Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
1.
Bioinformatics ; 38(5): 1385-1392, 2022 02 07.
Article in English | MEDLINE | ID: mdl-34888617

ABSTRACT

MOTIVATION: Multitargeting features of small molecules have been of increasing interest in recent years. Polypharmacological drugs that address several therapeutic targets may provide greater therapeutic benefits for patients. Furthermore, multitarget compounds can be used to address proteins of the same (or similar) protein families for their exploration as potential pharmacological targets. In addition, the knowledge of multitargeting features is of major importance in the drug selection process; particularly in ultra-large virtual screening procedures to gain high-quality compound collections. However, large-scale multitarget modulator landscapes are almost non-existent. RESULTS: We implemented a specific feature-driven computer-aided pattern analysis (C@PA) to extract molecular-structural features of inhibitors of the model protein family of ATP-binding cassette (ABC) transporters. New molecular-structural features have been identified that successfully expanded the known multitarget modulator landscape of pan-ABC transporter inhibitors. The prediction capability was biologically confirmed by the successful discovery of pan-ABC transporter inhibitors with a distinct inhibitory activity profile. AVAILABILITY AND IMPLEMENTATION: The multitarget dataset is available on the PANABC web page (http://www.panabc.info) and its use is free of charge. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
ATP-Binding Cassette Transporters , Humans , ATP-Binding Cassette Transporters/chemistry , ATP-Binding Cassette Transporters/metabolism
2.
J Chem Inf Model ; 62(6): 1538-1549, 2022 03 28.
Article in English | MEDLINE | ID: mdl-35285230

ABSTRACT

The human ATP- and UTP-activated P2Y2 receptor (P2Y2R) is a Gq protein-coupled receptor involved in several pathophysiological conditions including acute and chronic inflammation, cancer, and pain. Despite its potential as a novel drug target, only few P2Y2R antagonists have been developed so far, all of which suffer from severe drawbacks. These include (i) high polarity due to one or several negative charges resulting in low oral bioavailability, (ii) metabolic instability and generally poor pharmacokinetic properties, and/or (iii) lack of selectivity, which limits their utility for in vitro and in vivo studies aimed at target validation. In search of new druglike scaffolds for P2Y2R antagonists, we employed a structure-based virtual high-throughput screening approach utilizing the complex of a P2Y2R homology model with one of the most potent and selective orthosteric antagonists described so far, AR-C118925 (10). After virtual screening of 3.2 million molecules, 58 compounds were purchased and pharmacologically evaluated. Several novel antagonist scaffolds were discovered, and their binding modes at the human P2Y2R were analyzed by molecular docking studies. The investigated antagonists likely share a similar binding mode with 10 which includes accommodation of bulky, lipophilic groups in the putative orthosteric binding site of the P2Y2R. The discovered scaffolds and the elucidated structure-activity relationships provide a basis for the development of future drug candidates for the P2Y2R which have great potential as novel drugs.


Subject(s)
High-Throughput Screening Assays , Purinergic P2Y Receptor Antagonists , Signal Transduction , Binding Sites , Humans , Molecular Docking Simulation , Purinergic P2Y Receptor Antagonists/chemistry , Receptors, G-Protein-Coupled
3.
Pharmacol Res ; 173: 105880, 2021 11.
Article in English | MEDLINE | ID: mdl-34506902

