Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 4.539
1.
Cell Mol Life Sci ; 79(2): 113, 2022 Jan 31.
Article En | MEDLINE | ID: mdl-35099616

Induction of bone formation by Wnt ligands is inhibited when sclerostin (Scl), an osteocyte-produced antagonist, binds to its receptors, the low-density lipoprotein receptor-related proteins 5 or 6 (LRP5/6). Recently, it was shown that enhanced inhibition is achieved by Scl binding to the co-receptor LRP4. However, it is not clear if the binding of Scl to LRP4 facilitates Scl binding to LRP5/6 or inhibits the Wnt pathway in an LRP5/6-independent manner. Here, using the yeast display system, we demonstrate that Scl exhibits a stronger binding affinity for LRP4 than for LRP6. Moreover, we found stronger Scl binding to LRP6 in the presence of LRP4. We further show that a Scl mutant (SclN93A), which tightly binds LRP4 but not LRP6, does not inhibit the Wnt pathway on its own. We demonstrate that SclN93A competes with Scl for a common binding site on LRP4 and antagonizes Scl inhibition of the Wnt signaling pathway in osteoblasts in vitro. Finally, we demonstrate that 2 weeks of bi-weekly subcutaneous injections of SclN93A fused to the fragment crystallizable (Fc) domain of immunoglobulin (SclN93AFc), which retains the antagonistic activity of the mutant, significantly increases bone formation rate and enhances trabecular volumetric bone fraction, trabecular number, and bone length in developing mice. Our data show that LRP4 serves as an anchor that facilitates Scl-LRP6 binding and that inhibition of the Wnt pathway by Scl depends on its prior binding to LRP4. We further provide evidence that compounds that inhibit Scl-LRP4 interactions offer a potential strategy to promote anabolic bone functions.


Adaptor Proteins, Signal Transducing/metabolism , LDL-Receptor Related Proteins/metabolism , Osteogenesis/drug effects , Recombinant Proteins/pharmacology , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Animals , Binding, Competitive/drug effects , Binding, Competitive/genetics , Cells, Cultured , Female , HEK293 Cells , Humans , LDL-Receptor Related Proteins/antagonists & inhibitors , LDL-Receptor Related Proteins/chemistry , LDL-Receptor Related Proteins/genetics , Mice , Mice, Inbred C57BL , Mutant Proteins/chemistry , Mutant Proteins/pharmacology , Osteoblasts/drug effects , Osteoblasts/physiology , Osteogenesis/genetics , Protein Binding/drug effects , Protein Binding/genetics , Protein Interaction Domains and Motifs/drug effects , Protein Interaction Domains and Motifs/genetics , RNA, Small Interfering/pharmacology , Recombinant Proteins/chemistry
2.
Bioengineered ; 12(2): 11425-11440, 2021 12.
Article En | MEDLINE | ID: mdl-34866524

LncRNA DDX11 antisense RNA 1 (DDX11-AS1) is recognized as having an imperative oncogenic role in different types of human cancer. Nevertheless, the functions, as well as the basic mechanisms of DDX11-AS1 in the EMT process of esophageal squamous cell carcinoma (ESCC), are yet to be clarified. In this research, high DDX11-AS1 expression was detected in ESCC cells as well as tissues and was linked to the poor prognosis of patients with ESCC. DDX11-AS1 promoted cell proliferation, migration, invasion ability and epithelial mesenchymal transition (EMT) process in vitro. Mechanistic analysis depicted that DDX11-AS1 may function as a ceRNA through sponging miR-30d-5p to upregulate the expression of SNAI1 and ZEB2. Meanwhile, overexpression of DDX11-AS1 might cause the activation of the Wnt/ß-catenin signaling pathway via targeting miR-30d-5p. On the whole, the findings of this research illustrate that DDX11-AS1 may act as an EMT-related lncRNA to advance ESCC progression through sponging miR-30d-5p to regulate SNAI1/ZEB2 expression and activate the Wnt/ß-catenin pathway, which indicates that it might serve as a probable therapeutic target for ESCC.


Epithelial-Mesenchymal Transition/genetics , Esophageal Neoplasms/genetics , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/pathology , RNA, Long Noncoding/metabolism , Snail Family Transcription Factors/genetics , Zinc Finger E-box Binding Homeobox 2/genetics , Base Sequence , Binding, Competitive/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Esophageal Neoplasms/pathology , Gene Expression Regulation, Neoplastic/drug effects , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Multivariate Analysis , Neoplasm Invasiveness , Prognosis , RNA, Long Noncoding/genetics , Snail Family Transcription Factors/metabolism , Transforming Growth Factor beta/pharmacology , Up-Regulation/drug effects , Up-Regulation/genetics , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/genetics , Zinc Finger E-box Binding Homeobox 2/metabolism
3.
Comput Biol Chem ; 95: 107600, 2021 Dec.
Article En | MEDLINE | ID: mdl-34794076

Peroxisome proliferator-activated receptor gamma (PPARγ), a member of the nuclear receptor superfamily is an excellent example of targets that orchestrates cancer, inflammation, lipid and glucose metabolism. We report a protocol for the development of novel PPARγ antagonists by employing 3D QSAR based virtual screening for the identification of ligands with anticancer properties. The models are generated based on a large and diverse set of PPARγ antagonist ligands by the HYPOGEN algorithm using Discovery Studio 2019 drug design software. Among the 10 hypotheses generated, Hypotheses 2 showed the highest correlation coefficient values of 0.95 with less RMS deviation of 1.193. Validation of the developed pharmacophore model was performed by Fischer's randomization and screening against test and decoy set. The GH score or goodness score was found to be 0.81 indicating moderate to a good model. The selected pharmacophore model Hypo 2 was used as a query model for further screening of 11,145 compounds from the PubChem, sc-PDB structure database, and designed novel ligands. Based on fit values and ADMET filter, the final 10 compounds with the predicated activity of ≤ 3 nM were subjected for docking analysis. Docking analysis revealed the unique binding mode with hydrophobic amino acid that can cause destabilization of the H12 which is an important molecular mechanism to prove its antagonist action. Based on high CDocker scores, Cpd31 was synthesized, purified, analyzed and screened for PPARγ competitive binding by TR-FRET assay. The biochemical protein binding results matched the predicted results. Further, Cpd31 was screened against cancer cells and validated the results.


Anilides/pharmacology , Antineoplastic Agents/pharmacology , Drug Design , PPAR gamma/antagonists & inhibitors , Algorithms , Anilides/chemical synthesis , Anilides/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Binding, Competitive/drug effects , Humans , Molecular Docking Simulation , Molecular Structure , PPAR gamma/metabolism
4.
Biochem Biophys Res Commun ; 556: 156-162, 2021 06 04.
Article En | MEDLINE | ID: mdl-33839411

The transient receptor potential vanilloid 1 (TRPV1) channel is a polymodal receptor in sensory nerves and involved in pain sensation. TRPV1 has at least three distinct activation modes that are selectively induced by different stimuli capsaicin, noxious heat, and protons. Although many mode-selective TRPV1 antagonists have been developed for their anticipated analgesic effects, there have been few successful reports because of adverse effects due to burn injuries and hyperthermia. Eugenol is a vanilloid that has been used as an analgesic in the dental treatment, and its TRPV1 activation ability has been reported. However, our knowledge about the underlying mechanisms of the antagonistic effects of eugenol on TRPV1 activation induced by three different modes is limited. Here, we show that eugenol dose-dependently inhibited the capsaicin-activated inward currents of mouse TRPV1 expressed in human embryonic kidney 293 (HEK293) cells. Under low pH conditions, low concentrations of eugenol only enhanced the proton-induced TRPV1 currents, whereas high eugenol concentrations initially potentiated but then immediately abrogated TRPV1 currents. Finally, eugenol had no modulatory effects on heat-activated TRPV1 in electrophysiological and Fura-2-based Ca2+ imaging experiments. Our results demonstrate that eugenol is a mode-selective antagonist of TRPV1 and can be evaluated as a lead compound of analgesics targeting TRPV1 without serious side effects.


Eugenol/pharmacology , Hot Temperature , Protons , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/metabolism , Animals , Binding, Competitive/drug effects , Calcium/metabolism , Capsaicin/antagonists & inhibitors , Capsaicin/pharmacology , Dose-Response Relationship, Drug , Electric Conductivity , Eugenol/administration & dosage , HEK293 Cells , Humans , Mice
5.
PLoS One ; 16(2): e0246583, 2021.
Article En | MEDLINE | ID: mdl-33561155

We reported that bisphenol AF (BPAF) works as an agonist for estrogen receptor (ER) ERα but as an antagonist for ERß. Similar results were observed for bisphenol E analogs (BPE-X) such as BPE-F, BPE-Cl, and BPE-Br, each consisting of a series of a tri-halogenated methyl group CX3 in the central alkyl moiety. It was demonstrated that the electrostatic halogen bond based on the dispersion force of halogen atoms is a major driving force in the activities of bifunctional ERα-agonist and ERß-antagonist. Since the chlorine atoms present in bisphenol C (BPC) exist in a π-π conjugated system due to the presence of an adjacent C = C double bond, we intended to prove that BPC is also a bifunctional ERα-agonist and ERß-antagonist exhibiting greatly enhanced agonist/antagonist activities. BPC was evaluated for its ability to activate ERα and ERß in the luciferase reporter gene assay using HeLa cells. With high receptor-binding ability to both ERs, BPC was found to be fully active for ERα but inactive for ERß. BPC's definite antagonist activity in ERß was revealed by its inhibitory activity against 17ß-estradiol. Thus, BPC is a bifunctional ERα-agonist and ERß-antagonist. These agonist/antagonist activities were discovered to be extremely high among series of halogen-containing bisphenol compounds. This comparative structure-activity study revealed that the ascending order of ERα-agonist and ERß-antagonist activities was BPE-F ≪ BPE-Cl ≲ BPAF < BPE-Br ≪ BPC. The highly intensified receptor interaction of BPC is attributable to the presence of an n-π-π-n conjugation system mediated through the >C = CCl2 double bond.


Benzhydryl Compounds/pharmacology , Estrogen Receptor alpha/agonists , Estrogen Receptor beta/antagonists & inhibitors , Halogens/metabolism , Phenols/pharmacology , Benzhydryl Compounds/chemistry , Binding, Competitive/drug effects , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Genes, Reporter , HeLa Cells , Humans , Ligands , Luciferases/metabolism , Phenols/chemistry , Transcriptional Activation/drug effects , Transcriptional Activation/genetics
6.
Nat Commun ; 12(1): 582, 2021 01 25.
Article En | MEDLINE | ID: mdl-33495441

Tumour necrosis factor (TNF) is a trimeric protein which signals through two membrane receptors, TNFR1 and TNFR2. Previously, we identified small molecules that inhibit human TNF by stabilising a distorted trimer and reduce the number of receptors bound to TNF from three to two. Here we present a biochemical and structural characterisation of the small molecule-stabilised TNF-TNFR1 complex, providing insights into how a distorted TNF trimer can alter signalling function. We demonstrate that the inhibitors reduce the binding affinity of TNF to the third TNFR1 molecule. In support of this, we show by X-ray crystallography that the inhibitor-bound, distorted, TNF trimer forms a complex with a dimer of TNFR1 molecules. This observation, along with data from a solution-based network assembly assay, leads us to suggest a model for TNF signalling based on TNF-TNFR1 clusters, which are disrupted by small molecule inhibitors.


Protein Multimerization/drug effects , Receptors, Tumor Necrosis Factor, Type I/chemistry , Signal Transduction/drug effects , Small Molecule Libraries/pharmacology , Tumor Necrosis Factor-alpha/chemistry , Algorithms , Animals , Binding, Competitive/drug effects , Humans , Models, Molecular , Protein Binding/drug effects , Protein Conformation/drug effects , Receptors, Tumor Necrosis Factor, Type I/metabolism , Small Molecule Libraries/chemistry , Tumor Necrosis Factor-alpha/metabolism
7.
Anal Biochem ; 612: 113966, 2021 01 01.
Article En | MEDLINE | ID: mdl-32956692

Aberrant activation of the Wnt/ß-catenin signaling pathway is prominent in the development and metastasis of non-small cell lung cancer (NSCLC). Highly effective inhibition of this pathway highlights a therapeutic avenue against NSCLC. Moreover, ß-catenin/LEF1 interaction regulates ß-catenin nuclear transport as well as the transcriptions of the key oncogenes in Wnt/ß-catenin signaling pathway. Therefore, interruption of this interaction would be a promising therapeutic strategy for NSCLC metastasis. To date, no economical and rapid high-throughput screening (HTS) assay has been reported for the discovery of ß-catenin/LEF1 interaction inhibitors. In this study, we developed a novel fluorescence polarization (FP)-based HTS assay to identify ß-catenin/LEF1 interaction inhibitors. The FITC-LEF1 sequence, incubation time, temperature, and DMSO resistance were optimized, and then a high Z' factor of 0.77 was achieved. A pilot screening of a natural product library via this established FP screening assay identified sanguinarine analogues as potential ß-catenin/LEF1 interaction inhibitors. GST pull-down and surface plasmon resonance (SPR) assay demonstrated that ß-catenin/LEF1 interaction is a potential anticancer target of sanguinarine in vitro. This newly developed FP screening assay will be vital for the rapid discovery of novel Wnt inhibitors targeting ß-catenin/LEF1 interaction.


Fluorescence Polarization/methods , High-Throughput Screening Assays/methods , Lymphoid Enhancer-Binding Factor 1/antagonists & inhibitors , Lymphoid Enhancer-Binding Factor 1/metabolism , beta Catenin/antagonists & inhibitors , beta Catenin/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Benzophenanthridines/chemistry , Benzophenanthridines/metabolism , Benzophenanthridines/pharmacology , Binding, Competitive/drug effects , Biological Products/chemistry , Biological Products/metabolism , Biological Products/pharmacology , Cell Line, Tumor , Humans , Isoquinolines/chemistry , Isoquinolines/metabolism , Isoquinolines/pharmacology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Protein Binding/drug effects , Protein Stability , Recombinant Proteins , Structure-Activity Relationship , Surface Plasmon Resonance , Wnt Proteins/antagonists & inhibitors
8.
Article En | MEDLINE | ID: mdl-33310480

The recent emergence of the novel pathogenic coronavirus disease 2019 (COVID-19) is responsible for a worldwide pandemic. In sight of this, there has been growing interest in the use of chloroquine (CQ) and hydroxychloroquine (HCQ) as potential treatments. In this study, we use angiotensin converting enzyme 2 (ACE2) over-expressed cell membrane chromatography (CMC) to study the interaction of CQ and HCQ with ACE2 receptor. Both CQ and HCQ were retained on the ACE2/CMC column. Then we analyzed the binding character of CQ and HCQ to ACE2 by CMC frontal analysis, ionic force investigation and competitive binding experiment. Results showed that CQ and HCQ KD values obtained from the CMC frontal analysis method were 8.22(±0.61) × 10-7 M and 11.70(±2.44) × 10-7 M. Compare to CQ, HCQ has the weaker affinity with ACE2. The action force of CQ, HCQ and ACE2 is mainly ionic force. CQ and HCQ have different degrees of competitive binding relationship with ACE2. Our study revealed the interaction of CQ and HCQ with ACE2 receptor, which provides new insights for the use of CQ and HCQ in the treatment of COVID-19. Moreover, this biomimetic drug screening method is expected to open the door for rapid targeting and separating bioactive ingredients active towards ACE2 receptor.


Angiotensin-Converting Enzyme 2/drug effects , Antimalarials/pharmacology , Cell Membrane/chemistry , Chloroquine/pharmacology , Hydroxychloroquine/pharmacology , Angiotensin-Converting Enzyme 2/biosynthesis , Binding, Competitive/drug effects , COVID-19/metabolism , Chromatography/methods , Humans , Models, Molecular , Molecular Docking Simulation
9.
Biotechnol Lett ; 43(2): 369-382, 2021 Feb.
Article En | MEDLINE | ID: mdl-33141321

OBJECTIVE: Currently, there are two categories of epidermal growth factor receptor (EGFR) antagonists, small molecule antagonists and anti-EGFR antibodies. In the current study, we developed a new EGFR antagonist employing the anti-idiotypic antibodies strategy. RESULTS: First, using EGF as an antigen, through a series of immunological protocols and hybridoma technology, we obtained an anti-idiotypic antibody against EGF receptor-binding epitopes. On this basis, we screened and characterized the anti-idiotype antibodies against EGFR through competitive ELISA, co-localization analysis, competitive receptor binding analysis, and immunofluorescence. Finally, an internal image anti-idiotype antibody called FG8 was successfully prepared. Experiment result shows that FG8 inhibits EGFR-mediated signaling pathways in vitro. Additionally, FG8 inhibits liver tumor cell proliferation as well as induces tumor cell apoptosis. CONCLUSIONS: The present study suggests that FG8 is a potential therapeutic agent for liver cancer. In addition, this study provides a novel method for the preparation of EGFR antagonists.


Antibodies, Anti-Idiotypic/biosynthesis , Antineoplastic Agents/pharmacology , Liver Neoplasms/drug therapy , Antibodies, Anti-Idiotypic/drug effects , Antibodies, Anti-Idiotypic/immunology , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Binding, Competitive/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme-Linked Immunosorbent Assay , Epitopes/drug effects , Epitopes/genetics , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/immunology , Humans , Liver Neoplasms/genetics , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Protein Binding/genetics , Signal Transduction/drug effects
10.
Bioorg Chem ; 106: 104504, 2021 01.
Article En | MEDLINE | ID: mdl-33279247

A new series of 5-(2-aryloxy-4-nitrophenyl)-4H-1,2,4-triazoles and 5-(2-aryloxy-3-pyridyl)-4H-1,2,4-triazoles, possessing C-3 thio or alkylthio substituents, was synthesized and evaluated for their benzodiazepine receptor affinity and anti-seizure activity. These analogues revealed similar to significantly superior affinity to GABAA/benzodiazepine receptor complex (IC50 values of 0.04-4.1 nM), relative to diazepam as the reference drug (IC50 value of 2.4 nM). To determine the onset of anti-seizure activity, the time-dependent effectiveness of i.p. administration of compounds on pentylenetetrazole induced seizure threshold was studied and a very good relationship was observed between the lipophilicity (cLogP) and onset of action of studied analogues (r2 = 0.964). The minimum effective dose of the compounds, determined at the time the analogues showed their highest activity, was demonstrated to be 0.025-0.1 mg/kg, relative to diazepam (0.025 mg/kg).


Anticonvulsants/pharmacology , Benzodiazepines/pharmacology , Receptors, GABA-A/chemistry , Seizures/drug therapy , Triazoles/pharmacology , Animals , Anticonvulsants/chemical synthesis , Anticonvulsants/chemistry , Benzodiazepines/chemical synthesis , Benzodiazepines/chemistry , Binding, Competitive/drug effects , Dose-Response Relationship, Drug , Hydrophobic and Hydrophilic Interactions , Male , Molecular Structure , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Triazoles/chemical synthesis , Triazoles/chemistry
11.
J Nat Prod ; 83(10): 3191-3198, 2020 10 23.
Article En | MEDLINE | ID: mdl-33034450

Conioidine A (1), isolated in 1993 with unknown relative and absolute configuration, was suggested to be a DNA-binding compound by an indirect technique. Four stereoisomers of conioidine A have been synthesized from d- and l-proline, and the natural product has been identified as possessing (4R,6R) absolute configuration. Binding of the conioidine diastereomers to calf thymus DNA (CT DNA) and human serum albumin (HSA) has been investigated by fluorescence spectroscopy and isothermal titration calorimetry (ITC). All stereoisomers display at least an order of magnitude weaker binding to DNA than the control compound netropsin; however, a strong association with HSA was observed for the (4R,6S) stereoisomer.


Pyrrolidines/chemistry , Pyrrolidines/chemical synthesis , Solanaceous Alkaloids/chemistry , Solanaceous Alkaloids/chemical synthesis , Binding Sites , Binding, Competitive/drug effects , Calorimetry , Circular Dichroism , DNA/chemistry , Ethidium , Molecular Docking Simulation , Molecular Structure , Netropsin/chemistry , Netropsin/metabolism , Proline/chemistry , Serum Albumin, Human/chemistry , Spectrometry, Fluorescence , Stereoisomerism
12.
Sci Rep ; 10(1): 17232, 2020 10 14.
Article En | MEDLINE | ID: mdl-33057060

Lipoprotein associated phospholipase A2 (Lp-PLA2) has been characterized for its interfacial activation as well as inhibition by detergent micelles and lipoprotein particles. The enzyme has been shown to bind on the surfaces of hydrophobic aggregates, such as detergent micelles, lipoprotein particles and even polystyrene latex nanobeads. Binding to hydrophobic aggregates stimulates the activity of Lp-PLA2 but may not be the necessary step for catalysis. However, at higher concentrations, detergent micelles, latex nanobeads or lipoprotein particles inhibit Lp-PLA2 possibly by blocking the access of substrates to the active site. The competition mechanism also blocks inhibitors such as darapladib binding to Lp-PLA2 and reduces the efficacy of the drug. Darapladib has very low solubility and mainly exists in solutions as complexes with detergents or lipoprotein particles. The inhibition of Lp-PLA2 by darapladib is dependent on many factors such as concentrations of detergents or lipoproteins, incubation time, as well as the order of mixing reaction components. The in vitro Lp-PLA2 activity assays used in clinical studies may not accurately reflect the residual Lp-PLA2 activity in vivo. Darapladib has been found mainly bound on HDL and albumin when it is incubated with human serum. However, Lp-PLA2 is more sensitive to darapladib when bound on LDL and relatively resistant to darapladib when bound on HDL. Therefore, high cholesterol levels may decrease the efficacy of darapladip and cause the drug to be less effective in high risk patients. Our study will help to design better inhibitors for Lp-PLA2. The discoveries also contribute to understanding the mechanism of interfacial activation and inhibition for Lp-PLA2 and provide a new concept for researchers in building better kinetic model for interfacial enzymes.


Benzaldehydes/pharmacology , Binding, Competitive/drug effects , Catalytic Domain/drug effects , Detergents/pharmacology , Lipoproteins/pharmacology , Micelles , Oximes/pharmacology , Phospholipase A2 Inhibitors , Phospholipases A2/metabolism , Benzaldehydes/metabolism , Hydrolysis/drug effects , Hydrophobic and Hydrophilic Interactions , Lipoproteins, HDL/metabolism , Lipoproteins, LDL/metabolism , Oximes/metabolism , Protein Binding/drug effects , Solubility
13.
J Med Chem ; 63(20): 11920-11933, 2020 10 22.
Article En | MEDLINE | ID: mdl-32940040

Macrophage migration inhibitory factor (MIF) is a cytokine with key roles in inflammation and cancer, which qualifies it as a potential drug target. Apart from its cytokine activity, MIF also harbors enzyme activity for keto-enol tautomerization. MIF enzymatic activity has been used for identification of MIF binding molecules that also interfere with its biological activity. However, MIF tautomerase activity assays are troubled by irregularities, thus creating a need for alternative methods. In this study, we identified a 7-hydroxycoumarin fluorophore with high affinity for the MIF tautomerase active site (Ki = 18 ± 1 nM) that binds with concomitant quenching of its fluorescence. This property enabled development of a novel competition-based assay format to quantify MIF binding. We also demonstrated that the 7-hydroxycoumarin fluorophore interfered with the MIF-CD74 interaction and inhibited proliferation of A549 cells. Thus, we provide a high-affinity MIF binder as a novel tool to advance MIF-oriented research.


Enzyme Inhibitors/pharmacology , Fluorescent Dyes/pharmacology , Intramolecular Oxidoreductases/antagonists & inhibitors , Macrophage Migration-Inhibitory Factors/antagonists & inhibitors , Umbelliferones/pharmacology , Binding, Competitive/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/chemistry , Humans , Models, Molecular , Molecular Structure , Structure-Activity Relationship , Umbelliferones/chemical synthesis , Umbelliferones/chemistry
14.
Nature ; 585(7824): 303-308, 2020 09.
Article En | MEDLINE | ID: mdl-32879488

Most general anaesthetics and classical benzodiazepine drugs act through positive modulation of γ-aminobutyric acid type A (GABAA) receptors to dampen neuronal activity in the brain1-5. However, direct structural information on the mechanisms of general anaesthetics at their physiological receptor sites is lacking. Here we present cryo-electron microscopy structures of GABAA receptors bound to intravenous anaesthetics, benzodiazepines and inhibitory modulators. These structures were solved in a lipidic environment and are complemented by electrophysiology and molecular dynamics simulations. Structures of GABAA receptors in complex with the anaesthetics phenobarbital, etomidate and propofol reveal both distinct and common transmembrane binding sites, which are shared in part by the benzodiazepine drug diazepam. Structures in which GABAA receptors are bound by benzodiazepine-site ligands identify an additional membrane binding site for diazepam and suggest an allosteric mechanism for anaesthetic reversal by flumazenil. This study provides a foundation for understanding how pharmacologically diverse and clinically essential drugs act through overlapping and distinct mechanisms to potentiate inhibitory signalling in the brain.


Anesthetics, General/chemistry , Anesthetics, General/pharmacology , Barbiturates/chemistry , Barbiturates/pharmacology , Benzodiazepines/chemistry , Benzodiazepines/pharmacology , Cryoelectron Microscopy , Receptors, GABA-A/chemistry , Allosteric Regulation/drug effects , Anesthetics, General/metabolism , Barbiturates/metabolism , Benzodiazepines/metabolism , Bicuculline/chemistry , Bicuculline/metabolism , Bicuculline/pharmacology , Binding Sites , Binding, Competitive/drug effects , Diazepam/chemistry , Diazepam/metabolism , Diazepam/pharmacology , Electrophysiology , Etomidate/chemistry , Etomidate/metabolism , Etomidate/pharmacology , Flumazenil/pharmacology , GABA-A Receptor Antagonists/chemistry , GABA-A Receptor Antagonists/metabolism , GABA-A Receptor Antagonists/pharmacology , Humans , Ligands , Models, Molecular , Molecular Conformation , Molecular Dynamics Simulation , Phenobarbital/chemistry , Phenobarbital/metabolism , Phenobarbital/pharmacology , Picrotoxin/chemistry , Picrotoxin/metabolism , Picrotoxin/pharmacology , Propofol/chemistry , Propofol/metabolism , Propofol/pharmacology , Receptors, GABA-A/metabolism , Receptors, GABA-A/ultrastructure , gamma-Aminobutyric Acid/chemistry , gamma-Aminobutyric Acid/metabolism , gamma-Aminobutyric Acid/pharmacology
15.
Mol Med Rep ; 22(2): 1111-1118, 2020 08.
Article En | MEDLINE | ID: mdl-32626958

Neuromyelitis optica (NMO) is a severe neurological demyelinating autoimmune disease that affects the optic nerves and spinal cord. There is currently no effective cure or therapy. Aquaporin­4 (AQP4) is a known target of the autoimmune antibody NMO­IgG. Therefore, binding of NMO­IgG to AQP4, and subsequent activation of antibody­mediated and complement­dependent cytotoxicity (CDC), are thought to underlie the pathogenesis of NMO. In the present study, a cell­based high­throughput screening approach was developed to identify molecular inhibitors of NMO­IgG binding to AQP4. Using this approach, extracts from the herb Petroselinum crispum were shown to have inhibitory effects on NMO­IgG binding to AQP4, and the natural compound geraldol was purified from the herb extracts. Analytical high performance liquid chromatography, electrospray ionization­mass spectrometry and nuclear magnetic resonance analyses confirmed the identity of the isolated compound as geraldol, a flavonoid. Geraldol effectively blocked binding of NMO­IgG to AQP4 in immunofluorescence assays and decreased CDC in NMO­IgG/complement­treated FRTL­AQP4 cells and primary astrocytes. Geraldol exhibited low cytotoxicity, with no effect on proliferation or apoptosis of FRTL­AQP4 cells and primary astrocytes. Permeability assays indicated that geraldol did not alter the water transport function of AQP4 in either cell system. The present study suggests the potential therapeutic value of geraldol for NMO drug development.


Aquaporin 4/metabolism , Binding, Competitive/drug effects , Flavones/pharmacology , Immunoglobulin G/immunology , Neuromyelitis Optica/immunology , Animals , Antibody-Dependent Cell Cytotoxicity/drug effects , Aquaporin 4/drug effects , Astrocytes/metabolism , Autoantibodies/immunology , Autoantibodies/metabolism , Cell Survival/drug effects , Cells, Cultured , Cricetinae , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Flavones/analysis , Flavones/chemistry , Flavones/isolation & purification , Humans , Immunoglobulin G/metabolism , Male , Mice , Neuromyelitis Optica/metabolism , Permeability/drug effects , Petroselinum/chemistry , Primary Cell Culture , Rats , Thyroid Epithelial Cells/metabolism , Water/metabolism
16.
Cell Death Dis ; 11(6): 445, 2020 06 10.
Article En | MEDLINE | ID: mdl-32522979

SHQ1 was reported to control the biogenesis and assembly of H/ACA ribonucleoprotein particles (RNPs). It was independently isolated as a growth suppressor, GRIM1, in a genetic screen. Recent studies have indicated that SHQ1 inhibits prostate cancer growth and metastasis. SHQ1 facilitates MYC RNA splicing to promote T-acute lymphoblastic leukemia (T-ALL) development. Thus, the mechanisms of SHQ1 in cancers remain largely unknown. We report here that SHQ1 promotes tumor apoptosis and chemo-sensitivity in hepatocellular carcinoma (HCC) cells. In HCC tissues from patients, expression of SHQ1 was significantly decreased in the tumor compared to adjacent tissues. Experiments with HCC xenograft models revealed that restoring SHQ1 levels enhanced the anti-tumor activity of the endoplasmic reticulum (ER) stress inducer tunicamycin (TM) and common chemotherapy drug paclitaxel (PTX). Mechanistically, SHQ1 is an ER-stress response gene which is regulated by p50ATF6 and XBP1s through an ER stress response like element located on the SHQ1 promoter. SHQ1 interacts with the ER chaperone GRP78 to release ER sensors PERK/IRE1α/ATF6 from GRP78/ER-sensor complexes, leading to hyper-activation of unfolded protein response (UPR). In the persistent ER stress conditions of a HepG2 xenograft tumor model, SHQ1-mediated hyper-activation of ER-sensor signaling induces apoptosis. Our study thus demonstrates a SHQ1-mediated ER-stress response feedback loop that promotes tumor sensitivity to chemotherapeutics.


Antineoplastic Agents/pharmacology , Apoptosis , Endoplasmic Reticulum Stress/genetics , Intracellular Signaling Peptides and Proteins/genetics , Animals , Apoptosis/drug effects , Apoptosis/genetics , Binding, Competitive/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Female , Heat-Shock Proteins/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Mice, Inbred BALB C , Mice, Nude , Models, Biological , Paclitaxel/pharmacology , Protein Binding/drug effects , Transcription, Genetic/drug effects , Unfolded Protein Response/drug effects , Unfolded Protein Response/genetics , Up-Regulation/drug effects , Up-Regulation/genetics , Xenograft Model Antitumor Assays
17.
Molecules ; 25(10)2020 May 18.
Article En | MEDLINE | ID: mdl-32443441

Puerol A (1) from Amorpha fruticosa showed highly potent inhibition against both monophenolase (IC50 = 2.2 µM) and diphenolase (IC50 = 3.8 µM) of tyrosinase. We tried to obtain a full story of enzyme inhibitory behavior for inhibitor 1 because the butenolide skeleton has never been reported as a tyrosinase inhibitor. Puerol A was proved as a reversible, competitive, simple slow-binding inhibitor, according to the respective parameters; k3 = 0.0279 µM-1 min-1 and k4 = 0.003 min-1. A longer lag-phase and a reduced static-state activity of the enzyme explained that puerol A had a tight formation of the complex with Emet. Dose-dependent inhibition was also confirmed by high-performance liquid chromatography (HPLC) analysis using N-acetyl-l-tyrosine as a substrate, which was completely inhibited at 20 µM. A high binding affinity of 1 to tyrosinase was confirmed by fluorescence quenching analysis. Moreover, puerol A decreased melanin content in the B16 melanoma cell dose-dependently with an IC50 of 11.4 µM.


Enzyme Inhibitors/chemistry , Fabaceae/chemistry , Melanoma, Experimental/drug therapy , Monophenol Monooxygenase/antagonists & inhibitors , Animals , Binding, Competitive/drug effects , Catechol Oxidase/antagonists & inhibitors , Catechol Oxidase/chemistry , Cell Proliferation/drug effects , Chromatography, High Pressure Liquid , Enzyme Inhibitors/isolation & purification , Enzyme Inhibitors/pharmacology , Humans , Kinetics , Melanins/antagonists & inhibitors , Melanins/biosynthesis , Melanoma, Experimental/enzymology , Mice , Molecular Docking Simulation , Monophenol Monooxygenase/chemistry , Oxidoreductases/antagonists & inhibitors , Oxidoreductases/chemistry , Tyrosine/analogs & derivatives , Tyrosine/chemistry
18.
Article En | MEDLINE | ID: mdl-32360816

Lithium (Li) is a typical mood stabilizer and the first choice for treatment of bipolar disorder (BD). Despite an extensive clinical use of Li, its mechanisms of action remain widely different and debated. In this work, we studied the time-course of the therapeutic Li effects on ouabain-sensitive Na+/K+-ATPase in forebrain cortex and hippocampus of rats exposed to 3-day sleep deprivation (SD). We also monitored lipid peroxidation as malondialdehyde (MDA) production. In samples of plasma collected from all experimental groups of animals, Li concentrations were followed by ICP-MS. The acute (1 day), short-term (7 days) and chronic (28 days) treatment of rats with Li resulted in large decrease of Na+/K+-ATPase activity in both brain parts. At the same time, SD of control, Li-untreated rats increased Na+/K+-ATPase along with increased production of MDA. The SD-induced increase of Na+/K+-ATPase and MDA was attenuated in Li-treated rats. While SD results in a positive change of Na+/K+-ATPase, the inhibitory effect of Li treatment may be interpreted as a pharmacological mechanism causing a normalization of the stress-induced shift and return the Na+/K+-ATPase back to control level. We conclude that SD alone up-regulates Na+/K+-ATPase together with increased peroxidative damage of lipids. Chronic treatment of rats with Li before SD, protects the brain tissue against this type of damage and decreases Na+/K+-ATPase level back to control level.


Antimanic Agents/pharmacology , Hippocampus/drug effects , Lipid Peroxidation/drug effects , Lithium Carbonate/pharmacology , Prosencephalon/drug effects , Prosencephalon/metabolism , Sleep Deprivation/drug therapy , Sleep Deprivation/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Antimanic Agents/therapeutic use , Binding, Competitive/drug effects , Hippocampus/metabolism , Male , Malondialdehyde/metabolism , Ouabain/metabolism , Prosencephalon/enzymology , Rats , Rats, Wistar , Sleep Deprivation/enzymology
19.
Nat Commun ; 11(1): 2070, 2020 04 24.
Article En | MEDLINE | ID: mdl-32332765

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged in Wuhan, China, at the end of 2019, and there are currently no specific antiviral treatments or vaccines available. SARS-CoV-2 has been shown to use the same cell entry receptor as SARS-CoV, angiotensin-converting enzyme 2 (ACE2). In this report, we generate a recombinant protein by connecting the extracellular domain of human ACE2 to the Fc region of the human immunoglobulin IgG1. A fusion protein containing an ACE2 mutant with low catalytic activity is also used in this study. The fusion proteins are then characterized. Both fusion proteins have a high binding affinity for the receptor-binding domains of SARS-CoV and SARS-CoV-2 and exhibit desirable pharmacological properties in mice. Moreover, the fusion proteins neutralize virus pseudotyped with SARS-CoV or SARS-CoV-2 spike proteins in vitro. As these fusion proteins exhibit cross-reactivity against coronaviruses, they have potential applications in the diagnosis, prophylaxis, and treatment of SARS-CoV-2.


Betacoronavirus/drug effects , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin G/chemistry , Neutralization Tests , Peptidyl-Dipeptidase A/chemistry , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacology , Spike Glycoprotein, Coronavirus/antagonists & inhibitors , Angiotensin-Converting Enzyme 2 , Animals , Betacoronavirus/metabolism , Binding, Competitive/drug effects , Cross Reactions , Drug Design , Humans , Immunoglobulin Fc Fragments/metabolism , Immunoglobulin Fc Fragments/pharmacology , Immunoglobulin G/metabolism , Immunoglobulin G/pharmacology , In Vitro Techniques , Inhibitory Concentration 50 , Membrane Fusion/drug effects , Mice , Mice, Inbred BALB C , Mutation , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Peptidyl-Dipeptidase A/genetics , Peptidyl-Dipeptidase A/pharmacokinetics , Peptidyl-Dipeptidase A/pharmacology , Protein Domains/genetics , Protein Stability , Receptors, Virus/antagonists & inhibitors , Receptors, Virus/chemistry , Receptors, Virus/genetics , Receptors, Virus/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacokinetics , Severe acute respiratory syndrome-related coronavirus/drug effects , Severe acute respiratory syndrome-related coronavirus/metabolism , SARS-CoV-2 , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/metabolism
20.
J Med Chem ; 63(8): 3935-3955, 2020 04 23.
Article En | MEDLINE | ID: mdl-32212732

CD73 is an extracellular mediator of purinergic signaling. When upregulated in the tumor microenvironment, CD73 has been implicated in the inhibition of immune function through overproduction of adenosine. Traditional efforts to inhibit CD73 have involved antibody therapy or the development of small molecules, the most potent of which mimic the acidic and ionizable structure of the enzyme's natural substrate, adenosine 5'-monophosphate (AMP). Here, we report the systematic discovery of a novel class of non-nucleotide CD73 inhibitors that are more potent than all other nonphosphonate inhibitor classes reported to date. These efforts have culminated in the discovery of 4-({5-[4-fluoro-1-(2H-indazol-6-yl)-1H-1,2,3-benzotriazol-6-yl]-1H-pyrazol-1-yl}methyl)benzonitrile (73, IC50 = 12 nM) and 4-({5-[4-chloro-1-(2H-indazol-6-yl)-1H-1,2,3-benzotriazol-6-yl]-1H-pyrazol-1-yl}methyl)benzonitrile (74, IC50 = 19 nM). Cocrystallization of 74 with human CD73 demonstrates a competitive binding mode. These compounds show promise for the improvement of drug-like character via the attenuation of the acidity and low membrane permeability inherent to known nucleoside inhibitors of CD73.


5'-Nucleotidase/antagonists & inhibitors , Drug Discovery/methods , Triazoles/chemistry , Triazoles/pharmacology , 5'-Nucleotidase/metabolism , Animals , Binding, Competitive/drug effects , Binding, Competitive/physiology , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Crystallography, X-Ray/methods , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans
...