Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 131
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Am Chem Soc ; 144(7): 2905-2920, 2022 02 23.
Article in English | MEDLINE | ID: mdl-35142215

ABSTRACT

Drugs targeting SARS-CoV-2 could have saved millions of lives during the COVID-19 pandemic, and it is now crucial to develop inhibitors of coronavirus replication in preparation for future outbreaks. We explored two virtual screening strategies to find inhibitors of the SARS-CoV-2 main protease in ultralarge chemical libraries. First, structure-based docking was used to screen a diverse library of 235 million virtual compounds against the active site. One hundred top-ranked compounds were tested in binding and enzymatic assays. Second, a fragment discovered by crystallographic screening was optimized guided by docking of millions of elaborated molecules and experimental testing of 93 compounds. Three inhibitors were identified in the first library screen, and five of the selected fragment elaborations showed inhibitory effects. Crystal structures of target-inhibitor complexes confirmed docking predictions and guided hit-to-lead optimization, resulting in a noncovalent main protease inhibitor with nanomolar affinity, a promising in vitro pharmacokinetic profile, and broad-spectrum antiviral effect in infected cells.


Subject(s)
Antiviral Agents/pharmacology , Coronavirus 3C Proteases/metabolism , Cysteine Proteinase Inhibitors/pharmacology , SARS-CoV-2/drug effects , Small Molecule Libraries/pharmacology , Animals , Antiviral Agents/metabolism , Antiviral Agents/pharmacokinetics , Catalytic Domain , Chlorocebus aethiops , Coronavirus 3C Proteases/chemistry , Cysteine Proteinase Inhibitors/metabolism , Cysteine Proteinase Inhibitors/pharmacokinetics , Drug Evaluation, Preclinical , Humans , Microbial Sensitivity Tests , Microsomes, Liver/metabolism , Molecular Docking Simulation , Protein Binding , SARS-CoV-2/enzymology , Small Molecule Libraries/metabolism , Small Molecule Libraries/pharmacokinetics , Vero Cells
2.
Biochem Pharmacol ; 194: 114798, 2021 12.
Article in English | MEDLINE | ID: mdl-34678227

ABSTRACT

Drug affinity responsive target stability (DARTS) is a novel target discovery approach and is particularly adept at screening small molecule (SM) targets without requiring any structural modifications. The DARTS method is capable of revealing drug-target interactions from cells or tissues by tracking changes in the stability of proteins acting as receptors of bioactive SMs. Due to its simple operation and high efficiency, the DARTS method has been applied to uncover the drug-action mechanism. This review summarized analytical principles, protocols, validation approaches, applications, and challenges involved in the DARTS method. Due to the innate advantages of the DARTS method, it is expected to be a powerful tool to accelerate SM target discovery, especially for bioactive natural products with unknown mechanisms.


Subject(s)
Drug Delivery Systems/methods , Drug Discovery/methods , Small Molecule Libraries/administration & dosage , Animals , Drug Delivery Systems/trends , Drug Discovery/trends , Drug Evaluation, Preclinical/methods , Drug Evaluation, Preclinical/trends , Drug Stability , Humans , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Protein Binding/physiology , Small Molecule Libraries/metabolism
3.
Eur J Med Chem ; 223: 113660, 2021 Nov 05.
Article in English | MEDLINE | ID: mdl-34246853

ABSTRACT

TGFß is crucial for the homeostasis of epithelial and neural tissues, wound repair, and regulating immune responses. Its dysregulation is associated with a vast number of diseases, of which modifying the tumor microenvironment is one of vital clinical interest. Despite various attempts, there is still no FDA-approved therapy to inhibit the TGFß pathway. Major mainstream approaches involve impairment of the TGFß pathway via inhibition of the TGFßRI kinase. With the purpose to identify non-receptor kinase-based inhibitors to impair TGFß signaling, an in-house chemical library was enriched, through a computational study, to eliminate TGFßRI kinase activity. Selected compounds were screened against a cell line engineered with a firefly luciferase gene under TGFß-Smad-dependent transcriptional control. Results indicated moderate potency for a molecule with phthalazine core against TGFß-Smad signaling. A series of phthalazine compounds were synthesized and evaluated for potency. The most promising compound (10p) exhibited an IC50 of 0.11 ± 0.02 µM and was confirmed to be non-cytotoxic up to 12 µM, with a selectivity index of approximately 112-fold. Simultaneously, 10p was confirmed to reduce the Smad phosphorylation using Western blot without exhibiting inhibition on the TGFßRI enzyme. This study identified a novel small-molecule scaffold that targets the TGFß pathway via a non-receptor-kinase mechanism.