ABSTRACT

G proteins represent intracellular switches that transduce signals relayed from G protein-coupled receptors. The structurally related macrocyclic depsipeptides FR900359 (FR) and YM-254890 (YM) are potent, selective inhibitors of the Gαq protein family. We recently discovered that radiolabeled FR and YM display strongly divergent residence times, which translates into significantly longer antiasthmatic effects of FR. The present study is aimed at investigating the molecular basis for this observed disparity. Based on docking studies, we mutated amino acid residues of the Gαq protein predicted to interact with FR or YM, and recombinantly expressed the mutated Gαq proteins in cells in which the native Gαq proteins had been knocked out by CRISPR-Cas9. Both radioligands showed similar association kinetics, and their binding followed a conformational selection mechanism, which was rationalized by molecular dynamics simulation studies. Several mutations of amino acid residues near the putative binding site of the "lipophilic anchors" of FR, especially those predicted to interact with the isopropyl group present in FR but not in YM, led to dramatically accelerated dissociation kinetics. Our data indicate that the long residence time of FR depends on lipophilic interactions within its binding site. The observed structure-kinetic relationships point to a complex binding mechanism of FR, which likely involves snap-lock- or dowel-like conformational changes of either ligand or protein, or both. These experimental data will be useful for the design of compounds with a desired residence time, a parameter that has now been recognized to be of utmost importance in drug development.


Subject(s)
Depsipeptides/pharmacology , GTP-Binding Protein alpha Subunits, Gq-G11/antagonists & inhibitors , Peptides, Cyclic/pharmacology , GTP-Binding Protein alpha Subunits, Gq-G11/chemistry , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , HEK293 Cells , Humans , Kinetics , Models, Molecular , Protein Binding
4.
Purinergic Signal ; 17(4): 633-648, 2021 12.
Article in English | MEDLINE | ID: mdl-34476721

ABSTRACT

This review article presents a collection of tool compounds that selectively block and are recommended for studying P2Y and P2X receptor subtypes, investigating their roles in physiology and validating them as future drug targets. Moreover, drug candidates and approved drugs for P2 receptors will be discussed.


Subject(s)
Adenosine Triphosphate/metabolism , Purinergic P2X Receptor Antagonists/pharmacology , Purinergic P2Y Receptor Antagonists/pharmacology , Receptors, Purinergic P2X/metabolism , Receptors, Purinergic P2Y/metabolism , Animals , Humans
5.
J Nat Prod ; 84(7): 1941-1953, 2021 07 23.
Article in English | MEDLINE | ID: mdl-34197116

ABSTRACT

Both the soil bacterium Chromobacterium vaccinii and the bacterial endosymbiont Candidatus Burkholderia crenata of the plant Ardisia crenata are producers of FR900359 (FR). This cyclic depsipeptide is a potent and selective Gq protein inhibitor used extensively to investigate the intracellular signaling of G protein coupled receptors (GPCRs). In this study, the metabolomes of both FR producers were investigated and compared using feature-based molecular networking (FBMN). As a result, 30 previously unknown FR derivatives were identified, one-third being unique to C. vaccinii. Guided by MS, a novel FR derivative, FR-6 (compound 1), was isolated, and its structure unambiguously established. In a whole-cell biosensing assay based on detection of dynamic mass redistribution (DMR) as readout for Gq inhibition, FR-6 suppressed Gq signaling with micromolar potency (pIC50 = 5.56). This functional activity was confirmed in radioligand binding assays (pKi = 7.50). This work demonstrates the power of molecular networking, guiding the way to a novel Gq-inhibiting FR derivative and underlining the potency of FR as a Gq inhibitor.


Subject(s)
Depsipeptides/pharmacology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Signal Transduction/drug effects , Ardisia/chemistry , Chromobacterium/chemistry , HEK293 Cells , Humans , Molecular Docking Simulation , Molecular Structure , Plant Leaves/chemistry
6.
Adv Exp Med Biol ; 1322: 63-95, 2021.
Article in English | MEDLINE | ID: mdl-34258737

ABSTRACT

Human immunodeficiency virus (HIV) is a deadly virus that attacks the body's immune system, subsequently leading to AIDS (acquired immunodeficiency syndrome) and ultimately death. Currently, there is no vaccine or effective cure for this infection; however, antiretrovirals that act at various phases of the virus life cycle have been useful to control the viral load in patients. One of the major problems with antiretroviral therapies involves drug resistance. The three-dimensional structure from crystallography studies are instrumental in understanding the structural basis of drug binding to various targets. This chapter provides key insights into different targets and drugs used in the treatment from a structural perspective. Specifically, an insight into the binding characteristics of drugs at the active and allosteric sites of different targets and the importance of targeting allosteric sites for design of new-generation antiretrovirals to overcome complex and resistant forms of the virus has been reviewed.


