1
|
Dashevskii D, Luginina A, Maslov I, Shevelyova M, Khorn P, Dmitrieva D, Kapranov I, Belousov A, Permyakov S, Cherezov V, Borshchevskiy V, Mishin A. Unlocking GPCR-ligand interactions: Measuring binding affinities with thermal shift assay. Protein Sci 2025; 34:e70120. [PMID: 40247825 PMCID: PMC12006757 DOI: 10.1002/pro.70120] [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: 12/25/2024] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 04/19/2025]
Abstract
G protein-coupled receptors (GPCRs) constitute the largest transmembrane protein superfamily, with over 800 representatives in the human genome. Recognized as pivotal targets in pharmacological research and drug discovery, these receptors play a crucial role in advancing therapeutics. Understanding the molecular mechanisms of receptor-ligand interactions is imperative for drug discovery applications. However, experimental procedures for measuring ligand binding are complicated by various factors, including the transmembrane nature of the receptors and the high cost associated with specialized instruments and consumables. Here we introduce an application of the thermal shift assay (TSA) to measuring ligand binding affinities for GPCRs. TSA is a cost-effective and user-friendly method that detects changes in protein stability induced by alterations in environmental conditions. Employing the human A2A adenosine receptor as a representative GPCR, we determined binding constants for four orthosteric ligands and allosteric sodium using three mathematical models for TSA data approximation and analysis. Models were additionally validated by two antagonists of cysteinyl leukotriene GPCR (CysLT1R), used as antiasthmatic drugs. Our results suggest that the TSA approach demonstrates a high degree of reproducibility and agreement with existing literature data, thereby affirming its suitability for investigating GPCR interactions with various types of ligands.
Collapse
Affiliation(s)
- Dmitrii Dashevskii
- Research Center for Molecular Mechanisms of Aging and Age‐Related DiseasesMoscow Institute of Physics and TechnologyMoscowRussia
| | - Aleksandra Luginina
- Research Center for Molecular Mechanisms of Aging and Age‐Related DiseasesMoscow Institute of Physics and TechnologyMoscowRussia
| | - Ivan Maslov
- Research Center for Molecular Mechanisms of Aging and Age‐Related DiseasesMoscow Institute of Physics and TechnologyMoscowRussia
| | - Marina Shevelyova
- Pushchino Scientific Center for Biological Research of the Russian Academy of ScienceInstitute for Biological InstrumentationPushchinoRussia
| | - Polina Khorn
- Research Center for Molecular Mechanisms of Aging and Age‐Related DiseasesMoscow Institute of Physics and TechnologyMoscowRussia
| | - Daria Dmitrieva
- Research Center for Molecular Mechanisms of Aging and Age‐Related DiseasesMoscow Institute of Physics and TechnologyMoscowRussia
| | - Ivan Kapranov
- Research Center for Molecular Mechanisms of Aging and Age‐Related DiseasesMoscow Institute of Physics and TechnologyMoscowRussia
| | - Anatolii Belousov
- Research Center for Molecular Mechanisms of Aging and Age‐Related DiseasesMoscow Institute of Physics and TechnologyMoscowRussia
- Sao Carlos Institute of PhysicsUniversity of Sao PauloSao CarlosSão PaoloBrazil
| | - Sergei Permyakov
- Pushchino Scientific Center for Biological Research of the Russian Academy of ScienceInstitute for Biological InstrumentationPushchinoRussia
| | - Vadim Cherezov
- Bridge Institute, Department of ChemistryUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Valentin Borshchevskiy
- Research Center for Molecular Mechanisms of Aging and Age‐Related DiseasesMoscow Institute of Physics and TechnologyMoscowRussia
- Frank Laboratory of Neutron PhysicsDubnaRussia
| | - Alexey Mishin
- Research Center for Molecular Mechanisms of Aging and Age‐Related DiseasesMoscow Institute of Physics and TechnologyMoscowRussia
| |
Collapse
|
2
|
Hassan HM, El Safadi M, Hayat MF, Al-Emam A. Prevention of fenitrothion induced hepatic toxicity by saponarin via modulating TLR4/MYD88, JAK1/STAT3 and NF-κB signaling pathways. Int J Biochem Cell Biol 2025; 179:106716. [PMID: 39645143 DOI: 10.1016/j.biocel.2024.106716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 11/27/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