Subject(s)
Phthalazines/chemistry , Transforming Growth Factor beta/antagonists & inhibitors , Cell Survival/drug effects , Drug Evaluation, Preclinical , HEK293 Cells , Humans , Phosphorylation/drug effects , Phthalazines/metabolism , Phthalazines/pharmacology , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/drug effects , Smad Proteins/chemistry , Smad Proteins/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism , Small Molecule Libraries/pharmacology , Structure-Activity Relationship , Transforming Growth Factor beta/metabolism
4.
Angew Chem Int Ed Engl ; 60(35): 19191-19200, 2021 08 23.
Article in English | MEDLINE | ID: mdl-34161644

ABSTRACT

SARS-CoV-2 contains a positive single-stranded RNA genome of approximately 30 000 nucleotides. Within this genome, 15 RNA elements were identified as conserved between SARS-CoV and SARS-CoV-2. By nuclear magnetic resonance (NMR) spectroscopy, we previously determined that these elements fold independently, in line with data from in vivo and ex-vivo structural probing experiments. These elements contain non-base-paired regions that potentially harbor ligand-binding pockets. Here, we performed an NMR-based screening of a poised fragment library of 768 compounds for binding to these RNAs, employing three different 1 H-based 1D NMR binding assays. The screening identified common as well as RNA-element specific hits. The results allow selection of the most promising of the 15 RNA elements as putative drug targets. Based on the identified hits, we derive key functional units and groups in ligands for effective targeting of the RNA of SARS-CoV-2.


Subject(s)
Genome , RNA, Viral/metabolism , SARS-CoV-2/genetics , Small Molecule Libraries/metabolism , Drug Evaluation, Preclinical , Ligands , Molecular Structure , Nucleic Acid Conformation , Proton Magnetic Resonance Spectroscopy , RNA, Viral/chemistry , Small Molecule Libraries/chemistry
5.
Bioorg Med Chem Lett ; 42: 128067, 2021 06 15.
Article in English | MEDLINE | ID: mdl-33957246

ABSTRACT

The outbreak of coronavirus (CoV) disease 2019 (COVID-19) caused by the severe acute respiratory syndrome CoV-2 (SARS-CoV-2) has turned into a pandemic. The enzyme 3C-like protease (3CLpro) is essential for the maturation of viral polyproteins in SARS-CoV-2 and is therefore regarded as a key drug target for treating the disease. To identify 3CLpro inhibitors that can suppress SARS-CoV-2 replication, we performed a virtual screening of 500,282 compounds in a Korean compound bank. We then subjected the top computational hits to inhibitory assays against 3CLpro in vitro, leading to the identification of a class of non-covalent inhibitors. Among these inhibitors, compound 7 showed an EC50 of 39.89 µM against SARS-CoV-2 and CC50 of 453.5 µM. This study provides candidates for the optimization of potent 3CLpro inhibitors showing antiviral effects against SARS-CoV-2.


Subject(s)
Antiviral Agents/pharmacology , Coronavirus 3C Proteases/antagonists & inhibitors , Protease Inhibitors/pharmacology , SARS-CoV-2/enzymology , Small Molecule Libraries/pharmacology , Animals , Antiviral Agents/metabolism , Chlorocebus aethiops , Coronavirus 3C Proteases/metabolism , Drug Evaluation, Preclinical , Microbial Sensitivity Tests , Molecular Docking Simulation , Protease Inhibitors/metabolism , Protein Binding , Republic of Korea , Small Molecule Libraries/metabolism , Vero Cells
6.
Chem Biol Drug Des ; 98(3): 395-404, 2021 09.
Article in English | MEDLINE | ID: mdl-33963664