Subject(s)
Acquired Immunodeficiency Syndrome , Anti-HIV Agents , HIV Infections , HIV-1 , Anti-HIV Agents/therapeutic use , HIV Infections/drug therapy , Humans
7.
Arch Pharm (Weinheim) ; 354(12): e2100300, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34697820

ABSTRACT

The antithrombotic prodrugs ticlopidine and clopidogrel are thienotetrahydro-pyridine derivatives that are metabolized in the liver to produce thiols that irreversibly block adenosine diphosphate (ADP)-activated P2Y12 receptors on thrombocytes. In their native, nonmetabolized form, both drugs were reported to act as inhibitors of ectonucleoside triphosphate diphosphohydrolase-1 (NTPDase1, CD39). CD39 catalyzes the extracellular hydrolysis of nucleoside tri- and diphosphates, mainly adenosine 5'-triphosphate (ATP) and ADP, yielding adenosine monophosphate, which is further hydrolyzed by ecto-5'-nucleotidase (CD73) to produce adenosine. While ATP has proinflammatory effects, adenosine is a potent anti-inflammatory, immunosuppressive agent. Inhibitors of CD39 and CD73 have potential as novel checkpoint inhibitors for the immunotherapy of cancer and infection. In the present study, we investigated 2-substituted thienotetrahydropyridine derivatives, structurally related to ticlopidine, as CD39 inhibitors. Due to their substituent on the 2-position, they will not be metabolically transformed into reactive thiols and can, therefore, be expected to be devoid of P2Y12 receptor-antagonistic activity in vivo. Several of the investigated 2-substituted thienotetrahydropyridine derivatives showed concentration-dependent inhibition of CD39. The most potent derivative, 32, showed similar CD39-inhibitory potency to ticlopidine, both acting as allosteric inhibitors. Compound 32 showed an improved selectivity profile: While ticlopidine blocked several NTPDase isoenzymes, 32 was characterized as a novel dual inhibitor of CD39 and CD73.


Subject(s)
5'-Nucleotidase/antagonists & inhibitors , Apyrase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Thienopyridines/pharmacology , Allosteric Regulation/drug effects , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , GPI-Linked Proteins/antagonists & inhibitors , Humans , Structure-Activity Relationship , Thienopyridines/chemical synthesis , Thienopyridines/chemistry , Ticlopidine/pharmacology
8.
Angew Chem Int Ed Engl ; 60(18): 10423-10429, 2021 04 26.
Article in English | MEDLINE | ID: mdl-33655614

ABSTRACT

The main protease of SARS-CoV-2 (Mpro ), the causative agent of COVID-19, constitutes a significant drug target. A new fluorogenic substrate was kinetically compared to an internally quenched fluorescent peptide and shown to be ideally suitable for high throughput screening with recombinantly expressed Mpro . Two classes of protease inhibitors, azanitriles and pyridyl esters, were identified, optimized and subjected to in-depth biochemical characterization. Tailored peptides equipped with the unique azanitrile warhead exhibited concomitant inhibition of Mpro and cathepsin L, a protease relevant for viral cell entry. Pyridyl indole esters were analyzed by a positional scanning. Our focused approach towards Mpro inhibitors proved to be superior to virtual screening. With two irreversible inhibitors, azanitrile 8 (kinac /Ki =37 500 m-1 s-1 , Ki =24.0 nm) and pyridyl ester 17 (kinac /Ki =29 100 m-1 s-1 , Ki =10.0 nm), promising drug candidates for further development have been discovered.


