1
|
Gilles N. Natural Peptide Toxins as an Option for Renewed Treatment of Type 2 Vasopressin Receptor-Related Diseases. BIOLOGY 2023; 12:544. [PMID: 37106745 PMCID: PMC10136000 DOI: 10.3390/biology12040544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 03/31/2023] [Accepted: 03/31/2023] [Indexed: 04/07/2023]
Abstract
The type 2 vasopressin receptor (V2R) is expressed in the kidneys, and it is the keystone of water homeostasis. Under the control of the antidiuretic hormone vasopressin, the V2R ensures vital functions, and any disturbance has dramatic consequences. Despite decades of research to develop drugs capable of activating or blocking V2R function to meet real medical needs, only one agonist and one antagonist are virtually used today. These two drugs cover only a small portion of patients' needs, leaving millions of patients without treatment. Natural peptide toxins known to act selectively and at low doses on their receptor target could offer new therapeutic options.
Collapse
Affiliation(s)
- Nicolas Gilles
- CEA, SIMoS, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris-Saclay, 91191 Gif-sur-Yvette, France
| |
Collapse
|
2
|
Papageorgiou L, Christou E, Louka E, Papakonstantinou E, Diakou I, Pierouli K, Dragoumani K, Bacopoulou F, Chrousos GP, Eliopoulos E, Vlachakis D. ADRA2B and HTR1A: An Updated Study of the Biogenic Amine Receptors Reveals Novel Conserved Motifs Which Play Key Role in Mental Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1423:79-99. [PMID: 37525034 DOI: 10.1007/978-3-031-31978-5_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Abstract
Mental disorders are strongly connected with several psychiatric conditions including depression, bipolar disorder, schizophrenia, eating disorder, and suicides. There are many biological conditions and pathways that define these complicated illnesses. For example, eating disorders are complex mental health conditions that require the intervention of geneticists, psychiatrists, and medical experts in order to alleviate their symptoms. A patient with suicidal ideation should first be identified and consequently monitored by a similar team of specialists. Both genetics and epigenetics can shed light on eating disorders and suicides as they are found in the main core of such investigations. In the present study, an analysis has been performed on two specific members of the GPCR family toward drawing conclusions regarding their functionality and implementation in mental disorders. Specifically, evolutionary and structural studies on the adrenoceptor alpha 2b (ADRA2B) and the 5-hydroxytryptamine receptor 1A (HTR1A) have been carried out. Both receptors are classified in the biogenic amine receptors sub-cluster of the GPCRs and have been connected in many studies with mental diseases and malnutrition conditions. The major goal of this study is the investigation of conserved motifs among biogenic amine receptors that play an important role in this family signaling pathway, through an updated evolutionary analysis and the correlation of this information with the structural features of the HTR1A and ADRA2B. Furthermore, the structural comparison of ADRA2B, HTR1A, and other members of GPCRs related to mental disorders is performed.
Collapse
Affiliation(s)
- Louis Papageorgiou
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, Athens, Greece
| | - Evangelia Christou
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, Athens, Greece
| | - Effrosyni Louka
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, Athens, Greece
| | - Eleni Papakonstantinou
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, Athens, Greece
| | - Io Diakou
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, Athens, Greece
| | - Katerina Pierouli
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, Athens, Greece
| | - Konstantina Dragoumani
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, Athens, Greece
| | - Flora Bacopoulou
- University Research Institute of Maternal and Child Health & Precision Medicine, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - George P Chrousos
- University Research Institute of Maternal and Child Health & Precision Medicine, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Elias Eliopoulos
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, Athens, Greece
| | - Dimitrios Vlachakis
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, Athens, Greece.
- University Research Institute of Maternal and Child Health & Precision Medicine, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens, Greece.
- Division of Endocrinology and Metabolism, Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| |
Collapse
|
3
|
Using filters in virtual screening: A comprehensive guide to minimize errors and maximize efficiency. ANNUAL REPORTS IN MEDICINAL CHEMISTRY 2022. [DOI: 10.1016/bs.armc.2022.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
4
|
Chen J, Campbell AP, Wakelin LPG, Finch AM. Characterisation of bis(4-aminoquinoline)s as α 1A adrenoceptor allosteric modulators. Eur J Pharmacol 2021; 916:174659. [PMID: 34871559 DOI: 10.1016/j.ejphar.2021.174659] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 11/19/2021] [Accepted: 11/29/2021] [Indexed: 11/30/2022]
Abstract
The development of sub-type selective α1 adrenoceptor ligands has been hampered by the high sequence similarity of the amino acids forming the orthosteric binding pocket of the three α1 adrenoceptor subtypes, along with other biogenic amine receptors. One possible approach to overcome this issue is to target allosteric sites on the α1 adrenoceptors. Previous docking studies suggested that one of the quinoline moieties of a bis(4-aminoquinoline), comprising a 9-carbon methylene linker attached via the amine groups, could interact with residues outside of the orthosteric binding site while, simultaneously, the other quinoline moiety bound within the orthosteric site. We therefore hypothesized that this compound could act in a bitopic manner, displaying both orthosteric and allosteric binding properties. To test this proposition, we investigated the allosteric activity of a series of bis(4-aminoquinoline)s with linker lengths ranging from 2 to 12 methylene units (designated C2-C12). A linear trend of increasing [3H]prazosin dissociation rate with increasing linker length between C7 and C11 was observed, confirming their action as allosteric modulators. These data suggest that the optimal linker length for the bis(4-aminoquinoline)s to occupy the allosteric site of the α1A adrenoceptor is between 7 and 11 methylene units. In addition, the ability of C9 bis(4-aminoquinoline) to modulate the activation of the α1A adrenoceptor by norepinephrine was subsequently examined, showing that C9 acted as a non-competitive antagonist. Our findings indicate that the bis(4-aminoquinolines) are acting as allosteric modulators of orthosteric ligand binding, but not efficacy, in a bitopic manner.
Collapse
Affiliation(s)
- Junli Chen
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, Sydney, NSW, 2052, Australia
| | - Adrian P Campbell
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, Sydney, NSW, 2052, Australia
| | - Laurence P G Wakelin
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, Sydney, NSW, 2052, Australia
| | - Angela M Finch
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, Sydney, NSW, 2052, Australia.
| |
Collapse
|
5
|
Kumar S, Kim MH. SMPLIP-Score: predicting ligand binding affinity from simple and interpretable on-the-fly interaction fingerprint pattern descriptors. J Cheminform 2021; 13:28. [PMID: 33766140 PMCID: PMC7993508 DOI: 10.1186/s13321-021-00507-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 03/16/2021] [Indexed: 12/13/2022] Open
Abstract
In drug discovery, rapid and accurate prediction of protein–ligand binding affinities is a pivotal task for lead optimization with acceptable on-target potency as well as pharmacological efficacy. Furthermore, researchers hope for a high correlation between docking score and pose with key interactive residues, although scoring functions as free energy surrogates of protein–ligand complexes have failed to provide collinearity. Recently, various machine learning or deep learning methods have been proposed to overcome the drawbacks of scoring functions. Despite being highly accurate, their featurization process is complex and the meaning of the embedded features cannot directly be interpreted by human recognition without an additional feature analysis. Here, we propose SMPLIP-Score (Substructural Molecular and Protein–Ligand Interaction Pattern Score), a direct interpretable predictor of absolute binding affinity. Our simple featurization embeds the interaction fingerprint pattern on the ligand-binding site environment and molecular fragments of ligands into an input vectorized matrix for learning layers (random forest or deep neural network). Despite their less complex features than other state-of-the-art models, SMPLIP-Score achieved comparable performance, a Pearson’s correlation coefficient up to 0.80, and a root mean square error up to 1.18 in pK units with several benchmark datasets (PDBbind v.2015, Astex Diverse Set, CSAR NRC HiQ, FEP, PDBbind NMR, and CASF-2016). For this model, generality, predictive power, ranking power, and robustness were examined using direct interpretation of feature matrices for specific targets. ![]()
Collapse
Affiliation(s)
- Surendra Kumar
- Gachon Institute of Pharmaceutical Science & Department of Pharmacy, College of Pharmacy, Gachon University, 191 Hambakmoeiro, Yeonsu-gu, Incheon, Republic of Korea
| | - Mi-Hyun Kim
- Gachon Institute of Pharmaceutical Science & Department of Pharmacy, College of Pharmacy, Gachon University, 191 Hambakmoeiro, Yeonsu-gu, Incheon, Republic of Korea.
| |
Collapse
|
6
|
Chemogenomic Analysis of the Druggable Kinome and Its Application to Repositioning and Lead Identification Studies. Cell Chem Biol 2019; 26:1608-1622.e6. [DOI: 10.1016/j.chembiol.2019.08.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 07/18/2019] [Accepted: 08/21/2019] [Indexed: 02/06/2023]
|
7
|
Boyles F, Deane CM, Morris GM. Learning from the ligand: using ligand-based features to improve binding affinity prediction. Bioinformatics 2019; 36:758-764. [DOI: 10.1093/bioinformatics/btz665] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 08/14/2019] [Accepted: 08/21/2019] [Indexed: 12/27/2022] Open
Abstract
Abstract
Motivation
Machine learning scoring functions for protein–ligand binding affinity prediction have been found to consistently outperform classical scoring functions. Structure-based scoring functions for universal affinity prediction typically use features describing interactions derived from the protein–ligand complex, with limited information about the chemical or topological properties of the ligand itself.
Results
We demonstrate that the performance of machine learning scoring functions are consistently improved by the inclusion of diverse ligand-based features. For example, a Random Forest (RF) combining the features of RF-Score v3 with RDKit molecular descriptors achieved Pearson correlation coefficients of up to 0.836, 0.780 and 0.821 on the PDBbind 2007, 2013 and 2016 core sets, respectively, compared to 0.790, 0.746 and 0.814 when using the features of RF-Score v3 alone. Excluding proteins and/or ligands that are similar to those in the test sets from the training set has a significant effect on scoring function performance, but does not remove the predictive power of ligand-based features. Furthermore a RF using only ligand-based features is predictive at a level similar to classical scoring functions and it appears to be predicting the mean binding affinity of a ligand for its protein targets.
Availability and implementation
Data and code to reproduce all the results are freely available at http://opig.stats.ox.ac.uk/resources.
Supplementary information
Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Fergus Boyles
- Department of Statistics, University of Oxford, Oxford, UK
| | | | | |
Collapse
|
8
|
Ravikumar B, Alam Z, Peddinti G, Aittokallio T. C-SPADE: a web-tool for interactive analysis and visualization of drug screening experiments through compound-specific bioactivity dendrograms. Nucleic Acids Res 2019; 45:W495-W500. [PMID: 28472495 PMCID: PMC5570255 DOI: 10.1093/nar/gkx384] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 04/25/2017] [Indexed: 12/20/2022] Open
Abstract
The advent of polypharmacology paradigm in drug discovery calls for novel chemoinformatic tools for analyzing compounds’ multi-targeting activities. Such tools should provide an intuitive representation of the chemical space through capturing and visualizing underlying patterns of compound similarities linked to their polypharmacological effects. Most of the existing compound-centric chemoinformatics tools lack interactive options and user interfaces that are critical for the real-time needs of chemical biologists carrying out compound screening experiments. Toward that end, we introduce C-SPADE, an open-source exploratory web-tool for interactive analysis and visualization of drug profiling assays (biochemical, cell-based or cell-free) using compound-centric similarity clustering. C-SPADE allows the users to visually map the chemical diversity of a screening panel, explore investigational compounds in terms of their similarity to the screening panel, perform polypharmacological analyses and guide drug-target interaction predictions. C-SPADE requires only the raw drug profiling data as input, and it automatically retrieves the structural information and constructs the compound clusters in real-time, thereby reducing the time required for manual analysis in drug development or repurposing applications. The web-tool provides a customizable visual workspace that can either be downloaded as figure or Newick tree file or shared as a hyperlink with other users. C-SPADE is freely available at http://cspade.fimm.fi/.
Collapse
Affiliation(s)
- Balaguru Ravikumar
- Institute for Molecular Medicine Finland, FIMM, University of Helsinki, Helsinki, Finland
| | - Zaid Alam
- Institute for Molecular Medicine Finland, FIMM, University of Helsinki, Helsinki, Finland
| | - Gopal Peddinti
- Institute for Molecular Medicine Finland, FIMM, University of Helsinki, Helsinki, Finland
| | - Tero Aittokallio
- Institute for Molecular Medicine Finland, FIMM, University of Helsinki, Helsinki, Finland.,Department of Mathematics and Statistics, University of Turku, Turku, Finland
| |
Collapse
|
9
|
Advantages and shortcomings of cell-based electrical impedance measurements as a GPCR drug discovery tool. Biosens Bioelectron 2019; 137:33-44. [PMID: 31077988 DOI: 10.1016/j.bios.2019.04.041] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 04/05/2019] [Accepted: 04/20/2019] [Indexed: 12/13/2022]
Abstract
G Protein-Coupled Receptors (GPCRs) transduce extracellular signals and activate intracellular pathways, usually through activating associated G proteins. Due to their involvement in many human diseases, they are recognized worldwide as valuable drug targets. Many experimental approaches help identify small molecules that target GPCRs, including in vitro cell-based reporter assays and binding studies. Most cell-based assays use one signaling pathway or reporter as an assay readout. Moreover, they often require cell labeling or the integration of reporter systems. Over the last decades, cell-based electrical impedance biosensors have been explored for drug discovery. This label-free method holds many advantages over other cellular assays in GPCR research. The technology requires no cell manipulation and offers real-time kinetic measurements of receptor-mediated cellular changes. Instead of measuring the activity of a single reporter, the impedance readout includes information on multiple signaling events. This is beneficial when screening for ligands targeting orphan GPCRs since the signaling cascade(s) of the majority of these receptors are unknown. Due to its sensitivity, the method also applies to cellular models more relevant to disease, including patient-derived cell cultures. Despite its advantages, remaining issues regarding data comparability and interpretability has limited implementation of cell-based electrical impedance (CEI) in drug discovery. Future optimization must include both full exploitation of CEI response data using various ways of analysis as well as further exploration of its potential to detect biased activities early on in drug discovery. Here, we review the contribution of CEI technology to GPCR research, discuss its comparative benefits, and provide recommendations.