Fenitrothion (FEN) is an organophosphate insecticidal agent that is considered as major source of organs toxicity. Saponarin (SAP) is a naturally occurring novel flavone that exhibits a wide range of medicinal properties. The current trial was conducted to evaluate the ameliorative potential of SAP against FEN instigated liver toxicity in rats. Thirty-two male albino rats were apportioned into four groups including control, FEN (10 mg/kg), FEN (10 mg/kg) + SAP (80 mg/kg), and SAP (80 mg/kg) alone treated group. It was revealed that FEN administration upregulated the gene expression of TNF-α, TLR4, IL-1β, MYD88, IL-6, TRAF6, COX-2, NF-κB, JAK1 and STAT3 while reducing the gene expression of IκB. Moreover, the levels of reactive oxygen species (ROS) and malondialdehyde (MDA) were increased while the activities of catalase (CAT), glutathione peroxidase (GPx), superoxide dismutase (SOD), heme-oxygenase-1 (HO-1) and glutathione reductase (GSR) were decreased after FEN exposure. Furthermore, FEN administration notably escalated the levels of hepatic enzymes including alanine transaminase (ALT), aspartate aminotransferase (AST), gamma-glutamyl transferase (GGT) and alkaline phosphatase (ALP) whereas reduced the levels of total proteins and albumin. Besides, FEN intake upregulated the levels of Caspase-9, Bax and Caspase-3 while reducing the levels of Bcl-2. Hepatic histology was impaired after FEN intoxication. Nonetheless, SAP treatment remarkably protected the normal state of liver via regulating abovementioned irregularities. Our in-silico analysis confirmed that SAP hold that potential to interact with binding pocket of these proteins, highlighting its ability as a therapeutic compound to alleviate FEN-induced liver damage.
Collapse
Affiliation(s)
- Hesham M Hassan
- Department of pathology, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Mahmoud El Safadi
- Department of Chemistry, College of Science, United Arab Emirates University, P.O. Box 15551, Al Ain, Abu Dhabi , United Arab Emirates
| | - Muhammad Faisal Hayat
- Department of Zoology, Wildlife and Fisheries, University of Agriculture, Faisalabad, Pakistan.
| | - Ahmed Al-Emam
- Department of Pathology, College of Medicine, King Khalid University, Asir 61421, Saudi Arabia; Department of Forensic Medicine and Clinical Toxicology, Mansoura University, Egypt
| |
Collapse
|
3
|
Miguel-Ruano V, Acebrón I, Lee M, Martín-Galiano AJ, Freton C, de José UP, Ramachandran B, Gago F, Kjos M, Hesek D, Grangeasse C, Håvarstein LS, Straume D, Mobashery S, Hermoso JA. Characterization of VldE (Spr1875), a Pneumococcal Two-State l,d-Endopeptidase with a Four-Zinc Cluster in the Active Site. ACS Catal 2024; 14:18786-18798. [PMID: 39722888 PMCID: PMC11667670 DOI: 10.1021/acscatal.4c05090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 12/28/2024]
Abstract
Remodeling of the pneumococcal cell wall, carried out by peptidoglycan (PG) hydrolases, is imperative for maintaining bacterial cell shape and ensuring survival, particularly during cell division or stress response. The Streptococcus pneumoniae protein Spr1875 plays a role in stress response, both regulated by the VicRK two-component system (analogous to the WalRK TCS found in Firmicutes). Modular Spr1875 presents a putative cell-wall binding module at the N-terminus and a catalytic C-terminal module (Spr1875MT3) connected by a long linker. Assays of the full-length protein and Spr1875MT3 with PG-based synthetic substrates by liquid chromatography/mass spectrometry revealed Spr1875 as an l,d-endopeptidase, renamed VldE (for VicRK-regulated l,d-endopeptidase), which hydrolyzed the cross-linked stem peptide in the PG. Remarkably, we observed asymmetric turnover with specific recognition of the acceptor peptide strand. Localization experiments showed that the protein is directed to the septum, which suggests that muralytic activity could be required for pneumococcal growth under stress conditions. Our findings, based on six high-resolution X-ray crystallographic structures and molecular-dynamics simulations, reveal two states for VldEMT3. The protein transitions between a noncatalytic state that binds up to four zinc ions, thus behaving as a Zn2+ reservoir, and a catalytic state that performs the hydrolytic reaction with a single zinc ion. Furthermore, computational studies provide insight into the mechanism of catalytic-water activation and nucleophilic attack on the specific scissile peptide bond of the asymmetric cross-linked PG.
Collapse
Affiliation(s)
- Vega Miguel-Ruano
- Department
of Crystallography and Structural Biology, Consejo Superior de Investigaciones
Científicas, Instituto de Química-Física
“Blas Cabrera”, Madrid 28006, Spain
| | - Iván Acebrón
- Department
of Crystallography and Structural Biology, Consejo Superior de Investigaciones
Científicas, Instituto de Química-Física
“Blas Cabrera”, Madrid 28006, Spain
| | - Mijoon Lee
- Department
of Chemistry and Biochemistry, University
of Notre Dame, Notre
Dame, Indiana 46556, United States
| | | | - Celine Freton
- Molecular
Microbiology and Structural Biochemistry, CNRS UMR, Université de Lyon, Lyon 69367, France
| | - Uxía P. de José
- Department
of Crystallography and Structural Biology, Consejo Superior de Investigaciones
Científicas, Instituto de Química-Física
“Blas Cabrera”, Madrid 28006, Spain
| | - Balajee Ramachandran
- Department
of Chemistry and Biochemistry, University
of Notre Dame, Notre
Dame, Indiana 46556, United States
| | - Federico Gago
- Department
of Biomedical Sciences and IQM-CSIC Associate Unit, School of Medicine
and Health Sciences, University of Alcalá, Alcalá de Henares 28805, Spain
| | - Morten Kjos
- Department
of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås 1430, Norway
| | - Dusan Hesek
- Department
of Chemistry and Biochemistry, University
of Notre Dame, Notre
Dame, Indiana 46556, United States
| | - Christophe Grangeasse
- Molecular
Microbiology and Structural Biochemistry, CNRS UMR, Université de Lyon, Lyon 69367, France
| | - Leiv Sigve Håvarstein
- Department
of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås 1430, Norway
| | - Daniel Straume
- Department
of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås 1430, Norway
| | - Shahriar Mobashery
- Department
of Chemistry and Biochemistry, University
of Notre Dame, Notre
Dame, Indiana 46556, United States
| | - Juan A. Hermoso
- Department
of Crystallography and Structural Biology, Consejo Superior de Investigaciones
Científicas, Instituto de Química-Física
“Blas Cabrera”, Madrid 28006, Spain
| |
Collapse
|
4
|
Li X, Zhang J, Xiao Y, Song H, Li Y, Li W, Cao R, Li S, Qin Y, Wang C, Zhong W. Chemoproteomics enables identification of coatomer subunit zeta-1 targeted by a small molecule for enterovirus A71 inhibition. MedComm (Beijing) 2024; 5:e587. [PMID: 38840773 PMCID: PMC11151152 DOI: 10.1002/mco2.587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 05/02/2024] [Accepted: 05/06/2024] [Indexed: 06/07/2024] Open
Abstract
Human enterovirus A71 (EV-A71) is a significant etiological agent responsible for epidemics of hand, foot, and mouth disease (HFMD) in Asia-Pacific regions. There are presently no licensed antivirals against EV-A71, and the druggable target for EV-A71 remains very limited. The phenotypic hit 10,10'-bis(trifluoromethyl) marinopyrrole A derivative, herein termed MPA-CF3, is a novel potent small-molecule inhibitor against EV-A71, but its pharmacological target(s) and antiviral mechanisms are not defined. Here, quantitative chemoproteomics deciphered the antiviral target of MAP-CF3 as host factor coatomer subunit zeta-1 (COPZ1). Mechanistically, MPA-CF3 disrupts the interaction of COPZ1 with the EV-A71 nonstructural protein 2C by destabilizing COPZ1 upon binding. The destruction of this interaction blocks the coatomer-mediated transport of 2C to endoplasmic reticulum, and ultimately inhibits EV-A71 replication. Taken together, our study disclosed that MPA-CF3 can be a structurally novel host-targeting anti-EV-A71 agent, providing a structural basis for developing the COPZ1-targeting broad-spectrum antivirals against enteroviruses. The mechanistic elucidation of MPA-CF3 against EV-A71 may offer an alternative COPZ1-involved therapeutic pathway for enterovirus infection.