Subject(s)
Antiviral Agents/pharmacology , COVID-19 Drug Treatment , Coronavirus 3C Proteases/antagonists & inhibitors , Nitriles/pharmacology , Protease Inhibitors/pharmacology , SARS-CoV-2/drug effects , Antiviral Agents/chemistry , COVID-19/metabolism , COVID-19/virology , Coronavirus 3C Proteases/metabolism , Drug Design , Drug Discovery , HEK293 Cells , High-Throughput Screening Assays , Humans , Molecular Docking Simulation , Nitriles/chemistry , Protease Inhibitors/chemistry , Pyridines/chemistry , Pyridines/pharmacology , SARS-CoV-2/enzymology , SARS-CoV-2/physiology , Virus Internalization/drug effects
9.
Bioorg Med Chem ; 27(18): 4110-4123, 2019 09 15.
Article in English | MEDLINE | ID: mdl-31378598

ABSTRACT

The sulfonamidophenylethylamide analogues were explored for finding novel and potent cardiac myosin activators. Among them, N-(4-(N,N-dimethylsulfamoyl)phenethyl-N-methyl-5-phenylpentanamide (13, CMA at 10 µM = 48.5%; FS = 26.21%; EF = 15.28%) and its isomer, 4-(4-(N,N-dimethylsulfamoyl)phenyl-N-methyl-N-(3-phenylpropyl)butanamide (27, CMA at 10 µM = 55.0%; FS = 24.69%; EF = 14.08%) proved to be efficient cardiac myosin activators both in in vitro and in vivo studies. Compounds 13 (88.2 + 3.1% at 5 µM) and 27 (46.5 + 2.8% at 5 µM) showed positive inotropic effect in isolated rat ventricular myocytes. The potent compounds 13 and 27 were highly selective for cardiac myosin over skeletal and smooth muscle myosin, and therefore these potent and selective amide derivatives could be considered a new class of cardiac myosin activators for the treatment of systolic heart failure.


Subject(s)
Amides/therapeutic use , Cardiac Myosins/drug effects , Amides/pharmacology , Humans , Structure-Activity Relationship
10.
Analyst ; 143(22): 5417-5430, 2018 Nov 05.
Article in English | MEDLINE | ID: mdl-30303204

ABSTRACT

Ecto-nucleoside triphosphate diphosphohydrolase1 (NTPDase1, CD39) is a major ectonucleotidase that hydrolyzes proinflammatory ATP via ADP to AMP, which is subsequently converted by ecto-5'-nucleotidase (CD73) to immunosuppressive adenosine. Activation of CD39 has potential for treating inflammatory diseases, while inhibition was suggested as a novel strategy for the immunotherapy of cancer. In the present study, we developed a selective and highly sensitive capillary electrophoresis (CE) assay using a novel fluorescent CD39 substrate, a fluorescein-labelled ATP (PSB-170621A) that is converted to its AMP derivative. To accelerate the assays, a two-directional (forward and reverse) CE system was implemented using 96-well plates, which is suitable for the screening of compound libraries (Z'-factor: 0.7). The detection limits for the forward and reverse operation were 11.7 and 2.00 pM, respectively, indicating a large enhancement in sensitivity as compared to previous methods (e.g. malachite-green assay: 1 000 000-fold, CE-UV assay: 500 000-fold, fluorescence polarization immunoassay: 12 500-fold). Enzyme kinetic studies at human CD39 revealed a Km value of 19.6 µM, and a kcat value of 119 × 10-3 s-1 for PSB-170621A, which shows similar substrate properties as ATP (11.4 µM and 82.5 × 10-3 s-1). The compound displayed similar properties at rat and mouse CD39. Subsequent docking studies into a homology model of human CD39 revealed a hydrophobic pocket that accommodates the fluorescein tag. PSB-170621A was found to be preferably hydrolyzed by CD39 as compared to other ectonucleotidases. The new assay was validated by performing inhibition assays with several standard CD39 inhibitors yielding results that were consonant with data using the natural substrates.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/chemistry , Antigens, CD/analysis , Apyrase/analysis , Electrophoresis, Capillary/methods , Enzyme Assays/methods , Fluoresceins/chemistry , Fluorescent Dyes/chemistry , Animals , Antigens, CD/chemistry , Antigens, CD/isolation & purification , Apyrase/antagonists & inhibitors , Apyrase/chemistry , Apyrase/isolation & purification , Humans , Kinetics , Limit of Detection , Mice , Molecular Docking Simulation , Rats , Sequence Homology, Amino Acid
11.
Bioorg Med Chem ; 26(16): 4650-4663, 2018 09 01.
Article in English | MEDLINE | ID: mdl-30104122