ABSTRACT

"Candidatus Liberibacter asiaticus" (Ca. L. asiaticus) is the causal agent of Huanglongbing disease of citrus and current study focuses on the discovery of novel small-molecule inhibitors against SecA protein of Ca. L. asiaticus. In this study, homologous modeling was used to construct the three-dimensional structure of SecA. Then, molecular docking-based virtual screening and two rounds of in vitro bacteriostatic experiments were utilized to identify novel small-molecule inhibitors of SecA. Encouragingly, 93 compounds were obtained and two of them (P684-2850, P684-3808) showed strong antimicrobial activities against Liberibacter crescens BT-1 in bacteriostatic experiments. Finally, molecular dynamics simulations were employed to explore the binding modes of the receptor-ligand complexes. Results in MD simulations showed that compound P684-3808 was relatively stable during simulation, while compound P684-2850 left the binding pocket. Compound P684-3808 might be suitable as a lead compound for further development of antimicrobial compounds against SecA of Ca. L. asiaticus.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , Liberibacter/metabolism , SecA Proteins/antagonists & inhibitors , Small Molecule Libraries/chemistry , Bacterial Proteins/metabolism , Binding Sites , Drug Evaluation, Preclinical , Inhibitory Concentration 50 , Liberibacter/drug effects , Microbial Sensitivity Tests , Molecular Docking Simulation , Molecular Dynamics Simulation , SecA Proteins/metabolism , Small Molecule Libraries/metabolism , Small Molecule Libraries/pharmacology
7.
J Med Chem ; 64(9): 5577-5592, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33886285

ABSTRACT

The central melanocortin-3 and melanocortin-4 receptors (MC3R, MC4R) are key regulators of body weight and energy homeostasis. Herein, the discovery and characterization of first-in-class small molecule melanocortin agonists with selectivity for the melanocortin-3 receptor over the melanocortin-4 receptor are reported. Identified via "unbiased" mixture-based high-throughput screening approaches, pharmacological evaluation of these pyrrolidine bis-cyclic guanidines resulted in nanomolar agonist activity at the melanocortin-3 receptor. The pharmacological profiles at the remaining melanocortin receptor subtypes tested indicated similar agonist potencies at both the melanocortin-1 and melanocortin-5 receptors and antagonist or micromolar agonist activities at the melanocortin-4 receptor. This group of small molecules represents a new area of chemical space for the melanocortin receptors with mixed receptor pharmacology profiles that may serve as novel lead compounds to modulate states of dysregulated energy balance.


Subject(s)
Guanidine/metabolism , Pyrrolidines/chemistry , Receptor, Melanocortin, Type 3/agonists , Algorithms , Animals , Drug Evaluation, Preclinical , Energy Metabolism/drug effects , Guanidine/analogs & derivatives , Guanidine/pharmacology , Guanidine/therapeutic use , High-Throughput Screening Assays , Humans , Mice , Mice, Knockout , Protein Isoforms/agonists , Protein Isoforms/genetics , Protein Isoforms/metabolism , Pyrrolidines/metabolism , Pyrrolidines/pharmacology , Pyrrolidines/therapeutic use , Receptor, Melanocortin, Type 3/genetics , Receptor, Melanocortin, Type 3/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism , Small Molecule Libraries/pharmacology , Small Molecule Libraries/therapeutic use , Structure-Activity Relationship
9.
Future Med Chem ; 13(4): 341-361, 2021 02.
Article in English | MEDLINE | ID: mdl-33427493

ABSTRACT

Background: Discovery of effective autophagy-initiating kinase ULK1 inhibitors has attracted more and more attention in cancer treatment. Methodology & results: The present study describes the application of a pharmacophore-based virtual screening and structure-based docking approach guided drug design. Compound U-2 exhibited a nanomolar range of IC50 against the ULK1 target. Molecular dynamics simulation was used to assess the quality of docking studies. The determinants of binding affinity were investigated, and a different binding pattern was observed. Subsequently, prediction properties of ADMET (absorption, distribution, metabolism, excretion and toxicity) and hepatotoxicity in vitro studies indicated that U-2 possessed good drug-like properties. Moreover, western blot analysis indicated that the compound inhibited autophagic flux in cells. Conclusion: The present study provides an appropriate guideline for discovering novel ULK1 inhibitors. The novel compound may serve as a good starting point for further development and optimizations.


