| | - Emanuele M. Gargano
- Boehringer
Ingelheim Pharma GmbH & Co. KG, 88397 Biberach an der Riß, Germany
| | - Daniel Grabowski
- Boehringer
Ingelheim Pharma GmbH & Co. KG, 88397 Biberach an der Riß, Germany
| | - Christoph Hoenke
- Boehringer
Ingelheim Pharma GmbH & Co. KG, 88397 Biberach an der Riß, Germany
| | - Damian Matera
- Boehringer
Ingelheim Pharmaceuticals, Inc., 900 Ridgebury Road, PO Box 368, Ridgefield, Connecticut 06877-0368, United States
| | - Katja Mueck
- Boehringer
Ingelheim Pharma GmbH & Co. KG, 88397 Biberach an der Riß, Germany
| | - Stefan Peters
- Boehringer
Ingelheim Pharma GmbH & Co. KG, 88397 Biberach an der Riß, Germany
| | - Sophia Reindl
- Boehringer
Ingelheim Pharma GmbH & Co. KG, 88397 Biberach an der Riß, Germany
| | - Doris Riether
- Boehringer
Ingelheim Pharma GmbH & Co. KG, 88397 Biberach an der Riß, Germany
| | - Matthias Schmid
- Boehringer
Ingelheim Pharma GmbH & Co. KG, 88397 Biberach an der Riß, Germany
| | | | - Aaron M. Teitelbaum
- Boehringer
Ingelheim Pharma GmbH & Co. KG, 88397 Biberach an der Riß, Germany
| | - Cornelius Trünkle
- Boehringer
Ingelheim Pharma GmbH & Co. KG, 88397 Biberach an der Riß, Germany
| | - Thomas Veser
- Boehringer
Ingelheim Pharma GmbH & Co. KG, 88397 Biberach an der Riß, Germany
| | - Martin Winter
- Boehringer
Ingelheim Pharma GmbH & Co. KG, 88397 Biberach an der Riß, Germany
| | - Lars Wortmann
- Boehringer
Ingelheim Pharma GmbH & Co. KG, 88397 Biberach an der Riß, Germany,
| |
Collapse
|
15
|
Quantitative Measurements of Pharmacological and Toxicological Activity of Molecules. CHEMISTRY 2022. [DOI: 10.3390/chemistry4040097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Toxicity and pharmacological activity scales of molecules, in particular toxicants, xenobiotics, drugs, nutraceuticals, etc., are described by multiples indicators, and the most popular is the median lethal dose (LD50). At the molecular level, reversible inhibition or binding constants provide unique information on the potential activity of molecules. The important problem concerning the meaningfulness of IC50 for irreversible ligands/inhibitors is emphasized. Definitions and principles for determination of these quantitative parameters are briefly introduced in this article. Special attention is devoted to the relationships between these indicators. Finally, different approaches making it possible to link pharmacological and toxicological properties of molecules in terms of molecular interactions (or chemical reactions) with their biological targets are briefly examined. Experimental trends for future high-throughput screening of active molecules are pointed out.
Collapse
|
16
|
Saccoccia F, Pozzetti L, Gimmelli R, Butini S, Guidi A, Papoff G, Giannaccari M, Brogi S, Scognamiglio V, Gemma S, Ruberti G, Campiani G. Crystal structures of Schistosoma mansoni histone deacetylase 8 reveal a novel binding site for allosteric inhibitors. J Biol Chem 2022; 298:102375. [PMID: 35970392 PMCID: PMC9486128 DOI: 10.1016/j.jbc.2022.102375] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 08/10/2022] [Accepted: 08/11/2022] [Indexed: 11/30/2022] Open
Abstract
Parasitic diseases cause significant global morbidity and mortality particularly in the poorest regions of the world. Schistosomiasis, one of the most widespread neglected tropical diseases, affects more than 200 million people worldwide. Histone deacetylase (HDAC) inhibitors are prominent epigenetic drugs that are being investigated in the treatment of several diseases, including cancers and parasitic diseases. Schistosoma mansoni HDAC8 (SmHDAC8) is highly expressed in all life cycle stages of the parasite and selective inhibition is required in order to avoid undesirable off-target effects in the host. Herein, by X-ray crystal structures of SmHDAC8-inhibitor complexes, biochemical and phenotypic studies, we found two schistosomicidal spiroindoline-derivatives binding a novel site, next to Trp198, on the enzyme surface. We determined that by acting on this site, either by mutation of the Trp198 or by compound binding, a decrease in the activity of the enzyme is achieved. Remarkably, this allosteric site differs from the human counterpart; rather, it is conserved in all Schistosoma spp., as well as Rhabidoptera and Trematoda classes, thus paving the way for the design of HDAC8-selective allosteric inhibitors with improved properties.
Collapse
Affiliation(s)
- Fulvio Saccoccia
- Institute of Biochemistry and Cell Biology, Italian National Research Council (IBBC-CNR), Adriano Buzzati-Traverso Campus, Via Ercole Ramarini 32, 00015 Monterotondo Scalo, Rome, Italy.
| | - Luca Pozzetti
- Department of Biotechnology, Chemistry and Pharmacy, DoE Department of Excellence 2018-2022, University of Siena, via Aldo Moro 2, I-53100 Siena, Italy
| | - Roberto Gimmelli
- Institute of Biochemistry and Cell Biology, Italian National Research Council (IBBC-CNR), Adriano Buzzati-Traverso Campus, Via Ercole Ramarini 32, 00015 Monterotondo Scalo, Rome, Italy
| | - Stefania Butini
- Department of Biotechnology, Chemistry and Pharmacy, DoE Department of Excellence 2018-2022, University of Siena, via Aldo Moro 2, I-53100 Siena, Italy
| | - Alessandra Guidi
- Institute of Biochemistry and Cell Biology, Italian National Research Council (IBBC-CNR), Adriano Buzzati-Traverso Campus, Via Ercole Ramarini 32, 00015 Monterotondo Scalo, Rome, Italy
| | - Giuliana Papoff
- Institute of Biochemistry and Cell Biology, Italian National Research Council (IBBC-CNR), Adriano Buzzati-Traverso Campus, Via Ercole Ramarini 32, 00015 Monterotondo Scalo, Rome, Italy
| | - Marialaura Giannaccari
- Institute of Biochemistry and Cell Biology, Italian National Research Council (IBBC-CNR), Adriano Buzzati-Traverso Campus, Via Ercole Ramarini 32, 00015 Monterotondo Scalo, Rome, Italy
| | - Simone Brogi
- Department of Pharmacy, University of Pisa, Via Bonanno 6, I-56126 Pisa, Italy
| | - Viviana Scognamiglio
- Institute of Crystallography, Italian National Research Council, Department of Chemical Sciences and Materials Technologies, Via Salaria km 29.300, 00015 Monterotondo, Italy
| | - Sandra Gemma
- Department of Biotechnology, Chemistry and Pharmacy, DoE Department of Excellence 2018-2022, University of Siena, via Aldo Moro 2, I-53100 Siena, Italy
| | - Giovina Ruberti
- Institute of Biochemistry and Cell Biology, Italian National Research Council (IBBC-CNR), Adriano Buzzati-Traverso Campus, Via Ercole Ramarini 32, 00015 Monterotondo Scalo, Rome, Italy.
| | - Giuseppe Campiani
- Department of Biotechnology, Chemistry and Pharmacy, DoE Department of Excellence 2018-2022, University of Siena, via Aldo Moro 2, I-53100 Siena, Italy.
| |
Collapse
|
17
|
Andrade JM, Pachar P, Trujillo L, Cartuche L. Suillin: A mixed-type acetylcholinesterase inhibitor from Suillus luteus which is used by Saraguros indigenous, southern Ecuador. PLoS One 2022; 17:e0268292. [PMID: 35576219 PMCID: PMC9109927 DOI: 10.1371/journal.pone.0268292] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 04/27/2022] [Indexed: 11/19/2022] Open
Abstract
Suillus luteus (L.) Roussel is an edible mushroom commonly known as slippery jack or “Kallampa” by indigenous people from Loja province. It is used in traditional medicine to manage gastrointestinal disorders and headaches. In addition, edible mushrooms have been used for neurodegenerative diseases; however, there is no report about the anticholinesterase effect produced by this species. The aim of this work was to isolate the main secondary metabolite of Suillus luteus and characterize its inhibitory potential against acetylcholinesterase. Fruiting bodies were extracted with ethanol (EtOH) and ethyl acetate (EtOAc). From the EtOAc, suillin, is reported as the major compound. The cholinesterase inhibitory potential of extracts and the major isolated compound was assessed by Ellman´s method and progression curves were recorded at 405 nm for 60 min. Donepezil hydroclhoride was used as a positive control. The samples were dissolved in methanol at 10 mg/mL and two more 10× dilutions were included to obtain final concentrations of 1, 0.1 and 0.01 mg/mL at the mix of reaction. IC50, Km, Vmax, and Ki were calculated for suillin. Suillin (200 mg) along with linoleic acid, ergosterol peroxide and ergosterol were isolated. The EtOH and EtOAc extracts exerted a moderate inhibitory effect (IC50 > 200 μg/mL. In adittion, suillin exerted a non-competitive mixed mechanism. against AChE with an IC50 value of 31.50 μM and Ki of 17.25 μM. To the best of our knowledge, this is the first report of the anticholinesterase effect of Suillus luteus and suillin. The kinetic parameters and the moderate potency of the compound determined in this study, encourage us to propose suillin as a promising chemopreventing agent for the treatment of neurodegenerative diseases such as Alzheimer.
Collapse
Affiliation(s)
- José Miguel Andrade
- Departamento de Química, Universidad Técnica Particular de Loja (UTPL), Loja, Ecuador
| | - Pamela Pachar
- Departamento de Química, Universidad Técnica Particular de Loja (UTPL), Loja, Ecuador
| | - Luisa Trujillo
- Departamento de Química, Universidad Técnica Particular de Loja (UTPL), Loja, Ecuador
| | - Luis Cartuche
- Departamento de Química, Universidad Técnica Particular de Loja (UTPL), Loja, Ecuador
- * E-mail:
| |
Collapse
|
18
|
The Relationship between the IC50 Values and the Apparent Inhibition Constant in the Study of Inhibitors of Tyrosinase Diphenolase Activity Helps Confirm the Mechanism of Inhibition. Molecules 2022; 27:molecules27103141. [PMID: 35630619 PMCID: PMC9142954 DOI: 10.3390/molecules27103141] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 05/03/2022] [Accepted: 05/11/2022] [Indexed: 12/02/2022] Open
Abstract
Tyrosinase is the enzyme involved in melanization and is also responsible for the browning of fruits and vegetables. Control of its activity can be carried out using inhibitors, which is interesting in terms of quantitatively understanding the action of these regulators. In the study of the inhibition of the diphenolase activity of tyrosinase, it is intriguing to know the strength and type of inhibition. The strength is indicated by the value of the inhibition constant(s), and the type can be, in a first approximation: competitive, non-competitive, uncompetitive and mixed. In this work, it is proposed to calculate the degree of inhibition (iD), varying the concentration of inhibitor to a fixed concentration of substrate, L-dopa (D). The non-linear regression adjustment of iD with respect to the initial inhibitor concentration [I]0 allows for the calculation of the inhibitor concentration necessary to inhibit the activity by 50%, at a given substrate concentration (IC50), thus avoiding making interpolations between different values of iD. The analytical expression of the IC50, for the different types of inhibition, are related to the apparent inhibition constant (KIapp). Therefore, this parameter can be used: (a) To classify a series of inhibitors of an enzyme by their power. Determining these values at a fixed substrate concentration, the lower IC50, the more potent the inhibitor. (b) Checking an inhibitor for which the type and the inhibition constant have been determined (using the usual methods), must confirm the IC50 value according to the corresponding analytical expression. (c) The type and strength of an inhibitor can be analysed from the study of the variation in iD and IC50 with substrate concentration. The dependence of IC50 on the substrate concentration allows us to distinguish between non-competitive inhibition (iD does not depend on [D]0) and the rest. In the case of competitive inhibition, this dependence of iD on [D]0 leads to an ambiguity between competitive inhibition and type 1 mixed inhibition. This is solved by adjusting the data to the possible equations; in the case of a competitive inhibitor, the calculation of KI1app is carried out from the IC50 expression. The same occurs with uncompetitive inhibition and type 2 mixed inhibition. The representation of iD vs. n, with n=[D]0/KmD, allows us to distinguish between them. A hyperbolic iD vs. n representation that passes through the origin of coordinates is a characteristic of uncompetitive inhibition; the calculation of KI2app is immediate from the IC50 value. In the case of mixed inhibitors, the values of the apparent inhibition constant of meta-tyrosinase (Em) and oxy-tyrosinase (Eox), KI1app and the apparent inhibition constant of metatyrosinase/Dopa complexes (EmD) and oxytyrosinase/Dopa (EoxD), KI2app are obtained from the dependence of iD vs. n, and the results obtained must comply with the IC50 value.
Collapse
|
19
|
Satapati S, Downes DP, Metzger D, Shankaran H, Talukdar S, Zhou Y, Ren Z, Chen M, Lim YH, Hatcher NG, Wen X, Sheth PR, McLaren DG, Previs SF. Using measures of metabolic flux to align screening and clinical development: Avoiding pitfalls to enable translational studies. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2022; 27:20-28. [PMID: 35058172 DOI: 10.1016/j.slasd.2021.10.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Screening campaigns, especially those aimed at modulating enzyme activity, often rely on measuring substrate→product conversions. Unfortunately, the presence of endogenous substrates and/or products can limit one's ability to measure conversions. As well, coupled detection systems, often used to facilitate optical readouts, are subject to interference. Stable isotope labeled substrates can overcome background contamination and yield a direct readout of enzyme activity. Not only can isotope kinetic assays enable early screening, but they can also be used to follow hit progression in translational (pre)clinical studies. Herein, we consider a case study surrounding lipid biology to exemplify how metabolic flux analyses can connect stages of drug development, caveats are highlighted to ensure reliable data interpretations. For example, when measuring enzyme activity in early biochemical screening it may be enough to quantify the formation of a labeled product. In contrast, cell-based and in vivo studies must account for variable exposure to a labeled substrate (or precursor) which occurs via tracer dilution and/or isotopic exchange. Strategies are discussed to correct for these complications. We believe that measures of metabolic flux can help connect structure-activity relationships with pharmacodynamic mechanisms of action and determine whether mechanistically differentiated biophysical interactions lead to physiologically relevant outcomes. Adoption of this logic may allow research programs to (i) build a critical bridge between primary screening and (pre)clinical development, (ii) elucidate biology in parallel with screening and (iii) suggest a strategy aimed at in vivo biomarker development.
Collapse
Affiliation(s)
- Santhosh Satapati
- Merck & Co., Inc, 213 E. Grand Ave, South San Francisco, CA, 94080, USA
| | - Daniel P Downes
- Merck & Co., Inc, 2000 Galloping Hill Rd, Kenilworth, NJ, 07033, USA
| | - Daniel Metzger
- Merck & Co., Inc, 213 E. Grand Ave, South San Francisco, CA, 94080, USA
| | - Harish Shankaran
- Merck & Co., Inc, 213 E. Grand Ave, South San Francisco, CA, 94080, USA
| | - Saswata Talukdar
- Merck & Co., Inc, 213 E. Grand Ave, South San Francisco, CA, 94080, USA
| | - Yingjiang Zhou
- Merck & Co., Inc, 213 E. Grand Ave, South San Francisco, CA, 94080, USA
| | - Zhao Ren
- Merck & Co., Inc, 213 E. Grand Ave, South San Francisco, CA, 94080, USA
| | - Michelle Chen
- Merck & Co., Inc, 213 E. Grand Ave, South San Francisco, CA, 94080, USA
| | - Yeon-Hee Lim
- Merck & Co., Inc, 213 E. Grand Ave, South San Francisco, CA, 94080, USA
| | - Nathan G Hatcher
- Merck & Co., Inc, 770 Sumneytown Pike, West Point, PA, 19486, USA
| | - Xiujuan Wen
- Merck & Co., Inc, 2000 Galloping Hill Rd, Kenilworth, NJ, 07033, USA
| | - Payal R Sheth
- Merck & Co., Inc, 2000 Galloping Hill Rd, Kenilworth, NJ, 07033, USA
| | - David G McLaren
- Merck & Co., Inc, 2000 Galloping Hill Rd, Kenilworth, NJ, 07033, USA
| | - Stephen F Previs
- Merck & Co., Inc, 2000 Galloping Hill Rd, Kenilworth, NJ, 07033, USA.
| |
Collapse
|
20
|
Lloyd MD, Yevglevskis M, Nathubhai A, James TD, Threadgill MD, Woodman TJ. Racemases and epimerases operating through a 1,1-proton transfer mechanism: reactivity, mechanism and inhibition. Chem Soc Rev 2021; 50:5952-5984. [PMID: 34027955 PMCID: PMC8142540 DOI: 10.1039/d0cs00540a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Indexed: 12/12/2022]
Abstract
Racemases and epimerases catalyse changes in the stereochemical configurations of chiral centres and are of interest as model enzymes and as biotechnological tools. They also occupy pivotal positions within metabolic pathways and, hence, many of them are important drug targets. This review summarises the catalytic mechanisms of PLP-dependent, enolase family and cofactor-independent racemases and epimerases operating by a deprotonation/reprotonation (1,1-proton transfer) mechanism and methods for measuring their catalytic activity. Strategies for inhibiting these enzymes are reviewed, as are specific examples of inhibitors. Rational design of inhibitors based on substrates has been extensively explored but there is considerable scope for development of transition-state mimics and covalent inhibitors and for the identification of inhibitors by high-throughput, fragment and virtual screening approaches. The increasing availability of enzyme structures obtained using X-ray crystallography will facilitate development of inhibitors by rational design and fragment screening, whilst protein models will facilitate development of transition-state mimics.
Collapse
Affiliation(s)
- Matthew D Lloyd
- Drug & Target Discovery, Department of Pharmacy & Pharmacology, University of Bath, Claverton Down, Bath BA2 7AY, UK.
| | - Maksims Yevglevskis
- Drug & Target Discovery, Department of Pharmacy & Pharmacology, University of Bath, Claverton Down, Bath BA2 7AY, UK. and CatSci Ltd., CBTC2, Capital Business Park, Wentloog, Cardiff CF3 2PX, UK
| | - Amit Nathubhai
- Drug & Target Discovery, Department of Pharmacy & Pharmacology, University of Bath, Claverton Down, Bath BA2 7AY, UK. and University of Sunderland, School of Pharmacy & Pharmaceutical Sciences, Sciences Complex, Sunderland SR1 3SD, UK
| | - Tony D James
- Department of Chemistry, University of Bath, Claverton Down, Bath BA2 7AY, UK and School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, People's Republic of China
| | - Michael D Threadgill
- Drug & Target Discovery, Department of Pharmacy & Pharmacology, University of Bath, Claverton Down, Bath BA2 7AY, UK. and Institute of Biological, Environmental & Rural Sciences, Aberystwyth University, Aberystwyth SY23 3BY, UK
| | - Timothy J Woodman
- Drug & Target Discovery, Department of Pharmacy & Pharmacology, University of Bath, Claverton Down, Bath BA2 7AY, UK.
| |
Collapse
|
21
|
Whitfield H, Hemmings AM, Mills SJ, Baker K, White G, Rushworth S, Riley AM, Potter BVL, Brearley CA. Allosteric Site on SHIP2 Identified Through Fluorescent Ligand Screening and Crystallography: A Potential New Target for Intervention. J Med Chem 2021; 64:3813-3826. [PMID: 33724834 PMCID: PMC7610569 DOI: 10.1021/acs.jmedchem.0c01944] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Src homology 2 domain-containing inositol phosphate phosphatase 2 (SHIP2) is one of the 10 human inositol phosphate 5-phosphatases. One of its physiological functions is dephosphorylation of phosphatidylinositol 3,4,5-trisphosphate, PtdIns(3,4,5)P3. It is therefore a therapeutic target for pathophysiologies dependent on PtdIns(3,4,5)P3 and PtdIns(3,4)P2. Therapeutic interventions are limited by the dearth of crystallographic data describing ligand/inhibitor binding. An active site-directed fluorescent probe facilitated screening of compound libraries for SHIP2 ligands. With two additional orthogonal assays, several ligands including galloflavin were identified as low micromolar Ki inhibitors. One ligand, an oxo-linked ethylene-bridged dimer of benzene 1,2,4-trisphosphate, was shown to be an uncompetitive inhibitor that binds to a regulatory site on the catalytic domain. We posit that binding of ligands to this site restrains L4 loop motions that are key to interdomain communications that accompany high catalytic activity with phosphoinositide substrate. This site may, therefore, be a future druggable target for medicinal chemistry.
Collapse
Affiliation(s)
- Hayley Whitfield
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, U.K
| | - Andrew M Hemmings
- School of Chemistry, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, U.K
| | - Stephen J Mills
- Medicinal Chemistry & Drug Discovery, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, U.K
| | - Kendall Baker
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, U.K
| | - Gaye White
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, U.K
| | - Stuart Rushworth
- Department of Molecular Haematology; Norwich Medical School, University of East Anglia, Norwich NR4 7TJ, U.K
| | - Andrew M Riley
- Medicinal Chemistry & Drug Discovery, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, U.K
| | - Barry V L Potter
- Medicinal Chemistry & Drug Discovery, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, U.K
| | - Charles A Brearley
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, U.K
| |
Collapse
|
22
|
Davies G, Semple H, McCandless M, Cairns J, Holdgate GA. High-Throughput Mechanism of Inhibition. SLAS DISCOVERY 2021; 26:248-256. [PMID: 33482076 DOI: 10.1177/2472555220983809] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Enzymes represent a significant proportion of the druggable genome and constitute a rich source of drug targets. Delivery of a successful program for developing a modulator of enzyme activity requires an understanding of the enzyme's mechanism and the mode of interaction of compounds. This allows an understanding of how physiological conditions in disease-relevant cells will affect inhibitor potency. As a result, there is increasing interest in evaluating hit compounds from high-throughput screens to determine their mode of interaction with the target. This work revisits the common inhibition modalities and illustrates the impact of substrate concentration relative to Km upon the pattern of changes in IC50 that are expected for increasing substrate concentration. It proposes a new, high-throughput approach for assessing mode of inhibition, incorporating analyses based on a minimal descriptive model, to deliver a workflow that allows rapid and earlier compound classification immediately after high-throughput screening.
Collapse
Affiliation(s)
- Gareth Davies
- Hit Discovery, Discovery Sciences, R&D, AstraZeneca, Alderley Park UK
| | - Hannah Semple
- Hit Discovery, Discovery Sciences, R&D, AstraZeneca, Alderley Park UK
| | - Megan McCandless
- Hit Discovery, Discovery Sciences, R&D, AstraZeneca, Alderley Park UK
| | - Jonathan Cairns
- Data Sciences and Quantitative Biology, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | | |
Collapse
|
23
|
Srinivasan B. Explicit Treatment of Non-Michaelis-Menten and Atypical Kinetics in Early Drug Discovery*. ChemMedChem 2020; 16:899-918. [PMID: 33231926 DOI: 10.1002/cmdc.202000791] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Indexed: 12/27/2022]
Abstract
Biological systems are highly regulated. They are also highly resistant to sudden perturbations enabling them to maintain the dynamic equilibrium essential to sustain life. This robustness is conferred by regulatory mechanisms that influence the activity of enzymes/proteins within their cellular context to adapt to changing environmental conditions. However, the initial rules governing the study of enzyme kinetics were mostly tested and implemented for cytosolic enzyme systems that were easy to isolate and/or recombinantly express. Moreover, these enzymes lacked complex regulatory modalities. Now, with academic labs and pharmaceutical companies turning their attention to more-complex systems (for instance, multiprotein complexes, oligomeric assemblies, membrane proteins and post-translationally modified proteins), the initial axioms defined by Michaelis-Menten (MM) kinetics are rendered inadequate, and the development of a new kind of kinetic analysis to study these systems is required. This review strives to present an overview of enzyme kinetic mechanisms that are atypical and, oftentimes, do not conform to the classical MM kinetics. Further, it presents initial ideas on the design and analysis of experiments in early drug-discovery for such systems, to enable effective screening and characterisation of small-molecule inhibitors with desirable physiological outcomes.
Collapse
Affiliation(s)
- Bharath Srinivasan
- Mechanistic Biology and Profiling Discovery Sciences, R&D, AstraZeneca, 310, Milton Rd, Milton CB4 0WG, Cambridge, UK
| |
Collapse
|
24
|
Srinivasan B, Kantae V, Robinson J. Resurrecting the phoenix: When an assay fails. Med Res Rev 2020; 40:1776-1793. [DOI: 10.1002/med.21670] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 02/18/2020] [Accepted: 03/27/2020] [Indexed: 11/10/2022]
Affiliation(s)
- Bharath Srinivasan
- Mechanistic Biology and Profiling, Discovery Sciences, R&D, AstraZeneca Cambridge UK
| | - Vasudev Kantae
- Mechanistic Biology and Profiling, Discovery Sciences, R&D, AstraZeneca Cambridge UK
| | - James Robinson
- Mechanistic Biology and Profiling, Discovery Sciences, R&D, AstraZeneca Cambridge UK
| |
Collapse
|
25
|
Walsh R. Response to the Article "Enzyme-Inhibitor Interactions and a Simple, Rapid Method for Determining Inhibition Modality". SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2020; 25:7. [PMID: 31513471 DOI: 10.1177/2472555219872211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
|
26
|
Buker SM, Gurard-Levin ZA, Wheeler BD, Scholle MD, Case AW, Hirsch JL, Ribich S, Copeland RA, Boriack-Sjodin PA. A Mass Spectrometric Assay of METTL3/METTL14 Methyltransferase Activity. SLAS DISCOVERY 2019; 25:361-371. [PMID: 31585521 DOI: 10.1177/2472555219878408] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A variety of covalent modifications of RNA have been identified and demonstrated to affect RNA processing, stability, and translation. Methylation of adenosine at the N6 position (m6A) in messenger RNA (mRNA) is currently the most well-studied RNA modification and is catalyzed by the RNA methyltransferase complex METTL3/METTL14. Once generated, m6A can modulate mRNA splicing, export, localization, degradation, and translation. Although potent and selective inhibitors exist for several members of the Type I S-adenosylmethionine (SAM)-dependent methyltransferase family, no inhibitors have been reported for METTL3/METTL14 to date. To facilitate drug discovery efforts, a sensitive and robust mass spectrometry-based assay for METTL3/METTL14 using self-assembled monolayer desorption/ionization (SAMDI) technology has been developed. The assay uses an 11-nucleotide single-stranded RNA compared to a previously reported 27-nucleotide substrate. IC50 values of mechanism-based inhibitors S-adenosylhomocysteine (SAH) and sinefungin (SFG) are comparable between the SAMDI and radiometric assays that use the same substrate. This work demonstrates that SAMDI technology is amenable to RNA substrates and can be used for high-throughput screening and compound characterization for RNA-modifying enzymes.
Collapse
Affiliation(s)
| | | | - Benjamin D Wheeler
- Confluence Discovery Technologies, St. Louis, MO, USA.,Biomedical Science Program, University of California, San Francisco, CA, USA
| | | | | | | | | | | | | |
Collapse
|