ABSTRACT

On the basis of a pyrazine core structure, three new adenosine A2B receptor ligands (7a-c) were synthesized containing a 2-fluoropyridine moiety suitable for 18F-labeling. Compound 7a was docked into a homology model of the A2B receptor based on X-ray structures of the related A2A receptor, and its interactions with the adenosine binding site were rationalized. Binding affinity data were determined at the four human adenosine receptor subtypes. Despite a rather low selectivity regarding the A1 receptor, 7a was radiolabeled as the most suitable candidate (Ki(A2B) = 4.24 nM) in order to perform in vivo studies in mice with the aim to estimate fundamental pharmacokinetic characteristics of the compound class. Organ distribution studies and a single PET study demonstrated brain uptake of [18F]7a with a standardized uptake value (SUV) of ≈1 at 5 min post injection followed by a fast wash out. Metabolism studies of [18F]7a in mice revealed the formation of a blood-brain barrier penetrable radiometabolite, which could be structurally identified. The results of this study provide an important basis for the design of new derivatives with improved binding properties and metabolic stability in vivo.


Subject(s)
Contrast Media/chemical synthesis , Positron-Emission Tomography , Pyrazines/chemistry , Radiopharmaceuticals/chemical synthesis , Receptor, Adenosine A2B/metabolism , Animals , Binding Sites , Blood-Brain Barrier/metabolism , Brain/diagnostic imaging , Brain/metabolism , Contrast Media/chemistry , Contrast Media/metabolism , Female , Fluorine Radioisotopes/chemistry , Humans , Mice , Molecular Dynamics Simulation , Protein Structure, Tertiary , Pyrazines/chemical synthesis , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/metabolism , Receptor, Adenosine A2B/chemistry
12.
J Enzyme Inhib Med Chem ; 32(1): 403-425, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28097901

ABSTRACT

Melanogenesis is a process to synthesize melanin, which is a primary responsible for the pigmentation of human skin, eye and hair. Although numerous enzymatic catalyzed and chemical reactions are involved in melanogenesis process, the enzymes such as tyrosinase and tyrosinase-related protein-1 (TRP-1) and TRP-2 played a major role in melanin synthesis. Specifically, tyrosinase is a key enzyme, which catalyzes a rate-limiting step of the melanin synthesis, and the downregulation of tyrosinase is the most prominent approach for the development of melanogenesis inhibitors. Therefore, numerous inhibitors that target tyrosinase have been developed in recent years. The review focuses on the recent discovery of tyrosinase inhibitors that are directly involved in the inhibition of tyrosinase catalytic activity and functionality from all sources, including laboratory synthetic methods, natural products, virtual screening and structure-based molecular docking studies.


Subject(s)
Enzyme Inhibitors/pharmacology , Monophenol Monooxygenase/antagonists & inhibitors , Skin Lightening Preparations , Animals , Catalysis , Humans , Melanins/antagonists & inhibitors , Melanins/biosynthesis
13.
Arch Pharm (Weinheim) ; 350(3-4)2017 Apr.
Article in English | MEDLINE | ID: mdl-28220535