Subject(s)
Autophagy-Related Protein-1 Homolog/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Molecular Docking Simulation , Protein Kinase Inhibitors/chemistry , Autophagy-Related Protein-1 Homolog/metabolism , Binding Sites , Cell Line, Tumor , Cell Survival/drug effects , Drug Evaluation, Preclinical , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Ligands , Molecular Dynamics Simulation , Protein Binding , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism , Small Molecule Libraries/pharmacology
10.
Eur J Med Chem ; 209: 112909, 2021 Jan 01.
Article in English | MEDLINE | ID: mdl-33081987

ABSTRACT

17beta-Hydroxysteroid dehydrogenase type 10 (17ß-HSD10) is the only mitochondrial member of 17ß-HSD family. This enzyme can oxidize estradiol (E2) into estrone (E1), thus reducing concentration of this neuroprotective steroid. Since 17ß-HSD10 possesses properties that suggest a possible role in Alzheimer's disease, its inhibition appears to be a therapeutic strategy. After we identified the androsterone (ADT) derivative 1 as a first steroidal inhibitor of 17ß-HSD10, new analogs were synthesized to increase the metabolic stability, to improve the selectivity of inhibition over 17ß-HSD3 and to optimize the inhibitory potency. From six D-ring derivatives of 1 (17-CO), two compounds (17ß-H/17α-OH and 17ß-OH/17α-CCH) were more metabolically stable and did not inhibit the 17ß-HSD3. Moreover, solid phase synthesis was used to extend the molecular diversity on the 3ß-piperazinylmethyl group of the steroid base core. Eight over 120 new derivatives were more potent inhibitors than 1 for the transformation of E2 to E1, with the 4-(4-trifluoromethyl-3-methoxybenzyl)piperazin-1-ylmethyl-ADT (D-3,7) being 16 times more potent (IC50 = 0.14 µM). Finally, D-ring modification of D-3,7 provided 17ß-OH/17α-CCH derivative 25 and 17ß-H/17α-OH derivative 26, which were more potent inhibitor than 1 (1.8 and 2.4 times, respectively).


Subject(s)
17-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Alzheimer Disease/drug therapy , Enzyme Inhibitors/chemical synthesis , Small Molecule Libraries/chemistry , Steroids/chemical synthesis , Biocatalysis , Drug Evaluation, Preclinical , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Estradiol/chemistry , Estrone/chemistry , HEK293 Cells , Humans , Piperazine/chemistry , Small Molecule Libraries/metabolism , Small Molecule Libraries/pharmacology , Solid-Phase Synthesis Techniques , Steroids/metabolism , Steroids/pharmacology , Structure-Activity Relationship
11.
Proc Natl Acad Sci U S A ; 117(51): 32711-32721, 2020 12 22.
Article in English | MEDLINE | ID: mdl-33277431

ABSTRACT

CLC-2 is a voltage-gated chloride channel that is widely expressed in mammalian tissues. In the central nervous system, CLC-2 appears in neurons and glia. Studies to define how this channel contributes to normal and pathophysiological function in the central nervous system raise questions that remain unresolved, in part due to the absence of precise pharmacological tools for modulating CLC-2 activity. Herein, we describe the development and optimization of AK-42, a specific small-molecule inhibitor of CLC-2 with nanomolar potency (IC50 = 17 ± 1 nM). AK-42 displays unprecedented selectivity (>1,000-fold) over CLC-1, the closest CLC-2 homolog, and exhibits no off-target engagement against a panel of 61 common channels, receptors, and transporters expressed in brain tissue. Computational docking, validated by mutagenesis and kinetic studies, indicates that AK-42 binds to an extracellular vestibule above the channel pore. In electrophysiological recordings of mouse CA1 hippocampal pyramidal neurons, AK-42 acutely and reversibly inhibits CLC-2 currents; no effect on current is observed on brain slices taken from CLC-2 knockout mice. These results establish AK-42 as a powerful tool for investigating CLC-2 neurophysiology.