Collapse
|
10
|
Pottel J, Levit A, Korczynska M, Fischer M, Shoichet BK. The Recognition of Unrelated Ligands by Identical Proteins. ACS Chem Biol 2018; 13:2522-2533. [PMID: 30095890 DOI: 10.1021/acschembio.8b00443] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Unrelated ligands, often found in drug discovery campaigns, can bind to the same receptor, even with the same protein residues. To investigate how this might occur, and whether it might be typically possible to find unrelated ligands for the same drug target, we sought examples of topologically unrelated ligands that bound to the same protein in the same site. Seventy-six pairs of ligands, each bound to the same protein (152 complexes total), were considered, classified into three groups. In the first (31 pairs of complexes), unrelated ligands interacted largely with the same pocket residues through different functional groups. In the second group (39 pairs), the unrelated ligand in each pair engaged different residues, though still within the same pocket. The smallest group (6 pairs) contained ligands with different scaffolds but with shared functional groups interacting with the same residues. We found that there are multiple chemically unrelated but physically similar functional groups that can complement any given local protein pocket; when these functional group substitutions are combined within a single molecule, they lead to topologically unrelated ligands that can each well-complement a site. It may be that many active and orthosteric sites can recognize topologically unrelated ligands.
Collapse
Affiliation(s)
- Joshua Pottel
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California 94158, United States
| | - Anat Levit
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California 94158, United States
| | - Magdalena Korczynska
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California 94158, United States
| | - Marcus Fischer
- Department of Chemical Biology and Therapeutics & Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee 38105, United States
| | - Brian K. Shoichet
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California 94158, United States
| |
Collapse
|
11
|
Weiss D, Karpiak J, Huang XP, Sassano MF, Lyu J, Roth BL, Shoichet BK. Selectivity Challenges in Docking Screens for GPCR Targets and Antitargets. J Med Chem 2018; 61:6830-6845. [PMID: 29990431 PMCID: PMC6105036 DOI: 10.1021/acs.jmedchem.8b00718] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Indexed: 12/14/2022]
Abstract
To investigate large library docking's ability to find molecules with joint activity against on-targets and selectivity versus antitargets, the dopamine D2 and serotonin 5-HT2A receptors were targeted, seeking selectivity against the histamine H1 receptor. In a second campaign, κ-opioid receptor ligands were sought with selectivity versus the μ-opioid receptor. While hit rates ranged from 40% to 63% against the on-targets, they were just as good against the antitargets, even though the molecules were selected for their putative lack of binding to the off-targets. Affinities, too, were often as good or better for the off-targets. Even though it was occasionally possible to find selective molecules, such as a mid-nanomolar D2/5-HT2A ligand with 21-fold selectivity versus the H1 receptor, this was the exception. Whereas false-negatives are tolerable in docking screens against on-targets, they are intolerable against antitargets; addressing this problem may demand new strategies in the field.
Collapse
Affiliation(s)
- Dahlia
R. Weiss
- Department
of Pharmaceutical Chemistry, University
of California—San Francisco, San Francisco, California 94158-2550, United States
| | - Joel Karpiak
- Department
of Pharmaceutical Chemistry, University
of California—San Francisco, San Francisco, California 94158-2550, United States
| | - Xi-Ping Huang
- Department
of Pharmacology and National Institute of Mental Health Psychoactive
Drug Screening Program, School of Medicine, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Maria F. Sassano
- Department
of Pharmacology and National Institute of Mental Health Psychoactive
Drug Screening Program, School of Medicine, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Jiankun Lyu
- Department
of Pharmaceutical Chemistry, University
of California—San Francisco, San Francisco, California 94158-2550, United States
| | - Bryan L. Roth
- Department
of Pharmacology and National Institute of Mental Health Psychoactive
Drug Screening Program, School of Medicine, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Brian K. Shoichet
- Department
of Pharmaceutical Chemistry, University
of California—San Francisco, San Francisco, California 94158-2550, United States
| |
Collapse
|
12
|
Rabal O, Castellar A, Oyarzabal J. Novel pharmacological maps of protein lysine methyltransferases: key for target deorphanization. J Cheminform 2018; 10:32. [PMID: 30032331 PMCID: PMC6054832 DOI: 10.1186/s13321-018-0288-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 07/14/2018] [Indexed: 11/30/2022] Open
Abstract
Epigenetic therapies are being investigated for the treatment of cancer, cognitive disorders, metabolic alterations and autoinmune diseases. Among the different epigenetic target families, protein lysine methyltransferases (PKMTs), are especially interesting because it is believed that their inhibition may be highly specific at the functional level. Despite its relevance, there are currently known inhibitors against only 10 out of the 50 SET-domain containing members of the PKMT family. Accordingly, the identification of chemical probes for the validation of the therapeutic impact of epigenetic modulation is key. Moreover, little is known about the mechanisms that dictate their substrate specificity and ligand selectivity. Consequently, it is desirable to explore novel methods to characterize the pharmacological similarity of PKMTs, going beyond classical phylogenetic relationships. Such characterization would enable the prediction of ligand off-target effects caused by lack of ligand selectivity and the repurposing of known compounds against alternative targets. This is particularly relevant in the case of orphan targets with unreported inhibitors. Here, we first perform a systematic study of binding modes of cofactor and substrate bound ligands with all available SET domain-containing PKMTs. Protein ligand interaction fingerprints were applied to identify conserved hot spots and contact-specific residues across subfamilies at each binding site; a relevant analysis for guiding the design of novel, selective compounds. Then, a recently described methodology (GPCR-CoINPocket) that incorporates ligand contact information into classical alignment-based comparisons was applied to the entire family of 50 SET-containing proteins to devise pharmacological similarities between them. The main advantage of this approach is that it is not restricted to proteins for which crystallographic data with bound ligands is available. The resulting family organization from the separate analysis of both sites (cofactor and substrate) was retrospectively and prospectively validated. Of note, three hits (inhibition > 50% at 10 µM) were identified for the orphan NSD1.
Collapse
Affiliation(s)
- Obdulia Rabal
- Small Molecule Discovery Platform. Molecular Therapeutics Program, Center for Applied Medical Research, CIMA, University of Navarra, Pio XII, 55, 31008, Pamplona, Spain.
| | - Andrea Castellar
- Small Molecule Discovery Platform. Molecular Therapeutics Program, Center for Applied Medical Research, CIMA, University of Navarra, Pio XII, 55, 31008, Pamplona, Spain
| | - Julen Oyarzabal
- Small Molecule Discovery Platform. Molecular Therapeutics Program, Center for Applied Medical Research, CIMA, University of Navarra, Pio XII, 55, 31008, Pamplona, Spain.
| |
Collapse
|
13
|
The G protein-coupled receptors deorphanization landscape. Biochem Pharmacol 2018; 153:62-74. [PMID: 29454621 DOI: 10.1016/j.bcp.2018.02.016] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 02/13/2018] [Indexed: 12/14/2022]
Abstract
G protein-coupled receptors (GPCRs) are usually highlighted as being both the largest family of membrane proteins and the most productive source of drug targets. However, most of the GPCRs are understudied and hence cannot be used immediately for innovative therapeutic strategies. Besides, there are still around 100 orphan receptors, with no described endogenous ligand and no clearly defined function. The race to discover new ligands for these elusive receptors seems to be less intense than before. Here, we present an update of the various strategies employed to assign a function to these receptors and to discover new ligands. We focus on the recent advances in the identification of endogenous ligands with a detailed description of newly deorphanized receptors. Replication being a key parameter in these endeavors, we also discuss the latest controversies about problematic ligand-receptor pairings. In this context, we propose several recommendations in order to strengthen the reporting of new ligand-receptor pairs.
Collapse
|
14
|
Calhoun S, Korczynska M, Wichelecki DJ, San Francisco B, Zhao S, Rodionov DA, Vetting MW, Al-Obaidi NF, Lin H, O'Meara MJ, Scott DA, Morris JH, Russel D, Almo SC, Osterman AL, Gerlt JA, Jacobson MP, Shoichet BK, Sali A. Prediction of enzymatic pathways by integrative pathway mapping. eLife 2018; 7:31097. [PMID: 29377793 PMCID: PMC5788505 DOI: 10.7554/elife.31097] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 12/18/2017] [Indexed: 01/17/2023] Open
Abstract
The functions of most proteins are yet to be determined. The function of an enzyme is often defined by its interacting partners, including its substrate and product, and its role in larger metabolic networks. Here, we describe a computational method that predicts the functions of orphan enzymes by organizing them into a linear metabolic pathway. Given candidate enzyme and metabolite pathway members, this aim is achieved by finding those pathways that satisfy structural and network restraints implied by varied input information, including that from virtual screening, chemoinformatics, genomic context analysis, and ligand -binding experiments. We demonstrate this integrative pathway mapping method by predicting the L-gulonate catabolic pathway in Haemophilus influenzae Rd KW20. The prediction was subsequently validated experimentally by enzymology, crystallography, and metabolomics. Integrative pathway mapping by satisfaction of structural and network restraints is extensible to molecular networks in general and thus formally bridges the gap between structural biology and systems biology.
Collapse
Affiliation(s)
- Sara Calhoun
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, United States
| | - Magdalena Korczynska
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, United States
| | - Daniel J Wichelecki
- Institute for Genomic Biology, University of Illinois, Urbana, United States.,Department of Biochemistry, University of Illinois, Urbana, United States.,Department of Chemistry, University of Illinois, Urbana, United States
| | - Brian San Francisco
- Institute for Genomic Biology, University of Illinois, Urbana, United States
| | - Suwen Zhao
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, United States
| | - Dmitry A Rodionov
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, United States.,A.A. Kharkevich Institute for Information Transmission Problems, Russian Academy of Sciences, Moscow, Russia
| | - Matthew W Vetting
- Department of Biochemistry, Albert Einstein College of Medicine, New York, United States
| | - Nawar F Al-Obaidi
- Department of Biochemistry, Albert Einstein College of Medicine, New York, United States
| | - Henry Lin
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, United States
| | - Matthew J O'Meara
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, United States
| | - David A Scott
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, United States
| | - John H Morris
- Resource for Biocomputing, Visualization and Informatics, Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, United States
| | - Daniel Russel
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, United States
| | - Steven C Almo
- Department of Biochemistry, Albert Einstein College of Medicine, New York, United States
| | - Andrei L Osterman
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, United States
| | - John A Gerlt
- Institute for Genomic Biology, University of Illinois, Urbana, United States.,Department of Biochemistry, University of Illinois, Urbana, United States.,Department of Chemistry, University of Illinois, Urbana, United States
| | - Matthew P Jacobson
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, United States
| | - Brian K Shoichet
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, United States
| | - Andrej Sali
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, United States.,Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, United States.,California Institute for Quantitative Biosciences, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
15
|
Vass M, Kooistra AJ, Verhoeven S, Gloriam D, de Esch IJP, de Graaf C. A Structural Framework for GPCR Chemogenomics: What's In a Residue Number? Methods Mol Biol 2018; 1705:73-113. [PMID: 29188559 DOI: 10.1007/978-1-4939-7465-8_4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The recent surge of crystal structures of G protein-coupled receptors (GPCRs), as well as comprehensive collections of sequence, structural, ligand bioactivity, and mutation data, has enabled the development of integrated chemogenomics workflows for this important target family. This chapter will focus on cross-family and cross-class studies of GPCRs that have pinpointed the need for, and the implementation of, a generic numbering scheme for referring to specific structural elements of GPCRs. Sequence- and structure-based numbering schemes for different receptor classes will be introduced and the remaining caveats will be discussed. The use of these numbering schemes has facilitated many chemogenomics studies such as consensus binding site definition, binding site comparison, ligand repurposing (e.g. for orphan receptors), sequence-based pharmacophore generation for homology modeling or virtual screening, and class-wide chemogenomics studies of GPCRs.
Collapse
Affiliation(s)
- Márton Vass
- Department of Medicinal Chemistry, Amsterdam Institute for Molecules Medicines and Systems, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HV, Amsterdam, The Netherlands
| | - Albert J Kooistra
- Department of Medicinal Chemistry, Amsterdam Institute for Molecules Medicines and Systems, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HV, Amsterdam, The Netherlands
- Centre for Molecular and Biomolecular Informatics (CMBI), Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
| | - Stefan Verhoeven
- Netherlands eScience Center, 1098 XG, Amsterdam, The Netherlands
| | - David Gloriam
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Iwan J P de Esch
- Department of Medicinal Chemistry, Amsterdam Institute for Molecules Medicines and Systems, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HV, Amsterdam, The Netherlands
| | - Chris de Graaf
- Department of Medicinal Chemistry, Amsterdam Institute for Molecules Medicines and Systems, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
16
|
Tresadern G, Trabanco AA, Pérez-Benito L, Overington JP, van Vlijmen HWT, van Westen GJP. Identification of Allosteric Modulators of Metabotropic Glutamate 7 Receptor Using Proteochemometric Modeling. J Chem Inf Model 2017; 57:2976-2985. [PMID: 29172488 PMCID: PMC5755953 DOI: 10.1021/acs.jcim.7b00338] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Indexed: 01/07/2023]
Abstract
Proteochemometric modeling (PCM) is a computational approach that can be considered an extension of quantitative structure-activity relationship (QSAR) modeling, where a single model incorporates information for a family of targets and all the associated ligands instead of modeling activity versus one target. This is especially useful for situations where bioactivity data exists for similar proteins but is scarce for the protein of interest. Here we demonstrate the application of PCM to identify allosteric modulators of metabotropic glutamate (mGlu) receptors. Given our long-running interest in modulating mGlu receptor function we compiled a matrix of compound-target bioactivity data. Some members of the mGlu family are well explored both internally and in the public domain, while there are much fewer examples of ligands for other targets such as the mGlu7 receptor. Using a PCM approach mGlu7 receptor hits were found. In comparison to conventional single target modeling the identified hits were more diverse, had a better confirmation rate, and provide starting points for further exploration. We conclude that the robust structure-activity relationship from well explored target family members translated to better quality hits for PCM compared to virtual screening (VS) based on a single target.