ABSTRACT

Levetiracetam (LEV) and its recently approved derivative brivaracetam are anti-epileptic drugs with a unique mechanism of action. The synaptic vesicle protein 2A (SV2A) was previously identified as their main target. In the current study, we tested a collection of 500 approved drugs for interaction with the human SV2A protein expressed in Chinese hamster ovary cells. Competition binding studies were performed using cell lysates with high SV2A expression and [3 H]brivaracetam as a radioligand. A hit rate of 3% was obtained, defined as compounds that inhibited radioligand binding by more than 90% at a screening concentration of 20 µM. Subsequent concentration-inhibition curves revealed the antihistaminic prodrug loratadine (Ki = 1.16 µM) and the antimalarial drug quinine (Ki = 2.03 µM) to be the most potent SV2A protein ligands of the investigated drug library. Both compounds were similarly potent as LEV (Ki = 1.74 µM), providing structurally novel scaffolds for SV2A ligands. A pharmacophore model was established, which indicated steric and electronic conformities of brivaracetam with the new SV2A ligands, and preliminary structure-activity relationships were determined. The anti-convulsive effects of the natural product quinine may - at least in part - be explained by interaction with SV2A. Loratadine and quinine represent new lead structures for anti-epileptic drug development.


Subject(s)
Anticonvulsants/pharmacology , Loratadine/pharmacology , Membrane Glycoproteins/antagonists & inhibitors , Nerve Tissue Proteins/antagonists & inhibitors , Piracetam/analogs & derivatives , Pyrrolidinones/pharmacology , Quinine/pharmacology , Animals , Anticonvulsants/chemistry , CHO Cells , Cricetulus , Dose-Response Relationship, Drug , Humans , Levetiracetam , Ligands , Loratadine/chemistry , Membrane Glycoproteins/chemistry , Molecular Structure , Nerve Tissue Proteins/chemistry , Piracetam/chemistry , Piracetam/pharmacology , Pyrrolidinones/chemistry , Quinine/chemistry , Structure-Activity Relationship
14.
Purinergic Signal ; 12(2): 313-29, 2016 06.
Article in English | MEDLINE | ID: mdl-26969588

ABSTRACT

The G protein-coupled A2A adenosine receptor represents an important drug target. Crystal structures and modeling studies indicated that three disulfide bonds are formed between ECL1 and ECL2 (I, Cys71(2.69)-Cys159(45.43); II, Cys74(3.22)-Cys146(45.30), and III, Cys77(3.25)-Cys166(45.50)). However, the A2BAR subtype appears to require only disulfide bond III for proper function. In this study, each of the three disulfide bonds in the A2AAR was disrupted by mutation of one of the cysteine residues to serine. The mutant receptors were stably expressed in Chinese hamster ovary cells and analyzed in cyclic adenosine monophosphate (cAMP) accumulation and radioligand binding studies using structurally diverse agonists: adenosine, NECA, CGS21680, and PSB-15826. Results were rationalized by molecular modeling. The observed effects were dependent on the investigated agonist. Loss of disulfide bond I led to a widening of the orthosteric binding pocket resulting in a strong reduction in the potency of adenosine, but not of NECA or 2-substituted nucleosides. Disruption of disulfide bond II led to a significant reduction in the agonists' efficacy indicating its importance for receptor activation. Disulfide bond III disruption reduced potency and affinity of the small adenosine agonists and NECA, but not of the larger 2-substituted agonists. While all the three disulfide bonds were essential for high potency or efficacy of adenosine, structural modification of the nucleoside could rescue affinity or efficacy at the mutant receptors. At present, it cannot be excluded that formation of the extracellular disulfide bonds in the A2AAR is dynamic. This might add another level of G protein-coupled receptor (GPCR) modulation, in particular for the cysteine-rich A2A and A2BARs.


Subject(s)
Cysteine/chemistry , Models, Molecular , Receptor, Adenosine A2A/chemistry , Receptor, Adenosine A2A/metabolism , Animals , CHO Cells , Cricetulus , Cysteine/metabolism , Extracellular Space/chemistry , Extracellular Space/metabolism , Humans , Molecular Docking Simulation , Mutagenesis, Site-Directed
15.
Arch Pharm (Weinheim) ; 349(5): 383-97, 2016 May.
Article in English | MEDLINE | ID: mdl-27135906