Collapse
Affiliation(s)
- Xiaoyong Li
- Key Laboratory of Drug‐Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant‐Sourced Drug, and Sichuan Research Center for Drug Precision Industrial Technology, West China School of PharmacySichuan UniversityChengduChina
- National Engineering Research Center for the Emergence DrugsBeijing Institute of Pharmacology and ToxicologyBeijingChina
| | - Jin Zhang
- College of Chemistry and Molecular EngineeringPeking UniversityBeijingChina
| | - Yaxin Xiao
- Key Laboratory of Drug‐Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant‐Sourced Drug, and Sichuan Research Center for Drug Precision Industrial Technology, West China School of PharmacySichuan UniversityChengduChina
| | - Hao Song
- Key Laboratory of Drug‐Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant‐Sourced Drug, and Sichuan Research Center for Drug Precision Industrial Technology, West China School of PharmacySichuan UniversityChengduChina
| | - Yuexiang Li
- National Engineering Research Center for the Emergence DrugsBeijing Institute of Pharmacology and ToxicologyBeijingChina
| | - Wei Li
- National Engineering Research Center for the Emergence DrugsBeijing Institute of Pharmacology and ToxicologyBeijingChina
| | - Ruiyuan Cao
- National Engineering Research Center for the Emergence DrugsBeijing Institute of Pharmacology and ToxicologyBeijingChina
| | - Song Li
- National Engineering Research Center for the Emergence DrugsBeijing Institute of Pharmacology and ToxicologyBeijingChina
| | - Yong Qin
- Key Laboratory of Drug‐Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant‐Sourced Drug, and Sichuan Research Center for Drug Precision Industrial Technology, West China School of PharmacySichuan UniversityChengduChina
| | - Chu Wang
- College of Chemistry and Molecular EngineeringPeking UniversityBeijingChina
| | - Wu Zhong
- National Engineering Research Center for the Emergence DrugsBeijing Institute of Pharmacology and ToxicologyBeijingChina
| |
Collapse
|
5
|
Strobel HM, Labador SD, Basu D, Sane M, Corbett KD, Meyer JR. Viral Receptor-Binding Protein Evolves New Function through Mutations That Cause Trimer Instability and Functional Heterogeneity. Mol Biol Evol 2024; 41:msae056. [PMID: 38586942 PMCID: PMC10999833 DOI: 10.1093/molbev/msae056] [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: 02/14/2023] [Revised: 02/07/2024] [Accepted: 03/11/2024] [Indexed: 04/09/2024] Open
Abstract
When proteins evolve new activity, a concomitant decrease in stability is often observed because the mutations that confer new activity can destabilize the native fold. In the conventional model of protein evolution, reduced stability is considered a purely deleterious cost of molecular innovation because unstable proteins are prone to aggregation and are sensitive to environmental stressors. However, recent work has revealed that nonnative, often unstable protein conformations play an important role in mediating evolutionary transitions, raising the question of whether instability can itself potentiate the evolution of new activity. We explored this question in a bacteriophage receptor-binding protein during host-range evolution. We studied the properties of the receptor-binding protein of bacteriophage λ before and after host-range evolution and demonstrated that the evolved protein is relatively unstable and may exist in multiple conformations with unique receptor preferences. Through a combination of structural modeling and in vitro oligomeric state analysis, we found that the instability arises from mutations that interfere with trimer formation. This study raises the intriguing possibility that protein instability might play a previously unrecognized role in mediating host-range expansions in viruses.
Collapse
Affiliation(s)
- Hannah M Strobel
- School of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Sweetzel D Labador
- School of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Dwaipayan Basu
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA, USA
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Mrudula Sane
- School of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Kevin D Corbett
- School of Biological Sciences, University of California San Diego, La Jolla, CA, USA
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Justin R Meyer
- School of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
6
|
Wang J, Song Y, Huang Z, Lin W, Yu G, Xiong Y, Jiang G, Tan Y, Wang J, Liao X. Coupling a Virulence-Targeting Moiety with Ru-Based AMP Mimics Efficiently Improved Its Anti-Infective Potency and Therapeutic Index. J Med Chem 2023; 66:13304-13318. [PMID: 37704628 DOI: 10.1021/acs.jmedchem.3c01282] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
The surge of antibiotic resistance in Staphylococcus aureus calls for novel drugs that attack new targets. Developing antimicrobial peptides (AMPs) or antivirulence agents (AvAs) is a promising strategy to tackle this challenge. However, AMPs, which kill bacteria by disrupting cell membranes, suffer from low stability and high synthesis cost, while AvAs, which inhibit toxin secretion, have relatively poor bactericidal activity. Here, to address their respective shortcomings, we combined these two different antibacterial activities on the same molecular scaffold and developed a Ru-based metalloantibiotic, termed Ru1. Notably, Ru1 exerted remarkable bactericidal activity (MICS = 460 nM) and attenuated bacterial virulence as well. Mechanistic studies demonstrated that Ru1 had two independent targets: CcpA and bacterial membrane integrity. Based on its dual mechanism of action, Ru1 effectively overcame S. aureus resistance and showed high efficacy in a mouse infection model against S. aureus. This study provides a promising approach to confronting bacterial infections.
Collapse
Affiliation(s)
- Jing Wang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Yun Song
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Ziying Huang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Wenjing Lin
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Guangying Yu
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Yanshi Xiong
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Guijuan Jiang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Yanhui Tan
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Jintao Wang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Xiangwen Liao
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| |
Collapse
|
7
|
Ford A, Breitgoff F, Pasquini M, MacKenzie A, McElroy S, Baker S, Abrusci P, Varzandeh S, Bird L, Gavard A, Damerell D, Redhead M. Application of particle swarm optimization to understand the mechanism of action of allosteric inhibitors of the enzyme HSD17β13. PATTERNS (NEW YORK, N.Y.) 2023; 4:100733. [PMID: 37223265 PMCID: PMC10201303 DOI: 10.1016/j.patter.2023.100733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 09/06/2022] [Accepted: 03/24/2023] [Indexed: 05/25/2023]
Abstract
Understanding a drug candidate's mechanism of action is crucial for its further development. However, kinetic schemes are often complex and multi-parametric, especially for proteins in oligomerization equilibria. Here, we demonstrate the use of particle swarm optimization (PSO) as a method to select between different sets of parameters that are too far apart in the parameter space to be found by conventional approaches. PSO is based upon the swarming of birds: each bird in the flock assesses multiple landing spots while at the same time sharing that information with its neighbors. We applied this approach to the kinetics of HSD17β13 enzyme inhibitors, which displayed unusually large thermal shifts. Thermal shift data for HSD17β13 indicated that the inhibitor shifted the oligomerization equilibrium toward the dimeric state. Validation of the PSO approach was provided by experimental mass photometry data. These results encourage further exploration of multi-parameter optimization algorithms as tools in drug discovery.
Collapse
Affiliation(s)
- Amy Ford
- Exscientia, The Schrödinger Building, Oxford Science Park, Oxford OX4 4GE, UK
| | - Frauke Breitgoff
- Exscientia, The Schrödinger Building, Oxford Science Park, Oxford OX4 4GE, UK
| | - Miriam Pasquini
- Exscientia, The Schrödinger Building, Oxford Science Park, Oxford OX4 4GE, UK
| | | | - Stuart McElroy
- Bioascent, Bo'Ness Road, Chapelhall, Motherwell ML1 5SH, UK
| | - Steve Baker
- Exscientia, The Schrödinger Building, Oxford Science Park, Oxford OX4 4GE, UK
| | - Patrizia Abrusci
- Exscientia, The Schrödinger Building, Oxford Science Park, Oxford OX4 4GE, UK
| | - Simon Varzandeh
- Exscientia, The Schrödinger Building, Oxford Science Park, Oxford OX4 4GE, UK
| | - Louise Bird
- Exscientia, The Schrödinger Building, Oxford Science Park, Oxford OX4 4GE, UK
| | - Angeline Gavard
- Exscientia, The Schrödinger Building, Oxford Science Park, Oxford OX4 4GE, UK
| | - David Damerell
- Exscientia, The Schrödinger Building, Oxford Science Park, Oxford OX4 4GE, UK
| | - Martin Redhead
- Exscientia, The Schrödinger Building, Oxford Science Park, Oxford OX4 4GE, UK
| |
Collapse
|
8
|
Umegawa Y, Kawatake S, Murata M, Matsuoka S. Combined effect of the head groups and alkyl chains of archaea lipids when interacting with bacteriorhodopsin. Biophys Chem 2023; 294:106959. [PMID: 36709544 DOI: 10.1016/j.bpc.2023.106959] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/12/2023] [Accepted: 01/16/2023] [Indexed: 01/21/2023]
Abstract
Bacteriorhodopsin (bR), a transmembrane protein with seven α-helices, is highly expressed in the purple membrane (PM) of archaea such as Halobacterium salinarum. It is well known that bR forms two-dimensional crystals with acidic lipids such as phosphatidylglycerol phosphate methyl ester (PGP-Me)-a major component of PM lipids bearing unique chemical structures-methyl-branched alkyl chains, ether linkages, and divalent anionic head groups with two phosphodiester groups. Therefore, we aimed to determine which functional groups of PGP-Me are essential for the boundary lipids of bR and how these functionalities interact with bR. To this end, we compared various well-known phospholipids (PLs) that carry one of the structural features of PGP-Me, and evaluated the affinity of PLs to bR using the centerband-only analysis of rotor-unsynchronized spin echo (COARSE) method in solid-state NMR measurements and thermal shift assays. The results clearly showed that the branched methyl groups of alkyl chains and double negative charges in the head groups are important for PL interactions with bR. We then examined the effect of phospholipids on the monomer-trimer exchange of bR using circular dichroism (CD) spectra. The results indicated that the divalent negative charge in a head group stabilizes the trimer structure, while the branched methyl chains significantly enhance the PLs' affinity for bR, thus dispersing bR trimers in the PM even at high concentrations. Finally, we investigated the effects of PL on the proton-pumping activity of bR based on the decay rate constant of the M intermediate of a bR photocycle. The findings showed that bR activities decreased to 20% in 1,2-dimyristoyl-sn-glycero-3-phosphate (DMPA), and in 1,2-diphytanoyl-sn-glycero-3-phosphocholine (DPhPC) bilayers as compared to that in PM. Meanwhile, 1,2-Diphytanoyl-sn-glycero-3-phosphate (DPhPA) bilayers bearing both negative charges and branched methyl groups preserved over 80% of the activity. These results strongly suggest that the head groups and alkyl chains of phospholipids are essential for boundary lipids and greatly influence the biological function of bR.
Collapse
Affiliation(s)
- Yuichi Umegawa
- JST ERATO, Lipid Active Structure Project, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan; Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan; Forefront Research Center, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan.
| | - Satoshi Kawatake
- JST ERATO, Lipid Active Structure Project, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan; Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan; Forefront Research Center, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | - Michio Murata
- JST ERATO, Lipid Active Structure Project, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan; Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | - Shigeru Matsuoka
- JST ERATO, Lipid Active Structure Project, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan; Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan; Forefront Research Center, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| |
Collapse
|
9
|
Zakšauskas A, Čapkauskaitė E, Paketurytė-Latvė V, Smirnov A, Leitans J, Kazaks A, Dvinskis E, Stančaitis L, Mickevičiūtė A, Jachno J, Jezepčikas L, Linkuvienė V, Sakalauskas A, Manakova E, Gražulis S, Matulienė J, Tars K, Matulis D. Methyl 2-Halo-4-Substituted-5-Sulfamoyl-Benzoates as High Affinity and Selective Inhibitors of Carbonic Anhydrase IX. Int J Mol Sci 2021; 23:130. [PMID: 35008553 PMCID: PMC8745178 DOI: 10.3390/ijms23010130] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/14/2021] [Accepted: 12/18/2021] [Indexed: 01/01/2023] Open
Abstract
Among the twelve catalytically active carbonic anhydrase isozymes present in the human body, the CAIX is highly overexpressed in various solid tumors. The enzyme acidifies the tumor microenvironment enabling invasion and metastatic processes. Therefore, many attempts have been made to design chemical compounds that would exhibit high affinity and selective binding to CAIX over the remaining eleven catalytically active CA isozymes to limit undesired side effects. It has been postulated that such drugs may have anticancer properties and could be used in tumor treatment. Here we have designed a series of compounds, methyl 5-sulfamoyl-benzoates, which bear a primary sulfonamide group, a well-known marker of CA inhibitors, and determined their affinities for all twelve CA isozymes. Variations of substituents on the benzenesulfonamide ring led to compound 4b, which exhibited an extremely high observed binding affinity to CAIX; the Kd was 0.12 nM. The intrinsic dissociation constant, where the binding-linked protonation reactions have been subtracted, reached 0.08 pM. The compound also exhibited more than 100-fold selectivity over the remaining CA isozymes. The X-ray crystallographic structure of compound 3b bound to CAIX showed the structural position, while several structures of compounds bound to other CA isozymes showed structural reasons for compound selectivity towards CAIX. Since this series of compounds possess physicochemical properties suitable for drugs, they may be developed for anticancer therapeutic purposes.
Collapse
Affiliation(s)
- Audrius Zakšauskas
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio al. 7, LT-10257 Vilnius, Lithuania; (A.Z.); (E.Č.); (V.P.-L.); (A.S.); (L.S.); (A.M.); (J.J.); (L.J.); (V.L.); (A.S.); (J.M.)
| | - Edita Čapkauskaitė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio al. 7, LT-10257 Vilnius, Lithuania; (A.Z.); (E.Č.); (V.P.-L.); (A.S.); (L.S.); (A.M.); (J.J.); (L.J.); (V.L.); (A.S.); (J.M.)
| | - Vaida Paketurytė-Latvė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio al. 7, LT-10257 Vilnius, Lithuania; (A.Z.); (E.Č.); (V.P.-L.); (A.S.); (L.S.); (A.M.); (J.J.); (L.J.); (V.L.); (A.S.); (J.M.)
| | - Alexey Smirnov
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio al. 7, LT-10257 Vilnius, Lithuania; (A.Z.); (E.Č.); (V.P.-L.); (A.S.); (L.S.); (A.M.); (J.J.); (L.J.); (V.L.); (A.S.); (J.M.)
| | - Janis Leitans
- Latvian Biomedical Research and Study Centre, Ratsupites 1 k-1, LV-1067 Riga, Latvia; (J.L.); (A.K.); (E.D.); (K.T.)
| | - Andris Kazaks
- Latvian Biomedical Research and Study Centre, Ratsupites 1 k-1, LV-1067 Riga, Latvia; (J.L.); (A.K.); (E.D.); (K.T.)
| | - Elviss Dvinskis
- Latvian Biomedical Research and Study Centre, Ratsupites 1 k-1, LV-1067 Riga, Latvia; (J.L.); (A.K.); (E.D.); (K.T.)
| | - Laimonas Stančaitis
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio al. 7, LT-10257 Vilnius, Lithuania; (A.Z.); (E.Č.); (V.P.-L.); (A.S.); (L.S.); (A.M.); (J.J.); (L.J.); (V.L.); (A.S.); (J.M.)
| | - Aurelija Mickevičiūtė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio al. 7, LT-10257 Vilnius, Lithuania; (A.Z.); (E.Č.); (V.P.-L.); (A.S.); (L.S.); (A.M.); (J.J.); (L.J.); (V.L.); (A.S.); (J.M.)
| | - Jelena Jachno
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio al. 7, LT-10257 Vilnius, Lithuania; (A.Z.); (E.Č.); (V.P.-L.); (A.S.); (L.S.); (A.M.); (J.J.); (L.J.); (V.L.); (A.S.); (J.M.)
| | - Linas Jezepčikas
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio al. 7, LT-10257 Vilnius, Lithuania; (A.Z.); (E.Č.); (V.P.-L.); (A.S.); (L.S.); (A.M.); (J.J.); (L.J.); (V.L.); (A.S.); (J.M.)
| | - Vaida Linkuvienė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio al. 7, LT-10257 Vilnius, Lithuania; (A.Z.); (E.Č.); (V.P.-L.); (A.S.); (L.S.); (A.M.); (J.J.); (L.J.); (V.L.); (A.S.); (J.M.)
| | - Andrius Sakalauskas
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio al. 7, LT-10257 Vilnius, Lithuania; (A.Z.); (E.Č.); (V.P.-L.); (A.S.); (L.S.); (A.M.); (J.J.); (L.J.); (V.L.); (A.S.); (J.M.)
| | - Elena Manakova
- Department of Protein—DNA Interactions, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio al. 7, LT-10257 Vilnius, Lithuania; (E.M.); (S.G.)
| | - Saulius Gražulis
- Department of Protein—DNA Interactions, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio al. 7, LT-10257 Vilnius, Lithuania; (E.M.); (S.G.)
| | - Jurgita Matulienė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio al. 7, LT-10257 Vilnius, Lithuania; (A.Z.); (E.Č.); (V.P.-L.); (A.S.); (L.S.); (A.M.); (J.J.); (L.J.); (V.L.); (A.S.); (J.M.)
| | - Kaspars Tars
- Latvian Biomedical Research and Study Centre, Ratsupites 1 k-1, LV-1067 Riga, Latvia; (J.L.); (A.K.); (E.D.); (K.T.)
| | - Daumantas Matulis
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio al. 7, LT-10257 Vilnius, Lithuania; (A.Z.); (E.Č.); (V.P.-L.); (A.S.); (L.S.); (A.M.); (J.J.); (L.J.); (V.L.); (A.S.); (J.M.)
| |
Collapse
|
10
|
Baranauskiene L, Škiudaitė L, Michailovienė V, Petrauskas V, Matulis D. Thiazide and other Cl-benzenesulfonamide-bearing clinical drug affinities for human carbonic anhydrases. PLoS One 2021; 16:e0253608. [PMID: 34166457 PMCID: PMC8224972 DOI: 10.1371/journal.pone.0253608] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 06/08/2021] [Indexed: 11/27/2022] Open
Abstract
Twelve carbonic anhydrase (CA) isoforms catalyze carbon dioxide hydration to bicarbonate and acid protons and are responsible for many biological functions in human body. Despite their vital functions, they are also responsible for, or implicated in, numerous ailments and diseases such as glaucoma, high altitude sickness, and cancer. Because CA isoforms are highly homologous, clinical drugs designed to inhibit enzymatic activity of a particular isoform, can also bind to others with similar affinity causing toxic side effects. In this study, the affinities of twelve CA isoforms have been determined for nineteen clinically used drugs used to treat hypertension related diseases, i.e. thiazides, indapamide, and metolazone. Their affinities were determined using a fluorescent thermal shift assay. Stopped flow assay and isothermal titration calorimetry were also employed on a subset of compounds and proteins to confirm inhibition of CA enzymatic activity and verify the quantitative agreement between different assays. The findings of this study showed that pharmaceuticals could bind to human CA isoforms with variable affinities and inhibit their catalytic activity, even though the drug was intended to interact with a different (non-CA) protein target. Relatively minor structural changes of the compounds may cause significant changes in affinity and selectivity for a particular CA isoform.
Collapse
Affiliation(s)
- Lina Baranauskiene
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Lina Škiudaitė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
- Pharmacy Center, Institute of Biomedical Science, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Vilma Michailovienė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Vytautas Petrauskas
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Daumantas Matulis
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
11
|
England P, Jowitt TA. Community-building and promotion of technological excellence in molecular biophysics: the ARBRE-MOBIEU network. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2021; 50:307-311. [PMID: 34057541 DOI: 10.1007/s00249-021-01550-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Patrick England
- Molecular Biophysics Facility, Institut Pasteur, 25-28 rue du Docteur Roux, 75724, Paris cedex 15, France.
| | - Thomas A Jowitt
- Biomolecular Analysis Core Facility, Faculty of Biology Medicine and Health, University of Manchester, Oxford Road, Manchester, M13 9PT, UK.
| |
Collapse
|