Collapse
Affiliation(s)
- Gary Tresadern
- Computational
Chemistry and Neuroscience Medicinal Chemistry, Janssen
Research & Development, Janssen-Cilag
S.A., Jarama 75A, 45007 Toledo, Spain
| | - Andres A. Trabanco
- Computational
Chemistry and Neuroscience Medicinal Chemistry, Janssen
Research & Development, Janssen-Cilag
S.A., Jarama 75A, 45007 Toledo, Spain
| | - Laura Pérez-Benito
- Computational
Chemistry and Neuroscience Medicinal Chemistry, Janssen
Research & Development, Janssen-Cilag
S.A., Jarama 75A, 45007 Toledo, Spain
| | - John P. Overington
- ChEMBL Group, EMBL-EBI,
Wellcome Trust Genome Campus, CB10 1SD Hinxton, United Kingdom
| | - Herman W. T. van Vlijmen
- Computational
Chemistry, Janssen Research & Development, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | | |
Collapse
|
17
|
Hounsou C, Baehr C, Gasparik V, Alili D, Belhocine A, Rodriguez T, Dupuis E, Roux T, Mann A, Heissler D, Pin JP, Durroux T, Bonnet D, Hibert M. From the Promiscuous Asenapine to Potent Fluorescent Ligands Acting at a Series of Aminergic G-Protein-Coupled Receptors. J Med Chem 2017; 61:174-188. [PMID: 29219316 DOI: 10.1021/acs.jmedchem.7b01220] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Monoamine neurotransmitters such as serotonin, dopamine, histamine, and noradrenaline have important and varied physiological functions and similar chemical structures. Representing important pharmaceutical drug targets, the corresponding G-protein-coupled receptors (termed aminergic GPCRs) belong to the class of cell membrane receptors and share many levels of similarity as well. Given their pharmacological and structural closeness, one could hypothesize the possibility to derivatize a ubiquitous ligand to afford rapidly fluorescent probes for a large set of GPCRs to be used for instance in FRET-based binding assays. Here we report fluorescent derivatives of the nonselective agent asenapine which were designed, synthesized, and evaluated as ligands of 34 serotonin, dopamine, histamine, melatonin, acetylcholine, and adrenergic receptors. It appears that this strategy led rapidly to the discovery and development of nanomolar affinity fluorescent probes for 14 aminergic GPCRs. Selected probes were tested in competition binding assays with unlabeled competitors in order to demonstrate their suitability for drug discovery purposes.
Collapse
Affiliation(s)
- Candide Hounsou
- Institut de Génomique Fonctionnelle, CNRS UMR5203, INSERM U661, Université de Montpellier (IFR3) , 141 Rue de la Cardonille, F-34094 Montpellier Cedex 5, France
| | - Corinne Baehr
- Laboratoire d'Innovation Thérapeutique, Faculté de Pharmacie, UMR7200 CNRS, Université de Strasbourg , 74 Route du Rhin, 67412 Illkirch, France
| | - Vincent Gasparik
- Laboratoire d'Innovation Thérapeutique, Faculté de Pharmacie, UMR7200 CNRS, Université de Strasbourg , 74 Route du Rhin, 67412 Illkirch, France
| | - Doria Alili
- Institut de Génomique Fonctionnelle, CNRS UMR5203, INSERM U661, Université de Montpellier (IFR3) , 141 Rue de la Cardonille, F-34094 Montpellier Cedex 5, France
| | - Abderazak Belhocine
- Institut de Génomique Fonctionnelle, CNRS UMR5203, INSERM U661, Université de Montpellier (IFR3) , 141 Rue de la Cardonille, F-34094 Montpellier Cedex 5, France
| | - Thiéric Rodriguez
- Institut de Génomique Fonctionnelle, CNRS UMR5203, INSERM U661, Université de Montpellier (IFR3) , 141 Rue de la Cardonille, F-34094 Montpellier Cedex 5, France
| | - Elodie Dupuis
- Cisbio Bioassays , Parc Marcel Boiteux, BP84175, 30200 Codolet, France
| | - Thomas Roux
- Cisbio Bioassays , Parc Marcel Boiteux, BP84175, 30200 Codolet, France
| | - André Mann
- Laboratoire d'Innovation Thérapeutique, Faculté de Pharmacie, UMR7200 CNRS, Université de Strasbourg , 74 Route du Rhin, 67412 Illkirch, France
| | - Denis Heissler
- Laboratoire d'Innovation Thérapeutique, Faculté de Pharmacie, UMR7200 CNRS, Université de Strasbourg , 74 Route du Rhin, 67412 Illkirch, France.,LabEx Medalis, Université de Strasbourg , 67000 Strasbourg, France
| | - Jean-Philippe Pin
- Institut de Génomique Fonctionnelle, CNRS UMR5203, INSERM U661, Université de Montpellier (IFR3) , 141 Rue de la Cardonille, F-34094 Montpellier Cedex 5, France
| | - Thierry Durroux
- Institut de Génomique Fonctionnelle, CNRS UMR5203, INSERM U661, Université de Montpellier (IFR3) , 141 Rue de la Cardonille, F-34094 Montpellier Cedex 5, France
| | - Dominique Bonnet
- Laboratoire d'Innovation Thérapeutique, Faculté de Pharmacie, UMR7200 CNRS, Université de Strasbourg , 74 Route du Rhin, 67412 Illkirch, France.,LabEx Medalis, Université de Strasbourg , 67000 Strasbourg, France
| | - Marcel Hibert
- Laboratoire d'Innovation Thérapeutique, Faculté de Pharmacie, UMR7200 CNRS, Université de Strasbourg , 74 Route du Rhin, 67412 Illkirch, France.,LabEx Medalis, Université de Strasbourg , 67000 Strasbourg, France
| |
Collapse
|
18
|
Ravikumar B, Aittokallio T. Improving the efficacy-safety balance of polypharmacology in multi-target drug discovery. Expert Opin Drug Discov 2017; 13:179-192. [DOI: 10.1080/17460441.2018.1413089] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Balaguru Ravikumar
- Institute for Molecular Medicine Finland, FIMM, University of Helsinki, Helsinki, Finland
| | - Tero Aittokallio
- Institute for Molecular Medicine Finland, FIMM, University of Helsinki, Helsinki, Finland
- Department of Mathematics and Statistics, University of Turku, Turku, Finland
| |
Collapse
|
19
|
Fish I, Stößel A, Eitel K, Valant C, Albold S, Huebner H, Möller D, Clark MJ, Sunahara RK, Christopoulos A, Shoichet BK, Gmeiner P. Structure-Based Design and Discovery of New M 2 Receptor Agonists. J Med Chem 2017; 60:9239-9250. [PMID: 29094937 DOI: 10.1021/acs.jmedchem.7b01113] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Muscarinic receptor agonists are characterized by apparently strict restraints on their tertiary or quaternary amine and their distance to an ester or related center. On the basis of the active state crystal structure of the muscarinic M2 receptor in complex with iperoxo, we explored potential agonists that lacked the highly conserved functionalities of previously known ligands. Using structure-guided pharmacophore design followed by docking, we found two agonists (compounds 3 and 17), out of 19 docked and synthesized compounds, that fit the receptor well and were predicted to form a hydrogen-bond conserved among known agonists. Structural optimization led to compound 28, which was 4-fold more potent than its parent 3. Fortified by the discovery of this new scaffold, we sought a broader range of chemotypes by docking 2.2 million fragments, which revealed another three micromolar agonists unrelated either to 28 or known muscarinics. Even pockets as tightly defined and as deeply studied as that of the muscarinic reveal opportunities for the structure-based design and the discovery of new chemotypes.
Collapse
Affiliation(s)
- Inbar Fish
- Department of Pharmaceutical Chemistry, University of California, San Francisco , San Francisco, California 94158, United States.,Department of Biochemistry and Molecular Biology, George S. Wise Faculty of Life Sciences, Tel-Aviv University , Ramat Aviv, Israel
| | - Anne Stößel
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich Alexander University , Schuhstraße 19, 91052 Erlangen, Germany
| | - Katrin Eitel
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich Alexander University , Schuhstraße 19, 91052 Erlangen, Germany
| | - Celine Valant
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University , Parkville Victoria 3052, Australia
| | - Sabine Albold
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University , Parkville Victoria 3052, Australia
| | - Harald Huebner
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich Alexander University , Schuhstraße 19, 91052 Erlangen, Germany
| | - Dorothee Möller
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich Alexander University , Schuhstraße 19, 91052 Erlangen, Germany
| | - Mary J Clark
- Department of Pharmacology, University of California, San Diego , La Jolla, California 92093, United States
| | - Roger K Sunahara
- Department of Pharmacology, University of California, San Diego , La Jolla, California 92093, United States
| | - Arthur Christopoulos
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University , Parkville Victoria 3052, Australia
| | - Brian K Shoichet
- Department of Pharmaceutical Chemistry, University of California, San Francisco , San Francisco, California 94158, United States
| | - Peter Gmeiner
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich Alexander University , Schuhstraße 19, 91052 Erlangen, Germany
| |
Collapse
|
20
|
Rare Diseases: Drug Discovery and Informatics Resource. Interdiscip Sci 2017; 10:195-204. [PMID: 29094320 DOI: 10.1007/s12539-017-0270-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 10/19/2017] [Accepted: 10/23/2017] [Indexed: 12/13/2022]
Abstract
A rare disease refers to any disease with very low prevalence individually. Although the impacted population is small for a single disease, more than 6000 rare diseases affect millions of people across the world. Due to the small market size, high cost and possibly low return on investment, only in recent years, the research and development of rare disease drugs have gradually risen globally, in several domains including gene therapy, enzyme replacement therapy, and drug repositioning. Due to the complex etiology and heterogeneous symptoms, there is a large gap between basic research and patient unmet needs for rare disease drug discovery. As computational biology increasingly arises researchers' awareness, the informatics database on rare disease have grown rapidly in the recent years, including drug targets, genetic variant and mutation, phenotype and ontology and patient registries. Along with the advances of informatics database and networks, new computational models will help accelerate the target identification and lead optimization process for rare disease pre-clinical drug development.
Collapse
|
21
|
Petrucci V, Chicca A, Glasmacher S, Paloczi J, Cao Z, Pacher P, Gertsch J. Pepcan-12 (RVD-hemopressin) is a CB2 receptor positive allosteric modulator constitutively secreted by adrenals and in liver upon tissue damage. Sci Rep 2017; 7:9560. [PMID: 28842619 PMCID: PMC5573408 DOI: 10.1038/s41598-017-09808-8] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 07/28/2017] [Indexed: 12/21/2022] Open
Abstract
Pepcan-12 (RVD-hemopressin; RVDPVNFKLLSH) is the major peptide of a family of endogenous peptide endocannabinoids (pepcans) shown to act as negative allosteric modulators (NAM) of cannabinoid CB1 receptors. Noradrenergic neurons have been identified to be a specific site of pepcan production. However, it remains unknown whether pepcans occur in the periphery and interact with peripheral CB2 cannabinoid receptors. Here, it is shown that pepcan-12 acts as a potent (K i value ~50 nM) hCB2 receptor positive allosteric modulator (PAM). It significantly potentiated the effects of CB2 receptor agonists, including the endocannabinoid 2-arachidonoyl glycerol (2-AG), for [35S]GTPγS binding and cAMP inhibition (5-10 fold). In mice, the putative precursor pepcan-23 (SALSDLHAHKLRVDPVNFKLLSH) was identified with pepcan-12 in brain, liver and kidney. Pepcan-12 was increased upon endotoxemia and ischemia reperfusion damage where CB2 receptors play a protective role. The adrenals are a major endocrine site of production/secretion of constitutive pepcan-12, as shown by its marked loss after adrenalectomy. However, upon I/R damage pepcan-12 was strongly increased in the liver (from ~100 pmol/g to ~500 pmol/g) independent of adrenals. The wide occurrence of this endogenous hormone-like CB2 receptor PAM, with unforeseen opposite allosteric effects on cannabinoid receptors, suggests its potential role in peripheral pathophysiological processes.