ABSTRACT

Twenty-seven hybridized pyrazolone analogs were designed, docked, synthesized in two series and evaluated for their in vitro antimycobacterial properties. In the first series, four Schiff base derivatives, 6b, 7b, 7h, and 7i, show good antitubercular activity with minimum inhibition concentration (MIC) values in the range of 32.56-42.55 µM. In the second series, two compounds, 8b and 8c, possessed significant antitubercular activity with MIC <0.37 and <0.44 µM, respectively; they were even more potent than the standards pyrazinamide (12.99 µM), ciprofloxacin (4.82 µM), and streptomycin (5.36 µM), with a selectivity index of >630. Compounds 8b and 8c showed shikimate kinase inhibition activity at 5.84 and 6.93 µM, respectively. The activity and docking results lead to the conclusion that the compounds without double bond in the imine side chain and hydrophobic clashes at the pyrazolone end are necessary for good accommodation in the binding pocket and for imparting flexibility. All the compounds were also tested for antimicrobial activity (antibacterial and antifungal) and show highly significant activities against all the microorganisms tested.


Subject(s)
Antitubercular Agents/chemical synthesis , Antitubercular Agents/pharmacology , Drug Design , Mycobacterium tuberculosis/drug effects , Pyrazolones/chemistry , Pyrazolones/pharmacology , Antitubercular Agents/chemistry , Ciprofloxacin/pharmacology , Microbial Sensitivity Tests , Molecular Docking Simulation , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Pyrazinamide/pharmacology , Pyrazolones/chemical synthesis , Schiff Bases/chemistry , Schiff Bases/pharmacology , Streptomycin/pharmacology , Structure-Activity Relationship
16.
J Chem Inf Model ; 54(5): 1301-10, 2014 May 27.
Article in English | MEDLINE | ID: mdl-24803014

ABSTRACT

Active compounds can participate in different local structure-activity relationship (SAR) environments and introduce different degrees of local SAR discontinuity, depending on their structural and potency relationships in data sets. Such SAR features have thus far mostly been analyzed using descriptive approaches, in particular, on the basis of activity landscape modeling. However, compounds in different local SAR environments have not yet been predicted. Herein, we adapt the emerging chemical patterns (ECP) method, a machine learning approach for compound classification, to systematically predict compounds with different local SAR characteristics. ECP analysis is shown to accurately assign many compounds to different local SAR environments across a variety of activity classes covering the entire range of observed local SARs. Control calculations using random forests and multiclass support vector machines were carried out and a variety of statistical performance measures were applied. In all instances, ECP calculations yielded comparable or better performance than controls. The approach presented herein can be applied to predict compounds that complement local SARs or prioritize compounds with different SAR characteristics.


Subject(s)
Artificial Intelligence , Models, Chemical , Structure-Activity Relationship
17.
Sci Data ; 11(1): 530, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38783061

ABSTRACT

The identification of lead molecules and the exploration of novel pharmacological drug targets are major challenges of medical life sciences today. Genome-wide association studies, multi-omics, and systems pharmacology steadily reveal new protein networks, extending the known and relevant disease-modifying proteome. Unfortunately, the vast majority of the disease-modifying proteome consists of 'orphan targets' of which intrinsic ligands/substrates, (patho)physiological roles, and/or modulators are unknown. Undruggability is a major challenge in drug development today, and medicinal chemistry efforts cannot keep up with hit identification and hit-to-lead optimization studies. New 'thinking-outside-the-box' approaches are necessary to identify structurally novel and functionally distinctive ligands for orphan targets. Here we present a unique dataset that includes critical information on the orphan target ABCA1, from which a novel cheminformatic workflow - computer-aided pattern scoring (C@PS) - for the identification of novel ligands was developed. Providing a hit rate of 95.5% and molecules with high potency and molecular-structural diversity, this dataset represents a suitable template for general deorphanization studies.