Subject(s)
Chloride Channels/antagonists & inhibitors , Chloride Channels/chemistry , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Animals , Binding Sites , CHO Cells , CLC-2 Chloride Channels , Cell Line , Chloride Channels/genetics , Chloride Channels/metabolism , Cricetulus , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Hippocampus/metabolism , Humans , Mice, Inbred C57BL , Mice, Knockout , Molecular Docking Simulation , Organ Culture Techniques , Patch-Clamp Techniques , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Small Molecule Libraries/metabolism , Structure-Activity Relationship
12.
Biomolecules ; 10(12)2020 12 03.
Article in English | MEDLINE | ID: mdl-33287369

ABSTRACT

Cysteinyl leukotriene G protein-coupled receptors, CysLT1R and CysLT2R, regulate bronchoconstrictive and pro-inflammatory effects and play a key role in allergic disorders, cardiovascular diseases, and cancer. CysLT1R antagonists have been widely used to treat asthma disorders, while CysLT2R is a potential target against uveal melanoma. However, very few selective antagonist chemotypes for CysLT receptors are available, and the design of such ligands has proved to be challenging. To overcome this obstacle, we took advantage of recently solved crystal structures of CysLT receptors and an ultra-large Enamine REAL library, representing a chemical space of 680 M readily available compounds. Virtual ligand screening employed 4D docking models comprising crystal structures of CysLT1R and CysLT2R and their corresponding ligand-optimized models. Functional assessment of the candidate hits yielded discovery of five novel antagonist chemotypes with sub-micromolar potencies and the best Ki = 220 nM at CysLT1R. One of the hits showed inverse agonism at the L129Q constitutively active mutant of CysLT2R, with potential utility against uveal melanoma.


Subject(s)
Drug Evaluation, Preclinical , Receptors, Leukotriene/metabolism , Small Molecule Libraries/pharmacology , Humans , Ligands , Molecular Docking Simulation , Protein Conformation , Receptors, Leukotriene/chemistry , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism , User-Computer Interface
13.
Bioorg Chem ; 105: 104452, 2020 12.
Article in English | MEDLINE | ID: mdl-33212311

ABSTRACT

The resistance of microbes to commonly used antibiotics has become a worldwide health problem. A major underlying mechanism of microbial antibiotic resistance is the export of drugs from bacterial cells. Drug efflux is mediated through the action of multidrug resistance efflux pumps located in the bacterial cell membranes. The critical role of bacterial efflux pumps in antibiotic resistance has directed research efforts to the identification of novel efflux pump inhibitors that can be used alongside antibiotics in clinical settings. Here, we aimed to find potential inhibitors of the archetypical ATP-binding cassette (ABC) efflux pump BmrA of Bacillus subtilis via virtual screening of the Mu.Ta.Lig. Chemotheca small molecule library. Molecular docking calculations targeting the nucleotide-binding domain of BmrA were performed using AutoDock Vina. Following a further drug-likeness filtering step based on Lipinski's Rule of Five, top 25 scorers were identified. These ligands were then clustered into separate groups based on their contact patterns with the BmrA nucleotide-binding domain. Six ligands with distinct contact patterns were used for further in vitro inhibition assays based on intracellular ethidium bromide accumulation. Using this methodology, we identified two novel inhibitors of BmrA from the Chemotheca small molecule library.


Subject(s)
ATP-Binding Cassette Transporters/antagonists & inhibitors , Bacillus subtilis/chemistry , Bacterial Proteins/chemistry , Membrane Transport Proteins/chemistry , Molecular Docking Simulation , Small Molecule Libraries/chemistry , Amino Acid Sequence , Drug Evaluation, Preclinical , Ethidium/chemistry , Humans , Ligands , Protein Conformation , Protein Multimerization , Small Molecule Libraries/metabolism
14.
Essays Biochem ; 64(6): 955-966, 2020 12 07.
Article in English | MEDLINE | ID: mdl-33078198

ABSTRACT

RNA is crucial for gene expression and regulation. Recent advances in understanding of RNA biochemistry, structure and molecular biology have revealed the importance of RNA structure in cellular processes and diseases. Various approaches to discovering drug-like small molecules that target RNA structure have been developed. This review provides a brief introduction to RNA structural biology and how RNA structures function as disease regulators. We summarize approaches to targeting RNA with small molecules and highlight their advantages, shortcomings and therapeutic potential.


Subject(s)
Drug Discovery/methods , Molecular Targeted Therapy/methods , RNA/chemistry , RNA/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism , Disease , Drug Design , Drug Evaluation, Preclinical/methods , High-Throughput Screening Assays/methods , Humans , Molecular Docking Simulation
15.
Curr Top Med Chem ; 20(26): 2344-2361, 2020.
Article in English | MEDLINE | ID: mdl-32819246

ABSTRACT

Embryonic stem cells (ESCs) are stem cells (SCs) that can self-renew and differentiate into a myriad of cell types. The process of developing stemness is determined by signaling molecules that drive stem cells to a specific lineage. For example, ESCs can differentiate into mature cells (e.g., cardiomyocytes) and mature cardiomyocytes can be characterized for cell beating, action potential, and ion channel function. A goal of this Perspective is to show how small molecules can be used to differentiate ESCs into cardiomyocytes and how this can reveal novel aspects of SC biology. This approach can also lead to the discovery of new molecules of use in cardiovascular disease. Human induced pluripotent stem cells (hiPSCs) afford the ability to produce unlimited numbers of normal human cells. The creation of patient-specific hiPSCs provides an opportunity to study cell models of human disease. The second goal is to show that small molecules can stimulate hiPSC commitment to cardiomyocytes. How iPSCs can be used in an approach to discover new molecules of use in cardiovascular disease will also be shown in this study. Adult SCs, including mesenchymal stem cells (MSCs), can likewise participate in self-renewal and multilineage differentiation. MSCs are capable of differentiating into osteoblasts, adipocytes or chondrocytes. A third goal of this Perspective is to describe differentiation of MSCs into chondrogenic and osteogenic lineages. Small molecules can stimulate MSCs to specific cell fate both in vitro and in vivo. In this Perspective, some recent examples of applying small molecules for osteogenic and chondrogenic cell fate determination are summarized. Underlying molecular mechanisms and signaling pathways involved are described. Small molecule-based modulation of stem cells shows insight into cell regulation and potential approaches to therapeutic strategies for MSC-related diseases.


Subject(s)
Bone and Bones/metabolism , Chondrocytes/metabolism , Embryonic Stem Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Mesenchymal Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Small Molecule Libraries/metabolism , Adipocytes/metabolism , Animals , Ascorbic Acid/metabolism , Bone and Bones/cytology , Cell Differentiation , Cells, Cultured , Chondrocytes/cytology , Dimethyl Sulfoxide/metabolism , Drug Evaluation, Preclinical , Humans , Hydrazones/metabolism , Hyperbaric Oxygenation , Induced Pluripotent Stem Cells/cytology , Ion Channels/metabolism , Mesenchymal Stem Cells/cytology , Myocytes, Cardiac/cytology , Osteoblasts/metabolism , Serine/metabolism , Small Molecule Libraries/chemistry , Structure-Activity Relationship
16.
J Med Chem ; 63(17): 10030-10044, 2020 09 10.
Article in English | MEDLINE | ID: mdl-32787113

ABSTRACT

To discover novel BChE inhibitors, a hierarchical virtual screening protocol followed by biochemical evaluation was applied. The most potent compound 8012-9656 (eqBChE IC50 = 0.18 ± 0.03 µM, hBChE IC50 = 0.32 ± 0.07 µM) was purchased and synthesized. It inhibited BChE in a noncompetitive manner and could occupy the binding pocket forming diverse interactions with the target. 8012-9656 was proven to be safe in vivo and in vitro and showed comparable performance in ameliorating the scopolamine-induced cognition impairment to tacrine. Additionally, treatment with 8012-9656 could almost entirely recover the Aß1-42 (icv)-impaired cognitive function to the normal level and showed better behavioral performance than donepezil. The evaluation of the Aß1-42 total amount confirmed its anti-amyloidogenic profile. Moreover, 8012-9656 possessed blood-brain barrier (BBB) penetrating ability, a long T1/2, and low intrinsic clearance. Hence, the novel potential BChE inhibitor 8012-9656 can be considered as a promising lead compound for further investigation of anti-AD agents.


Subject(s)
Aminoquinolines/pharmacology , Benzimidazoles/pharmacology , Butyrylcholinesterase/metabolism , Cholinesterase Inhibitors/pharmacology , Neuroprotective Agents/pharmacology , Aminoquinolines/chemical synthesis , Aminoquinolines/metabolism , Aminoquinolines/toxicity , Animals , Benzimidazoles/chemical synthesis , Benzimidazoles/metabolism , Benzimidazoles/toxicity , Cell Line, Tumor , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/metabolism , Cholinesterase Inhibitors/toxicity , Drug Discovery , Drug Evaluation, Preclinical , Female , Humans , Male , Mice, Inbred ICR , Microsomes, Liver/metabolism , Molecular Docking Simulation , Molecular Dynamics Simulation , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/metabolism , Neuroprotective Agents/toxicity , Protein Binding , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/metabolism , Small Molecule Libraries/pharmacology , Small Molecule Libraries/toxicity
17.
Bioorg Med Chem Lett ; 30(19): 127464, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32768646

ABSTRACT

Two critical steps in drug development are 1) the discovery of molecules that have the desired effects on a target, and 2) the optimization of such molecules into lead compounds with the required potency and pharmacokinetic properties for translation. DNA-encoded chemical libraries (DECLs) can nowadays yield hits with unprecedented ease, and lead-optimization is becoming the limiting step. Here we integrate DECL screening with structure-based computational methods to streamline the development of lead compounds. The presented workflow consists of enumerating a virtual combinatorial library (VCL) derived from a DECL screening hit and using computational binding prediction to identify molecules with enhanced properties relative to the original DECL hit. As proof-of-concept demonstration, we applied this approach to identify an inhibitor of PARP10 that is more potent and druglike than the original DECL screening hit.


Subject(s)
DNA/chemistry , Proto-Oncogene Proteins/antagonists & inhibitors , Small Molecule Libraries/chemistry , Combinatorial Chemistry Techniques , Drug Discovery , Drug Evaluation, Preclinical , Enzyme Assays , Humans , Molecular Docking Simulation , Poly(ADP-ribose) Polymerases/metabolism , Proof of Concept Study , Protein Binding , Proto-Oncogene Proteins/metabolism , Small Molecule Libraries/metabolism
18.
Sci Rep ; 10(1): 13093, 2020 08 04.
Article in English | MEDLINE | ID: mdl-32753646

ABSTRACT

A novel coronavirus, named SARS-CoV-2, emerged in 2019 in China and rapidly spread worldwide. As no approved therapeutics exists to treat COVID-19, the disease associated to SARS-Cov-2, there is an urgent need to propose molecules that could quickly enter into clinics. Repurposing of approved drugs is a strategy that can bypass the time-consuming stages of drug development. In this study, we screened the PRESTWICK CHEMICAL LIBRARY composed of 1,520 approved drugs in an infected cell-based assay. The robustness of the screen was assessed by the identification of drugs that already demonstrated in vitro antiviral effect against SARS-CoV-2. Thereby, 90 compounds were identified as positive hits from the screen and were grouped according to their chemical composition and their known therapeutic effect. Then EC50 and CC50 were determined for a subset of 15 compounds from a panel of 23 selected drugs covering the different groups. Eleven compounds such as macrolides antibiotics, proton pump inhibitors, antiarrhythmic agents or CNS drugs emerged showing antiviral potency with 2 < EC50 ≤ 20 µM. By providing new information on molecules inhibiting SARS-CoV-2 replication in vitro, this study provides information for the selection of drugs to be further validated in vivo. Disclaimer: This study corresponds to the early stages of antiviral development and the results do not support by themselves the use of the selected drugs to treat SARS-CoV-2 infection.


Subject(s)
Betacoronavirus/physiology , Small Molecule Libraries/chemistry , Animals , Antiviral Agents/chemistry , Antiviral Agents/metabolism , Antiviral Agents/pharmacology , Betacoronavirus/isolation & purification , COVID-19 , Caco-2 Cells , Cell Survival/drug effects , Chlorocebus aethiops , Coronavirus Infections/pathology , Coronavirus Infections/virology , Drug Approval , Drug Evaluation, Preclinical , Drug Repositioning , Humans , Pandemics , Pneumonia, Viral/pathology , Pneumonia, Viral/virology , SARS-CoV-2 , Small Molecule Libraries/metabolism , Small Molecule Libraries/pharmacology , Vero Cells , Virus Replication/drug effects
19.
PLoS One ; 15(7): e0236710, 2020.
Article in English | MEDLINE | ID: mdl-32735606

ABSTRACT

Argonaute (AGO) proteins are the key component of the RNA interference machinery that suppresses gene expression by forming an RNA-induced silencing complex (RISC) with microRNAs (miRNAs). Each miRNA is involved in various cellular processes, such as development, differentiation, tumorigenesis, and viral infection. Thus, molecules that regulate miRNA function are expected to have therapeutic potential. In addition, the biogenesis of miRNA is a multistep process involving various proteins, although the complete pathway remains to be elucidated. Therefore, identification of molecules that can specifically modulate each step will help understand the mechanism of gene suppression. To date, several AGO2 inhibitors have been identified. However, these molecules were identified through a single screening method, and no studies have specifically evaluated a combinatorial strategy. Here, we demonstrated a combinatorial screening (SCR) approach comprising an in silico molecular docking study, surface plasmon resonance (SPR) analysis, and nuclear magnetic resonance (NMR) analysis, focusing on the strong binding between the 5'-terminal phosphate of RNA and the AGO2 middle (MID) domain. By combining SPR and NMR, we identified binding modes of amino acid residues binding to AGO2. First, using a large chemical library (over 6,000,000 compounds), 171 compounds with acidic functional groups were screened using in silico SCR. Next, we constructed an SPR inhibition system that could analyze only the 5'-terminal binding site of RNA, and nine molecules that strongly bound to the AGO2 MID domain were selected. Finally, using NMR, three molecules that bound to the desired site were identified. The RISC inhibitory ability of the "hit" compounds was analyzed in human cell lysate, and all three hit compounds strongly inhibited the binding between double-stranded RNA and AGO2.


Subject(s)
Argonaute Proteins/antagonists & inhibitors , Drug Evaluation, Preclinical/methods , Magnetic Resonance Spectroscopy , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Surface Plasmon Resonance , Argonaute Proteins/chemistry , Argonaute Proteins/metabolism , Computer Simulation , HeLa Cells , Humans , Molecular Docking Simulation , Protein Conformation , Small Molecule Libraries/metabolism
20.
ChemMedChem ; 15(20): 1921-1931, 2020 10 19.
Article in English | MEDLINE | ID: mdl-32700795

ABSTRACT

Coronavirus disease 2019 (COVID-19) has spread out as a pandemic threat affecting over 2 million people. The infectious process initiates via binding of SARS-CoV-2 Spike (S) glycoprotein to host angiotensin-converting enzyme 2 (ACE2). The interaction is mediated by the receptor-binding domain (RBD) of S glycoprotein, promoting host receptor recognition and binding to ACE2 peptidase domain (PD), thus representing a promising target for therapeutic intervention. Herein, we present a computational study aimed at identifying small molecules potentially able to target RBD. Although targeting PPI remains a challenge in drug discovery, our investigation highlights that interaction between SARS-CoV-2 RBD and ACE2 PD might be prone to small molecule modulation, due to the hydrophilic nature of the bi-molecular recognition process and the presence of druggable hot spots. The fundamental objective is to identify, and provide to the international scientific community, hit molecules potentially suitable to enter the drug discovery process, preclinical validation and development.


Subject(s)
Betacoronavirus/chemistry , Peptidyl-Dipeptidase A/metabolism , Protein Binding/drug effects , Small Molecule Libraries/metabolism , Spike Glycoprotein, Coronavirus/metabolism , Angiotensin-Converting Enzyme 2 , Antiviral Agents/metabolism , Betacoronavirus/metabolism , COVID-19 , Coronavirus Infections/drug therapy , Drug Evaluation, Preclinical , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Pandemics , Pneumonia, Viral/drug therapy , Protein Domains , SARS-CoV-2
SELECTION OF CITATIONS
SEARCH DETAIL