Collapse
Affiliation(s)
- Vanessa Petrucci
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, 3012, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Andrea Chicca
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, 3012, Bern, Switzerland
| | - Sandra Glasmacher
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, 3012, Bern, Switzerland
| | - Janos Paloczi
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institutes of Health/NIAAA, Bethesda, MD, USA
| | - Zongxian Cao
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institutes of Health/NIAAA, Bethesda, MD, USA
| | - Pal Pacher
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institutes of Health/NIAAA, Bethesda, MD, USA
| | - Jürg Gertsch
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, 3012, Bern, Switzerland.
| |
Collapse
|
22
|
Template selection and refinement considerations for modelling aminergic GPCR-ligand complexes. J Mol Graph Model 2017; 76:488-503. [PMID: 28818718 DOI: 10.1016/j.jmgm.2017.07.030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Revised: 07/21/2017] [Accepted: 07/25/2017] [Indexed: 11/24/2022]
Abstract
G protein-coupled receptors (GPCRs) are important targets for development of drugs for the treatment of many diseases. However, crystal structures are available for only a small fraction of these membrane bound proteins. Accurate homology models will provide opportunities for effective drug design targeting GPCRs. Recently, several serotonin receptor crystal structures were solved and needed to be evaluated as potential templates. In the first part of this work different measures of similarity in template selection were explored and methods for homology modelling, docking and refinement of aminergic GPCR-ligand complexes were developed and evaluated by comparing models of the D3-R/eticlopride complex with the crystal structure. Homology models of the three α1 adrenergic receptor subtypes and of a serotonin receptor subtype were then constructed using these methods These models were evaluated by docking a range of antagonists into them.
Collapse
|
23
|
Zhang C, Shao YM, Ma X, Cheong SL, Qin C, Tao L, Zhang P, Chen S, Zeng X, Liu H, Pastorin G, Jiang Y, Chen YZ. Pharmacological relationships and ligand discovery of G protein-coupled receptors revealed by simultaneous ligand and receptor clustering. J Mol Graph Model 2017; 76:136-142. [PMID: 28728042 DOI: 10.1016/j.jmgm.2017.06.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Revised: 06/17/2017] [Accepted: 06/19/2017] [Indexed: 12/18/2022]
Abstract
Conventional ligand and receptor similarity methods have been extensively used for exposing pharmacological relationships and drug lead discovery. They may in some cases neglect minor relationships useful for target hopping particularly against the remote family members. To complement the conventional methods for capturing these minor relationships, we developed a new method that uses a SLARC (Simultaneous Ligand And Receptor Clustering) 2D map to simultaneously characterize the ligand structural and receptor binding-site sequence relationships of a receptor family. The SLARC maps of the rhodopsin-like GPCR family comprehensively revealed scaffold hopping, target hopping, and multi-target relationships for the ligands of both homologous and remote family members. Their usefulness in new ligand discovery was validated by guiding the prospective discovery of novel indole piperazinylpyrimidine dual-targeting adenosine A2A receptor antagonist and dopamine D2 agonist compounds. The SLARC approach is useful for revealing pharmacological relationships and discovering new ligands at target family levels.
Collapse
Affiliation(s)
- Cheng Zhang
- Ministry-Province Jointly Constructed Base for State Key Lab and Shenzhen Technology and Engineering Lab for Personalized Cancer Diagnostics and Therapeutics, Tsinghua University Shenzhen Graduate School, and Shenzhen Kivita Innovative Drug Discovery Institute, Shenzhen 518055, PR China; Department of Molecular Pharmacology and Experimental Therapeutics, Center for Individualized Medicine, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Yi-Ming Shao
- Department of Pharmacy, National University of Singapore, Singapore 117543, Singapore
| | - Xiaohua Ma
- School of Materials Science and Engineering, Nanyang Technological University, 639798, Singapore
| | - Siew Lee Cheong
- Department of Pharmacy, National University of Singapore, Singapore 117543, Singapore
| | - Chu Qin
- Department of Pharmacy, National University of Singapore, Singapore 117543, Singapore
| | - Lin Tao
- Department of Pharmacy, National University of Singapore, Singapore 117543, Singapore
| | - Peng Zhang
- Department of Pharmacy, National University of Singapore, Singapore 117543, Singapore
| | - Shangying Chen
- Department of Pharmacy, National University of Singapore, Singapore 117543, Singapore
| | - Xian Zeng
- Department of Pharmacy, National University of Singapore, Singapore 117543, Singapore
| | - Hongxia Liu
- Ministry-Province Jointly Constructed Base for State Key Lab and Shenzhen Technology and Engineering Lab for Personalized Cancer Diagnostics and Therapeutics, Tsinghua University Shenzhen Graduate School, and Shenzhen Kivita Innovative Drug Discovery Institute, Shenzhen 518055, PR China
| | - Giorgia Pastorin
- Department of Pharmacy, National University of Singapore, Singapore 117543, Singapore; NUS Graduate School for Integrative Sciences and Engineering, 117456, Singapore.
| | - Yuyang Jiang
- Ministry-Province Jointly Constructed Base for State Key Lab and Shenzhen Technology and Engineering Lab for Personalized Cancer Diagnostics and Therapeutics, Tsinghua University Shenzhen Graduate School, and Shenzhen Kivita Innovative Drug Discovery Institute, Shenzhen 518055, PR China.
| | - Yu Zong Chen
- Department of Pharmacy, National University of Singapore, Singapore 117543, Singapore; NUS Graduate School for Integrative Sciences and Engineering, 117456, Singapore.
| |
Collapse
|
24
|
Duran-Frigola M, Siragusa L, Ruppin E, Barril X, Cruciani G, Aloy P. Detecting similar binding pockets to enable systems polypharmacology. PLoS Comput Biol 2017; 13:e1005522. [PMID: 28662117 PMCID: PMC5490940 DOI: 10.1371/journal.pcbi.1005522] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 04/15/2017] [Indexed: 01/19/2023] Open
Abstract
In the era of systems biology, multi-target pharmacological strategies hold promise for tackling disease-related networks. In this regard, drug promiscuity may be leveraged to interfere with multiple receptors: the so-called polypharmacology of drugs can be anticipated by analyzing the similarity of binding sites across the proteome. Here, we perform a pairwise comparison of 90,000 putative binding pockets detected in 3,700 proteins, and find that 23,000 pairs of proteins have at least one similar cavity that could, in principle, accommodate similar ligands. By inspecting these pairs, we demonstrate how the detection of similar binding sites expands the space of opportunities for the rational design of drug polypharmacology. Finally, we illustrate how to leverage these opportunities in protein-protein interaction networks related to several therapeutic classes and tumor types, and in a genome-scale metabolic model of leukemia. Traditionally, the fact that most drugs are promiscuous binders has been a major concern in pharmacology, due to the occurrence of undesired off-target clinical events. In the recent years, however, the realization that many diseases are the result of complex biological processes has encouraged rethinking of drug promiscuity as a promising feature, since it is sometimes necessary to interfere with multiple receptors in order to overcome the robustness of disease-related networks. One way to identify groups of proteins that could be targeted simultaneously is to look for similar binding sites. We have massively done so for all human proteins with a known high-resolution three-dimensional structure, unveiling a vast space of ‘polypharmacology’ opportunities. Of these, we know, a great majority is not of therapeutic interest. To pinpoint promising multi-target combinations, we advocate for the use of computational tools that are able to rapidly simulate the effect of drug-target interactions on biological networks.
Collapse
Affiliation(s)
- Miquel Duran-Frigola
- Joint IRB-BSC-CRG Program in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Catalonia, Spain
| | | | - Eytan Ruppin
- Department of Computer Science & Center for Bioinformatics and Computational Biology, University of Maryland, College Park, Maryland, United States of America
- School of Computer Sciences, Tel Aviv University, Tel Aviv, Israel
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Xavier Barril
- Departament de Fisicoquímica, Facultat de Farmàcia, Universitat de Barcelona, Barcelona, Catalonia, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia, Spain
| | - Gabriele Cruciani
- Molecular Discovery Limited, London, United Kingdom
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Patrick Aloy
- Joint IRB-BSC-CRG Program in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Catalonia, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia, Spain
- * E-mail:
| |
Collapse
|
25
|
da Silva Junior ED, Sato M, Merlin J, Broxton N, Hutchinson DS, Ventura S, Evans BA, Summers RJ. Factors influencing biased agonism in recombinant cells expressing the human α 1A -adrenoceptor. Br J Pharmacol 2017; 174:2318-2333. [PMID: 28444738 DOI: 10.1111/bph.13837] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 04/06/2017] [Accepted: 04/12/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND PURPOSE Agonists acting at GPCRs promote biased signalling via Gα or Gβγ subunits, GPCR kinases and β-arrestins. Since the demonstration of biased agonism has implications for drug discovery, it is essential to consider confounding factors contributing to bias. We have examined bias at human α1A -adrenoceptors stably expressed at low levels in CHO-K1 cells, identifying off-target effects at endogenous receptors that contribute to ERK1/2 phosphorylation in response to the agonist oxymetazoline. EXPERIMENTAL APPROACH Intracellular Ca2+ mobilization was monitored in a Flexstation® using Fluo 4-AM. The accumulation of cAMP and ERK1/2 phosphorylation were measured using AlphaScreen® proximity assays, and mRNA expression was measured by RT-qPCR. Ligand bias was determined using the operational model of agonism. KEY RESULTS Noradrenaline, phenylephrine, methoxamine and A61603 increased Ca2+ mobilization, cAMP accumulation and ERK1/2 phosphorylation. However, oxymetazoline showed low efficacy for Ca+2 mobilization, no effect on cAMP generation and high efficacy for ERK1/2 phosphorylation. The apparent functional selectivity of oxymetazoline towards ERK1/2 was related to off-target effects at 5-HT1B receptors endogenously expressed in CHO-K1 cells. Phenylephrine and methoxamine showed genuine bias towards ERK1/2 phosphorylation compared to Ca2+ and cAMP pathways, whereas A61603 displayed bias towards cAMP accumulation compared to ERK1/2 phosphorylation. CONCLUSION AND IMPLICATIONS We have shown that while adrenergic agonists display bias at human α1A -adrenoceptors, the marked bias of oxymetazoline for ERK1/2 phosphorylation originates from off-target effects. Commonly used cell lines express a repertoire of endogenous GPCRs that may confound studies on biased agonism at recombinant receptors.
Collapse
Affiliation(s)
| | - Masaaki Sato
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Jon Merlin
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Natalie Broxton
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Dana S Hutchinson
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Sabatino Ventura
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Bronwyn A Evans
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Roger J Summers
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| |
Collapse
|
26
|
Rasolohery I, Moroy G, Guyon F. PatchSearch: A Fast Computational Method for Off-Target Detection. J Chem Inf Model 2017; 57:769-777. [PMID: 28282119 DOI: 10.1021/acs.jcim.6b00529] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Many therapeutic molecules are known to bind several proteins, which can be different from the initially targeted one. Such unexpected interactions with proteins called off-targets can lead to adverse effects. Potential off-target identification is important to predict to avoid drug side effects or to discover new targets for existing drugs. We propose a new program named PatchSearch that implements local nonsequential searching for similar binding sites on protein surfaces with a controlled amount of flexibility. It is based on detection of quasi-cliques in product graphs representing all the possible matchings between two compared structures. This method has been benchmarked on a large diversity of ligands and on five data sets ranging from 12 to more than 7000 protein structures. The experiments conducted in this study show that the PatchSearch method could be useful in the early identification of off-targets. The program and the benchmarks presented in this paper are available as an R package at https://github.com/MTiPatchSearch .
Collapse
Affiliation(s)
- Inès Rasolohery
- Molécules Thérapeutiques in Silico, UMRS 973, Université Paris Diderot, INSERM , F-75013 Paris, France
| | - Gautier Moroy
- Molécules Thérapeutiques in Silico, UMRS 973, Université Paris Diderot, INSERM , F-75013 Paris, France
| | - Frédéric Guyon
- Molécules Thérapeutiques in Silico, UMRS 973, Université Paris Diderot, INSERM , F-75013 Paris, France
| |
Collapse
|
27
|
In silico design of novel probes for the atypical opioid receptor MRGPRX2. Nat Chem Biol 2017; 13:529-536. [PMID: 28288109 PMCID: PMC5391270 DOI: 10.1038/nchembio.2334] [Citation(s) in RCA: 212] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 12/22/2016] [Indexed: 12/19/2022]
Abstract
The primate-exclusive MRGPRX2 G protein-coupled receptor (GPCR) has been suggested to modulate pain and itch. Despite putative peptide and small molecule MRGPRX2 agonists, selective nanomolar potency probes have not yet been reported. To identify a MRGPRX2 probe, we first screened 5,695 small molecules and found many opioid compounds activated MRGPRX2, including (−)- and (+)-morphine, hydrocodone, sinomenine, dextromethorphan and the prodynorphin-derived peptides, dynorphin A, dynorphin B, and α- and β-neoendorphin. We used these to select for mutagenesis-validated homology models and docked almost 4 million small molecules. From this docking, we predicted ZINC-3573, which represents a potent MRGPRX2-selective agonist, showing little activity against 315 other GPCRs and 97 representative kinases, and an essentially inactive enantiomer. ZINC-3573 activates endogenous MRGPRX2 in a human mast cell line inducing degranulation and calcium release. MRGPRX2 is a unique atypical opioid-like receptor important for modulating mast cell degranulation, which can now be specifically modulated with ZINC-3573.