Subject(s)
Drug Design , Drug Discovery , Humans , Ligands , Workflow
18.
Commun Biol ; 7(1): 52, 2024 01 06.
Article in English | MEDLINE | ID: mdl-38184723

ABSTRACT

Patients with idiopathic pulmonary fibrosis show a strongly upregulated expression of chemokine CXCL14, whose target is still unknown. Screening of CXCL14 in a panel of human G protein-coupled receptors (GPCRs) revealed its potent and selective activation of the orphan MAS-related GPCR X2 (MRGPRX2). This receptor is expressed on mast cells and - like CXCL14 - upregulated in bronchial inflammation. CXCL14 induces robust activation of MRGPRX2 and its putative mouse ortholog MRGPRB2 in G protein-dependent and ß-arrestin recruitment assays that is blocked by a selective MRGPRX2/B2 antagonist. Truncation combined with mutagenesis and computational studies identified the pharmacophoric sequence of CXCL14 and its presumed interaction with the receptor. Intriguingly, C-terminal domain sequences of CXCL14 consisting of 4 to 11 amino acids display similar or increased potency and efficacy compared to the full CXCL14 sequence (77 amino acids). These results provide a rational basis for the future development of potential idiopathic pulmonary fibrosis therapies.


Subject(s)
Chemokines , Idiopathic Pulmonary Fibrosis , Animals , Humans , Mice , Amino Acids , Biological Assay , Chemokines, CXC , Idiopathic Pulmonary Fibrosis/genetics , Nerve Tissue Proteins/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, Neuropeptide
19.
J Med Chem ; 67(12): 9896-9926, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38885438

ABSTRACT

The human orphan G protein-coupled receptor GPR18, activated by Δ9-tetrahydrocannabinol (THC), constitutes a promising drug target in immunology and cancer. However, studies on GPR18 are hampered by the lack of suitable tool compounds. In the present study, potent and selective GPR18 agonists were developed showing low nanomolar potency at human and mouse GPR18, determined in ß-arrestin recruitment assays. Structure-activity relationships were analyzed, and selectivity versus cannabinoid (CB) and CB-like receptors was assessed. Compound 51 (PSB-KK1415, EC50 19.1 nM) was the most potent GPR18 agonist showing at least 25-fold selectivity versus CB receptors. The most selective GPR18 agonist 50 (PSB-KK1445, EC50 45.4 nM) displayed >200-fold selectivity versus both CB receptor subtypes, GPR55, and GPR183. The new GPR18 agonists showed minimal species differences, while THC acted as a weak partial agonist at the mouse receptor. The newly discovered compounds represent the most potent and selective GPR18 agonists reported to date.


Subject(s)
Receptors, G-Protein-Coupled , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Humans , Animals , Structure-Activity Relationship , Mice , Neoplasms/drug therapy , Neoplasms/metabolism , HEK293 Cells , Receptors, Cannabinoid/metabolism , Dronabinol/pharmacology , Dronabinol/analogs & derivatives , Dronabinol/chemistry
20.
J Chem Inf Model ; 53(12): 3131-9, 2013 Dec 23.
Article in English | MEDLINE | ID: mdl-24304008

ABSTRACT

Activity cliffs are formed by structurally similar or analogous compounds having large potency differences. In medicinal chemistry, pairs or groups of compounds forming activity cliffs are of interest for structure-activity relationship (SAR) analysis and compound optimization. Thus far, activity cliff assessment has mostly been descriptive, i.e., compound data sets and activity landscape representations have been searched for activity cliffs in the context of SAR analysis. Only recently, first attempts have also been made to depart from descriptive analysis and predict activity cliffs. This has been done by building computational models that distinguish compound pairs forming activity cliffs from non-cliff pairs. However, it is principally more challenging to predict single compounds that participate in activity cliffs. Here, we show that individual compounds having high or low potency can be accurately predicted to form activity cliffs on the basis of emerging chemical patterns.


Subject(s)
Carbonic Anhydrases/chemistry , Drug Design , Receptors, Cell Surface/chemistry , Serotonin Plasma Membrane Transport Proteins/chemistry , Small Molecule Libraries/chemistry , Chemistry, Pharmaceutical , Databases, Factual , Drug Discovery , Humans , Ligands , Receptors, Cell Surface/antagonists & inhibitors , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL