51
|
Wang J, Do HN, Koirala K, Miao Y. Predicting Biomolecular Binding Kinetics: A Review. J Chem Theory Comput 2023; 19:2135-2148. [PMID: 36989090 DOI: 10.1021/acs.jctc.2c01085] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Biomolecular binding kinetics including the association (kon) and dissociation (koff) rates are critical parameters for therapeutic design of small-molecule drugs, peptides, and antibodies. Notably, the drug molecule residence time or dissociation rate has been shown to correlate with their efficacies better than binding affinities. A wide range of modeling approaches including quantitative structure-kinetic relationship models, Molecular Dynamics simulations, enhanced sampling, and Machine Learning has been developed to explore biomolecular binding and dissociation mechanisms and predict binding kinetic rates. Here, we review recent advances in computational modeling of biomolecular binding kinetics, with an outlook for future improvements.
Collapse
Affiliation(s)
- Jinan Wang
- Center for Computational Biology and Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas 66047, United States
| | - Hung N Do
- Center for Computational Biology and Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas 66047, United States
| | - Kushal Koirala
- Center for Computational Biology and Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas 66047, United States
| | - Yinglong Miao
- Center for Computational Biology and Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas 66047, United States
| |
Collapse
|
52
|
Bansode AH, Bhoopal B, Gollapelli KK, Damuka N, Krizan I, Miller M, Craft S, Mintz A, Solingapuram Sai KK. Binding Parameters of [ 11C]MPC-6827, a Microtubule-Imaging PET Radiopharmaceutical in Rodents. Pharmaceuticals (Basel) 2023; 16:495. [PMID: 37111252 PMCID: PMC10140836 DOI: 10.3390/ph16040495] [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: 02/24/2023] [Revised: 03/18/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023] Open
Abstract
Impairment and/or destabilization of neuronal microtubules (MTs) resulting from hyper-phosphorylation of the tau proteins is implicated in many pathologies, including Alzheimer's disease (AD), Parkinson's disease and other neurological disorders. Increasing scientific evidence indicates that MT-stabilizing agents protect against the deleterious effects of neurodegeneration in treating AD. To quantify these protective benefits, we developed the first brain-penetrant PET radiopharmaceutical, [11C]MPC-6827, for in vivo quantification of MTs in rodent and nonhuman primate models of AD. Mechanistic insights revealed from recently reported studies confirm the radiopharmaceutical's high selectivity for destabilized MTs. To further translate it to clinical settings, its metabolic stability and pharmacokinetic parameters must be determined. Here, we report in vivo plasma and brain metabolism studies establishing the radiopharmaceutical-binding constants of [11C]MPC-6827. Binding constants were extrapolated from autoradiography experiments; pretreatment with a nonradioactive MPC-6827 decreased the brain uptake >70%. It exhibited ideal binding characteristics (typical of a CNS radiopharmaceutical) including LogP (2.9), Kd (15.59 nM), and Bmax (11.86 fmol/mg). Most important, [11C]MPC-6827 showed high serum and metabolic stability (>95%) in rat plasma and brain samples.
Collapse
Affiliation(s)
- Avinash H. Bansode
- Department of Radiology, Wake Forest School of Medicine, Winston Salem, NC 27157, USA
| | | | | | - Naresh Damuka
- Department of Radiology, Wake Forest School of Medicine, Winston Salem, NC 27157, USA
| | - Ivan Krizan
- Department of Radiology, Wake Forest School of Medicine, Winston Salem, NC 27157, USA
| | - Mack Miller
- Department of Radiology, Wake Forest School of Medicine, Winston Salem, NC 27157, USA
| | - Suzanne Craft
- Department of Gerontology, Wake Forest School of Medicine, Winston Salem, NC 27157, USA
| | - Akiva Mintz
- Department of Radiology, Columbia Medical Center, New York, NY 10032, USA
| | | |
Collapse
|
53
|
Baptista S, Pereira JR, Guerreiro BM, Baptista F, Silva JC, Freitas F. Cosmetic emulsion based on the fucose-rich polysaccharide FucoPol: Bioactive properties and sensorial evaluation. Colloids Surf B Biointerfaces 2023; 225:113252. [PMID: 36931042 DOI: 10.1016/j.colsurfb.2023.113252] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 02/15/2023] [Accepted: 03/04/2023] [Indexed: 03/08/2023]
Abstract
In this study, the physicochemical characteristics, bioactive properties, and sensorial evaluation of a O/W cosmetic formulation containing FucoPol, a fucose-containing bacterial polysaccharide, were assessed. The stability of the FucoPol-based cream, named F-cream, was demonstrated over a period of 2 months at different temperatures (4, 20 and 30 °C), during which it maintained the organoleptic characteristics and pH (5.88-6.19), with minimal variations on the apparent viscosity. Furthermore, no breaking mechanisms occurred upon centrifuging the samples (accelerated stability test) kept at 4 °C and at 30 °C for 60 days. The F-cream presented a shear-thinning and solid-liquid behavior consistent with its envisaged use for topical applications, proving to be a suitable candidate for an anti-aging application due to its antioxidant capacity and effective photoprotection, maintaining cellular preservation. Moreover, the formulation was proven non-cytotoxic for HaCaT cells at concentrations between 0.78 and 12.5 mg/mL, promoting HFFF2 cell migration (46-70 % of wound closure) at a concentration of 2.5 mg/mL, and HaCaT cell migration at a concentration of 10 mg/mL (95-98 % of wound closure). Upon application over the skin, the F-cream provided a hydration and softness with desired spreadability with no residues after application. These findings show that FucoPol has good potential to be used as a functional and/or active ingredient in cosmetic formulations, forming an emulsified cream with appealing sensorial properties that can act as a moisturizer with photoprotection, antioxidant, and regeneration properties.
Collapse
Affiliation(s)
- Sílvia Baptista
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, School of Science and Technology, NOVA University Lisbon, Caparica, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Department of Chemistry, School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal; 73100, Lda. Edifício Arcis, Rua Ivone Silva, 6, 4º piso, 1050-124 Lisboa, Portugal
| | - João R Pereira
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, School of Science and Technology, NOVA University Lisbon, Caparica, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Department of Chemistry, School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal; CENIMAT/I3N, Department of Physics, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
| | - Bruno M Guerreiro
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, School of Science and Technology, NOVA University Lisbon, Caparica, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Department of Chemistry, School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal; CENIMAT/I3N, Department of Physics, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
| | - Filipa Baptista
- UCIBIO - Applied Molecular Biosciences Unit, Department of Chemistry, School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
| | - Jorge C Silva
- CENIMAT/I3N, Department of Physics, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
| | - Filomena Freitas
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, School of Science and Technology, NOVA University Lisbon, Caparica, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Department of Chemistry, School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal.
| |
Collapse
|
54
|
Parolo C, Idili A, Heikenfeld J, Plaxco KW. Conformational-switch biosensors as novel tools to support continuous, real-time molecular monitoring in lab-on-a-chip devices. LAB ON A CHIP 2023; 23:1339-1348. [PMID: 36655710 PMCID: PMC10799767 DOI: 10.1039/d2lc00716a] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Recent years have seen continued expansion of the functionality of lab on a chip (LOC) devices. Indeed LOCs now provide scientists and developers with useful and versatile platforms across a myriad of chemical and biological applications. The field still fails, however, to integrate an often important element of bench-top analytics: real-time molecular measurements that can be used to "guide" a chemical response. Here we describe the analytical techniques that could provide LOCs with such real-time molecular monitoring capabilities. It appears to us that, among the approaches that are general (i.e., that are independent of the reactive or optical properties of their targets), sensing strategies relying on binding-induced conformational change of bioreceptors are most likely to succeed in such applications.
Collapse
Affiliation(s)
- Claudio Parolo
- Barcelona Institute for Global Health, Hospital Clínic Universitat de Barcelona, 08036, Barcelona, Spain
| | - Andrea Idili
- Department of Chemical Science and Technologies, University of Rome, Tor Vergata, 00133 Rome, Italy
| | - Jason Heikenfeld
- Novel Devices Laboratory, University of Cincinnati, Cincinnati, Ohio, USA
| | - Kevin W Plaxco
- Department of Chemistry and Biochemistry, University of California Santa Barbara, Santa Barbara, California, USA.
- Interdepartmental Program in Biomolecular Science and Engineering, University of California Santa Barbara, Santa Barbara, California, USA
| |
Collapse
|
55
|
Özyurt C, Uludağ İ, Sezgintürk MK. An ultrasensitive and disposable electrochemical aptasensor for prostate-specific antigen (PSA) detection in real serum samples. Anal Bioanal Chem 2023; 415:1123-1136. [PMID: 36155829 DOI: 10.1007/s00216-022-04309-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/22/2022] [Accepted: 08/29/2022] [Indexed: 02/07/2023]
Abstract
In this study, we constructed a disposable indium tin oxide polyethylene terephthalate film (ITO-PET)-based electrochemical aptasensor for analyzing prostate-specific antigen (PSA), one of the most important biomarkers of prostate cancer. Because of their clinical importance, building PSA biosensing systems with high sensitivity and stability is essential. However, it still presents significant difficulties, such as low detection limits. We designed a platform to covalently bind the amino-terminated aptamer by modifying the ITO-PET surface with carboxyethylsilanetriol (CTES) to obtain a self-assembled monolayer (SAM). We also evaluated the potential for use in real human serum samples by investigating the optimal operating conditions and analytical performance characteristics of the developed biosensor. The design we present here exhibits excellent precision, with a limit of detection (LOD) as low as 8.74 fg/mL PSA. The broad linear detection range of the biosensor under optimal conditions was determined as 1.0-1500 fg/mL. The dissociation constant (Kd) for the aptamer was also calculated as 46.28 ± 5.63 nM by evaluating the impedimetric response as a function of PSA concentration. The aptasensor displayed considerable repeatability (1.3% RSD) and reproducibility (7.51% RSD) and good storage stability (98.34% of the initial activity for 8 weeks). Additionally, we demonstrated that the technique we developed was quite efficient in estimating the kinetics of aptamer-analyte interactions by determining the Kd and single-frequency impedance (SFI) data. In conclusion, we proposed a selective and sensitive biosensor with the potential for clinical application and superior performance in real serum samples.
Collapse
Affiliation(s)
- Canan Özyurt
- Department of Chemistry and Chemical Processing Technologies, Lapseki Vocational School, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| | - İnci Uludağ
- Bioengineering Department, Engineering Faculty, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| | - Mustafa Kemal Sezgintürk
- Bioengineering Department, Engineering Faculty, Çanakkale Onsekiz Mart University, Çanakkale, Turkey.
| |
Collapse
|
56
|
Repeated exposure to 4-methyl-2,4-bis(4-hydroxyphenyl)pent-1-ene (MBP) accelerates ligand-independent activation of estrogen receptors in long-term estradiol-deprived MCF-7 cells. Toxicol Lett 2023; 378:31-38. [PMID: 36863540 DOI: 10.1016/j.toxlet.2023.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 02/16/2023] [Accepted: 02/26/2023] [Indexed: 03/02/2023]
Abstract
It was previously identified that there may be an active metabolite of bisphenol A (BPA), 4-methyl-2,4-bis(4-hydroxyphenyl)pent-1-ene (MBP). An in vitro system was developed to detect MBP toxicity to the Michigan Cancer Foundation-7 (MCF-7) cells that had been repeatedly exposed to a low dose of the metabolite. MBP profoundly activated estrogen receptor (ER)-dependent transcription as a ligand, with an EC50 of 2.8 nM. Women are continuously exposed to numerous estrogenic environmental chemicals; but their susceptibility to these chemicals may be significantly altered after menopause. Long-term estrogen-deprived (LTED) cells, which display ligand-independent ER activation, are a postmenopausal breast cancer model derived from MCF-7 cells. In this study, we investigated the estrogenic effects of MBP on LTED cells in a repeated exposure in vitro model. The results suggest that i) nanomolar levels of MBP reciprocally disrupt the balanced expression of ERα and ERβ proteins, leading to the dominant expression of ERβ, ii) MBP stimulates ERs-mediated transcription without acting as an ERβ ligand, and iii) MBP utilizes mitogen-activated protein kinase and phosphatidylinositol-3 kinase signaling to evoke its estrogenic action. Moreover, the repeated exposure strategy was effective for detecting low-dose estrogenic-like effects caused by MBP in LTED cells.
Collapse
|
57
|
Rukundo JL, Latimer J, Jain S, Kochmann S, Krylov SN. Streamlined Data Processing for Determination of Equilibrium Dissociation Constants with Accurate Constant via Transient Incomplete Separation (ACTIS). Anal Chem 2023; 95:3563-3568. [PMID: 36763923 DOI: 10.1021/acs.analchem.2c05607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
The determination of accurate equilibrium dissociation constants, Kd, of protein-small molecule complexes is important but challenging as all established methods have inherent sources of inaccuracy. Accurate Constant via Transient Incomplete Separation (ACTIS) is a new method for Kd determination using transient incomplete separation of the complex from the unbound small molecule in a pressure-driven flow inside a capillary. ACTIS is accurate, and its accuracy is invariant to variations in geometries of both the fluidic system and the flow. Furthermore, ACTIS is implemented using a simple fluidic system supporting its accuracy and providing a simple-to-follow/copy template for instrumentation. Despite the simple and robust instrumentation/acquisition, the current data processing workflow is cumbersome, time consuming, and prone to hard-to-trace human errors therefore hindering ACTIS' ability to become a practical reference method for Kd determination. This technical note describes a streamlined workflow for processing ACTIS data; the workflow is implemented as a set of open-source software tools called prACTISed (https://github.com/prACTISedProgram/prACTISed). These tools allow all steps of data processing to be performed in a fast and straightforward fashion. These practical software tools complement the simple instrumentation serving both developers and users of ACTIS.
Collapse
Affiliation(s)
- Jean-Luc Rukundo
- Department of Chemistry and Centre for Research on Biomolecular Interactions, York University, Toronto, Ontario M3J 1P3, Canada
| | - Jessica Latimer
- Department of Chemistry and Centre for Research on Biomolecular Interactions, York University, Toronto, Ontario M3J 1P3, Canada
| | - Shiv Jain
- Department of Chemistry and Centre for Research on Biomolecular Interactions, York University, Toronto, Ontario M3J 1P3, Canada
| | - Sven Kochmann
- Department of Chemistry, University of Natural Resources and Life Sciences, 1190 Vienna, Austria
| | - Sergey N Krylov
- Department of Chemistry and Centre for Research on Biomolecular Interactions, York University, Toronto, Ontario M3J 1P3, Canada
| |
Collapse
|
58
|
Lopez-Morales J, Vanella R, Kovacevic G, Santos MS, Nash MA. Titrating Avidity of Yeast-Displayed Proteins Using a Transcriptional Regulator. ACS Synth Biol 2023; 12:419-431. [PMID: 36728831 PMCID: PMC9942200 DOI: 10.1021/acssynbio.2c00351] [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: 07/04/2022] [Indexed: 02/03/2023]
Abstract
Yeast surface display is a valuable tool for protein engineering and directed evolution; however, significant variability in the copy number (i.e., avidity) of displayed variants on the yeast cell wall complicates screening and selection campaigns. Here, we report an engineered titratable display platform that modulates the avidity of Aga2-fusion proteins on the yeast cell wall dependent on the concentration of the anhydrotetracycline (aTc) inducer. Our design is based on a genomic Aga1 gene copy and an episomal Aga2-fusion construct both under the control of an aTc-dependent transcriptional regulator that enables stoichiometric and titratable expression, secretion, and display of Aga2-fusion proteins. We demonstrate tunable display levels over 2-3 orders of magnitude for various model proteins, including glucose oxidase enzyme variants, mechanostable dockerin-binding domains, and anti-PDL1 affibody domains. By regulating the copy number of displayed proteins, we demonstrate the effects of titratable avidity levels on several specific phenotypic activities, including enzyme activity and cell adhesion to surfaces under shear flow. Finally, we show that titrating down the display level allows yeast-based binding affinity measurements to be performed in a regime that avoids ligand depletion effects while maintaining small sample volumes, avoiding a well-known artifact in yeast-based binding assays. The ability to titrate the multivalency of proteins on the yeast cell wall through simple inducer control will benefit protein engineering and directed evolution methodology relying on yeast display for broad classes of therapeutic and diagnostic proteins of interest.
Collapse
Affiliation(s)
- Joanan Lopez-Morales
- Department
of Chemistry, University of Basel, Basel 4058, Switzerland
- Swiss
Nanoscience Institute, University of Basel, Basel 4056, Switzerland
- Department
of Biosystems Science and Engineering, ETH
Zurich, Basel 4058, Switzerland
| | - Rosario Vanella
- Department
of Chemistry, University of Basel, Basel 4058, Switzerland
- Department
of Biosystems Science and Engineering, ETH
Zurich, Basel 4058, Switzerland
| | - Gordana Kovacevic
- Department
of Chemistry, University of Basel, Basel 4058, Switzerland
- Department
of Biosystems Science and Engineering, ETH
Zurich, Basel 4058, Switzerland
| | - Mariana Sá Santos
- Department
of Chemistry, University of Basel, Basel 4058, Switzerland
- Department
of Biosystems Science and Engineering, ETH
Zurich, Basel 4058, Switzerland
| | - Michael A. Nash
- Department
of Chemistry, University of Basel, Basel 4058, Switzerland
- Swiss
Nanoscience Institute, University of Basel, Basel 4056, Switzerland
- Department
of Biosystems Science and Engineering, ETH
Zurich, Basel 4058, Switzerland
| |
Collapse
|
59
|
Villaluenga JPG, Brunete D, Cao-García FJ. Competitive ligand binding kinetics to linear polymers. Phys Rev E 2023; 107:024401. [PMID: 36932540 DOI: 10.1103/physreve.107.024401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 01/10/2023] [Indexed: 02/04/2023]
Abstract
Different types of ligands compete in binding to polymers with different consequences for the physical and chemical properties of the resulting complex. Here, we derive a general kinetic model for the competitive binding kinetics of different types of ligands to a linear polymer, using the McGhee and von Hippel detailed binding-site counting procedure. The derived model allows the description of the competitive binding process in terms of the size of the ligand, binding, and release rates, and cooperativity parameters. We illustrate the implications of the general theory showing the equations for the competitive binding of two ligands. The size of the ligand, given by the number of monomers occluded, is shown to have a great impact on competitive binding. Ligands requiring a large available gap for binding are strongly inhibited by smaller ligands. Ligand size then has a leading role compared to binding affinity or cooperativity. For ligands that can bind in different modes (i.e., different number of monomers), this implies that they are more effective in covering or passivating the polymer in lower modes, if the different modes have similar binding energies.
Collapse
Affiliation(s)
- Juan P G Villaluenga
- Departamento de Estructura de la Materia, Física Térmica y Electrónica, Universidad Complutense de Madrid, Plaza de Ciencias, 1, 28040 Madrid, Spain
| | - David Brunete
- Departamento de Estructura de la Materia, Física Térmica y Electrónica, Universidad Complutense de Madrid, Plaza de Ciencias, 1, 28040 Madrid, Spain
| | - Francisco Javier Cao-García
- Departamento de Estructura de la Materia, Física Térmica y Electrónica, Universidad Complutense de Madrid, Plaza de Ciencias, 1, 28040 Madrid, Spain.,Instituto Madrileño de Estudios Avanzados en Nanociencia, IMDEA Nanociencia, Calle Faraday, 9, 28049 Madrid, Spain
| |
Collapse
|
60
|
Olaleye O, Spanov B, Bults P, van der Voort A, Govorukhina N, Sonke GS, Horvatovich P, van de Merbel NC, Bischoff R. Biotransformation of Trastuzumab and Pertuzumab in Breast Cancer Patients Assessed by Affinity Enrichment and Ion-Exchange Chromatography. Drug Metab Dispos 2023; 51:249-256. [PMID: 36379709 DOI: 10.1124/dmd.122.001094] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/19/2022] [Accepted: 10/21/2022] [Indexed: 11/16/2022] Open
Abstract
Therapeutic proteins (TPs) are known to be heterogeneous due to modifications that occur during the production process and storage. Modifications may also occur in TPs after their administration to patients due to in vivo biotransformation. Ligand binding assays, which are widely used in the bioanalysis of TPs in body fluids, are typically unable to distinguish such modifications. Liquid chromatography coupled to mass spectrometry is being increasingly used to study modifications in TPs, but its use to study in vivo biotransformation has been limited until now. We present a novel approach that combines affinity enrichment using Affimer reagents with ion-exchange chromatography (IEX) to analyze charge variants of the TPs trastuzumab and pertuzumab in plasma of patients undergoing therapy for HER2-positive breast cancer. Affimer reagents were immobilized via engineered Cys tags to maleimide beads, and the TPs were eluted under acidic conditions followed by rapid neutralization. The enriched TPs were analyzed by cation-exchange chromatography (IEX) using pH-gradient elution, resulting in the separation of about 20 charge variants for trastuzumab and about five charge variants for pertuzumab. A comparison between in vitro stressed TPs spiked into plasma, and TPs enriched from patient plasma showed that the observed profiles were highly similar. This indicates that in vitro stress testing in plasma can mimic the situation in patient plasma, as far as the generation of charge variants is concerned. SIGNIFICANCE STATEMENT: This research attempts to elucidate the modifications that occur in therapeutic proteins (TPs) after they have been administered to patients. This is important because there is little knowledge about the fate of TPs in this regard, and certain modifications could affect their efficiency. Our results show that the modifications discovered are most likely due to a chemical process and are not patient specific.
Collapse
Affiliation(s)
- Oladapo Olaleye
- Department of Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Baubek Spanov
- Department of Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Peter Bults
- Department of Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Anna van der Voort
- Department of Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Natalia Govorukhina
- Department of Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Gabe S Sonke
- Department of Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Peter Horvatovich
- Department of Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Nico C van de Merbel
- Department of Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Rainer Bischoff
- Department of Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
61
|
Decker AM, Rahman MT, Kormos CM, Hesk D, Darcq E, Kieffer BL, Jin C. Synthesis and pharmacological validation of a novel radioligand for the orphan GPR88 receptor. Bioorg Med Chem Lett 2023; 80:129120. [PMID: 36587872 PMCID: PMC9852087 DOI: 10.1016/j.bmcl.2022.129120] [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: 10/24/2022] [Revised: 12/16/2022] [Accepted: 12/27/2022] [Indexed: 12/30/2022]
Abstract
GPR88 is an orphan G protein-coupled receptor which has been implicated in a number of striatal-associated disorders. Herein we describe the synthesis and pharmacological characterization of the first GPR88 radioligand, [3H]RTI-33, derived from a synthetic agonist RTI-13951-33. [3H]RTI-33 has a specific activity of 83.4 Ci/mmol and showed one-site, saturable binding (KD of 85 nM) in membranes prepared from stable PPLS-HA-hGPR88-CHO cells. A competition binding assay was developed to determine binding affinities of several known GPR88 agonists. This radioligand represents a powerful tool for future mechanistic and cell-based ligand-receptor interaction studies of GPR88.
Collapse
Affiliation(s)
- Ann M Decker
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, NC 27709, USA.
| | - Md Toufiqur Rahman
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, NC 27709, USA.
| | - Chad M Kormos
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, NC 27709, USA.
| | - David Hesk
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, NC 27709, USA.
| | - Emmanuel Darcq
- INSERM U1114, University of Strasbourg, Strasbourg 67085, France.
| | | | - Chunyang Jin
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
62
|
de Sousa Lacerda CM, Almeida NBF, Dos Santos VCF, Plentz F, de Andrade ASR. Foot-and-mouth disease virus: DNA aptamer selection for the 3ABC protein. Virus Res 2023; 323:199008. [PMID: 36414190 PMCID: PMC10194325 DOI: 10.1016/j.virusres.2022.199008] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/04/2022] [Accepted: 11/18/2022] [Indexed: 11/21/2022]
Abstract
Foot-and-mouth disease (FMD) is a devastating livestock disease caused by foot-and-mouth disease virus (FMDV), a member of the Picornaviridae family. The 3ABC is a non-structural protein of FMDV, produced during viral replication and absent from inactivated FMD vaccines. Nucleic acid aptamers are DNA or RNA oligonucleotides capable of binding with high specificity and affinity to a molecular target. The aim of this study was to obtain DNA aptamers specific for 3ABC protein with a view of their application in the FMD diagnosis. Aptamers are usually obtained through SELEX (Systematic Evolution of Ligands by EXponential enrichment) procedure. In this study, an aptamer (termed FMDV1) was selected by a variation of this technique called Capillary Electrophoresis SELEX (CE-SELEX). The FMDV1 aptamer showed high binding affinity to the 3ABC protein with Kd value in the nano molar range: 22.69 ± 1.79 nM. The FMDV1 aptamer binding to 3ABC was significantly higher when compared with the BSA protein, used as control, demonstrating its specificity.
Collapse
Affiliation(s)
- Camila Maria de Sousa Lacerda
- Centro de Desenvolvimento da Tecnologia Nuclear, CDTN, Belo Horizonte, MG 31270-901, Brasil; MedicOnChip, Parque Tecnológico de Belo Horizonte-BH-TEC, Belo Horizonte, MG, Brasil
| | - Nathalie Bonatti Franco Almeida
- Centro de Desenvolvimento da Tecnologia Nuclear, CDTN, Belo Horizonte, MG 31270-901, Brasil; MedicOnChip, Parque Tecnológico de Belo Horizonte-BH-TEC, Belo Horizonte, MG, Brasil
| | - Viviane Cristina Fernandes Dos Santos
- Centro de Desenvolvimento da Tecnologia Nuclear, CDTN, Belo Horizonte, MG 31270-901, Brasil; MedicOnChip, Parque Tecnológico de Belo Horizonte-BH-TEC, Belo Horizonte, MG, Brasil
| | - Flávio Plentz
- Departamento de Física, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil; MedicOnChip, Parque Tecnológico de Belo Horizonte-BH-TEC, Belo Horizonte, MG, Brasil
| | | |
Collapse
|
63
|
Holt A. Conventional Receptor Radioligand Binding Techniques Applied to the Study of Monoamine Oxidase. Methods Mol Biol 2023; 2558:75-96. [PMID: 36169857 DOI: 10.1007/978-1-0716-2643-6_7] [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: 06/16/2023]
Abstract
Designed to measure binding interactions between small molecules and receptor proteins, radioligand binding approaches may also be applied to interactions between monoamine oxidase (MAO) and its ligands. The technique may be used with tissue homogenates or with mitochondrial membranes and can provide information about binding site density, ligand affinity, binding rate constants, and binding events at sites that do not impact absorbance characteristics of the flavin cofactor and that may not be amenable to spectrophotometric studies. This overview describes the use of a cell harvester in a common filtration approach to measure binding to MAO of radiolabeled substrates, inhibitors, or allosteric ligands in saturation analyses and to take advantage of the principles of competition to obtain quantitative binding data for unlabeled ligands that may bind with much lower affinity. The quality and reproducibility of data are impacted by factors such as choice of ligand concentrations, pipetting technique, graphing and regression approaches, and scintillation counting parameters, and consideration is given to these and other factors that may influence the results.
Collapse
Affiliation(s)
- Andrew Holt
- Neurochemical Research Unit, Department of Psychiatry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
64
|
Papay RS, Macdonald JD, Stauffer SR, Perez DM. Characterization of a novel positive allosteric modulator of the α 1A-Adrenergic receptor. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2022; 4:100142. [PMID: 36544813 PMCID: PMC9762201 DOI: 10.1016/j.crphar.2022.100142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/27/2022] [Accepted: 11/15/2022] [Indexed: 12/05/2022] Open
Abstract
α1-Adrenergic Receptors (ARs) are G-protein Coupled Receptors (GPCRs) that regulate the sympathetic nervous system via the binding and activation of norepinephrine (NE) and epinephrine (Epi). α1-ARs control various aspects of neurotransmission, cognition, cardiovascular functions as well as other organ systems. However, therapeutic drug development for these receptors, particularly agonists, has been stagnant due to unwanted effects on blood pressure regulation. We report the synthesis and characterization of the first positive allosteric modulator (PAM) for the α1-AR based upon the derivation of the α1A-AR selective imidazoline agonist, cirazoline. Compound 3 (Cmpd-3) binds the α1A-AR with high and low affinity sites (0.13pM; 54 nM) typical of GPCR agonists, and reverts to a single low affinity site of 100 nM upon the addition of GTP. Comparison of Cmpd-3 versus other orthosteric α1A-AR-selective imidazoline ligands reveal unique properties that are consistent with a type I PAM. Cmpd-3 is both conformationally and ligand-selective for the α1A-AR subtype. In competition binding studies, Cmpd-3 potentiates NE-binding at the α1A-AR only on the high affinity state of NE with no effect on the Epi-bound α1A-AR. Moreover, Cmpd-3 demonstrates signaling-bias and potentiates the NE-mediated cAMP response of the α1A-AR at nM concentrations with no effects on the NE-mediated inositol phosphate response. There are no effects of Cmpd-3 on the signaling at the α1B- or α1D-AR subtypes. Cmpd-3 displays characteristics of a pure PAM with no intrinsic agonist properties. Specific derivation of Cmpd-3 at the R1 ortho-position recapitulated PAM characteristics. Our results characterize the first PAM for the α1-AR and holds promise for a first-in-class therapeutic to treat various diseases without the side effect of increasing blood pressure intrinsic to classical orthosteric agonists.
Collapse
Affiliation(s)
- Robert S. Papay
- The Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, The Cleveland Clinic Foundation, 9500 Euclid Ave, Cleveland, OH, 44195, USA
| | - Jonathan D. Macdonald
- Center for Therapeutics Discovery, Lerner Research Institute, The Cleveland Clinic Foundation, 9500 Euclid Ave, Cleveland, OH, 44195, USA
| | - Shaun R. Stauffer
- Center for Therapeutics Discovery, Lerner Research Institute, The Cleveland Clinic Foundation, 9500 Euclid Ave, Cleveland, OH, 44195, USA
| | - Dianne M. Perez
- The Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, The Cleveland Clinic Foundation, 9500 Euclid Ave, Cleveland, OH, 44195, USA
| |
Collapse
|
65
|
De Silva NIU, Fargason T, Zhang Z, Wang T, Zhang J. Inter-domain Flexibility of Human Ser/Arg-Rich Splicing Factor 1 Allows Variable Spacer Length in Cognate RNA’s Bipartite Motifs. Biochemistry 2022; 61:2922-2932. [DOI: 10.1021/acs.biochem.2c00565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Affiliation(s)
- Naiduwadura Ivon Upekala De Silva
- Department of Chemistry, College of Arts and Sciences, University of Alabama at Birmingham, CH266, 901 14th Street South, Birmingham, Alabama35294-1240, United States
| | - Talia Fargason
- Department of Chemistry, College of Arts and Sciences, University of Alabama at Birmingham, CH266, 901 14th Street South, Birmingham, Alabama35294-1240, United States
| | - Zihan Zhang
- Department of Chemistry, College of Arts and Sciences, University of Alabama at Birmingham, CH266, 901 14th Street South, Birmingham, Alabama35294-1240, United States
| | - Ting Wang
- Department of Chemistry, College of Arts and Sciences, University of Alabama at Birmingham, CH266, 901 14th Street South, Birmingham, Alabama35294-1240, United States
| | - Jun Zhang
- Department of Chemistry, College of Arts and Sciences, University of Alabama at Birmingham, CH266, 901 14th Street South, Birmingham, Alabama35294-1240, United States
| |
Collapse
|
66
|
Pavan M, Menin S, Bassani D, Sturlese M, Moro S. Qualitative Estimation of Protein-Ligand Complex Stability through Thermal Titration Molecular Dynamics Simulations. J Chem Inf Model 2022; 62:5715-5728. [PMID: 36315402 PMCID: PMC9709921 DOI: 10.1021/acs.jcim.2c00995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The prediction of ligand efficacy has long been linked to thermodynamic properties such as the equilibrium dissociation constant, which considers both the association and the dissociation rates of a defined protein-ligand complex. In the last 15 years, there has been a paradigm shift, with an increased interest in the determination of kinetic properties such as the drug-target residence time since they better correlate with ligand efficacy compared to other parameters. In this article, we present thermal titration molecular dynamics (TTMD), an alternative computational method that combines a series of molecular dynamics simulations performed at progressively increasing temperatures with a scoring function based on protein-ligand interaction fingerprints for the qualitative estimation of protein-ligand-binding stability. The protocol has been applied to four different pharmaceutically relevant test cases, including protein kinase CK1δ, protein kinase CK2, pyruvate dehydrogenase kinase 2, and SARS-CoV-2 main protease, on a variety of ligands with different sizes, structures, and experimentally determined affinity values. In all four cases, TTMD was successfully able to distinguish between high-affinity compounds (low nanomolar range) and low-affinity ones (micromolar), proving to be a useful screening tool for the prioritization of compounds in a drug discovery campaign.
Collapse
|
67
|
Yang H, Micovic N, Monaghan JR, Clark HA. Click Chemistry-Enabled Conjugation Strategy for Producing Dibenzodiazepinone-Type Fluorescent Probes To Target M 2 Acetylcholine Receptors. Bioconjug Chem 2022; 33:2223-2233. [PMID: 36327428 DOI: 10.1021/acs.bioconjchem.2c00446] [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] [Indexed: 11/06/2022]
Abstract
The development of fluorescently labeled receptor-targeting compounds represents a powerful pharmacological tool to study and characterize ligand-receptor interactions. Despite significant advances in developing sub-type-specific antagonists for muscarinic acetylcholine receptors (mAChRs), reports on antagonists feasible for click chemistry are less common. Here, we designed and synthesized an antagonist suitable for probe attachment through click chemistry, namely, dibenzodiazepinone (DIBA)-alkyne, based on a previously reported DIBA scaffold with a high binding affinity to type-2 mAChR (M2R). To demonstrate the versatility of DIBA-alkyne as a building block for bioconjugates, we assembled DIBA-alkyne with Cyanine5 fluorophores (Cy5) and polyethylene glycol (PEG) biomolecules to obtain fluorescent DIBA antagonist (DIBA-Cy5) and fluorescent DIBA PEG derivatives. Flow cytometric analysis showed that DIBA-Cy5 possessed a high binding affinity to M2R (Kd = 1.80 nM), a two-order magnitude higher binding affinity than M1R. Fluorescent DIBA PEG derivatives maintained a potent binding to the M2R (Kd ≤ 4 nM), confirmed by confocal microscopic imaging. Additionally, DIBA-Cy5 can serve as a fluorescent ligand in the receptor-ligand competitive binding assay for other mAChR ligands, an attractive alternative to the traditional radioligand-based assay. The competitive binding mode between DIBA-Cy5 and orthosteric antagonist atropine/allosteric modulator LY2119620 indicated a dualsteric binding mode of the DIBA-type antagonist to M2R. Lastly, we demonstrated the direct staining of DIBA-Cy5 to M2R receptors in the sinoatrial node of a mouse heart. The adaptability of the clickable DIBA antagonist to a wide range of fluorophores and biomolecules can facilitate its use in various biomedical applications such as binding assays that screen compounds for M2R as the receptor target.
Collapse
Affiliation(s)
- Hongrong Yang
- Department of Bioengineering, Northeastern University, Boston, Massachusetts02115, United States
| | - Nicholas Micovic
- Department of Bioengineering, Northeastern University, Boston, Massachusetts02115, United States
| | - James R Monaghan
- Department of Biology, Northeastern University, Boston, Massachusetts02115, United States
- Institute for Chemical Imaging of Living Systems, Northeastern University, Boston, Massachusetts02115, United States
| | - Heather A Clark
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona85281, United States
| |
Collapse
|
68
|
Stockley ML, Ferdinand A, Benedetti G, Blencowe P, Boyd SM, Calder M, Charles MD, Edwardes LV, Ekwuru T, Finch H, Galbiati A, Geo L, Grande D, Grinkevich V, Holliday ND, Krajewski WW, MacDonald E, Majithiya JB, McCarron H, McWhirter CL, Patel V, Pedder C, Rajendra E, Ranzani M, Rigoreau LJM, Robinson HMR, Schaedler T, Sirina J, Smith GCM, Swarbrick ME, Turnbull AP, Willis S, Heald RA. Discovery, Characterization, and Structure-Based Optimization of Small-Molecule In Vitro and In Vivo Probes for Human DNA Polymerase Theta. J Med Chem 2022; 65:13879-13891. [PMID: 36200480 DOI: 10.1021/acs.jmedchem.2c01142] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Human DNA polymerase theta (Polθ), which is essential for microhomology-mediated DNA double strand break repair, has been proposed as an attractive target for the treatment of BRCA deficient and other DNA repair pathway defective cancers. As previously reported, we recently identified the first selective small molecule Polθ in vitro probe, 22 (ART558), which recapitulates the phenotype of Polθ loss, and in vivo probe, 43 (ART812), which is efficacious in a model of PARP inhibitor resistant TNBC in vivo. Here we describe the discovery, biochemical and biophysical characterization of these probes including small molecule ligand co-crystal structures with Polθ. The crystallographic data provides a basis for understanding the unique mechanism of inhibition of these compounds which is dependent on stabilization of a "closed" enzyme conformation. Additionally, the structural biology platform provided a basis for rational optimization based primarily on reduced ligand conformational flexibility.
Collapse
Affiliation(s)
- Martin L Stockley
- Artios Pharma Ltd., B940, Babraham Research Campus, CambridgeCB22 3FH, U. K
| | - Amanda Ferdinand
- Cancer Research Horizons Therapeutic Innovation, Jonas Webb Building, Babraham Research Campus, CambridgeCB22 3AT, U. K
| | - Giovanni Benedetti
- Artios Pharma Ltd., B940, Babraham Research Campus, CambridgeCB22 3FH, U. K
| | - Peter Blencowe
- Cancer Research Horizons Therapeutic Innovation, Jonas Webb Building, Babraham Research Campus, CambridgeCB22 3AT, U. K
| | - Susan M Boyd
- CompChem Solutions Ltd, St John's Innovation Centre, Cowley Rd, CambridgeCB4 0WS, U. K
| | - Mat Calder
- Cancer Research Horizons Therapeutic Innovation, Jonas Webb Building, Babraham Research Campus, CambridgeCB22 3AT, U. K
| | - Mark D Charles
- Cancer Research Horizons Therapeutic Innovation, Jonas Webb Building, Babraham Research Campus, CambridgeCB22 3AT, U. K
| | - Lucy V Edwardes
- Artios Pharma Ltd., B940, Babraham Research Campus, CambridgeCB22 3FH, U. K
| | - Tennyson Ekwuru
- Cancer Research Horizons Therapeutic Innovation, Jonas Webb Building, Babraham Research Campus, CambridgeCB22 3AT, U. K
| | - Harry Finch
- Artios Pharma Ltd., B940, Babraham Research Campus, CambridgeCB22 3FH, U. K
| | | | - Lerin Geo
- Artios Pharma Ltd., B940, Babraham Research Campus, CambridgeCB22 3FH, U. K
| | - Diego Grande
- Artios Pharma Ltd., B940, Babraham Research Campus, CambridgeCB22 3FH, U. K
| | - Vera Grinkevich
- Artios Pharma Ltd., B940, Babraham Research Campus, CambridgeCB22 3FH, U. K
| | - Nicholas D Holliday
- Excellerate Bioscience Ltd., BioCity, Pennyfoot Street, NottinghamNG1 1GF, U. K
| | - Wojciech W Krajewski
- Cancer Research Horizons Therapeutic Innovation, The Francis Crick Institute, 1 Midland Road, LondonNW1 1AT, U. K
| | - Ellen MacDonald
- Cancer Research Horizons Therapeutic Innovation, Jonas Webb Building, Babraham Research Campus, CambridgeCB22 3AT, U. K
| | - Jayesh B Majithiya
- Artios Pharma Ltd., B940, Babraham Research Campus, CambridgeCB22 3FH, U. K
| | - Hollie McCarron
- Cancer Research Horizons Therapeutic Innovation, Jonas Webb Building, Babraham Research Campus, CambridgeCB22 3AT, U. K
| | - Claire L McWhirter
- Artios Pharma Ltd., B940, Babraham Research Campus, CambridgeCB22 3FH, U. K
| | - Viral Patel
- Excellerate Bioscience Ltd., BioCity, Pennyfoot Street, NottinghamNG1 1GF, U. K
| | - Chris Pedder
- Cancer Research Horizons Therapeutic Innovation, Jonas Webb Building, Babraham Research Campus, CambridgeCB22 3AT, U. K
| | - Eeson Rajendra
- Artios Pharma Ltd., B940, Babraham Research Campus, CambridgeCB22 3FH, U. K
| | - Marco Ranzani
- Artios Pharma Ltd., B940, Babraham Research Campus, CambridgeCB22 3FH, U. K
| | - Laurent J M Rigoreau
- Cancer Research Horizons Therapeutic Innovation, Jonas Webb Building, Babraham Research Campus, CambridgeCB22 3AT, U. K
| | - Helen M R Robinson
- Artios Pharma Ltd., B940, Babraham Research Campus, CambridgeCB22 3FH, U. K
| | - Theresia Schaedler
- Artios Pharma Ltd., B940, Babraham Research Campus, CambridgeCB22 3FH, U. K
| | - Julija Sirina
- Excellerate Bioscience Ltd., BioCity, Pennyfoot Street, NottinghamNG1 1GF, U. K
| | - Graeme C M Smith
- Artios Pharma Ltd., B940, Babraham Research Campus, CambridgeCB22 3FH, U. K
| | - Martin E Swarbrick
- Cancer Research Horizons Therapeutic Innovation, Jonas Webb Building, Babraham Research Campus, CambridgeCB22 3AT, U. K
| | - Andrew P Turnbull
- Cancer Research Horizons Therapeutic Innovation, The Francis Crick Institute, 1 Midland Road, LondonNW1 1AT, U. K
| | - Simon Willis
- Cancer Research Horizons Therapeutic Innovation, The Francis Crick Institute, 1 Midland Road, LondonNW1 1AT, U. K
| | - Robert A Heald
- Artios Pharma Ltd., B940, Babraham Research Campus, CambridgeCB22 3FH, U. K
| |
Collapse
|
69
|
Reissig F, Zarschler K, Novy Z, Petrik M, Bendova K, Kurfurstova D, Bouchal J, Ludik MC, Brandt F, Kopka K, Khoylou M, Pietzsch HJ, Hajduch M, Mamat C. Modulating the pharmacokinetic profile of Actinium-225-labeled macropa-derived radioconjugates by dual targeting of PSMA and albumin. Theranostics 2022; 12:7203-7215. [PMID: 36438496 PMCID: PMC9691366 DOI: 10.7150/thno.78043] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 09/21/2022] [Indexed: 11/29/2022] Open
Abstract
Rationale: Small 225Ac-labeled prostate-specific membrane antigen (PSMA)-targeted radioconjugates have been described for targeted alpha therapy of metastatic castration-resistant prostate cancer. Transient binding to serum albumin as a highly abundant, inherent transport protein represents a commonly applied strategy to modulate the tissue distribution profile of such low-molecular-weight radiotherapeutics and to enhance radioactivity uptake into tumor lesions with the ultimate objective of improved therapeutic outcome. Methods: Two ligands mcp-M-alb-PSMA and mcp-D-alb-PSMA were synthesized by combining a macropa-derived chelator with either one or two lysine-ureido-glutamate-based PSMA- and 4-(p-iodophenyl)butyrate albumin-binding entities using multistep peptide-coupling chemistry. Both compounds were labeled with [225Ac]Ac3+ under mild conditions and their reversible binding to serum albumin was analyzed by an ultrafiltration assay as well as microscale thermophoresis measurements. Saturation binding studies and clonogenic survival assays using PSMA-expressing LNCaP cells were performed to evaluate PSMA-mediated cell binding and to assess the cytotoxic potency of the novel radioconjugates [225Ac]Ac-mcp-M-alb-PSMA and [225Ac]Ac-mcp-D-alb-PSMA, respectively. Biodistributions of both 225Ac-radioconjugates were investigated using LNCaP tumor-bearing SCID mice. Histological examinations of selected organs were performed to analyze the occurrence of necrosis using H&E staining, DNA damage via γH2AX staining and proliferation via Ki67 expression in the tissue samples. Results: Enhanced binding to serum components in general and to human serum albumin in particular was revealed for [225Ac]Ac-mcp-M-alb-PSMA and [225Ac]Ac-mcp-D-alb-PSMA, respectively. Moreover, the novel derivatives are highly potent PSMA ligands as their KD values in the nanomolar range (23.38 and 11.56 nM) are comparable to the reference radioconjugates [225Ac]Ac-mcp-M-PSMA (30.83 nM) and [225Ac]Ac-mcp-D-PSMA (10.20 nM) without albumin binders. The clonogenic activity of LNCaP cells after treatment with the 225Ac-labeled ligands was affected in a dose- and time-dependent manner, whereas the bivalent radioconjugate [225Ac]Ac-mcp-D-alb-PSMA has a stronger impact on the clonogenic cell survival than its monovalent counterpart [225Ac]Ac-mcp-M-alb-PSMA. Biodistribution studies performed in LNCaP tumor xenografts showed prolonged blood circulation times for both albumin-binding radioconjugates and a substantially increased tumor uptake (46.04 ± 7.77 %ID/g for [225Ac]Ac-mcp-M-alb-PSMA at 128 h p.i. and 153.48 ± 37.76 %ID/g at 168 h p.i. for [225Ac]Ac-mcp-D-alb-PSMA) with favorable tumor-to-background ratios. Consequently, a clear histological indication of DNA damage was discovered in the tumor tissues, whereas DNA double-strand break formation in kidney and liver sections was less pronounced. Conclusion: The modification of the PSMA-based 225Ac-radioconjugates with one or two albumin-binding entities resulted in an improved radiopharmacological behavior including a greatly enhanced tumor accumulation combined with a rather low uptake in most non-targeted organs combined with a high excretion via the kidneys.
Collapse
Affiliation(s)
- Falco Reissig
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, D‑01328 Dresden, Germany
- Technische Universität Dresden, Faculty of Chemistry and Food Chemistry, D-01062 Dresden, Germany
| | - Kristof Zarschler
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, D‑01328 Dresden, Germany
| | - Zbynek Novy
- Palacky University Olomouc, Faculty of Medicine and Dentistry, Institute of Molecular and Translational Medicine and Czech Advanced Technology and Research Institute, Hnevotinska 1333/5, 779 00 Olomouc, Czech Republic
| | - Milos Petrik
- Palacky University Olomouc, Faculty of Medicine and Dentistry, Institute of Molecular and Translational Medicine and Czech Advanced Technology and Research Institute, Hnevotinska 1333/5, 779 00 Olomouc, Czech Republic
| | - Katerina Bendova
- Palacky University Olomouc, Faculty of Medicine and Dentistry, Institute of Molecular and Translational Medicine and Czech Advanced Technology and Research Institute, Hnevotinska 1333/5, 779 00 Olomouc, Czech Republic
| | - Daniela Kurfurstova
- Palacky University Olomouc, Faculty of Medicine and Dentistry, Institute of Clinical and Molecular Pathology, Hnevotinska 976/3, 775 15 Olomouc, Czech Republic
| | - Jan Bouchal
- Palacky University Olomouc, Faculty of Medicine and Dentistry, Institute of Clinical and Molecular Pathology, Hnevotinska 976/3, 775 15 Olomouc, Czech Republic
| | - Marie-Charlotte Ludik
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, D‑01328 Dresden, Germany
| | - Florian Brandt
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, D‑01328 Dresden, Germany
- Technische Universität Dresden, Faculty of Chemistry and Food Chemistry, D-01062 Dresden, Germany
| | - Klaus Kopka
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, D‑01328 Dresden, Germany
- Technische Universität Dresden, Faculty of Chemistry and Food Chemistry, D-01062 Dresden, Germany
| | - Marta Khoylou
- Palacky University Olomouc, Faculty of Medicine and Dentistry, Institute of Molecular and Translational Medicine and Czech Advanced Technology and Research Institute, Hnevotinska 1333/5, 779 00 Olomouc, Czech Republic
| | - Hans-Jürgen Pietzsch
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, D‑01328 Dresden, Germany
| | - Marian Hajduch
- Palacky University Olomouc, Faculty of Medicine and Dentistry, Institute of Molecular and Translational Medicine and Czech Advanced Technology and Research Institute, Hnevotinska 1333/5, 779 00 Olomouc, Czech Republic
| | - Constantin Mamat
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, D‑01328 Dresden, Germany
- Technische Universität Dresden, Faculty of Chemistry and Food Chemistry, D-01062 Dresden, Germany
| |
Collapse
|
70
|
Vaidyanathan S, Reed A. Pipeline Impact of Radiolabeled Compounds in Drug Discovery and Development. ACS Med Chem Lett 2022; 13:1564-1567. [PMID: 36262403 PMCID: PMC9575178 DOI: 10.1021/acsmedchemlett.2c00281] [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: 06/17/2022] [Accepted: 09/07/2022] [Indexed: 11/30/2022] Open
Abstract
The long-lived radionuclides tritium and carbon-14 have been used for many years in pharmaceutical research and development for making key efficacy and toxicological decisions. Early discovery utilizes radiolabels for compound selection through radioligand binding assays and autoradiography. In preclinical safety evaluation, the use of labeled compounds for adsorption, distribution, metabolism, and excretion studies is often preferred for the added detection sensitivity. As the drug substance proceeds to the clinic, human metabolism studies are reliant on the use of labeled materials to fulfill required regulatory applications.
Collapse
Affiliation(s)
- Srirajan Vaidyanathan
- Department of Process Chemistry, Radiochemistry, AbbVie Inc., 1 North Waukegan Road, North
Chicago, Illinois 60064, United States
| | - Aimee Reed
- Department of Process Chemistry, Radiochemistry, AbbVie Inc., 1 North Waukegan Road, North
Chicago, Illinois 60064, United States
| |
Collapse
|
71
|
Shevelev AY, Kashirina NM, Lipatova LN, Yanushevskaya EV, Peklo MM, Rybalkin IN, Rutkevich PN, Chusovitina OK, Skoblova NA, Skoblov YS, Vlasik TN, Zykov KA. Radioligand Binding Assay for the Simultaneous Determination of β1- and β2-Adrenergic Receptors in Human Blood Cells. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2022. [DOI: 10.1134/s1068162022050211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
72
|
Fink EA, Xu J, Hübner H, Braz JM, Seemann P, Avet C, Craik V, Weikert D, Schmidt MF, Webb CM, Tolmachova NA, Moroz YS, Huang XP, Kalyanaraman C, Gahbauer S, Chen G, Liu Z, Jacobson MP, Irwin JJ, Bouvier M, Du Y, Shoichet BK, Basbaum AI, Gmeiner P. Structure-based discovery of nonopioid analgesics acting through the α 2A-adrenergic receptor. Science 2022; 377:eabn7065. [PMID: 36173843 PMCID: PMC10360211 DOI: 10.1126/science.abn7065] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Because nonopioid analgesics are much sought after, we computationally docked more than 301 million virtual molecules against a validated pain target, the α2A-adrenergic receptor (α2AAR), seeking new α2AAR agonists chemotypes that lack the sedation conferred by known α2AAR drugs, such as dexmedetomidine. We identified 17 ligands with potencies as low as 12 nanomolar, many with partial agonism and preferential Gi and Go signaling. Experimental structures of α2AAR complexed with two of these agonists confirmed the docking predictions and templated further optimization. Several compounds, including the initial docking hit '9087 [mean effective concentration (EC50) of 52 nanomolar] and two analogs, '7075 and PS75 (EC50 4.1 and 4.8 nanomolar), exerted on-target analgesic activity in multiple in vivo pain models without sedation. These newly discovered agonists are interesting as therapeutic leads that lack the liabilities of opioids and the sedation of dexmedetomidine.
Collapse
Affiliation(s)
- Elissa A. Fink
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
- Graduate Program in Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Jun Xu
- Kobilka Institute of Innovative Drug Discovery, School of Life and Health Sciences, Chinese University of Hong Kong, Shenzhen, Guangdong 518172, China
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Harald Hübner
- Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Joao M. Braz
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA
| | - Philipp Seemann
- Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Charlotte Avet
- Department of Biochemistry and Molecular Medicine, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Veronica Craik
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA
| | - Dorothee Weikert
- Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Maximilian F. Schmidt
- Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Chase M. Webb
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
- Graduate Program in Pharmaceutical Sciences and Pharmacogenomics, University of California, San Francisco, San Francisco, CA, USA
| | - Nataliya A. Tolmachova
- Enamine Ltd., 02094 Kyiv, Ukraine
- Institute of Bioorganic Chemistry and Petrochemistry, National Ukrainian Academy of Science, 02660 Kyiv, Ukraine
| | - Yurii S. Moroz
- National Taras Shevchenko University of Kyiv, 01601 Kyiv, Ukraine
- Chemspace, Riga LV-1082, Latvia
| | - Xi-Ping Huang
- National Institute of Mental Health Psychoactive Drug Screening Program (NIMH PDSP), School of Medicine, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Chakrapani Kalyanaraman
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Stefan Gahbauer
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Geng Chen
- Kobilka Institute of Innovative Drug Discovery, School of Life and Health Sciences, Chinese University of Hong Kong, Shenzhen, Guangdong 518172, China
| | - Zheng Liu
- Kobilka Institute of Innovative Drug Discovery, School of Life and Health Sciences, Chinese University of Hong Kong, Shenzhen, Guangdong 518172, China
| | - Matthew P. Jacobson
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - John J. Irwin
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Michel Bouvier
- Department of Biochemistry and Molecular Medicine, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Yang Du
- Kobilka Institute of Innovative Drug Discovery, School of Life and Health Sciences, Chinese University of Hong Kong, Shenzhen, Guangdong 518172, China
| | - Brian K. Shoichet
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Allan I. Basbaum
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA
| | - Peter Gmeiner
- Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany
| |
Collapse
|
73
|
Saumell-Esnaola M, Elejaga-Jimeno A, Echeazarra L, Borrega-Román L, Barrondo S, López de Jesús M, González-Burguera I, Gómez-Caballero A, Goicolea MA, Sallés J, García del Caño G. Design and validation of recombinant protein standards for quantitative Western blot analysis of cannabinoid CB1 receptor density in cell membranes: an alternative to radioligand binding methods. Microb Cell Fact 2022; 21:192. [PMID: 36109736 PMCID: PMC9479267 DOI: 10.1186/s12934-022-01914-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 08/27/2022] [Indexed: 11/16/2022] Open
Abstract
Background Replacement of radioligand binding assays with antibody-antigen interaction-based approaches for quantitative analysis of G protein-coupled receptor (GPCR) levels requires the use of purified protein standards containing the antigen. GPCRs in general and cannabinoid CB1 receptor in particular show a progressive tendency to aggregate and precipitate in aqueous solution outside of their biological context due to the low solubility that the hydrophobic nature imprinted by their seven transmembrane domains. This renders full-length recombinant GPCRs useless for analytical purposes, a problem that can be overcome by engineering soluble recombinant fragments of the receptor containing the antigen. Results Here we generated highly soluble and stable recombinant protein constructs GST-CB1414–472 and GST-CB1414-442 containing much of the human CB1 receptor C-terminal tail for use as standard and negative control, respectively, in quantitative Western blot analysis of CB1 receptor expression on crude synaptosomes of the adult rat brain cortex. To this end we used three different antibodies, all raised against a peptide comprising the C-terminal residues 443–473 of the mouse CB1 receptor that corresponds to residues 442–472 in the human homolog. Estimated values of CB1 receptor density obtained by quantitative Western blot were of the same order of magnitude but slightly higher than values obtained by the radioligand saturation binding assay. Conclusions Collectively, here we provide a suitable Western blot-based design as a simple, cost-effective and radioactivity-free alternative for the quantitative analysis of CB1 receptor expression, and potentially of any GPCR, in a variety of biological samples. The discrepancies between the results obtained by quantitative Western blot and radioligand saturation binding techniques are discussed in the context of their particular theoretical bases and methodological constraints. Supplementary Information The online version contains supplementary material available at 10.1186/s12934-022-01914-1.
Collapse
|
74
|
Almeida NBF, Sousa TASL, Santos VCF, Lacerda CMS, Silva TG, Grenfell RFQ, Plentz F, Andrade ASR. DNA aptamer selection and construction of an aptasensor based on graphene FETs for Zika virus NS1 protein detection. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2022; 13:873-881. [PMID: 36105684 PMCID: PMC9443353 DOI: 10.3762/bjnano.13.78] [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: 05/05/2022] [Accepted: 08/25/2022] [Indexed: 06/15/2023]
Abstract
Zika virus (ZIKV) is a mosquito-borne virus that is phylogenetically close to other medically important flaviviruses with high global public health significance, such as dengue (DENV) and yellow fever (YFV) viruses. Correct diagnosis of a flavivirus infection can be challenging, particularly in world regions where more than one flavivirus co-circulates and YFV vaccination is mandatory. Acid nucleic aptamers are oligonucleotides that bind to a specific target molecule with high affinity and specificity. Because of their unique characteristics, aptamers are promising tools for biosensor development. Aptamers are usually obtained through a procedure called "systematic evolution of ligands by exponential enrichment" (SELEX). In this study, we select an aptamer (termed ZIKV60) by capillary electrophoresis SELEX (CE-SELEX) to the Zika virus non-structural protein 1 (NS1) and counterselection against the NS1 proteins of DENV (serotypes 1, 2, 3, and 4) and YFV. The ZIKV60 dissociation constant (K d) is determined by enzyme-linked oligonucleotide assay (ELONA) and the aptamer specificity is evaluated by quantitative real-time polymerase chain reaction. ZIKV60 shows a high binding affinity to the ZIKV NS1 protein with a K d value of 2.28 ± 0.28 nM. The aptamer presents high specificity for ZIKV NS1 compared to NS1 of DENV and YFV. Furthermore, graphene field-effect transistor devices functionalized with ZIKV60 exhibit an evident identification of NS1 protein diluted in human serum. These results point to the applicability of biosensors based on the ZIKV60 aptamer for the differential diagnosis of the Zika virus.
Collapse
Affiliation(s)
- Nathalie B F Almeida
- Centro de Desenvolvimento da Tecnologia Nuclear (CDTN), Avenida Presidente Antônio Carlos 6627, Belo Horizonte, CEP 31270-901, Brazil
- Departamento de Física, ICEx, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos 6627, Belo Horizonte, CEP 31270-901, Brazil
- MedicOnChip, Parque Tecnológico de Belo Horizonte-BH-TEC, Rua Professor José Vieira de Mendonça 770, Belo Horizonte, CEP 31310-260, Brazil
| | - Thiago A S L Sousa
- Departamento de Física, ICEx, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos 6627, Belo Horizonte, CEP 31270-901, Brazil
- MedicOnChip, Parque Tecnológico de Belo Horizonte-BH-TEC, Rua Professor José Vieira de Mendonça 770, Belo Horizonte, CEP 31310-260, Brazil
- Current address: DTU Physics, Technical University of Denmark, Kongens Lyngby, 2800, Denmark
| | - Viviane C F Santos
- Centro de Desenvolvimento da Tecnologia Nuclear (CDTN), Avenida Presidente Antônio Carlos 6627, Belo Horizonte, CEP 31270-901, Brazil
- Departamento de Física, ICEx, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos 6627, Belo Horizonte, CEP 31270-901, Brazil
- MedicOnChip, Parque Tecnológico de Belo Horizonte-BH-TEC, Rua Professor José Vieira de Mendonça 770, Belo Horizonte, CEP 31310-260, Brazil
| | - Camila M S Lacerda
- Centro de Desenvolvimento da Tecnologia Nuclear (CDTN), Avenida Presidente Antônio Carlos 6627, Belo Horizonte, CEP 31270-901, Brazil
- Departamento de Física, ICEx, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos 6627, Belo Horizonte, CEP 31270-901, Brazil
- MedicOnChip, Parque Tecnológico de Belo Horizonte-BH-TEC, Rua Professor José Vieira de Mendonça 770, Belo Horizonte, CEP 31310-260, Brazil
| | - Thais G Silva
- Departamento de Física, ICEx, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos 6627, Belo Horizonte, CEP 31270-901, Brazil
- MedicOnChip, Parque Tecnológico de Belo Horizonte-BH-TEC, Rua Professor José Vieira de Mendonça 770, Belo Horizonte, CEP 31310-260, Brazil
| | - Rafaella F Q Grenfell
- Instituto René Rachou - Fundação Oswaldo Cruz, Avenida Augusto de Lima 1715, Belo Horizonte, Minas Gerais, 30190-002, Brazil
| | - Flavio Plentz
- Departamento de Física, ICEx, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos 6627, Belo Horizonte, CEP 31270-901, Brazil
- MedicOnChip, Parque Tecnológico de Belo Horizonte-BH-TEC, Rua Professor José Vieira de Mendonça 770, Belo Horizonte, CEP 31310-260, Brazil
| | - Antero S R Andrade
- Centro de Desenvolvimento da Tecnologia Nuclear (CDTN), Avenida Presidente Antônio Carlos 6627, Belo Horizonte, CEP 31270-901, Brazil
| |
Collapse
|
75
|
Redox potential determination of the Hell’s gate globin I protein facing multiple exogenous ligands. Microchem J 2022. [DOI: 10.1016/j.microc.2022.108027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
76
|
Mlčochová H, Ratih R, Michalcová L, Wätzig H, Glatz Z, Stein M. Comparison of mobility shift affinity capillary electrophoresis and capillary electrophoresis frontal analysis for binding constant determination between human serum albumin and small drugs. Electrophoresis 2022; 43:1724-1734. [DOI: 10.1002/elps.202100320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 05/30/2022] [Accepted: 06/06/2022] [Indexed: 11/08/2022]
Affiliation(s)
- Hana Mlčochová
- Institute of Medicinal and Pharmaceutical Chemistry TU Braunschweig Braunschweig Lower Saxony Germany
- Department of Biochemistry Faculty of Science Masaryk University Brno Czech Republic
| | - Ratih Ratih
- Institute of Medicinal and Pharmaceutical Chemistry TU Braunschweig Braunschweig Lower Saxony Germany
- Department of Pharmaceutical Chemistry Faculty of Pharmacy University of Surabaya Surabaya East Java Indonesia
| | - Lenka Michalcová
- Institute of Medicinal and Pharmaceutical Chemistry TU Braunschweig Braunschweig Lower Saxony Germany
- Department of Biochemistry Faculty of Science Masaryk University Brno Czech Republic
| | - Hermann Wätzig
- Institute of Medicinal and Pharmaceutical Chemistry TU Braunschweig Braunschweig Lower Saxony Germany
| | - Zdeněk Glatz
- Department of Biochemistry Faculty of Science Masaryk University Brno Czech Republic
| | - Matthias Stein
- Institute of Medicinal and Pharmaceutical Chemistry TU Braunschweig Braunschweig Lower Saxony Germany
| |
Collapse
|
77
|
Youness F, Jaafar A, Tehrani A, Bilbeisi RA. Functionalised electrospun membranes (TETA-PVC) for the removal of lead(ii) from water. RSC Adv 2022; 12:24607-24613. [PMID: 36128362 PMCID: PMC9426649 DOI: 10.1039/d2ra02946d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 08/16/2022] [Indexed: 11/27/2022] Open
Abstract
Driven by the need for delivering sustainable water purification solutions for the removal of heavy metals from water, electrospun PVC membranes were functionalised with triethylenetetramine (TETA) and were used to remove lead(ii) ions selectively from water. The membranes were characterised and their adsorption behavior towards the removal of lead from water was investigated. The incorporation of TETA on the membrane's surface significantly improved the removal efficiency of lead(ii) up to 99.8% in 30 minutes and under ambient conditions, with the lowest concentration of 50 ppm. The adsorption mechanism was investigated and kinetic data showed a better correlation with the pseudo-second-order model. Similarly, the equilibrium data best fitted with the Langmuir adsorption isotherm model with a relatively high maximum adsorption capacity of 1250 mg g-1 for lead(ii) ions, larger than recently reported adsorption capacities for similar membranes. The functionalised membrane also showed high selectivity to lead(ii) in a mixed solution containing lead(ii), mercury(ii), cadmium(ii), arsenic(iii), copper(ii), and zinc(ii). The functionalised membrane was regenerated, where desorption of lead(ii) was achieved, under mildly acidic conditions. The removal efficiency of the regenerated membrane after six cycles of adsorption/desorption was maintained at a high level of 98%. The proposed design offers a simple yet effective, sustainable, and environmentally friendly solution for water treatment.
Collapse
Affiliation(s)
- Fatima Youness
- American University of Beirut (AUB), Department of Civil and Environmental Engineering Riad El Solh Beirut 1107-2020 Lebanon
| | - Amani Jaafar
- American University of Beirut (AUB), Department of Civil and Environmental Engineering Riad El Solh Beirut 1107-2020 Lebanon
| | - Ali Tehrani
- Aalto University, Departments of Bioproducts and Biosystems Espoo Finland
| | - Rana A Bilbeisi
- American University of Beirut (AUB), Department of Civil and Environmental Engineering Riad El Solh Beirut 1107-2020 Lebanon
| |
Collapse
|
78
|
Ngougni Pokem P, Matzneller P, Vervaeke S, Wittebole X, Goeman L, Coessens M, Cottone E, Capron A, Wulkersdorfer B, Wallemacq P, Mouton JW, Muller AE, Zeitlinger M, Laterre PF, Tulkens PM, Van Bambeke F. Binding of temocillin to plasma proteins in vitro and in vivo: the importance of plasma protein levels in different populations and of co-medications. J Antimicrob Chemother 2022; 77:2742-2753. [PMID: 36018077 DOI: 10.1093/jac/dkac286] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 07/28/2022] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Temocillin plasma protein binding (PPB) in healthy individuals is reported to be ∼85% but had not been studied in patients. OBJECTIVES To obtain normative data on temocillin PPB in patients in relation to infection and impact of co-medications widely used in ICU. METHODS Plasma was obtained from healthy individuals (Group #1), non-ICU patients with UTI (Group #2), ICU patients with suspected/confirmed ventriculitis (Group #3) or with sepsis/septic shock (Group #4). Total and unbound temocillin concentrations were measured in spiked samples from temocillin-naive donors (in vitro) or in plasma from temocillin-treated subjects (in vivo). The impact of diluting plasma, using pharmaceutical albumin, or adding drugs potentially competing for PPB was tested in spiked samples. Data were analysed using a modified Hill-Langmuir equation taking ligand depletion into account. RESULTS Temocillin PPB was saturable in all groups, both in vitro and in vivo. Maximal binding capacity (Bmax) was 1.2-2-fold lower in patients. At 20 and 200 mg/L (total concentrations), the unbound fraction reached 12%-29%, 23%-42% and 32%-52% in Groups #2, #3, #4. The unbound fraction was inversely correlated with albumin and C-reactive protein concentrations. Binding to albumin was 2-3-fold lower than in plasma and non-saturable. Drugs with high PPB but active at lower molar concentrations than temocillin caused minimal displacement, while fluconazole (low PPB but similar plasma concentrations to temocillin) increased up to 2-fold its unbound fraction. CONCLUSIONS Temocillin PPB is saturable, 2-4-fold lowered in infected patients in relation to disease severity (ICU admission, hypoalbuminaemia, inflammation) and only partially reproducible with albumin. Competition with other drugs must be considered for therapeutic concentrations to be meaningful.
Collapse
Affiliation(s)
- Perrin Ngougni Pokem
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Peter Matzneller
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna Austria
| | - Steven Vervaeke
- Department of Laboratory Medicine, AZ Delta Hospital, Roeselare, Belgium
| | - Xavier Wittebole
- Department of Critical Care Medicine, Cliniques universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium
| | - Lieven Goeman
- Department of Urology, AZ Delta Hospital, Roeselare, Belgium
| | - Marie Coessens
- Department of Laboratory Medicine, AZ Delta Hospital, Roeselare, Belgium
| | - Eleonora Cottone
- Department of Laboratory Medicine, AZ Delta Hospital, Roeselare, Belgium
| | - Arnaud Capron
- Department of Clinical Chemistry, Cliniques universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium
| | | | - Pierre Wallemacq
- Department of Clinical Chemistry, Cliniques universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium
| | - Johan W Mouton
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC, Rotterdam, The Netherlands
| | - Anouk E Muller
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC, Rotterdam, The Netherlands.,Department of Medical Microbiology, Haaglanden Medical Centre, The Hague, The Netherlands
| | - Markus Zeitlinger
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna Austria
| | - Pierre François Laterre
- Department of Critical Care Medicine, Cliniques universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium
| | - Paul M Tulkens
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Françoise Van Bambeke
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
79
|
Tye MA, Payne NC, Johansson C, Singh K, Santos SA, Fagbami L, Pant A, Sylvester K, Luth MR, Marques S, Whitman M, Mota MM, Winzeler EA, Lukens AK, Derbyshire ER, Oppermann U, Wirth DF, Mazitschek R. Elucidating the path to Plasmodium prolyl-tRNA synthetase inhibitors that overcome halofuginone resistance. Nat Commun 2022; 13:4976. [PMID: 36008486 PMCID: PMC9403976 DOI: 10.1038/s41467-022-32630-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 08/10/2022] [Indexed: 02/07/2023] Open
Abstract
The development of next-generation antimalarials that are efficacious against the human liver and asexual blood stages is recognized as one of the world's most pressing public health challenges. In recent years, aminoacyl-tRNA synthetases, including prolyl-tRNA synthetase, have emerged as attractive targets for malaria chemotherapy. We describe the development of a single-step biochemical assay for Plasmodium and human prolyl-tRNA synthetases that overcomes critical limitations of existing technologies and enables quantitative inhibitor profiling with high sensitivity and flexibility. Supported by this assay platform and co-crystal structures of representative inhibitor-target complexes, we develop a set of high-affinity prolyl-tRNA synthetase inhibitors, including previously elusive aminoacyl-tRNA synthetase triple-site ligands that simultaneously engage all three substrate-binding pockets. Several compounds exhibit potent dual-stage activity against Plasmodium parasites and display good cellular host selectivity. Our data inform the inhibitor requirements to overcome existing resistance mechanisms and establish a path for rational development of prolyl-tRNA synthetase-targeted anti-malarial therapies.
Collapse
Affiliation(s)
- Mark A Tye
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Graduate School of Arts and Sciences, Cambridge, MA, USA
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - N Connor Payne
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Catrine Johansson
- Botnar Research Centre, NIHR Oxford Biomedical Research Unit, University of Oxford, Oxford, UK
- Centre for Medicines Discovery, University of Oxford, Oxford, UK
| | - Kritika Singh
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Sofia A Santos
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Lọla Fagbami
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Graduate School of Arts and Sciences, Cambridge, MA, USA
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Akansha Pant
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | | | - Madeline R Luth
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| | - Sofia Marques
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Malcolm Whitman
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA, USA
| | - Maria M Mota
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Elizabeth A Winzeler
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| | | | | | - Udo Oppermann
- Botnar Research Centre, NIHR Oxford Biomedical Research Unit, University of Oxford, Oxford, UK
- Centre for Medicines Discovery, University of Oxford, Oxford, UK
| | - Dyann F Wirth
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ralph Mazitschek
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA, USA.
- Harvard T.H. Chan School of Public Health, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
80
|
Baumgaertner P, Schmidt J, Costa-Nunes CM, Bordry N, Guillaume P, Luescher I, Speiser DE, Rufer N, Hebeisen M. CD8 T cell function and cross-reactivity explored by stepwise increased peptide-HLA versus TCR affinity. Front Immunol 2022; 13:973986. [PMID: 36032094 PMCID: PMC9399405 DOI: 10.3389/fimmu.2022.973986] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 07/22/2022] [Indexed: 12/05/2022] Open
Abstract
Recruitment and activation of CD8 T cells occur through specific triggering of T cell receptor (TCR) by peptide-bound human leucocyte antigen (HLA) ligands. Within the generated trimeric TCR-peptide:HLA complex, the molecular binding affinities between peptide and HLA, and between TCR and peptide:HLA both impact T cell functional outcomes. However, how their individual and combined effects modulate immunogenicity and overall T cell responsiveness has not been investigated systematically. Here, we established two panels of human tumor peptide variants differing in their affinity to HLA. For precise characterization, we developed the “blue peptide assay”, an upgraded cell-based approach to measure the peptide:HLA affinity. These peptide variants were then used to investigate the cross-reactivity of tumor antigen-specific CD8 T cell clonotypes derived from blood of cancer patients after vaccination with either the native or an affinity-optimized Melan-A/MART-1 epitope, or isolated from tumor infiltrated lymph nodes (TILNs). Vaccines containing the native tumor epitope generated T cells with better functionality, and superior cross-reactivity against potential low affinity escape epitopes, as compared to T cells induced by vaccines containing an HLA affinity-optimized epitope. Comparatively, Melan-A/MART-1-specific TILN cells displayed functional and cross-reactive profiles that were heterogeneous and clonotype-dependent. Finally, we took advantage of a collection of T cells expressing affinity-optimized NY-ESO-1-specific TCRs to interrogate the individual and combined impact of peptide:HLA and TCR-pHLA affinities on overall CD8 T cell responses. We found profound and distinct effects of both biophysical parameters, with additive contributions and absence of hierarchical dominance. Altogether, the biological impact of peptide:HLA and TCR-pHLA affinities on T cell responses was carefully dissected in two antigenic systems, frequently targeted in human cancer immunotherapy. Our technology and stepwise comparison open new insights into the rational design and selection of vaccine-associated tumor-specific epitopes and highlight the functional and cross-reactivity profiles that endow T cells with best tumor control capacity.
Collapse
Affiliation(s)
- Petra Baumgaertner
- Department of Oncology, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch - University of Lausanne, Epalinges, Switzerland
- *Correspondence: Michael Hebeisen, ; Petra Baumgaertner,
| | - Julien Schmidt
- Department of Oncology, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch - University of Lausanne, Epalinges, Switzerland
| | - Carla-Marisa Costa-Nunes
- Department of Oncology, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch - University of Lausanne, Epalinges, Switzerland
| | - Natacha Bordry
- Department of Oncology, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch - University of Lausanne, Epalinges, Switzerland
| | - Philippe Guillaume
- Department of Oncology, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch - University of Lausanne, Epalinges, Switzerland
| | - Immanuel Luescher
- Department of Oncology, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch - University of Lausanne, Epalinges, Switzerland
| | - Daniel E. Speiser
- Department of Oncology, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch - University of Lausanne, Epalinges, Switzerland
| | - Nathalie Rufer
- Department of Oncology, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch - University of Lausanne, Epalinges, Switzerland
| | - Michael Hebeisen
- Department of Oncology, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch - University of Lausanne, Epalinges, Switzerland
- *Correspondence: Michael Hebeisen, ; Petra Baumgaertner,
| |
Collapse
|
81
|
Andres AE, Mariano A, Rane D, Peterson BR. Quantification of Engagement of Microtubules by Small Molecules in Living Cells by Flow Cytometry. ACS BIO & MED CHEM AU 2022; 2:529-537. [PMID: 36281300 PMCID: PMC9585582 DOI: 10.1021/acsbiomedchemau.2c00031] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/25/2022] [Accepted: 07/27/2022] [Indexed: 11/29/2022]
Abstract
![]()
Drugs such as paclitaxel (Taxol) that bind microtubules
are widely
used for the treatment of cancer. Measurements of the affinity and
selectivity of these compounds for their targets are largely based
on studies of purified proteins, and only a few quantitative methods
for the analysis of interactions of small molecules with microtubules
in living cells have been reported. We describe here a novel method
for rapidly quantifying the affinities of compounds that bind polymerized
tubulin in living HeLa cells. This method uses the fluorescent molecular
probe Pacific Blue-GABA-Taxol in conjunction with verapamil to block
cellular efflux. Under physiologically relevant conditions of 37 °C,
this combination allowed quantification of equilibrium saturation
binding of this probe to cellular microtubules (Kd = 1.7 μM) using flow cytometry. Competitive binding
of the microtubule stabilizers paclitaxel (cellular Ki = 22 nM), docetaxel (cellular Ki = 16 nM), cabazitaxel (cellular Ki = 6 nM), and ixabepilone (cellular Ki = 10 nM) revealed intracellular affinities for microtubules that
closely matched previously reported biochemical affinities. By including
a cooperativity factor (α) for curve fitting of allosteric modulators,
this probe also allowed quantification of binding (Kb) of the microtubule destabilizers colchicine (Kb = 80 nM, α = 0.08), vinblastine (Kb = 7 nM, α = 0.18), and maytansine (Kb = 3 nM, α = 0.21). Screening of this
assay against 1008 NCI diversity compounds identified NSC 93427 as
a novel microtubule destabilizer (Kb =
485 nM, α = 0.02), illustrating the potential of this approach
for drug discovery.
Collapse
Affiliation(s)
- Angelo E. Andres
- Division of Medicinal Chemistry and Pharmacognosy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Andres Mariano
- Division of Medicinal Chemistry and Pharmacognosy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Digamber Rane
- Division of Medicinal Chemistry and Pharmacognosy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Blake R. Peterson
- Division of Medicinal Chemistry and Pharmacognosy, The Ohio State University, Columbus, Ohio 43210, United States
| |
Collapse
|
82
|
Puiu M, Bala C. Affinity Assays for Cannabinoids Detection: Are They Amenable to On-Site Screening? BIOSENSORS 2022; 12:608. [PMID: 36005003 PMCID: PMC9405638 DOI: 10.3390/bios12080608] [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] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 07/27/2022] [Accepted: 08/04/2022] [Indexed: 12/02/2022]
Abstract
Roadside testing of illicit drugs such as tetrahydrocannabinol (THC) requires simple, rapid, and cost-effective methods. The need for non-invasive detection tools has led to the development of selective and sensitive platforms, able to detect phyto- and synthetic cannabinoids by means of their main metabolites in breath, saliva, and urine samples. One may estimate the time passed from drug exposure and the frequency of use by corroborating the detection results with pharmacokinetic data. In this review, we report on the current detection methods of cannabinoids in biofluids. Fluorescent, electrochemical, colorimetric, and magnetoresistive biosensors will be briefly overviewed, putting emphasis on the affinity formats amenable to on-site screening, with possible applications in roadside testing and anti-doping control.
Collapse
Affiliation(s)
- Mihaela Puiu
- R&D Center LaborQ, University of Bucharest, 4-12 Regina Elisabeta Blvd., 030018 Bucharest, Romania
| | - Camelia Bala
- R&D Center LaborQ, University of Bucharest, 4-12 Regina Elisabeta Blvd., 030018 Bucharest, Romania
- Department of Analytical Chemistry, University of Bucharest, 4-12 Regina Elisabeta Blvd., 030018 Bucharest, Romania
| |
Collapse
|
83
|
Mattern A, Claßen R, Wolf A, Pouokam E, Schlüter KD, Wickleder MS, Diener M. Multivalent stimulation of β 1-, but not β 2-receptors by adrenaline functionalised gold nanoparticles. NANOSCALE ADVANCES 2022; 4:3182-3193. [PMID: 36132815 PMCID: PMC9418053 DOI: 10.1039/d2na00171c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 06/25/2022] [Indexed: 05/13/2023]
Abstract
In this study, we present a strategy for the synthesis of catecholamine functionalised gold nanoparticles and investigated their multivalent interactions with adrenergic receptors in different biological systems. The catecholamines adrenaline and noradrenaline represent key examples of adrenergic agonists. We used gold nanoparticles as carriers and functionalised them on their surface with a variety of these neurotransmitter molecules. For this purpose, we synthesised each ligand separately using mercaptoundecanoic acid as a bifunctional linking unit and adrenaline (or noradrenaline) as a biogenic amine. This ligand was then immobilised onto the surface of presynthesised spherical monodispersive gold nanoparticles in a ligand exchange reaction. After detailed analytical characterisations, the functionalised gold nanoparticles were investigated for their interactions with adrenergic receptors in intestinal, cardiac and respiratory tissues. Whereas the contractility of respiratory smooth muscle cells (regulated by β2-receptors) was not influenced, (nor)adrenaline functionalised nanoparticles administered in nanomolar concentrations induced epithelial K+ secretion (mediated via different β-receptors) and increased contractility of isolated rat cardiomyocytes (mediated by β1-receptors). The present results suggest differences in the accessibility of adrenergic agonists bound to gold nanoparticles to the binding pockets of different β-receptor subtypes.
Collapse
Affiliation(s)
- Annabelle Mattern
- Institute of Inorganic Chemistry, University of Cologne Greinstrasse 6 50939 Cologne Germany
| | - Rebecca Claßen
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen Frankfurter Strasse 100 35392 Giessen Germany
| | - Annemarie Wolf
- Institute of Physiology, Justus Liebig University Giessen Aulweg 129 35392 Giessen Germany
| | - Ervice Pouokam
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen Frankfurter Strasse 100 35392 Giessen Germany
| | - Klaus-Dieter Schlüter
- Institute of Physiology, Justus Liebig University Giessen Aulweg 129 35392 Giessen Germany
| | - Mathias S Wickleder
- Institute of Inorganic Chemistry, University of Cologne Greinstrasse 6 50939 Cologne Germany
| | - Martin Diener
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen Frankfurter Strasse 100 35392 Giessen Germany
| |
Collapse
|
84
|
Deng Y, Wang SY. Sorption of Cellulases in Biofilm Enhances Cellulose Degradation by Bacillus subtilis. Microorganisms 2022; 10:microorganisms10081505. [PMID: 35893563 PMCID: PMC9329931 DOI: 10.3390/microorganisms10081505] [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: 06/24/2022] [Revised: 07/11/2022] [Accepted: 07/22/2022] [Indexed: 02/04/2023] Open
Abstract
Biofilm commonly forms on the surfaces of cellulosic biomass but its roles in cellulose degradation remain largely unexplored. We used Bacillus subtilis to study possible mechanisms and the contributions of two major biofilm components, extracellular polysaccharides (EPS) and TasA protein, to submerged biofilm formation on cellulose and its degradation. We found that biofilm produced by B. subtilis is able to absorb exogenous cellulase added to the culture medium and also retain self-produced cellulase within the biofilm matrix. The bacteria that produced more biofilm degraded more cellulose compared to strains that produced less biofilm. Knockout strains that lacked both EPS and TasA formed a smaller amount of submerged biofilm on cellulose than the wild-type strain and also degraded less cellulose. Imaging of biofilm on cellulose suggests that bacteria, cellulose, and cellulases form cellulolytic biofilm complexes that facilitate synergistic cellulose degradation. This study brings additional insight into the important functions of biofilm in cellulose degradation and could potentiate the development of biofilm-based technology to enhance biomass degradation for biofuel production.
Collapse
|
85
|
Luo H, Xiang Y, Fang X, Lin W, Wang F, Wu H, Wang H. BatchDTA: implicit batch alignment enhances deep learning-based drug-target affinity estimation. Brief Bioinform 2022; 23:6632927. [PMID: 35794723 DOI: 10.1093/bib/bbac260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/23/2022] [Accepted: 06/03/2022] [Indexed: 11/14/2022] Open
Abstract
Candidate compounds with high binding affinities toward a target protein are likely to be developed as drugs. Deep neural networks (DNNs) have attracted increasing attention for drug-target affinity (DTA) estimation owning to their efficiency. However, the negative impact of batch effects caused by measure metrics, system technologies and other assay information is seldom discussed when training a DNN model for DTA. Suffering from the data deviation caused by batch effects, the DNN models can only be trained on a small amount of 'clean' data. Thus, it is challenging for them to provide precise and consistent estimations. We design a batch-sensitive training framework, namely BatchDTA, to train the DNN models. BatchDTA implicitly aligns multiple batches toward the same protein through learning the orders of candidate compounds with respect to the batches, alleviating the impact of the batch effects on the DNN models. Extensive experiments demonstrate that BatchDTA facilitates four mainstream DNN models to enhance the ability and robustness on multiple DTA datasets (BindingDB, Davis and KIBA). The average concordance index of the DNN models achieves a relative improvement of 4.0%. The case study reveals that BatchDTA can successfully learn the ranking orders of the compounds from multiple batches. In addition, BatchDTA can also be applied to the fused data collected from multiple sources to achieve further improvement.
Collapse
Affiliation(s)
- Hongyu Luo
- PaddleHelix team, Baidu Inc., 518000, Shenzhen, China
| | - Yingfei Xiang
- PaddleHelix team, Baidu Inc., 518000, Shenzhen, China
| | - Xiaomin Fang
- PaddleHelix team, Baidu Inc., 518000, Shenzhen, China
| | - Wei Lin
- PaddleHelix team, Baidu Inc., 518000, Shenzhen, China
| | - Fan Wang
- PaddleHelix team, Baidu Inc., 518000, Shenzhen, China
| | - Hua Wu
- Baidu Inc., 100000, Beijing, China
| | | |
Collapse
|
86
|
Guffick C, Hsieh PY, Ali A, Shi W, Howard J, Chinthapalli DK, Kong AC, Salaa I, Crouch LI, Ansbro MR, Isaacson SC, Singh H, Barrera NP, Nair AV, Robinson CV, Deery MJ, van Veen HW. Drug-dependent inhibition of nucleotide hydrolysis in the heterodimeric ABC multidrug transporter PatAB from Streptococcus pneumoniae. FEBS J 2022; 289:3770-3788. [PMID: 35066976 PMCID: PMC9541285 DOI: 10.1111/febs.16366] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 12/07/2021] [Accepted: 01/20/2022] [Indexed: 02/02/2023]
Abstract
The bacterial heterodimeric ATP‐binding cassette (ABC) multidrug exporter PatAB has a critical role in conferring antibiotic resistance in multidrug‐resistant infections by Streptococcus pneumoniae. As with other heterodimeric ABC exporters, PatAB contains two transmembrane domains that form a drug translocation pathway for efflux and two nucleotide‐binding domains that bind ATP, one of which is hydrolysed during transport. The structural and functional elements in heterodimeric ABC multidrug exporters that determine interactions with drugs and couple drug binding to nucleotide hydrolysis are not fully understood. Here, we used mass spectrometry techniques to determine the subunit stoichiometry in PatAB in our lactococcal expression system and investigate locations of drug binding using the fluorescent drug‐mimetic azido‐ethidium. Surprisingly, our analyses of azido‐ethidium‐labelled PatAB peptides point to ethidium binding in the PatA nucleotide‐binding domain, with the azido moiety crosslinked to residue Q521 in the H‐like loop of the degenerate nucleotide‐binding site. Investigation into this compound and residue’s role in nucleotide hydrolysis pointed to a reduction in the activity for a Q521A mutant and ethidium‐dependent inhibition in both mutant and wild type. Most transported drugs did not stimulate or inhibit nucleotide hydrolysis of PatAB in detergent solution or lipidic nanodiscs. However, further examples for ethidium‐like inhibition were found with propidium, novobiocin and coumermycin A1, which all inhibit nucleotide hydrolysis by a non‐competitive mechanism. These data cast light on potential mechanisms by which drugs can regulate nucleotide hydrolysis by PatAB, which might involve a novel drug binding site near the nucleotide‐binding domains.
Collapse
Affiliation(s)
| | - Pei-Yu Hsieh
- Department of Pharmacology, University of Cambridge, UK
| | - Anam Ali
- Department of Pharmacology, University of Cambridge, UK
| | - Wilma Shi
- Department of Pharmacology, University of Cambridge, UK
| | - Julie Howard
- Cambridge Centre for Proteomics, Department of Biochemistry, University of Cambridge, UK
| | | | - Alex C Kong
- Department of Pharmacology, University of Cambridge, UK
| | - Ihsene Salaa
- Department of Pharmacology, University of Cambridge, UK
| | - Lucy I Crouch
- Department of Pharmacology, University of Cambridge, UK
| | | | | | | | - Nelson P Barrera
- Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Asha V Nair
- Department of Pharmacology, University of Cambridge, UK
| | | | - Michael J Deery
- Cambridge Centre for Proteomics, Department of Biochemistry, University of Cambridge, UK
| | | |
Collapse
|
87
|
Alam MS, Azam S, Pham K, Leyva D, Fouque KJD, Fernandez-Lima F, Miksovska J. Nanomolar affinity of EF-hands in neuronal calcium sensor 1 for bivalent cations Pb2+, Mn2+ and Hg2. Metallomics 2022; 14:6601456. [PMID: 35657675 DOI: 10.1093/mtomcs/mfac039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 05/31/2022] [Indexed: 11/12/2022]
Abstract
Abiogenic metals Pb and Hg are highly toxic since chronic and/or acute exposure often leads to severe neuropathologies. Mn2+ is an essential metal ion but in excess can impair neuronal function. In this study, we address in vitro the interactions between neuronal calcium sensor 1 (NCS1) and divalent cations. Results showed that non-physiological ions (Pb2+, Mn2+ and Hg2+) bind to EF-hands in NCS1 with nanomolar affinity and lower equilibrium dissociation constant than the physiological Ca2+ ion. (Kd,Pb2+ = 7.0±1.0 nM; Kd,Mn2+ = 34.0±6.0 nM; Kd, Hg2+ = 0.5±0.1 nM and 27.0±13.0 nM and Kd,Ca2+ = 96.0±48.0 nM). Native ultra-high resolution mass spectrometry (FT-ICR MS) and trapped ion mobility spectrometry - mass spectrometry (nESI-TIMS-MS) studies provided the NCS1-metal complex compositions - up to four Ca2+ or Mn2+ ions and three Pb2+ ions (M⋅Pb1-3Ca1-3, M⋅Mn1-4Ca1-2, and M⋅Ca1-4) were observed in complex - and similarity across the mobility profiles suggests that the overall native structure is preserved regardless of the number and type of cations. However, the non-physiological metal ions (Pb2+, Mn2+, and Hg2+) binding to NCS1 leads to more efficient quenching of Trp emission and a decrease in W30 and W103 solvent exposure compared to the apo and Ca2+ bound form, although the secondary structural rearrangement and exposure of hydrophobic sites are analogous to those for Ca2+ bound protein. Only Pb2+ and Hg2+ binding to EF-hands leads to the NCS1 dimerization whereas Mn2+ bound NCS1 remains in the monomeric form, suggesting that other factors in addition to metal ion coordination, are required for protein dimerization.
Collapse
Affiliation(s)
- Md Shofiul Alam
- Department of Chemistry and Biochemistry, Florida International University, Miami FL 33199USA
| | - Samiol Azam
- Department of Chemistry and Biochemistry, Florida International University, Miami FL 33199USA
| | - Khoa Pham
- Department of Chemistry and Biochemistry, Florida International University, Miami FL 33199USA
| | - Dennys Leyva
- Department of Chemistry and Biochemistry, Florida International University, Miami FL 33199USA
| | - Kevin Jeanne Dit Fouque
- Department of Chemistry and Biochemistry, Florida International University, Miami FL 33199USA.,Biomolecular Sciences Institute, Florida International University, Miami, 33199USA
| | - Francisco Fernandez-Lima
- Department of Chemistry and Biochemistry, Florida International University, Miami FL 33199USA.,Biomolecular Sciences Institute, Florida International University, Miami, 33199USA
| | - Jaroslava Miksovska
- Department of Chemistry and Biochemistry, Florida International University, Miami FL 33199USA.,Biomolecular Sciences Institute, Florida International University, Miami, 33199USA
| |
Collapse
|
88
|
From affinity selection to kinetic selection in Germinal Centre modelling. PLoS Comput Biol 2022; 18:e1010168. [PMID: 35658003 PMCID: PMC9200358 DOI: 10.1371/journal.pcbi.1010168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 06/15/2022] [Accepted: 05/05/2022] [Indexed: 11/30/2022] Open
Abstract
Affinity maturation is an evolutionary process by which the affinity of antibodies (Abs) against specific antigens (Ags) increases through rounds of B-cell proliferation, somatic hypermutation, and positive selection in germinal centres (GC). The positive selection of B cells depends on affinity, but the underlying mechanisms of affinity discrimination and affinity-based selection are not well understood. It has been suggested that selection in GC depends on both rapid binding of B-cell receptors (BcRs) to Ags which is kinetically favourable and tight binding of BcRs to Ags, which is thermodynamically favourable; however, it has not been shown whether a selection bias for kinetic properties is present in the GC. To investigate the GC selection bias towards rapid and tight binding, we developed an agent-based model of GC and compared the evolution of founder B cells with initially identical low affinities but with different association/dissociation rates for Ag presented by follicular dendritic cells in three Ag collection mechanisms. We compared an Ag collection mechanism based on association/dissociation rates of B-cell interaction with presented Ag, which includes a probabilistic rupture of bonds between the B-cell and Ag (Scenario-1) with a reference scenario based on an affinity-based Ag collection mechanism (Scenario-0). Simulations showed that the mechanism of Ag collection affects the GC dynamics and the GC outputs concerning fast/slow (un)binding of B cells to FDC-presented Ags. In particular, clones with lower dissociation rates outcompete clones with higher association rates in Scenario-1, while remaining B cells from clones with higher association rates reach higher affinities. Accordingly, plasma cell and memory B cell populations were biased towards B-cell clones with lower dissociation rates. Without such probabilistic ruptures during the Ag extraction process (Scenario-2), the selective advantage for clones with very low dissociation rates diminished, and the affinity maturation level of all clones decreased to the reference level. Adaptive immunity is one of the vital defence mechanisms of the human body to fight virtually unlimited types of pathogens by producing antigen-specific high-affinity antibodies that bind to pathogens and neutralise them or mark them for further elimination. Affinity is a quantity used to measure and report the strength of interaction between antibodies and antigens that depends both on how fast antibodies bind to antigens (association rate) and how long the bond lasts (dissociation rate). The affinity of produced antibodies for a specific antigen increases in germinal centres through a process called affinity maturation, during which B cells with higher affinities have a competitive advantage and get positively selected to differentiate to antibody-producing plasma cells. Our research shows that the mechanism by which B cells capture Ag affects GC dynamics and GC output with respect to B-cell receptor kinetics. Notably, in a mechanism where rupture of CC-FDC bonds is possible during Ag extraction, B-cell clones with low dissociation rates outcompete clones with high association rates over time. Understanding how B cells get selected in germinal centres could help to develop an optimised and effective immune response against a disease through vaccination for a fast-operating and long-lasting immune response.
Collapse
|
89
|
Wollman AJM, Fournier C, Llorente-Garcia I, Harriman O, Payne-Dwyer AL, Shashkova S, Zhou P, Liu TC, Ouaret D, Wilding J, Kusumi A, Bodmer W, Leake MC. Critical roles for EGFR and EGFR-HER2 clusters in EGF binding of SW620 human carcinoma cells. J R Soc Interface 2022; 19:20220088. [PMID: 35612280 PMCID: PMC9131850 DOI: 10.1098/rsif.2022.0088] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Epidermal growth factor (EGF) signalling regulates normal epithelial and other cell growth, with EGF receptor (EGFR) overexpression reported in many cancers. However, the role of EGFR clusters in cancer and their dependence on EGF binding is unclear. We present novel single-molecule total internal reflection fluorescence microscopy of (i) EGF and EGFR in living cancer cells, (ii) the action of anti-cancer drugs that separately target EGFR and human EGFR2 (HER2) on these cells and (iii) EGFR–HER2 interactions. We selected human epithelial SW620 carcinoma cells for their low level of native EGFR expression, for stable transfection with fluorescent protein labelled EGFR, and imaged these using single-molecule localization microscopy to quantify receptor architectures and dynamics upon EGF binding. Prior to EGF binding, we observe pre-formed EGFR clusters. Unexpectedly, clusters likely contain both EGFR and HER2, consistent with co-diffusion of EGFR and HER2 observed in a different model CHO-K1 cell line, whose stoichiometry increases following EGF binding. We observe a mean EGFR : EGF stoichiometry of approximately 4 : 1 for plasma membrane-colocalized EGFR–EGF that we can explain using novel time-dependent kinetics modelling, indicating preferential ligand binding to monomers. Our results may inform future cancer drug developments.
Collapse
Affiliation(s)
- Adam J M Wollman
- Department of Physics, University of York, York, UK.,Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Charlotte Fournier
- Department of Physics, Clarendon Laboratory, University of Oxford, Oxford OX1 3PU, UK.,Science and Technology Group, Okinawa Institute of Science and Technology Graduate University (OIST), 1919 Tancha, Onna-son, Okinawa 904-0495, Japan
| | | | - Oliver Harriman
- Department of Physics, Clarendon Laboratory, University of Oxford, Oxford OX1 3PU, UK
| | | | | | - Peng Zhou
- Membrane Cooperativity Unit, OIST, 1919 Tancha, Onna-son, Okinawa 904-0495, Japan
| | - Ta-Chun Liu
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, UK
| | - Djamila Ouaret
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, UK
| | - Jenny Wilding
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, UK
| | - Akihiro Kusumi
- Membrane Cooperativity Unit, OIST, 1919 Tancha, Onna-son, Okinawa 904-0495, Japan
| | - Walter Bodmer
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, UK
| | - Mark C Leake
- Department of Physics, University of York, York, UK.,Department of Biology, University of York, York, UK.,Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| |
Collapse
|
90
|
Fallah HP, Ahuja E, Lin H, Qi J, He Q, Gao S, An H, Zhang J, Xie Y, Liang D. A Review on the Role of TRP Channels and Their Potential as Drug Targets_An Insight Into the TRP Channel Drug Discovery Methodologies. Front Pharmacol 2022; 13:914499. [PMID: 35685622 PMCID: PMC9170958 DOI: 10.3389/fphar.2022.914499] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 04/27/2022] [Indexed: 01/13/2023] Open
Abstract
Transient receptor potential (TRP) proteins are a large group of ion channels that control many physiological functions in our body. These channels are considered potential therapeutic drug targets for various diseases such as neurological disorders, cancers, cardiovascular disease, and many more. The Nobel Prize in Physiology/Medicine in the year 2021 was awarded to two scientists for the discovery of TRP and PIEZO ion channels. Improving our knowledge of technologies for their study is essential. In the present study, we reviewed the role of TRP channel types in the control of normal physiological functions as well as disease conditions. Also, we discussed the current and novel technologies that can be used to study these channels successfully. As such, Flux assays for detecting ionic flux through ion channels are among the core and widely used tools for screening drug compounds. Technologies based on these assays are available in fully automated high throughput set-ups and help detect changes in radiolabeled or non-radiolabeled ionic flux. Aurora's Ion Channel Reader (ICR), which works based on label-free technology of flux assay, offers sensitive, accurate, and reproducible measurements to perform drug ranking matching with patch-clamp (gold standard) data. The non-radiolabeled trace-based flux assay coupled with the ICR detects changes in various ion types, including potassium, calcium, sodium, and chloride channels, by using appropriate tracer ions. This technology is now considered one of the very successful approaches for analyzing ion channel activity in modern drug discovery. It could be a successful approach for studying various ion channels and transporters, including the different members of the TRP family of ion channels.
Collapse
Affiliation(s)
| | - Ekta Ahuja
- Aurora Biomed Inc., Vancouver, BC, Canada
| | | | - Jinlong Qi
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Qian He
- Aurora Discovery Inc., Foshan, China
| | - Shan Gao
- Aurora Discovery Inc., Foshan, China
| | | | | | | | - Dong Liang
- Aurora Biomed Inc., Vancouver, BC, Canada
- Aurora Discovery Inc., Foshan, China
- Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| |
Collapse
|
91
|
Shave S, Pham NT, Auer M. CLAffinity: A Software Tool for Identification of Optimum Ligand Affinity for Competition-Based Primary Screens. J Chem Inf Model 2022; 62:2264-2268. [PMID: 35442032 PMCID: PMC9131445 DOI: 10.1021/acs.jcim.2c00285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Indexed: 12/02/2022]
Abstract
A simplistic assumption in setting up a competition assay is that a low affinity labeled ligand can be more easily displaced from a target protein than a high affinity ligand, which in turn produces a more sensitive assay. An often-cited paper correctly rallies against this assumption and recommends the use of the highest affinity ligand available for experiments aiming to determine competitive inhibitor affinities. However, we have noted this advice being applied incorrectly to competition-based primary screens where the goal is optimum assay sensitivity, enabling a clear yes/no binding determination for even low affinity interactions. The published advice only applies to secondary, confirmatory assays intended for accurate affinity determination of primary screening hits. We demonstrate that using very high affinity ligands in competition-based primary screening can lead to reduced assay sensitivity and, ultimately, the discarding of potentially valuable active compounds. We build on techniques developed in our PyBindingCurve software for a mechanistic understanding of complex biological interaction systems, developing the "CLAffinity tool" for simulating competition experiments using protein, ligand, and inhibitor concentrations common to drug screening campaigns. CLAffinity reveals optimum labeled ligand affinity ranges based on assay parameters, rather than general rules to optimize assay sensitivity. We provide the open source CLAffinity software toolset to carry out assay simulations and a video summarizing key findings to aid in understanding, along with a simple lookup table allowing identification of optimal dynamic ranges for competition-based primary screens. The application of our freely available software and lookup tables will lead to the consistent creation of more performant competition-based primary screens identifying valuable hit compounds, particularly for difficult targets.
Collapse
Affiliation(s)
- Steven Shave
- School of Biological Sciences, University of Edinburgh, The King’s Buildings, Edinburgh, Scotland EH9
3BF, United Kingdom
| | - Nhan T. Pham
- School of Biological Sciences, University of Edinburgh, The King’s Buildings, Edinburgh, Scotland EH9
3BF, United Kingdom
| | - Manfred Auer
- School of Biological Sciences, University of Edinburgh, The King’s Buildings, Edinburgh, Scotland EH9
3BF, United Kingdom
| |
Collapse
|
92
|
Karvonen H, Raivola J, Ungureanu D. Cellular thermal shift assay (CETSA) for determining the drug binding affinity using Ba/F3 clones stably expressing receptor pseudokinases. Methods Enzymol 2022; 667:339-363. [PMID: 35525546 DOI: 10.1016/bs.mie.2022.03.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The majority of drug screening approaches are performed using recombinant proteins, however, drug binding to its target(s) in cells should be also assessed, especially for drugs aimed at modulating intracellular signaling pathways. As a result, the development of a cellular thermal shift assay (CETSA) has become an important tool for determining the binding affinity of drugs to their intracellular targets. Cell lines, such as Ba/F3, are an excellent model system to stably express and study a target protein when this protein is not endogenously expressed or only present at low levels. Together with CETSA, Ba/F3 clones allow study of the transforming properties of the protein in question, its downstream intracellular signaling activation pathways, as well as its drug binding kinetics. This chapter describes in detail the establishment of Ba/F3 clones stably expressing receptor pseudokinases, such as receptor tyrosine kinase-like orphan receptors (ROR1, ROR2) and protein tyrosine kinase 7 (PTK7), and the use thereof to evaluate binding of small molecule inhibitors to their intracellular (pseudo)kinase domain by CETSA.
Collapse
Affiliation(s)
- Hanna Karvonen
- Cancer Signaling, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Juuli Raivola
- Applied Tumor Genomics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Daniela Ungureanu
- Cancer Signaling, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland; Applied Tumor Genomics, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.
| |
Collapse
|
93
|
UnbiasedDTI: Mitigating Real-World Bias of Drug-Target Interaction Prediction by Using Deep Ensemble-Balanced Learning. Molecules 2022; 27:molecules27092980. [PMID: 35566330 PMCID: PMC9100109 DOI: 10.3390/molecules27092980] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 04/26/2022] [Accepted: 04/28/2022] [Indexed: 01/27/2023] Open
Abstract
Drug-target interaction (DTI) prediction through in vitro methods is expensive and time-consuming. On the other hand, computational methods can save time and money while enhancing drug discovery efficiency. Most of the computational methods frame DTI prediction as a binary classification task. One important challenge is that the number of negative interactions in all DTI-related datasets is far greater than the number of positive interactions, leading to the class imbalance problem. As a result, a classifier is trained biased towards the majority class (negative class), whereas the minority class (interacting pairs) is of interest. This class imbalance problem is not widely taken into account in DTI prediction studies, and the few previous studies considering balancing in DTI do not focus on the imbalance issue itself. Additionally, they do not benefit from deep learning models and experimental validation. In this study, we propose a computational framework along with experimental validations to predict drug-target interaction using an ensemble of deep learning models to address the class imbalance problem in the DTI domain. The objective of this paper is to mitigate the bias in the prediction of DTI by focusing on the impact of balancing and maintaining other involved parameters at a constant value. Our analysis shows that the proposed model outperforms unbalanced models with the same architecture trained on the BindingDB both computationally and experimentally. These findings demonstrate the significance of balancing, which reduces the bias towards the negative class and leads to better performance. It is important to note that leaning on computational results without experimentally validating them and by relying solely on AUROC and AUPRC metrics is not credible, particularly when the testing set remains unbalanced.
Collapse
|
94
|
Peters M, Bockfeld D, Tamm M. Cationic Iridium(I) NHC‐Phosphinidene Complexes and Their Application in Hydrogen Isotope Exchange Reactions. Eur J Inorg Chem 2022. [DOI: 10.1002/ejic.202200148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Marius Peters
- Technische Universität Braunschweig: Technische Universitat Braunschweig Institut für Anorganische und Analytische Chemie GERMANY
| | - Dirk Bockfeld
- Technische Universität Braunschweig: Technische Universitat Braunschweig Institut für Anorganische und Analytische Chemie GERMANY
| | - Matthias Tamm
- Technische Universität Braunschweig Institut für Anorganische und Analytische Chemie Hagenring 30 38106 Braunschweig GERMANY
| |
Collapse
|
95
|
Zhu Y, Schrecke S, Tang S, Odenkirk MT, Walker T, Stover L, Lyu J, Zhang T, Russell D, Baker ES, Yan X, Laganowsky A. Cupric Ions Selectively Modulate TRAAK-Phosphatidylserine Interactions. J Am Chem Soc 2022; 144:7048-7053. [PMID: 35421309 DOI: 10.1021/jacs.2c00612] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
TRAAK and TREK2 are two-pore domain K+ (K2P) channels and are modulated by diverse factors including temperature, membrane stretching, and lipids, such as phosphatidic acid. In addition, copper and zinc, both of which are essential for life, are known to regulate TREK2 and a number of other ion channels. However, the role of ions in the association of lipids with integral membrane proteins is poorly understood. Here, we discover cupric ions selectively modulate the binding of phosphatidylserine (PS) to TRAAK but not TREK2. Other divalent cations (Ca2+, Mg2+, and Zn2+) bind both channels but have no impact on binding PS and other lipids. Additionally, TRAAK binds more avidly to Cu2+ and Zn2+ than TREK2. In the presence of Cu2+, TRAAK binds similarly to PS with different acyl chains, indicating a crucial role of the serine headgroup in coordinating Cu2+. High-resolution native mass spectrometry (MS) enables the determination of equilibrium binding constants for distinct Cu2+-bound stoichiometries and uncovered the highest coupling factor corresponds to a 1:1 PS-to-Cu2+ ratio. Interestingly, the next three highest coupling factors had a ∼1.5:1 PS-to-Cu2+ ratio. Our findings bring forth the role of cupric ions as an essential cofactor in selective TRAAK-PS interactions.
Collapse
Affiliation(s)
- Yun Zhu
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Samantha Schrecke
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Shuli Tang
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Melanie T Odenkirk
- Department of Chemistry, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Thomas Walker
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Lauren Stover
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Jixing Lyu
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Tianqi Zhang
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - David Russell
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Erin S Baker
- Department of Chemistry, North Carolina State University, Raleigh, North Carolina 27695, United States.,Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Xin Yan
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Arthur Laganowsky
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| |
Collapse
|
96
|
Non-Peptide Opioids Differ in Effects on Mu-Opioid (MOP) and Serotonin 1A (5-HT 1A) Receptors Heterodimerization and Cellular Effectors (Ca 2+, ERK1/2 and p38) Activation. Molecules 2022; 27:molecules27072350. [PMID: 35408749 PMCID: PMC9000251 DOI: 10.3390/molecules27072350] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/24/2022] [Accepted: 04/02/2022] [Indexed: 11/17/2022] Open
Abstract
The importance of the dynamic interplay between the opioid and the serotonin neuromodulatory systems in chronic pain is well recognized. In this study, we investigated whether these two signalling pathways can be integrated at the single-cell level via direct interactions between the mu-opioid (MOP) and the serotonin 1A (5-HT1A) receptors. Using fluorescence cross-correlation spectroscopy (FCCS), a quantitative method with single-molecule sensitivity, we characterized in live cells MOP and 5-HT1A interactions and the effects of prolonged (18 h) exposure to selected non-peptide opioids: morphine, codeine, oxycodone and fentanyl, on the extent of these interactions. The results indicate that in the plasma membrane, MOP and 5-HT1A receptors form heterodimers that are characterized with an apparent dissociation constant Kdapp = (440 ± 70) nM). Prolonged exposure to all non-peptide opioids tested facilitated MOP and 5-HT1A heterodimerization and stabilized the heterodimer complexes, albeit to a different extent: Kd, Fentanylapp = (80 ± 70) nM), Kd,Morphineapp = (200 ± 70) nM, Kd, Codeineapp = (100 ± 70) nM and Kd, Oxycodoneapp = (200 ± 70) nM. The non-peptide opioids differed also in the extent to which they affected the mitogen-activated protein kinases (MAPKs) p38 and the extracellular signal-regulated kinase (Erk1/2), with morphine, codeine and fentanyl activating both pathways, whereas oxycodone activated p38 but not ERK1/2. Acute stimulation with different non-peptide opioids differently affected the intracellular Ca2+ levels and signalling dynamics. Hypothetically, targeting MOP−5-HT1A heterodimer formation could become a new strategy to counteract opioid induced hyperalgesia and help to preserve the analgesic effects of opioids in chronic pain.
Collapse
|
97
|
An original approach to measure ligand/receptor binding affinity in non-purified samples. Sci Rep 2022; 12:5400. [PMID: 35354858 PMCID: PMC8967833 DOI: 10.1038/s41598-022-09217-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 03/10/2022] [Indexed: 11/30/2022] Open
Abstract
Several biochemical and biophysical methods are available to determine ligand binding affinities between a biological target and its ligands, most of which require purification, labelling or surface immobilisation. These measurements, however, remain challenging in regards to membrane proteins, as purification processes require their extraction from their native lipid environment, which may in turn impact receptor conformation and functionality. In this study, we have developed a novel experimental procedure using microscale thermophoresis (MST) directly from cell membrane fragments, to determine different ligand binding affinities to a membrane protein, the dopamine D2 receptor (D2R). In order to achieve this, two main challenges had to be overcome: determining the concentration of dopamine D2R in the crude sample; finding ways to minimize or account for non-specific binding of the ligand to cell fragments. Using MST, we were able to determine the D2R concentration in cell membrane fragments to approximately 36.8 ± 2.6 pmol/mg. Next, the doses-responses curves allowed for the determination of KD, to approximately 5.3 ± 1.7 nM, which is very close to the reported value. Important details of the experimental procedure have been detailed in this paper to allow the application of this novel method to various membrane proteins.
Collapse
|
98
|
Nerella SG, Singh P, Sanam T, Digwal CS. PET Molecular Imaging in Drug Development: The Imaging and Chemistry Perspective. Front Med (Lausanne) 2022; 9:812270. [PMID: 35295604 PMCID: PMC8919964 DOI: 10.3389/fmed.2022.812270] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/18/2022] [Indexed: 11/13/2022] Open
Abstract
Positron emission tomography with selective radioligands advances the drug discovery and development process by revealing information about target engagement, proof of mechanism, pharmacokinetic and pharmacodynamic profiles. Positron emission tomography (PET) is an essential and highly significant tool to study therapeutic drug development, dose regimen, and the drug plasma concentrations of new drug candidates. Selective radioligands bring up target-specific information in several disease states including cancer, cardiovascular, and neurological conditions by quantifying various rates of biological processes with PET, which are associated with its physiological changes in living subjects, thus it reveals disease progression and also advances the clinical investigation. This study explores the major roles, applications, and advances of PET molecular imaging in drug discovery and development process with a wide range of radiochemistry as well as clinical outcomes of positron-emitting carbon-11 and fluorine-18 radiotracers.
Collapse
Affiliation(s)
- Sridhar Goud Nerella
- Department of Neuroimaging and Interventional Radiology, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | - Priti Singh
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Hyderabad, India
| | - Tulja Sanam
- Department of Microbiology and Applied Sciences, University of Agricultural Sciences, Bangalore, India
| | - Chander Singh Digwal
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Hyderabad, India
| |
Collapse
|
99
|
Yang H, Huang Z, Lehnherr D, Lam YH, Ren S, Strotman NA. Efficient Aliphatic Hydrogen-Isotope Exchange with Tritium Gas through the Merger of Photoredox and Hydrogenation Catalysts. J Am Chem Soc 2022; 144:5010-5022. [PMID: 35263094 DOI: 10.1021/jacs.1c13265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Employment of a combination of an organophotoredox catalyst with Wilkinson's catalyst (Rh(PPh3)3Cl) has given rise to an unprecedented method for hydrogen-isotope exchange (HIE) of aliphatic C(sp3)-H bonds of complex pharmaceuticals using T2 gas directly. Wilkinson's catalyst, commonly used for catalytic hydrogenations, was exploited as a precatalyst for activation of D2 or T2 and hydrogen atom transfer. In this combined methodology and mechanistic study, we demonstrate that by coupling photocatalysis with Rh catalysis, carbon-centered radicals generated via photoredox catalysis can be intercepted by Rh-hydride intermediates to deliver an effective hydrogen atom donor for hydrogen-isotope labeling of complex molecules in one step. By optimizing the ratio of the photocatalyst and Wilkinson's catalyst to balance the rate of the dual catalytic cycles, we can achieve efficient HIE and high recovery yield. This protocol was readily applied to direct HIE of C(sp3)-H bonds in 10 complex drug molecules, showing high isotope incorporation efficiency and exceptionally good functional group tolerance and demonstrating this approach as a practical and attractive labeling method for deuteration and tritiation.
Collapse
Affiliation(s)
- Haifeng Yang
- Process Research & Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Zheng Huang
- Process Research & Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Dan Lehnherr
- Process Research & Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Yu-Hong Lam
- Computational and Structural Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Sumei Ren
- Process Research & Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Neil A Strotman
- Process Research & Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| |
Collapse
|
100
|
Sikstus S, Benkherouf AY, Soini SL, Uusi-Oukari M. The Influence of AA29504 on GABA A Receptor Ligand Binding Properties and Its Implications on Subtype Selectivity. Neurochem Res 2022; 47:667-678. [PMID: 34727270 PMCID: PMC8847198 DOI: 10.1007/s11064-021-03475-y] [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: 06/04/2021] [Revised: 09/03/2021] [Accepted: 10/27/2021] [Indexed: 10/26/2022]
Abstract
The unique pharmacological properties of δ-containing γ-aminobutyric acid type A receptors (δ-GABAARs) make them an attractive target for selective and persistent modulation of neuronal excitability. However, the availability of selective modulators targeting δ-GABAARs remains limited. AA29504 ([2-amino-4-(2,4,6-trimethylbenzylamino)-phenyl]-carbamic acid ethyl ester), an analog of K+ channel opener retigabine, acts as an agonist and a positive allosteric modulator (Ago-PAM) of δ-GABAARs. Based on electrophysiological studies using recombinant receptors, AA29504 was found to be a more potent and effective agonist in δ-GABAARs than in γ2-GABAARs. In comparison, AA29504 positively modulated the activity of recombinant δ-GABAARs more effectively than γ2-GABAARs, with no significant differences in potency. The impact of AA29504's efficacy- and potency-associated GABAAR subtype selectivity on radioligand binding properties remain unexplored. Using [3H]4'-ethynyl-4-n-propylbicycloorthobenzoate ([3H]EBOB) binding assay, we found no difference in the modulatory potency of AA29504 on GABA- and THIP (4,5,6,7-tetrahydroisoxazolo[5,4-c]pyridin-3-ol)-induced responses between native forebrain GABAARs of wild type and δ knock-out mice. In recombinant receptors expressed in HEK293 cells, AA29504 showed higher efficacy on δ- than γ2-GABAARs in the GABA-independent displacement of [3H]EBOB binding. Interestingly, AA29504 showed a concentration-dependent stimulation of [3H]muscimol binding to γ2-GABAARs, which was absent in δ-GABAARs. This was explained by AA29504 shifting the low-affinity γ2-GABAAR towards a higher affinity desensitized state, thereby rising new sites capable of binding GABAAR agonists with low nanomolar affinity. Hence, the potential of AA29504 to act as a desensitization-modifying allosteric modulator of γ2-GABAARs deserves further investigation for its promising influence on shaping efficacy, duration and plasticity of GABAAR synaptic responses.
Collapse
Affiliation(s)
- Sylvia Sikstus
- Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, 20014, Turku, Finland
| | - Ali Y Benkherouf
- Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, 20014, Turku, Finland
| | - Sanna L Soini
- Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, 20014, Turku, Finland
| | - Mikko Uusi-Oukari
- Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, 20014, Turku, Finland.
| |
Collapse
|