Collapse
|
28
|
Gallion J, Koire A, Katsonis P, Schoenegge A, Bouvier M, Lichtarge O. Predicting phenotype from genotype: Improving accuracy through more robust experimental and computational modeling. Hum Mutat 2017; 38:569-580. [PMID: 28230923 PMCID: PMC5516182 DOI: 10.1002/humu.23193] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 01/25/2017] [Accepted: 02/04/2017] [Indexed: 11/11/2022]
Abstract
Computational prediction yields efficient and scalable initial assessments of how variants of unknown significance may affect human health. However, when discrepancies between these predictions and direct experimental measurements of functional impact arise, inaccurate computational predictions are frequently assumed as the source. Here, we present a methodological analysis indicating that shortcomings in both computational and biological data can contribute to these disagreements. We demonstrate that incomplete assaying of multifunctional proteins can affect the strength of correlations between prediction and experiments; a variant's full impact on function is better quantified by considering multiple assays that probe an ensemble of protein functions. Additionally, many variants predictions are sensitive to protein alignment construction and can be customized to maximize relevance of predictions to a specific experimental question. We conclude that inconsistencies between computation and experiment can often be attributed to the fact that they do not test identical hypotheses. Aligning the design of the computational input with the design of the experimental output will require cooperation between computational and biological scientists, but will also lead to improved estimations of computational prediction accuracy and a better understanding of the genotype–phenotype relationship.
Collapse
Affiliation(s)
- Jonathan Gallion
- Program in Structural and Computational Biology and Molecular BiophysicsBaylor College of MedicineHoustonTexas
| | - Amanda Koire
- Program in Structural and Computational Biology and Molecular BiophysicsBaylor College of MedicineHoustonTexas
| | - Panagiotis Katsonis
- Department of Molecular and Human GeneticsBaylor College of MedicineHoustonTexas
| | - Anne‐Marie Schoenegge
- Department of BiochemistryInstitute for Research in Immunology and CancerUniversité de MontrealQuebecCanada
| | - Michel Bouvier
- Department of BiochemistryInstitute for Research in Immunology and CancerUniversité de MontrealQuebecCanada
| | - Olivier Lichtarge
- Program in Structural and Computational Biology and Molecular BiophysicsBaylor College of MedicineHoustonTexas
- Department of Molecular and Human GeneticsBaylor College of MedicineHoustonTexas
| |
Collapse
|
29
|
Abstract
Designing drugs that can simultaneously interact with multiple targets is a promising approach for treating complicated diseases. Compared to using combinations of single target drugs, multitarget drugs have advantages of higher efficacy, improved safety profile, and simpler administration. Many in silico methods have been developed to approach different aspects of this polypharmacology-guided drug design, particularly for drug repurposing and multitarget drug design. In this review, we summarize recent progress in computational multitarget drug design and discuss their advantages and limitations. Perspectives for future drug development will also be discussed.
Collapse
Affiliation(s)
- Weilin Zhang
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies (AAIS), Peking University , Beijing 100871, People's Republic of China
| | - Jianfeng Pei
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies (AAIS), Peking University , Beijing 100871, People's Republic of China
| | - Luhua Lai
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies (AAIS), Peking University , Beijing 100871, People's Republic of China.,Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies (AAIS), Peking University , Beijing 100871, People's Republic of China.,BNLMS, State Key Laboratory for Structural Chemistry of Unstable and Stable Species, College of Chemistry and Molecular Engineering, Peking University , Beijing 100871, People's Republic of China
| |
Collapse
|
30
|
Castel H, Desrues L, Joubert JE, Tonon MC, Prézeau L, Chabbert M, Morin F, Gandolfo P. The G Protein-Coupled Receptor UT of the Neuropeptide Urotensin II Displays Structural and Functional Chemokine Features. Front Endocrinol (Lausanne) 2017; 8:76. [PMID: 28487672 PMCID: PMC5403833 DOI: 10.3389/fendo.2017.00076] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 03/28/2017] [Indexed: 12/16/2022] Open
Abstract
The urotensinergic system was previously considered as being linked to numerous physiopathological states, including atherosclerosis, heart failure, hypertension, pre-eclampsia, diabetes, renal disease, as well as brain vascular lesions. Thus, it turns out that the actions of the urotensin II (UII)/G protein-coupled receptor UT system in animal models are currently not predictive enough in regard to their effects in human clinical trials and that UII analogs, established to target UT, were not as beneficial as expected in pathological situations. Thus, many questions remain regarding the overall signaling profiles of UT leading to complex involvement in cardiovascular and inflammatory responses as well as cancer. We address the potential UT chemotactic structural and functional definition under an evolutionary angle, by the existence of a common conserved structural feature among chemokine receptorsopioïdergic receptors and UT, i.e., a specific proline position in the transmembrane domain-2 TM2 (P2.58) likely responsible for a kink helical structure that would play a key role in chemokine functions. Even if the last decade was devoted to the elucidation of the cardiovascular control by the urotensinergic system, we also attempt here to discuss the role of UII on inflammation and migration, likely providing a peptide chemokine status for UII. Indeed, our recent work established that activation of UT by a gradient concentration of UII recruits Gαi/o and Gα13 couplings in a spatiotemporal way, controlling key signaling events leading to chemotaxis. We think that this new vision of the urotensinergic system should help considering UT as a chemotactic therapeutic target in pathological situations involving cell chemoattraction.
Collapse
Affiliation(s)
- Hélène Castel
- Normandie University, UNIROUEN, INSERM, DC2N, Rouen, France
- Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
- *Correspondence: Hélène Castel,
| | - Laurence Desrues
- Normandie University, UNIROUEN, INSERM, DC2N, Rouen, France
- Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Jane-Eileen Joubert
- Normandie University, UNIROUEN, INSERM, DC2N, Rouen, France
- Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Marie-Christine Tonon
- Normandie University, UNIROUEN, INSERM, DC2N, Rouen, France
- Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Laurent Prézeau
- CNRS UMR 5203, INSERM U661, Institute of Functional Genomic (IGF), University of Montpellier 1 and 2, Montpellier, France
| | - Marie Chabbert
- UMR CNRS 6214, INSERM 1083, Faculté de Médecine 3, Angers, France
| | - Fabrice Morin
- Normandie University, UNIROUEN, INSERM, DC2N, Rouen, France
- Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Pierrick Gandolfo
- Normandie University, UNIROUEN, INSERM, DC2N, Rouen, France
- Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| |
Collapse
|
31
|
Orphan receptor ligand discovery by pickpocketing pharmacological neighbors. Nat Chem Biol 2016; 13:235-242. [PMID: 27992882 DOI: 10.1038/nchembio.2266] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Accepted: 10/11/2016] [Indexed: 12/20/2022]
Abstract
Understanding the pharmacological similarity of G protein-coupled receptors (GPCRs) is paramount for predicting ligand off-target effects, drug repurposing, and ligand discovery for orphan receptors. Phylogenetic relationships do not always correctly capture pharmacological similarity. Previous family-wide attempts to define pharmacological relationships were based on three-dimensional structures and/or known receptor-ligand pairings, both unavailable for orphan GPCRs. Here, we present GPCR-CoINPocket, a novel contact-informed neighboring pocket metric of GPCR binding-site similarity that is informed by patterns of ligand-residue interactions observed in crystallographically characterized GPCRs. GPCR-CoINPocket is applicable to receptors with unknown structure or ligands and accurately captures known pharmacological relationships between GPCRs, even those undetected by phylogeny. When applied to orphan receptor GPR37L1, GPCR-CoINPocket identified its pharmacological neighbors, and transfer of their pharmacology aided in discovery of the first surrogate ligands for this orphan with a 30% success rate. Although primarily designed for GPCRs, the method is easily transferable to other protein families.
Collapse
|
32
|
Lim H, Poleksic A, Yao Y, Tong H, He D, Zhuang L, Meng P, Xie L. Large-Scale Off-Target Identification Using Fast and Accurate Dual Regularized One-Class Collaborative Filtering and Its Application to Drug Repurposing. PLoS Comput Biol 2016; 12:e1005135. [PMID: 27716836 PMCID: PMC5055357 DOI: 10.1371/journal.pcbi.1005135] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 09/08/2016] [Indexed: 12/19/2022] Open
Abstract
Target-based screening is one of the major approaches in drug discovery. Besides the intended target, unexpected drug off-target interactions often occur, and many of them have not been recognized and characterized. The off-target interactions can be responsible for either therapeutic or side effects. Thus, identifying the genome-wide off-targets of lead compounds or existing drugs will be critical for designing effective and safe drugs, and providing new opportunities for drug repurposing. Although many computational methods have been developed to predict drug-target interactions, they are either less accurate than the one that we are proposing here or computationally too intensive, thereby limiting their capability for large-scale off-target identification. In addition, the performances of most machine learning based algorithms have been mainly evaluated to predict off-target interactions in the same gene family for hundreds of chemicals. It is not clear how these algorithms perform in terms of detecting off-targets across gene families on a proteome scale. Here, we are presenting a fast and accurate off-target prediction method, REMAP, which is based on a dual regularized one-class collaborative filtering algorithm, to explore continuous chemical space, protein space, and their interactome on a large scale. When tested in a reliable, extensive, and cross-gene family benchmark, REMAP outperforms the state-of-the-art methods. Furthermore, REMAP is highly scalable. It can screen a dataset of 200 thousands chemicals against 20 thousands proteins within 2 hours. Using the reconstructed genome-wide target profile as the fingerprint of a chemical compound, we predicted that seven FDA-approved drugs can be repurposed as novel anti-cancer therapies. The anti-cancer activity of six of them is supported by experimental evidences. Thus, REMAP is a valuable addition to the existing in silico toolbox for drug target identification, drug repurposing, phenotypic screening, and side effect prediction. The software and benchmark are available at https://github.com/hansaimlim/REMAP. High-throughput techniques have generated vast amounts of diverse omics and phenotypic data. However, these sets of data have not yet been fully explored to improve the effectiveness and efficiency of drug discovery, a process which has traditionally adopted a one-drug-one-gene paradigm. Consequently, the cost of bringing a drug to market is astounding and the failure rate is daunting. The failure of the target-based drug discovery is in large part due to the fact that a drug rarely interacts only with its intended receptor, but also generally binds to other receptors. To rationally design potent and safe therapeutics, we need to identify all the possible cellular proteins interacting with a drug in an organism. Existing experimental techniques are not sufficient to address this problem, and will benefit from computational modeling. However, it is a daunting task to reliably screen millions of chemicals against hundreds of thousands of proteins. Here, we introduce a fast and accurate method REMAP for large-scale predictions of drug-target interactions. REMAP outperforms state-of-the-art algorithms in terms of both speed and accuracy, and has been successfully applied to drug repurposing. Thus, REMAP may have broad applications in drug discovery.
Collapse
Affiliation(s)
- Hansaim Lim
- The Graduate Center, The City University of New York, New York, New York, United States
| | - Aleksandar Poleksic
- Department of Computer Science, University of Northern Iowa, Cedar Falls, Iowa, United States
| | - Yuan Yao
- Department of Computer Science and Technology, Nanjing University, Nanjing, Jiangsu, China
| | - Hanghang Tong
- School of Computing, Informatics and Decision Systems Engineering, Arizona State University, Tempe, Arizona, United States
| | - Di He
- The Graduate Center, The City University of New York, New York, New York, United States
| | - Luke Zhuang
- Academy for Information Technology, Union County Vocational-Technical Schools, Scotch Plains, New Jersey, United States
| | - Patrick Meng
- High Technology High School, Lincroft, New Jersey, United States
| | - Lei Xie
- The Graduate Center, The City University of New York, New York, New York, United States
- Department of Computer Science, Hunter College, The City University of New York, New York, New York, United States
- * E-mail:
| |
Collapse
|
33
|
Secondary pharmacology: screening and interpretation of off-target activities – focus on translation. Drug Discov Today 2016; 21:1232-42. [DOI: 10.1016/j.drudis.2016.04.021] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 03/22/2016] [Accepted: 04/22/2016] [Indexed: 12/19/2022]
|
34
|
Stern AM, Schurdak ME, Bahar I, Berg JM, Taylor DL. A Perspective on Implementing a Quantitative Systems Pharmacology Platform for Drug Discovery and the Advancement of Personalized Medicine. JOURNAL OF BIOMOLECULAR SCREENING 2016; 21:521-34. [PMID: 26962875 PMCID: PMC4917453 DOI: 10.1177/1087057116635818] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Drug candidates exhibiting well-defined pharmacokinetic and pharmacodynamic profiles that are otherwise safe often fail to demonstrate proof-of-concept in phase II and III trials. Innovation in drug discovery and development has been identified as a critical need for improving the efficiency of drug discovery, especially through collaborations between academia, government agencies, and industry. To address the innovation challenge, we describe a comprehensive, unbiased, integrated, and iterative quantitative systems pharmacology (QSP)-driven drug discovery and development strategy and platform that we have implemented at the University of Pittsburgh Drug Discovery Institute. Intrinsic to QSP is its integrated use of multiscale experimental and computational methods to identify mechanisms of disease progression and to test predicted therapeutic strategies likely to achieve clinical validation for appropriate subpopulations of patients. The QSP platform can address biological heterogeneity and anticipate the evolution of resistance mechanisms, which are major challenges for drug development. The implementation of this platform is dedicated to gaining an understanding of mechanism(s) of disease progression to enable the identification of novel therapeutic strategies as well as repurposing drugs. The QSP platform will help promote the paradigm shift from reactive population-based medicine to proactive personalized medicine by focusing on the patient as the starting and the end point.
Collapse
Affiliation(s)
- Andrew M. Stern
- Department of Computational and Systems Biology, Pittsburgh, PA, USA
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
| | - Mark E. Schurdak
- Department of Computational and Systems Biology, Pittsburgh, PA, USA
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
- The University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Ivet Bahar
- Department of Computational and Systems Biology, Pittsburgh, PA, USA
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
- The University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Jeremy M. Berg
- Department of Computational and Systems Biology, Pittsburgh, PA, USA
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
- University of Pittsburgh Institute for Personalized Medicine, Pittsburgh, PA, USA
| | - D. Lansing Taylor
- Department of Computational and Systems Biology, Pittsburgh, PA, USA
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
- The University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| |
Collapse
|
35
|
Wang Z, Liang L, Yin Z, Lin J. Improving chemical similarity ensemble approach in target prediction. J Cheminform 2016; 8:20. [PMID: 27110288 PMCID: PMC4842302 DOI: 10.1186/s13321-016-0130-x] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 04/04/2016] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND In silico target prediction of compounds plays an important role in drug discovery. The chemical similarity ensemble approach (SEA) is a promising method, which has been successfully applied in many drug-related studies. There are various models available analogous to SEA, because this approach is based on different types of molecular fingerprints. To investigate the influence of training data selection and the complementarity of different models, several SEA models were constructed and tested. RESULTS When we used a test set of 37,138 positive and 42,928 negative ligand-target interactions, among the five tested molecular fingerprint methods, at significance level 0.05, Topological-based model yielded the best precision rate (83.7 %) and [Formula: see text] (0.784) while Atom pair-based model yielded the best [Formula: see text] (0.694). By employing an election system to combine the five models, a flexible prediction scheme was achieved with precision range from 71 to 90.6 %, [Formula: see text] range from 0.663 to 0.684 and [Formula: see text] range from 0.696 to 0.817. CONCLUSIONS The overall effectiveness of all of the five models could be ranked in decreasing order as follows: Atom pair [Formula: see text] Topological > Morgan > MACCS > Pharmacophore. Combining multiple SEA models, which takes advantages of different models, could be used to improve the success rates of the models. Another possibility of improving the model could be using target-specific classes or more active compounds.
Collapse
Affiliation(s)
- Zhonghua Wang
- />State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Weijin Road, Tianjin, China
| | - Lu Liang
- />State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Weijin Road, Tianjin, China
- />High-Throughput Molecular Drug Discovery Center, Tianjin Joint Academy of Biomedicine and Technology, Tianjin, China
| | - Zheng Yin
- />State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Weijin Road, Tianjin, China
| | - Jianping Lin
- />State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Weijin Road, Tianjin, China
- />High-Throughput Molecular Drug Discovery Center, Tianjin Joint Academy of Biomedicine and Technology, Tianjin, China
| |
Collapse
|
36
|
Farrell MS, McCorvy JD, Huang XP, Urban DJ, White KL, Giguere PM, Doak AK, Bernstein AI, Stout KA, Park SM, Rodriguiz RM, Gray BW, Hyatt WS, Norwood AP, Webster KA, Gannon BM, Miller GW, Porter JH, Shoichet BK, Fantegrossi WE, Wetsel WC, Roth BL. In Vitro and In Vivo Characterization of the Alkaloid Nuciferine. PLoS One 2016; 11:e0150602. [PMID: 26963248 PMCID: PMC4786259 DOI: 10.1371/journal.pone.0150602] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 02/17/2016] [Indexed: 01/05/2023] Open
Abstract
Rationale The sacred lotus (Nelumbo nucifera) contains many phytochemicals and has a history of human use. To determine which compounds may be responsible for reported psychotropic effects, we used in silico predictions of the identified phytochemicals. Nuciferine, an alkaloid component of Nelumbo nucifera and Nymphaea caerulea, had a predicted molecular profile similar to antipsychotic compounds. Our study characterizes nuciferine using in vitro and in vivo pharmacological assays. Methods Nuciferine was first characterized in silico using the similarity ensemble approach, and was followed by further characterization and validation using the Psychoactive Drug Screening Program of the National Institute of Mental Health. Nuciferine was then tested in vivo in the head-twitch response, pre-pulse inhibition, hyperlocomotor activity, and drug discrimination paradigms. Results Nuciferine shares a receptor profile similar to aripiprazole-like antipsychotic drugs. Nuciferine was an antagonist at 5-HT2A, 5-HT2C, and 5-HT2B, an inverse agonist at 5-HT7, a partial agonist at D2, D5 and 5-HT6, an agonist at 5-HT1A and D4 receptors, and inhibited the dopamine transporter. In rodent models relevant to antipsychotic drug action, nuciferine blocked head-twitch responses and discriminative stimulus effects of a 5-HT2A agonist, substituted for clozapine discriminative stimulus, enhanced amphetamine induced locomotor activity, inhibited phencyclidine (PCP)-induced locomotor activity, and rescued PCP-induced disruption of prepulse inhibition without induction of catalepsy. Conclusions The molecular profile of nuciferine was similar but not identical to that shared with several approved antipsychotic drugs suggesting that nuciferine has atypical antipsychotic-like actions.
Collapse
Affiliation(s)
- Martilias S. Farrell
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, United States of America
- * E-mail:
| | - John D. McCorvy
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, United States of America
| | - Xi-Ping Huang
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, United States of America
- National Institute of Mental Health Psychoactive Drug Screening Program, University of North Carolina School of Medicine, Chapel Hill, North Carolina, United States of America
| | - Daniel J. Urban
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, United States of America
| | - Kate L. White
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, United States of America
| | - Patrick M. Giguere
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, United States of America
| | - Allison K. Doak
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, United States of America
| | - Alison I. Bernstein
- Department of Environmental Health, Rollins School of Public Health and Center for Neurodegenerative Diseases, Emory University, Atlanta, Georgia, United States of America
| | - Kristen A. Stout
- Department of Environmental Health, Rollins School of Public Health and Center for Neurodegenerative Diseases, Emory University, Atlanta, Georgia, United States of America
| | - Su Mi Park
- Departments of Psychiatry and Behavioral Sciences, Cell Biology, and Neurobiology, Mouse Behavioral and Neuroendocrine Analysis Core Facility, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Ramona M. Rodriguiz
- Departments of Psychiatry and Behavioral Sciences, Cell Biology, and Neurobiology, Mouse Behavioral and Neuroendocrine Analysis Core Facility, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Bradley W. Gray
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - William S. Hyatt
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Andrew P. Norwood
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Kevin A. Webster
- Department of Psychology, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Brenda M. Gannon
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Gary W. Miller
- Department of Environmental Health, Rollins School of Public Health and Center for Neurodegenerative Diseases, Emory University, Atlanta, Georgia, United States of America
| | - Joseph H. Porter
- Department of Psychology, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Brian K. Shoichet
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, United States of America
| | - William E. Fantegrossi
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - William C. Wetsel
- Departments of Psychiatry and Behavioral Sciences, Cell Biology, and Neurobiology, Mouse Behavioral and Neuroendocrine Analysis Core Facility, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Bryan L. Roth
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, United States of America
- Department of Psychiatry, University of North Carolina School of Medicine, Chapel Hill, North Carolina, United States of America
- Program in Neuroscience, University of North Carolina School of Medicine, Chapel Hill, North Carolina, United States of America
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina, United States of America
- Carolina Institute for Developmental Disabilities, University of North Carolina School of Medicine, Chapel Hill, North Carolina, United States of America
- Division of Chemical Biology and Medicinal Chemistry, School of Pharmacy, University of North Carolina School of Medicine, Chapel Hill, North Carolina, United States of America
- National Institute of Mental Health Psychoactive Drug Screening Program, University of North Carolina School of Medicine, Chapel Hill, North Carolina, United States of America
| |
Collapse
|
37
|
Hart T, Xie L. Providing data science support for systems pharmacology and its implications to drug discovery. Expert Opin Drug Discov 2016; 11:241-56. [PMID: 26689499 DOI: 10.1517/17460441.2016.1135126] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION The conventional one-drug-one-target-one-disease drug discovery process has been less successful in tracking multi-genic, multi-faceted complex diseases. Systems pharmacology has emerged as a new discipline to tackle the current challenges in drug discovery. The goal of systems pharmacology is to transform huge, heterogeneous, and dynamic biological and clinical data into interpretable and actionable mechanistic models for decision making in drug discovery and patient treatment. Thus, big data technology and data science will play an essential role in systems pharmacology. AREAS COVERED This paper critically reviews the impact of three fundamental concepts of data science on systems pharmacology: similarity inference, overfitting avoidance, and disentangling causality from correlation. The authors then discuss recent advances and future directions in applying the three concepts of data science to drug discovery, with a focus on proteome-wide context-specific quantitative drug target deconvolution and personalized adverse drug reaction prediction. EXPERT OPINION Data science will facilitate reducing the complexity of systems pharmacology modeling, detecting hidden correlations between complex data sets, and distinguishing causation from correlation. The power of data science can only be fully realized when integrated with mechanism-based multi-scale modeling that explicitly takes into account the hierarchical organization of biological systems from nucleic acid to proteins, to molecular interaction networks, to cells, to tissues, to patients, and to populations.
Collapse
Affiliation(s)
- Thomas Hart
- a The Rockefeller University , New York , NY , USA.,b Department of Biological Sciences, Hunter College , The City University of New York , New York , NY , USA
| | - Lei Xie
- c Department of Computer Science, Hunter College , The City University of New York , New York , NY , USA.,d The Graduate Center , The City University of New York , New York , NY , USA
| |
Collapse
|
38
|
Gaspar HA, Sidorov P, Horvath D, Baskin II, Marcou G, Varnek A. Generative Topographic Mapping Approach to Chemical Space Analysis. FRONTIERS IN MOLECULAR DESIGN AND CHEMICAL INFORMATION SCIENCE - HERMAN SKOLNIK AWARD SYMPOSIUM 2015: JÜRGEN BAJORATH 2016. [DOI: 10.1021/bk-2016-1222.ch011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Affiliation(s)
- Héléna A. Gaspar
- Laboratoire de Chemoinformatique, UMR 7140, Université de Strasbourg, 1 rue Blaise Pascal, Strasbourg 67000, France
- Faculty of Physics, M.V. Lomonosov Moscow State University, Leninskie Gory, Moscow 119991, Russia
- Laboratory of Chemoinformatics, Butlerov Institute of Chemistry, Kazan Federal University, Kazan, Russia
| | - Pavel Sidorov
- Laboratoire de Chemoinformatique, UMR 7140, Université de Strasbourg, 1 rue Blaise Pascal, Strasbourg 67000, France
- Faculty of Physics, M.V. Lomonosov Moscow State University, Leninskie Gory, Moscow 119991, Russia
- Laboratory of Chemoinformatics, Butlerov Institute of Chemistry, Kazan Federal University, Kazan, Russia
| | - Dragos Horvath
- Laboratoire de Chemoinformatique, UMR 7140, Université de Strasbourg, 1 rue Blaise Pascal, Strasbourg 67000, France
- Faculty of Physics, M.V. Lomonosov Moscow State University, Leninskie Gory, Moscow 119991, Russia
- Laboratory of Chemoinformatics, Butlerov Institute of Chemistry, Kazan Federal University, Kazan, Russia
| | - Igor I. Baskin
- Laboratoire de Chemoinformatique, UMR 7140, Université de Strasbourg, 1 rue Blaise Pascal, Strasbourg 67000, France
- Faculty of Physics, M.V. Lomonosov Moscow State University, Leninskie Gory, Moscow 119991, Russia
- Laboratory of Chemoinformatics, Butlerov Institute of Chemistry, Kazan Federal University, Kazan, Russia
| | - Gilles Marcou
- Laboratoire de Chemoinformatique, UMR 7140, Université de Strasbourg, 1 rue Blaise Pascal, Strasbourg 67000, France
- Faculty of Physics, M.V. Lomonosov Moscow State University, Leninskie Gory, Moscow 119991, Russia
- Laboratory of Chemoinformatics, Butlerov Institute of Chemistry, Kazan Federal University, Kazan, Russia
| | - Alexandre Varnek
- Laboratoire de Chemoinformatique, UMR 7140, Université de Strasbourg, 1 rue Blaise Pascal, Strasbourg 67000, France
- Faculty of Physics, M.V. Lomonosov Moscow State University, Leninskie Gory, Moscow 119991, Russia
- Laboratory of Chemoinformatics, Butlerov Institute of Chemistry, Kazan Federal University, Kazan, Russia
| |
Collapse
|
39
|
Barelier S, Sterling T, O’Meara MJ, Shoichet BK. The Recognition of Identical Ligands by Unrelated Proteins. ACS Chem Biol 2015; 10:2772-84. [PMID: 26421501 DOI: 10.1021/acschembio.5b00683] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The binding of drugs and reagents to off-targets is well-known. Whereas many off-targets are related to the primary target by sequence and fold, many ligands bind to unrelated pairs of proteins, and these are harder to anticipate. If the binding site in the off-target can be related to that of the primary target, this challenge resolves into aligning the two pockets. However, other cases are possible: the ligand might interact with entirely different residues and environments in the off-target, or wholly different ligand atoms may be implicated in the two complexes. To investigate these scenarios at atomic resolution, the structures of 59 ligands in 116 complexes (62 pairs in total), where the protein pairs were unrelated by fold but bound an identical ligand, were examined. In almost half of the pairs, the ligand interacted with unrelated residues in the two proteins (29 pairs), and in 14 of the pairs wholly different ligand moieties were implicated in each complex. Even in those 19 pairs of complexes that presented similar environments to the ligand, ligand superposition rarely resulted in the overlap of related residues. There appears to be no single pattern-matching "code" for identifying binding sites in unrelated proteins that bind identical ligands, though modeling suggests that there might be a limited number of different patterns that suffice to recognize different ligand functional groups.
Collapse
Affiliation(s)
- Sarah Barelier
- Department of Pharmaceutical
Chemistry, University of California San Francisco, 1700 Fourth
Street, Byers Hall, San Francisco, California 94158, United States
| | - Teague Sterling
- Department of Pharmaceutical
Chemistry, University of California San Francisco, 1700 Fourth
Street, Byers Hall, San Francisco, California 94158, United States
| | - Matthew J. O’Meara
- Department of Pharmaceutical
Chemistry, University of California San Francisco, 1700 Fourth
Street, Byers Hall, San Francisco, California 94158, United States
| | - Brian K. Shoichet
- Department of Pharmaceutical
Chemistry, University of California San Francisco, 1700 Fourth
Street, Byers Hall, San Francisco, California 94158, United States
| |
Collapse
|
40
|
Sidorov P, Gaspar H, Marcou G, Varnek A, Horvath D. Mappability of drug-like space: towards a polypharmacologically competent map of drug-relevant compounds. J Comput Aided Mol Des 2015; 29:1087-108. [PMID: 26564142 DOI: 10.1007/s10822-015-9882-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 11/06/2015] [Indexed: 11/30/2022]
Abstract
Intuitive, visual rendering--mapping--of high-dimensional chemical spaces (CS), is an important topic in chemoinformatics. Such maps were so far dedicated to specific compound collections--either limited series of known activities, or large, even exhaustive enumerations of molecules, but without associated property data. Typically, they were challenged to answer some classification problem with respect to those same molecules, admired for their aesthetical virtues and then forgotten--because they were set-specific constructs. This work wishes to address the question whether a general, compound set-independent map can be generated, and the claim of "universality" quantitatively justified, with respect to all the structure-activity information available so far--or, more realistically, an exploitable but significant fraction thereof. The "universal" CS map is expected to project molecules from the initial CS into a lower-dimensional space that is neighborhood behavior-compliant with respect to a large panel of ligand properties. Such map should be able to discriminate actives from inactives, or even support quantitative neighborhood-based, parameter-free property prediction (regression) models, for a wide panel of targets and target families. It should be polypharmacologically competent, without requiring any target-specific parameter fitting. This work describes an evolutionary growth procedure of such maps, based on generative topographic mapping, followed by the validation of their polypharmacological competence. Validation was achieved with respect to a maximum of exploitable structure-activity information, covering all of Homo sapiens proteins of the ChEMBL database, antiparasitic and antiviral data, etc. Five evolved maps satisfactorily solved hundreds of activity-based ligand classification challenges for targets, and even in vivo properties independent from training data. They also stood chemogenomics-related challenges, as cumulated responsibility vectors obtained by mapping of target-specific ligand collections were shown to represent validated target descriptors, complying with currently accepted target classification in biology. Therefore, they represent, in our opinion, a robust and well documented answer to the key question "What is a good CS map?"
Collapse
Affiliation(s)
- Pavel Sidorov
- Laboratoire de Chémoinformatique, UMR 7140, CNRS-Univ. Strasbourg, 1 rue Blaise Pascal, 67000, Strasbourg, France.,Laboratory of Chemoinformatics, Butlerov Institute of Chemistry, Kazan Federal University, Kazan, Russia
| | - Helena Gaspar
- Laboratoire de Chémoinformatique, UMR 7140, CNRS-Univ. Strasbourg, 1 rue Blaise Pascal, 67000, Strasbourg, France
| | - Gilles Marcou
- Laboratoire de Chémoinformatique, UMR 7140, CNRS-Univ. Strasbourg, 1 rue Blaise Pascal, 67000, Strasbourg, France
| | - Alexandre Varnek
- Laboratoire de Chémoinformatique, UMR 7140, CNRS-Univ. Strasbourg, 1 rue Blaise Pascal, 67000, Strasbourg, France.,Laboratory of Chemoinformatics, Butlerov Institute of Chemistry, Kazan Federal University, Kazan, Russia
| | - Dragos Horvath
- Laboratoire de Chémoinformatique, UMR 7140, CNRS-Univ. Strasbourg, 1 rue Blaise Pascal, 67000, Strasbourg, France.
| |
Collapse
|
41
|
Gamo AM, González-Vera JA, Rueda-Zubiaurre A, Alonso D, Vázquez-Villa H, Martín-Couce L, Palomares Ó, López JA, Martín-Fontecha M, Benhamú B, López-Rodríguez ML, Ortega-Gutiérrez S. Chemoproteomic Approach to Explore the Target Profile of GPCR ligands: Application to 5-HT1A
and 5-HT6
Receptors. Chemistry 2015; 22:1313-21. [DOI: 10.1002/chem.201503101] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Indexed: 12/20/2022]
Affiliation(s)
- Ana M. Gamo
- Departamento de Química Orgánica I; Facultad de Ciencias Químicas; Universidad Complutense de Madrid; 28040 Madrid Spain
| | - Juan A. González-Vera
- Departamento de Química Orgánica I; Facultad de Ciencias Químicas; Universidad Complutense de Madrid; 28040 Madrid Spain
| | - Ainoa Rueda-Zubiaurre
- Departamento de Química Orgánica I; Facultad de Ciencias Químicas; Universidad Complutense de Madrid; 28040 Madrid Spain
| | - Dulce Alonso
- Departamento de Química Orgánica I; Facultad de Ciencias Químicas; Universidad Complutense de Madrid; 28040 Madrid Spain
| | - Henar Vázquez-Villa
- Departamento de Química Orgánica I; Facultad de Ciencias Químicas; Universidad Complutense de Madrid; 28040 Madrid Spain
| | - Lidia Martín-Couce
- Departamento de Química Orgánica I; Facultad de Ciencias Químicas; Universidad Complutense de Madrid; 28040 Madrid Spain
| | - Óscar Palomares
- Departamento de Bioquímica y Biología Molecular I; Facultad de Ciencias Químicas; Universidad Complutense de Madrid; 28040 Madrid Spain
| | - Juan A. López
- Proteomics Unit; Centro Nacional de Investigaciones Cardiovasculares, CNIC; 28029 Madrid Spain
| | - Mar Martín-Fontecha
- Departamento de Química Orgánica I; Facultad de Ciencias Químicas; Universidad Complutense de Madrid; 28040 Madrid Spain
| | - Bellinda Benhamú
- Departamento de Química Orgánica I; Facultad de Ciencias Químicas; Universidad Complutense de Madrid; 28040 Madrid Spain
| | - María L. López-Rodríguez
- Departamento de Química Orgánica I; Facultad de Ciencias Químicas; Universidad Complutense de Madrid; 28040 Madrid Spain
| | - Silvia Ortega-Gutiérrez
- Departamento de Química Orgánica I; Facultad de Ciencias Químicas; Universidad Complutense de Madrid; 28040 Madrid Spain
| |
Collapse
|
42
|
Irwin JJ, Duan D, Torosyan H, Doak AK, Ziebart KT, Sterling T, Tumanian G, Shoichet BK. An Aggregation Advisor for Ligand Discovery. J Med Chem 2015; 58:7076-87. [PMID: 26295373 DOI: 10.1021/acs.jmedchem.5b01105] [Citation(s) in RCA: 323] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Colloidal aggregation of organic molecules is the dominant mechanism for artifactual inhibition of proteins, and controls against it are widely deployed. Notwithstanding an increasingly detailed understanding of this phenomenon, a method to reliably predict aggregation has remained elusive. Correspondingly, active molecules that act via aggregation continue to be found in early discovery campaigns and remain common in the literature. Over the past decade, over 12 thousand aggregating organic molecules have been identified, potentially enabling a precedent-based approach to match known aggregators with new molecules that may be expected to aggregate and lead to artifacts. We investigate an approach that uses lipophilicity, affinity, and similarity to known aggregators to advise on the likelihood that a candidate compound is an aggregator. In prospective experimental testing, five of seven new molecules with Tanimoto coefficients (Tc's) between 0.95 and 0.99 to known aggregators aggregated at relevant concentrations. Ten of 19 with Tc's between 0.94 and 0.90 and three of seven with Tc's between 0.89 and 0.85 also aggregated. Another three of the predicted compounds aggregated at higher concentrations. This method finds that 61 827 or 5.1% of the ligands acting in the 0.1 to 10 μM range in the medicinal chemistry literature are at least 85% similar to a known aggregator with these physical properties and may aggregate at relevant concentrations. Intriguingly, only 0.73% of all drug-like commercially available compounds resemble the known aggregators, suggesting that colloidal aggregators are enriched in the literature. As a percentage of the literature, aggregator-like compounds have increased 9-fold since 1995, partly reflecting the advent of high-throughput and virtual screens against molecular targets. Emerging from this study is an aggregator advisor database and tool ( http://advisor.bkslab.org ), free to the community, that may help distinguish between fruitful and artifactual screening hits acting by this mechanism.
Collapse
Affiliation(s)
- John J Irwin
- Department of Pharmaceutical Chemistry, University of California, San Francisco , Byers Hall, 1700 4th St, San Francisco, California 94158-2550, United States
| | - Da Duan
- Department of Pharmaceutical Chemistry, University of California, San Francisco , Byers Hall, 1700 4th St, San Francisco, California 94158-2550, United States
| | - Hayarpi Torosyan
- Department of Pharmaceutical Chemistry, University of California, San Francisco , Byers Hall, 1700 4th St, San Francisco, California 94158-2550, United States
| | - Allison K Doak
- Department of Pharmaceutical Chemistry, University of California, San Francisco , Byers Hall, 1700 4th St, San Francisco, California 94158-2550, United States
| | - Kristin T Ziebart
- Department of Pharmaceutical Chemistry, University of California, San Francisco , Byers Hall, 1700 4th St, San Francisco, California 94158-2550, United States
| | - Teague Sterling
- Department of Pharmaceutical Chemistry, University of California, San Francisco , Byers Hall, 1700 4th St, San Francisco, California 94158-2550, United States
| | - Gurgen Tumanian
- Department of Pharmaceutical Chemistry, University of California, San Francisco , Byers Hall, 1700 4th St, San Francisco, California 94158-2550, United States
| | - Brian K Shoichet
- Department of Pharmaceutical Chemistry, University of California, San Francisco , Byers Hall, 1700 4th St, San Francisco, California 94158-2550, United States
| |
Collapse
|
43
|
Abstract
INTRODUCTION Over the past three decades, the predominant paradigm in drug discovery was designing selective ligands for a specific target to avoid unwanted side effects. However, in the last 5 years, the aim has shifted to take into account the biological network in which they interact. Quantitative and Systems Pharmacology (QSP) is a new paradigm that aims to understand how drugs modulate cellular networks in space and time, in order to predict drug targets and their role in human pathophysiology. AREAS COVERED This review discusses existing computational and experimental QSP approaches such as polypharmacology techniques combined with systems biology information and considers the use of new tools and ideas in a wider 'systems-level' context in order to design new drugs with improved efficacy and fewer unwanted off-target effects. EXPERT OPINION The use of network biology produces valuable information such as new indications for approved drugs, drug-drug interactions, proteins-drug side effects and pathways-gene associations. However, we are still far from the aim of QSP, both because of the huge effort needed to model precisely biological network models and the limited accuracy that we are able to reach with those. Hence, moving from 'one molecule for one target to give one therapeutic effect' to the 'big systems-based picture' seems obvious moving forward although whether our current tools are sufficient for such a step is still under debate.
Collapse
Affiliation(s)
- Violeta I Pérez-Nueno
- a Harmonic Pharma, Espace Transfert , 615 rue du Jardin Botanique, 54600 Villers lès Nancy, France +33 354 958 604 ; +33 383 593 046 ;
| |
Collapse
|
44
|
Insel PA, Wilderman A, Zambon AC, Snead AN, Murray F, Aroonsakool N, McDonald DS, Zhou S, McCann T, Zhang L, Sriram K, Chinn AM, Michkov AV, Lynch RM, Overland AC, Corriden R. G Protein-Coupled Receptor (GPCR) Expression in Native Cells: "Novel" endoGPCRs as Physiologic Regulators and Therapeutic Targets. Mol Pharmacol 2015; 88:181-7. [PMID: 25737495 PMCID: PMC4468643 DOI: 10.1124/mol.115.098129] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 03/02/2015] [Indexed: 12/24/2022] Open
Abstract
G protein-coupled receptors (GPCRs), the largest family of signaling receptors in the human genome, are also the largest class of targets of approved drugs. Are the optimal GPCRs (in terms of efficacy and safety) currently targeted therapeutically? Especially given the large number (∼ 120) of orphan GPCRs (which lack known physiologic agonists), it is likely that previously unrecognized GPCRs, especially orphan receptors, regulate cell function and can be therapeutic targets. Knowledge is limited regarding the diversity and identity of GPCRs that are activated by endogenous ligands and that native cells express. Here, we review approaches to define GPCR expression in tissues and cells and results from studies using these approaches. We identify problems with the available data and suggest future ways to identify and validate the physiologic and therapeutic roles of previously unrecognized GPCRs. We propose that a particularly useful approach to identify functionally important GPCRs with therapeutic potential will be to focus on receptors that show selective increases in expression in diseased cells from patients and experimental animals.
Collapse
Affiliation(s)
- Paul A Insel
- Departments of Pharmacology (P.A.I., A.W., A.C.Z., A.N.S., N.A., D.S.M., S.Z., T.M., L.Z., K.S., A.M.C., A.V.M., R.M.L., A.C.O., R.C.) and Medicine (P.A.I., F.M.), University of California, San Diego, La Jolla, California
| | - Andrea Wilderman
- Departments of Pharmacology (P.A.I., A.W., A.C.Z., A.N.S., N.A., D.S.M., S.Z., T.M., L.Z., K.S., A.M.C., A.V.M., R.M.L., A.C.O., R.C.) and Medicine (P.A.I., F.M.), University of California, San Diego, La Jolla, California
| | - Alexander C Zambon
- Departments of Pharmacology (P.A.I., A.W., A.C.Z., A.N.S., N.A., D.S.M., S.Z., T.M., L.Z., K.S., A.M.C., A.V.M., R.M.L., A.C.O., R.C.) and Medicine (P.A.I., F.M.), University of California, San Diego, La Jolla, California
| | - Aaron N Snead
- Departments of Pharmacology (P.A.I., A.W., A.C.Z., A.N.S., N.A., D.S.M., S.Z., T.M., L.Z., K.S., A.M.C., A.V.M., R.M.L., A.C.O., R.C.) and Medicine (P.A.I., F.M.), University of California, San Diego, La Jolla, California
| | - Fiona Murray
- Departments of Pharmacology (P.A.I., A.W., A.C.Z., A.N.S., N.A., D.S.M., S.Z., T.M., L.Z., K.S., A.M.C., A.V.M., R.M.L., A.C.O., R.C.) and Medicine (P.A.I., F.M.), University of California, San Diego, La Jolla, California
| | - Nakon Aroonsakool
- Departments of Pharmacology (P.A.I., A.W., A.C.Z., A.N.S., N.A., D.S.M., S.Z., T.M., L.Z., K.S., A.M.C., A.V.M., R.M.L., A.C.O., R.C.) and Medicine (P.A.I., F.M.), University of California, San Diego, La Jolla, California
| | - Daniel S McDonald
- Departments of Pharmacology (P.A.I., A.W., A.C.Z., A.N.S., N.A., D.S.M., S.Z., T.M., L.Z., K.S., A.M.C., A.V.M., R.M.L., A.C.O., R.C.) and Medicine (P.A.I., F.M.), University of California, San Diego, La Jolla, California
| | - Shu Zhou
- Departments of Pharmacology (P.A.I., A.W., A.C.Z., A.N.S., N.A., D.S.M., S.Z., T.M., L.Z., K.S., A.M.C., A.V.M., R.M.L., A.C.O., R.C.) and Medicine (P.A.I., F.M.), University of California, San Diego, La Jolla, California
| | - Thalia McCann
- Departments of Pharmacology (P.A.I., A.W., A.C.Z., A.N.S., N.A., D.S.M., S.Z., T.M., L.Z., K.S., A.M.C., A.V.M., R.M.L., A.C.O., R.C.) and Medicine (P.A.I., F.M.), University of California, San Diego, La Jolla, California
| | - Lingzhi Zhang
- Departments of Pharmacology (P.A.I., A.W., A.C.Z., A.N.S., N.A., D.S.M., S.Z., T.M., L.Z., K.S., A.M.C., A.V.M., R.M.L., A.C.O., R.C.) and Medicine (P.A.I., F.M.), University of California, San Diego, La Jolla, California
| | - Krishna Sriram
- Departments of Pharmacology (P.A.I., A.W., A.C.Z., A.N.S., N.A., D.S.M., S.Z., T.M., L.Z., K.S., A.M.C., A.V.M., R.M.L., A.C.O., R.C.) and Medicine (P.A.I., F.M.), University of California, San Diego, La Jolla, California
| | - Amy M Chinn
- Departments of Pharmacology (P.A.I., A.W., A.C.Z., A.N.S., N.A., D.S.M., S.Z., T.M., L.Z., K.S., A.M.C., A.V.M., R.M.L., A.C.O., R.C.) and Medicine (P.A.I., F.M.), University of California, San Diego, La Jolla, California
| | - Alexander V Michkov
- Departments of Pharmacology (P.A.I., A.W., A.C.Z., A.N.S., N.A., D.S.M., S.Z., T.M., L.Z., K.S., A.M.C., A.V.M., R.M.L., A.C.O., R.C.) and Medicine (P.A.I., F.M.), University of California, San Diego, La Jolla, California
| | - Rebecca M Lynch
- Departments of Pharmacology (P.A.I., A.W., A.C.Z., A.N.S., N.A., D.S.M., S.Z., T.M., L.Z., K.S., A.M.C., A.V.M., R.M.L., A.C.O., R.C.) and Medicine (P.A.I., F.M.), University of California, San Diego, La Jolla, California
| | - Aaron C Overland
- Departments of Pharmacology (P.A.I., A.W., A.C.Z., A.N.S., N.A., D.S.M., S.Z., T.M., L.Z., K.S., A.M.C., A.V.M., R.M.L., A.C.O., R.C.) and Medicine (P.A.I., F.M.), University of California, San Diego, La Jolla, California
| | - Ross Corriden
- Departments of Pharmacology (P.A.I., A.W., A.C.Z., A.N.S., N.A., D.S.M., S.Z., T.M., L.Z., K.S., A.M.C., A.V.M., R.M.L., A.C.O., R.C.) and Medicine (P.A.I., F.M.), University of California, San Diego, La Jolla, California
| |
Collapse
|
45
|
Abstract
Adenosine is an ubiquitous nucleoside involved in various physiological and pathological functions by stimulating A1, A2A, A2B and A3 adenosine receptors (ARs). Allosteric enhancers to A1ARs may represent novel therapeutic agents because they increase the activity of these receptors by mediating a shift to their active form in the A1AR-G protein ternary complex. In this manner, they are able to amplify the action of endogenous adenosine, which is produced in high concentrations under conditions of metabolic stress. A1AR allosteric enhancers could be used as a justifiable alternative to the exogenous agonists that are characterized by receptor desensitization and downregulation. In this review, an analysis of some of the most interesting allosteric modulators of A1ARs has been reported.
Collapse
|
46
|
Roth BL, Kroeze WK. Integrated Approaches for Genome-wide Interrogation of the Druggable Non-olfactory G Protein-coupled Receptor Superfamily. J Biol Chem 2015; 290:19471-7. [PMID: 26100629 DOI: 10.1074/jbc.r115.654764] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
G-protein-coupled receptors (GPCRs) are frequent and fruitful targets for drug discovery and development, as well as being off-targets for the side effects of a variety of medications. Much of the druggable non-olfactory human GPCR-ome remains under-interrogated, and we present here various approaches that we and others have used to shine light into these previously dark corners of the human genome.
Collapse
Affiliation(s)
- Bryan L Roth
- From the Department of Pharmacology, University of North Carolina Chapel Hill School of Medicine, Chapel Hill, North Carolina 27514
| | - Wesley K Kroeze
- From the Department of Pharmacology, University of North Carolina Chapel Hill School of Medicine, Chapel Hill, North Carolina 27514
| |
Collapse
|
47
|
Öztürk H, Ozkirimli E, Özgür A. Classification of Beta-lactamases and penicillin binding proteins using ligand-centric network models. PLoS One 2015; 10:e0117874. [PMID: 25689853 PMCID: PMC4331424 DOI: 10.1371/journal.pone.0117874] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 01/03/2015] [Indexed: 01/28/2023] Open
Abstract
β-lactamase mediated antibiotic resistance is an important health issue and the discovery of new β-lactam type antibiotics or β-lactamase inhibitors is an area of intense research. Today, there are about a thousand β-lactamases due to the evolutionary pressure exerted by these ligands. While β-lactamases hydrolyse the β-lactam ring of antibiotics, rendering them ineffective, Penicillin-Binding Proteins (PBPs), which share high structural similarity with β-lactamases, also confer antibiotic resistance to their host organism by acquiring mutations that allow them to continue their participation in cell wall biosynthesis. In this paper, we propose a novel approach to include ligand sharing information for classifying and clustering β-lactamases and PBPs in an effort to elucidate the ligand induced evolution of these β-lactam binding proteins. We first present a detailed summary of the β-lactamase and PBP families in the Protein Data Bank, as well as the compounds they bind to. Then, we build two different types of networks in which the proteins are represented as nodes, and two proteins are connected by an edge with a weight that depends on the number of shared identical or similar ligands. These models are analyzed under three different edge weight settings, namely unweighted, weighted, and normalized weighted. A detailed comparison of these six networks showed that the use of ligand sharing information to cluster proteins resulted in modules comprising proteins with not only sequence similarity but also functional similarity. Consideration of ligand similarity highlighted some interactions that were not detected in the identical ligand network. Analysing the β-lactamases and PBPs using ligand-centric network models enabled the identification of novel relationships, suggesting that these models can be used to examine other protein families to obtain information on their ligand induced evolutionary paths.
Collapse
Affiliation(s)
- Hakime Öztürk
- Department of Computer Engineering, Bogazici University, Istanbul, Bebek, Turkey
| | - Elif Ozkirimli
- Department of Chemical Engineering, Bogazici University, Istanbul, Bebek, Turkey
- * E-mail: (EO), (AÖ)
| | - Arzucan Özgür
- Department of Computer Engineering, Bogazici University, Istanbul, Bebek, Turkey
- * E-mail: (EO), (AÖ)
| |
Collapse
|
48
|
Antolín AA, Mestres J. Linking off-target kinase pharmacology to the differential cellular effects observed among PARP inhibitors. Oncotarget 2015; 5:3023-8. [PMID: 24632590 PMCID: PMC4102788 DOI: 10.18632/oncotarget.1814] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
PARP inhibitors hold promise as a novel class of targeted anticancer drugs. However, their true mechanism of action is still not well understood following recent reports that show marked differences in cellular effects. Here, we demonstrate that three PARP drug candidates, namely, rucaparib, veliparib, and olaparib, have a clearly different in vitro affinity profile across a panel of diverse kinases selected using a computational approach that relates proteins by ligand similarity. In this respect, rucaparib inhibits nine kinases with micromolar affinity, including PIM1, PIM2, PRKD2, DYRK1A, CDK1, CDK9, HIPK2, CK2, and ALK. In contrast, olaparib does not inhibit any of the sixteen kinases tested. In between, veliparib inhibits only two, namely, PIM1 and CDK9. The differential kinase pharmacology observed among PARP inhibitors provides a plausible explanation to their different cellular effects and offers unexplored opportunities for this drug class, but alerts also on the risk associated to transferring directly both preclinical and clinical outcomes from one PARP drug candidate to another.
Collapse
Affiliation(s)
- Albert A Antolín
- Systems Pharmacology, Research Program on Biomedical Informatics, IMIM Hospital del Mar Medical Research Institute and Universitat Pompeu Fabra, Doctor Aiguader 88, 08003 Barcelona, Catalonia, Spain
| | | |
Collapse
|
49
|
Soriano-Ursúa MA, Trujillo-Ferrara JG, Arias-Montaño JA, Villalobos-Molina R. Insights into a defined secondary binding region on β-adrenoceptors and putative roles in ligand binding and drug design. MEDCHEMCOMM 2015; 6:991-1002. [DOI: 10.1039/c5md00011d] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Putative roles of a secondary binding region shared among beta-adrenoceptors.
Collapse
Affiliation(s)
- M. A. Soriano-Ursúa
- Posgraduate and Research Section
- Escuela Superior de Medicina
- Instituto Politécnico Nacional
- Mexico City
- Mexico
| | - J. G. Trujillo-Ferrara
- Posgraduate and Research Section
- Escuela Superior de Medicina
- Instituto Politécnico Nacional
- Mexico City
- Mexico
| | - J. A. Arias-Montaño
- Departamento de Fisiología
- Biofísica y Neurociencias. Centro de Investigación y de Estudios Avanzados del IPN
- Mexico City
- Mexico
| | - R. Villalobos-Molina
- Unidad de Investigación en Biomedicina
- Facultad de Estudios Superiores Iztacala
- Universidad Nacional Autónoma de México
- Tlalnepantla
- Mexico
| |
Collapse
|
50
|
Cortés-Ciriano I, Ain QU, Subramanian V, Lenselink EB, Méndez-Lucio O, IJzerman AP, Wohlfahrt G, Prusis P, Malliavin TE, van Westen GJP, Bender A. Polypharmacology modelling using proteochemometrics (PCM): recent methodological developments, applications to target families, and future prospects. MEDCHEMCOMM 2015. [DOI: 10.1039/c4md00216d] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Proteochemometric (PCM) modelling is a computational method to model the bioactivity of multiple ligands against multiple related protein targets simultaneously.
Collapse
Affiliation(s)
- Isidro Cortés-Ciriano
- Unité de Bioinformatique Structurale
- Institut Pasteur and CNRS UMR 3825
- Structural Biology and Chemistry Department
- 75 724 Paris
- France
| | - Qurrat Ul Ain
- Unilever Centre for Molecular Informatics
- Department of Chemistry
- CB2 1EW Cambridge
- UK
| | | | - Eelke B. Lenselink
- Division of Medicinal Chemistry
- Leiden Academic Centre for Drug Research
- Leiden
- The Netherlands
| | - Oscar Méndez-Lucio
- Unilever Centre for Molecular Informatics
- Department of Chemistry
- CB2 1EW Cambridge
- UK
| | - Adriaan P. IJzerman
- Division of Medicinal Chemistry
- Leiden Academic Centre for Drug Research
- Leiden
- The Netherlands
| | - Gerd Wohlfahrt
- Computer-Aided Drug Design
- Orion Pharma
- FIN-02101 Espoo
- Finland
| | - Peteris Prusis
- Computer-Aided Drug Design
- Orion Pharma
- FIN-02101 Espoo
- Finland
| | - Thérèse E. Malliavin
- Unité de Bioinformatique Structurale
- Institut Pasteur and CNRS UMR 3825
- Structural Biology and Chemistry Department
- 75 724 Paris
- France
| | - Gerard J. P. van Westen
- European Molecular Biology Laboratory
- European Bioinformatics Institute
- Wellcome Trust Genome Campus
- Hinxton
- UK
| | - Andreas Bender
- Unilever Centre for Molecular Informatics
- Department of Chemistry
- CB2 1EW Cambridge
- UK
| |
Collapse
|