Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Chem Asian J ; 16(22): 3729-3742, 2021 Nov 15.
Article in English | MEDLINE | ID: mdl-34549886

ABSTRACT

Half-sandwich RuII complexes, [(YZ)RuII (η6 -arene)(X)]+, (YZ=chelating bidentate ligand, X=halide), with N,N and N,O coordination (1-9) show significant antiproliferative activity against the metastatic triple-negative breast carcinoma (MDA-MB-231). 3-aminobenzoic acid or its methyl ester is used in all the ligands while varying the aldehyde for N,N and N,O coordination. In the N,N coordinated complex the coordinated halide(X) is varied for enhancing stability in solution (X=Cl, I). Rapid aquation and halide exchange of the pyridine analogues, 2 and 3, in solution are a major bane towards their antiproliferative activity. Presence of free -COOH group (1 and 4) make complexes hydrophilic and reduces toxicity. The imidazolyl 3-aminobenzoate based N,N coordinated 5 and 6 display better solution stability and efficient antiproliferative activity (IC50 ca. 2.3-2.5 µM) compared to the pyridine based 2 and 3 (IC50 >100 µM) or the N,O coordinated complexes (7-9) (IC50 ca. 7-10 µM). The iodido coordinated, 6, is resistant towards aquation and halide exchange. The N,O coordinated 7-9 underwent instantaneous aquation at pH 7.4 generating monoaquated complexes stable for at least 6 h. Complexes 5 and 6, bind to 9-ethylguanine (9-EtG) showing propensity to interact with DNA bases. The complexes may kill via apoptosis as displayed from the study of 8. The change in coordination mode and the aldehyde affected the solution stability, antiproliferative activity and mechanistic pathways. The N,N coordinated (5 and 6) exhibit arrest in the G2/M phase while the N,O coordinated 8 showed arrest in the G0/G1 phase.


Subject(s)
Antineoplastic Agents/pharmacology , Coordination Complexes/pharmacology , Ruthenium/pharmacology , Triple Negative Breast Neoplasms/drug therapy , meta-Aminobenzoates/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Coordination Complexes/chemical synthesis , Coordination Complexes/chemistry , Drug Screening Assays, Antitumor , Humans , Ruthenium/chemistry , Schiff Bases/chemistry , Schiff Bases/pharmacology , Triple Negative Breast Neoplasms/pathology , Tumor Cells, Cultured , meta-Aminobenzoates/chemistry
2.
PLoS One ; 15(10): e0240517, 2020.
Article in English | MEDLINE | ID: mdl-33052980

ABSTRACT

Mitochondrial diseases are a clinically heterogenous group of disorders caused by respiratory chain dysfunction and associated with progressive, multi-systemic phenotype. There is no effective treatment or cure, and no FDA-approved drug for treating mitochondrial disease. To identify and characterize potential therapeutic compounds, we developed an in vitro screening assay and identified a group of direct AMP-activated protein kinase (AMPK) activators originally developed for the treatment of diabetes and metabolic syndrome. Unlike previously investigated AMPK agonists such as AICAR, these compounds allosterically activate AMPK in an AMP-independent manner, thereby increasing specificity and decreasing pleiotropic effects. The direct AMPK activator PT1 significantly improved mitochondrial function in assays of cellular respiration, energy status, and cellular redox. PT1 also protected against retinal degeneration in a mouse model of photoreceptor degeneration associated with mitochondrial dysfunction and oxidative stress, further supporting the therapeutic potential of AMP-independent AMPK agonists in the treatment of mitochondrial disease.


Subject(s)
AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Mitochondrial Diseases/drug therapy , Thiazoles/administration & dosage , meta-Aminobenzoates/administration & dosage , Allosteric Regulation/drug effects , Animals , Biphenyl Compounds , Cell Respiration/drug effects , Cell Survival , Cells, Cultured , Disease Models, Animal , Enzyme Activation/drug effects , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Mice , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Oxidative Stress/drug effects , Oxygen Consumption/drug effects , Pyrimidines/administration & dosage , Pyrimidines/pharmacology , Pyrones/administration & dosage , Pyrones/pharmacology , Thiazoles/pharmacology , Thiophenes/administration & dosage , Thiophenes/pharmacology , meta-Aminobenzoates/pharmacology
3.
Virology ; 515: 223-234, 2018 02.
Article in English | MEDLINE | ID: mdl-29306785

ABSTRACT

The crystal structure of chikungunya (CHIKV) virus capsid protease domain has been determined at 2.2Å. Structure reveals a chymotrypsin-like protease fold with a conserved hydrophobic pocket in CHIKV capsid protein (CP) for interaction with the cytoplasmic tail of E2 (cdE2) similar to the capsid protein of other alphaviruses. Molecular contacts between CP-cdE2 were determined by fitting structures of CHIKV CP and cdE2 into the cryo-EM map of Venezuelan equine encephalitis virus (VEEV). Binding of (S)-(+)-Mandelic acid (MDA) and Ethyl 3-aminobenzoate (EAB) to the hydrophobic pocket of CP was evaluated by molecular docking. Surface plasmon resonance (SPR) and fluorescence spectroscopy experiments confirmed MDA and EAB binding to the CP. The binding constants (KD) obtained from SPR for MDA and EAB were 1.2 × 10-3 M and 0.2 × 10-9 M, respectively. This study adds to the understanding of chikungunya virus structural proteins and may serve as the basis for antiviral development against chikungunya disease.


Subject(s)
Capsid Proteins/chemistry , Capsid Proteins/metabolism , Chikungunya Fever/virology , Chikungunya virus/metabolism , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Capsid/chemistry , Capsid/drug effects , Capsid/metabolism , Capsid Proteins/genetics , Chikungunya virus/chemistry , Chikungunya virus/drug effects , Chikungunya virus/genetics , Humans , Hydrophobic and Hydrophilic Interactions , Kinetics , Mandelic Acids/chemistry , Mandelic Acids/pharmacology , Molecular Docking Simulation , Protein Domains , meta-Aminobenzoates/chemistry , meta-Aminobenzoates/pharmacology
4.
J Bacteriol ; 200(1)2018 01 01.
Article in English | MEDLINE | ID: mdl-29038259

ABSTRACT

The 1,125-bp mabB gene encoding 5-aminosalicylate (5ASA) 1,2-dioxygenase, a nonheme iron dioxygenase in the bicupin family that catalyzes the cleavage of the 5ASA aromatic ring to form cis-4-amino-6-carboxy-2-oxohexa-3,5-dienoate in the biodegradation of 3-aminobenzoate, was cloned from Comamonas sp. strain QT12 and characterized. The deduced amino acid sequence of the enzyme has low sequence identity with that of other reported ring-cleaving dioxygenases. MabB was heterologously expressed in Escherichia coli cells and purified as a His-tagged enzyme. The optimum pH and temperature for MabB are 8.0 and 10°C, respectively. FeII is required for the catalytic activity of the purified enzyme. The apparent Km and Vmax values of MabB for 5ASA are 52.0 ± 5.6 µM and 850 ± 33.2 U/mg, respectively. The two oxygen atoms incorporated into the product of the MabB-catalyzed reaction are both from the dioxygen molecule. Both 5ASA and gentisate could be converted by MabB; however, the catalytic efficiency of MabB for 5ASA was much higher (∼70-fold) than that for gentisate. The mabB-disrupted mutant lost the ability to grow on 3-aminobenzoate, and mabB expression was higher when strain QT12 was cultivated in the presence of 3-aminobenzoate. Thus, 5ASA is the physiological substrate of MabB.IMPORTANCE For several decades, 5-aminosalicylate (5ASA) has been advocated as the drug mesalazine to treat human inflammatory bowel disease and considered the key intermediate in the xenobiotic degradation of many aromatic organic pollutants. 5ASA biotransformation research will help us elucidate the microbial degradation of these pollutants. Most studies have reported that gentisate 1,2-dioxygenases (GDOs) can convert 5ASA with significantly high activity; however, the catalytic efficiency of these enzymes for gentisate is much higher than that for 5ASA. This study showed that MabB can convert 5ASA to cis-4-amino-6-carboxy-2-oxohexa-3,5-dienoate, incorporating two oxygen atoms from the dioxygen molecule into the product. Unlike GDOs, MabB uses 5ASA instead of gentisate as the primary substrate. mabB is the first reported 5-aminosalicylate 1,2-dioxygenase gene.


Subject(s)
Comamonas/enzymology , Dioxygenases/genetics , Dioxygenases/metabolism , Biocatalysis , Biodegradation, Environmental , Cloning, Molecular , Comamonas/drug effects , Comamonas/genetics , Comamonas/growth & development , Dioxygenases/chemistry , Dioxygenases/isolation & purification , Escherichia coli/genetics , Gentisates/metabolism , Kinetics , Mesalamine/metabolism , Mutation , Oxygen/metabolism , Substrate Specificity , meta-Aminobenzoates/metabolism , meta-Aminobenzoates/pharmacology
5.
J Med Chem ; 60(20): 8631-8646, 2017 10 26.
Article in English | MEDLINE | ID: mdl-28991459

ABSTRACT

Previously, we reported a hypoxia-inducible factor (HIF)-1 inhibitor LW6 containing an (aryloxyacetylamino)benzoic acid moiety inhibits malate dehydrogenase 2 (MDH2) using a chemical biology approach. Structure-activity relationship studies on a series of (aryloxyacetylamino)benzoic acids identified selective MDH1, MDH2, and dual inhibitors, which were used to study the relationship between MDH enzyme activity and HIF-1 inhibition. We hypothesized that dual inhibition of MDH1 and MDH2 might be a powerful approach to target cancer metabolism and selected methyl-3-(3-(4-(2,4,4-trimethylpentan-2-yl)phenoxy)propanamido)-benzoate (16c) as the most potent dual inhibitor. Kinetic studies revealed that compound 16c competitively inhibited MDH1 and MDH2. Compound 16c inhibited mitochondrial respiration and hypoxia-induced HIF-1α accumulation. In xenograft assays using HCT116 cells, compound 16c demonstrated significant in vivo antitumor efficacy. This finding provides concrete evidence that inhibition of both MDH1 and MDH2 may provide a valuable platform for developing novel therapeutics that target cancer metabolism and tumor growth.


Subject(s)
Anilides/pharmacology , Enzyme Inhibitors/pharmacology , Isoenzymes/antagonists & inhibitors , Malate Dehydrogenase/antagonists & inhibitors , Neoplasms/metabolism , meta-Aminobenzoates/pharmacology , Animals , Cell Line, Tumor , Humans , Mice , Neoplasms/pathology , Xenograft Model Antitumor Assays
6.
J Med Chem ; 60(16): 6942-6990, 2017 08 24.
Article in English | MEDLINE | ID: mdl-28699740

ABSTRACT

A novel class of therapeutic drug candidates for heart failure, highly potent and selective GRK2 inhibitors, exhibit potentiation of ß-adrenergic signaling in vitro studies. Hydrazone derivative 5 and 1,2,4-triazole derivative 24a were identified as hit compounds by HTS. New scaffold generation and SAR studies of all parts resulted in a 4-methyl-1,2,4-triazole derivative with an N-benzylcarboxamide moiety with highly potent activity toward GRK2 and selectivity over other kinases. In terms of subtype selectivity, these compounds showed enough selectivity against GRK1, 5, 6, and 7 with almost equipotent inhibition to GRK3. Our medicinal chemistry efforts led to the discovery of 115h (GRK2 IC50 = 18 nM), which was obtained the cocrystal structure with human GRK2 and an inhibitor of GRK2 that potentiates ß-adrenergic receptor (ßAR)-mediated cAMP accumulation and prevents internalization of ßARs in ß2AR-expressing HEK293 cells treated with isoproterenol. Therefore, 115h appears to be a novel class of therapeutic for heart failure treatment.


Subject(s)
G-Protein-Coupled Receptor Kinase 2/antagonists & inhibitors , Heart Failure/drug therapy , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Triazoles/pharmacology , meta-Aminobenzoates/pharmacology , Crystallography, X-Ray , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 CYP3A Inhibitors/chemical synthesis , Cytochrome P-450 CYP3A Inhibitors/chemistry , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Drug Design , HEK293 Cells , High-Throughput Screening Assays , Humans , Hydrazones/chemical synthesis , Hydrazones/chemistry , Hydrazones/pharmacology , Protein Kinase C-alpha/antagonists & inhibitors , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Pyridines/chemical synthesis , Pyridines/chemistry , Receptors, Adrenergic, beta/metabolism , Structure-Activity Relationship , Triazoles/chemical synthesis , Triazoles/chemistry , meta-Aminobenzoates/chemical synthesis , meta-Aminobenzoates/chemistry , rho-Associated Kinases/antagonists & inhibitors
7.
PLoS One ; 12(1): e0169882, 2017.
Article in English | MEDLINE | ID: mdl-28118359

ABSTRACT

Voltage-gated sodium channels (VGSC) regulate neuronal excitability by governing action potential (AP) generation and propagation. Recent studies have revealed that AMP-activated protein kinase (AMPK) activators decrease sensory neuron excitability, potentially by preventing sodium (Na+) channel phosphorylation by kinases such as ERK or via modulation of translation regulation pathways. The direct positive allosteric modulator A769662 displays substantially greater efficacy than other AMPK activators in decreasing sensory neuron excitability suggesting additional mechanisms of action. Here, we show that A769662 acutely inhibits AP firing stimulated by ramp current injection in rat trigeminal ganglion (TG) neurons. PT1, a structurally dissimilar AMPK activator that reduces nerve growth factor (NGF) -induced hyperexcitability, has no influence on AP firing in TG neurons upon acute application. In voltage-clamp recordings, application of A769662 reduces VGSC current amplitudes. These findings, based on acute A769662 application, suggest a direct channel blocking effect. Indeed, A769662 dose-dependently blocks VGSC in rat TG neurons and in Nav1.7-transfected cells with an IC50 of ~ 10 µM. A769662 neither displayed use-dependent inhibition nor interacted with the local anesthetic (LA) binding site. Popliteal fossa administration of A769662 decreased noxious thermal responses with a peak effect at 5 mins demonstrating an analgesic effect. These data indicate that in addition to AMPK activation, A769662 acts as a direct blocker/modulator of VGSCs, a potential mechanism enhancing the analgesic property of this compound.


Subject(s)
AMP-Activated Protein Kinases/drug effects , Analgesics/pharmacology , NAV1.7 Voltage-Gated Sodium Channel/drug effects , Pyrones/pharmacology , Sensory Receptor Cells/drug effects , Sodium Channel Blockers/pharmacology , Thiophenes/pharmacology , Anesthetics, Local/metabolism , Animals , Binding Sites/genetics , Biphenyl Compounds , Drug Evaluation, Preclinical , HEK293 Cells , Hot Temperature/adverse effects , Humans , Male , Metformin/pharmacology , NAV1.7 Voltage-Gated Sodium Channel/genetics , Neural Conduction/drug effects , Pain/drug therapy , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Recombinant Fusion Proteins/drug effects , Recombinant Fusion Proteins/metabolism , Resveratrol , Sensory Receptor Cells/enzymology , Stilbenes/pharmacology , Thiazoles/pharmacology , Trigeminal Ganglion/drug effects , meta-Aminobenzoates/pharmacology
8.
Eur J Med Chem ; 123: 673-683, 2016 Nov 10.
Article in English | MEDLINE | ID: mdl-27517812

ABSTRACT

Through structure-based virtual screening and subsequent activity assays of selected natural products, Lavendustin B was previously identified as an inhibitor of HIV-1 integrase (IN) interaction with its cognate cellular cofactor, lens epithelium-derived growth factor (LEDGF/p75). In order to improve the inhibitory potency we have employed in silico-based approaches. Particularly, a series of new analogues was designed and docked into the LEDGF/p75 binding pocket of HIV-1 IN. To identify promising leads we used the Molecular Mechanics energies combined with the Generalized Born and Surface Area continuum solvation (MM-GBSA) method, molecular dynamics simulations and analysis of hydrogen bond occupancies. On the basis of these studies, six analogues of Lavendustine B, containing the benzylamino-hydroxybenzoic scaffold, were selected for synthesis and structure activity-relationship (SAR) studies. Our results demonstrated a good correlation between computational and experimental data, and all six analogues displayed an improved potency for inhibiting IN binding to LEDGF/p75 in vitro to respect to the parent compound Lavendustin B. Additionally, these analogs show to inhibit weakly LEDGF/p75-independent IN catalytic activity suggesting a multimodal allosteric mechanism of action. Nevertheless, for the synthesized compounds similar profiles for HIV-1 inhibition and cytoxicity were highlighted. Taken together, our studies elucidated the mode of action of Lavendustin B analogs and provided a path for their further development as a new promising class of HIV-1 integrase inhibitors.


Subject(s)
Drug Design , HIV Integrase/metabolism , Molecular Docking Simulation , Molecular Dynamics Simulation , Salicylates/chemical synthesis , Salicylates/pharmacology , meta-Aminobenzoates/chemical synthesis , meta-Aminobenzoates/pharmacology , Allosteric Regulation/drug effects , Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/chemistry , Anti-HIV Agents/metabolism , Anti-HIV Agents/pharmacology , Chemistry Techniques, Synthetic , HIV Integrase/chemistry , HIV-1/drug effects , HIV-1/enzymology , HeLa Cells , Humans , Protein Conformation , Salicylates/chemistry , Salicylates/metabolism , meta-Aminobenzoates/chemistry , meta-Aminobenzoates/metabolism
9.
Cell Rep ; 16(1): 28-36, 2016 06 28.
Article in English | MEDLINE | ID: mdl-27320923

ABSTRACT

The Wnt/ß-catenin signaling pathway plays a major role in tissue homeostasis, and its dysregulation can lead to various human diseases. Aberrant activation of ß-catenin is oncogenic and is a critical driver in the development and progression of human cancers. Despite the significant potential of targeting the oncogenic ß-catenin pathway for cancer therapy, the development of specific inhibitors remains insufficient. Using a T cell factor (TCF)-dependent luciferase-reporter system, we screened for small-molecule compounds that act against Wnt/ß-catenin signaling and identified MSAB (methyl 3-{[(4-methylphenyl)sulfonyl]amino}benzoate) as a selective inhibitor of Wnt/ß-catenin signaling. MSAB shows potent anti-tumor effects selectively on Wnt-dependent cancer cells in vitro and in mouse cancer models. MSAB binds to ß-catenin, promoting its degradation, and specifically downregulates Wnt/ß-catenin target genes. Our findings might represent an effective therapeutic strategy for cancers addicted to the Wnt/ß-catenin signaling pathway.


Subject(s)
Benzoates/pharmacology , Oncogenes , Proteasome Endopeptidase Complex/metabolism , Proteolysis/drug effects , Small Molecule Libraries/pharmacology , Sulfonamides/pharmacology , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism , meta-Aminobenzoates/pharmacology , Animals , Benzoates/chemistry , Cell Line, Tumor , Mice , Sulfonamides/chemistry , Xenograft Model Antitumor Assays , meta-Aminobenzoates/chemistry
10.
J Cardiovasc Pharmacol Ther ; 21(1): 70-81, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25868658

ABSTRACT

Myocardial ischemia has become one of the main causes of sudden cardiac death worldwide. Autophagy has been demonstrated to protect cardiomyocytes from ischemia/reperfusion (I/R)-induced damage. A novel small molecule compound 2-Chloro-5-[[5-[[5-(4,5-Dimethyl-2-nitrophenyl)-2-furanyl]methylene]-4,5-dihydro-4-oxo-2-thiazolyl]amino]benzoic acid (PT1) has been previously shown to specifically activate 5'-adenosine monophosphate-activated protein kinase (AMPK). Because AMPK activation effectively induces autophagy, we tested the protective efficacy of PT1 on cardiomyocytes after oxygen glucose deprivation/reoxygenation (OGD/R) in vitro. Mouse neonatal cardiomyocytes were treated with PT1 after OGD/R. 3-[4-(1,3-benzodioxol-5-yl)-2-oxo-3-buten-1-yl]-3-hydroxy-1,3-dihydro-2H-indol-2-one (3HOI-BA-01), a novel small compound showing potent inhibitory effect on mammalian target of rapamycin (mTOR) activation, was also tested for its cardioprotective effect, based on the established relationship between mTOR signaling and autophagy. Cell survival and autophagy-related signal pathways were examined after treatment with these agents. Our data indicate that both PT1 and 3HOI-BA-01 enhance cell survival after OGD/R. As expected, both PT1 and 3HOI-BA-01 induced autophagy in cardiomyocytes through activating AMPK pathway and inhibiting mTOR signaling, respectively. Induction of autophagy by PT1 and 3HOI-BA-01 was responsible for their cardioprotective effect, since inhibition of autophagy abolished the protective efficacy. Furthermore, simultaneous administration of PT1 and 3HOI-BA-01 profoundly upregulated autophagy after OGD/R and significantly promoted survival of cardiomyocytes. In vivo administration of PT1 and 3HOI-BA-01 in a murine myocardial (I/R injury model remarkably reduced infarct size and induced autophagy. Taken together, our research suggests that PT1 and 3HOI-BA-01 could be promising therapeutic agents for myocardial ischemia.


Subject(s)
AMP-Activated Protein Kinases/antagonists & inhibitors , Autophagy/drug effects , Benzodioxoles/pharmacology , Indoles/pharmacology , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/drug effects , Protein Kinase Inhibitors/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Thiazoles/pharmacology , meta-Aminobenzoates/pharmacology , AMP-Activated Protein Kinases/metabolism , Animals , Animals, Newborn , Apoptosis/drug effects , Cell Hypoxia , Cells, Cultured , Cytoprotection , Disease Models, Animal , Glucose/deficiency , Male , Mice , Mice, Inbred C57BL , Myocardial Infarction/enzymology , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/enzymology , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Oxygen/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
11.
J Med Chem ; 58(10): 4230-41, 2015 May 28.
Article in English | MEDLINE | ID: mdl-25938266

ABSTRACT

A series of indenopyrazoles was synthesized from the corresponding indanones and phenyl isothiocyanates in two steps. Among the compounds synthesized, methyl 3-((6-methoxy-1,4-dihydroindeno[1,2-c]pyrazol-3-yl)amino)benzoate 6m (GN39482) was found to possess a promising antiproliferative activity toward human cancer cells without affecting any antimicrobial and antimalarial activities at 100 nM. Both a methoxy group at R(1) position and a methoxycarbonyl group at R(2) position of the anilinoquinazoline framework are essential for the high cell growth inhibition. Both MorphoBase and ChemProteoBase profiling analyses suggested that compound 6m was classified as a tubulin inhibitor. Indeed, compound 6m inhibited the acetylated tubulin accumulation and the microtubule formation and induced G2/M cell cycle arrest in HeLa cells, revealing that a promising antiproliferative activity of compound 6m toward human cancer cells is probably caused by the tubulin polymerization inhibition.


Subject(s)
Drug Evaluation, Preclinical/methods , Pyrazoles/pharmacology , Structure-Activity Relationship , Tubulin Modulators/pharmacology , meta-Aminobenzoates/pharmacology , Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Antimalarials/chemistry , Antimalarials/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Chemistry Techniques, Synthetic , HeLa Cells/drug effects , Humans , Microbial Sensitivity Tests , Microtubules/drug effects , Microtubules/metabolism , Plasmodium falciparum/drug effects , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Pyrazoles/chemistry , Tubulin Modulators/chemistry , meta-Aminobenzoates/chemistry
12.
Chembiochem ; 16(2): 302-11, 2015 Jan 19.
Article in English | MEDLINE | ID: mdl-25572106

ABSTRACT

Streptomyces hygroscopicus is a natural producer of geldanamycin. Mutasynthetic supplementation of an AHBA-blocked mutant with all possible monofluoro 3-aminobenzoic acids provided new fluorogeldanamycins. These showed strong antiproliferative activity and inhibitory effects on human heat shock protein Hsp90. Binding to Hsp90 in the low nanomolar range was determined from molecular modelling, AFM analysis and by calorimetric studies.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Benzoquinones/chemistry , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Lactams, Macrocyclic/chemistry , Streptomyces/metabolism , Antineoplastic Agents/metabolism , Calorimetry/methods , Cell Line, Tumor/drug effects , Cell Proliferation/drug effects , Fluorobenzenes/metabolism , Fluorobenzenes/pharmacology , HSP90 Heat-Shock Proteins/metabolism , Humans , Magnetic Resonance Spectroscopy , Microscopy, Atomic Force , Models, Molecular , Quinones/chemistry , Streptomyces/genetics , meta-Aminobenzoates/metabolism , meta-Aminobenzoates/pharmacology
13.
Biochem J ; 466(2): 337-46, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25486442

ABSTRACT

Preventing histone recognition by bromodomains emerges as an attractive therapeutic approach in cancer. Overexpression of ATAD2 (ATPase family AAA domain-containing 2 isoform A) in cancer cells is associated with poor prognosis making the bromodomain of ATAD2 a promising epigenetic therapeutic target. In the development of an in vitro assay and identification of small molecule ligands, we conducted structure-guided studies which revealed a conformationally flexible ATAD2 bromodomain. Structural studies on apo-, peptide-and small molecule-ATAD2 complexes (by co-crystallization) revealed that the bromodomain adopts a 'closed', histone-compatible conformation and a more 'open' ligand-compatible conformation of the binding site respectively. An unexpected conformational change of the conserved asparagine residue plays an important role in driving the peptide-binding conformation remodelling. We also identified dimethylisoxazole-containing ligands as ATAD2 binders which aided in the validation of the in vitro screen and in the analysis of these conformational studies.


Subject(s)
Adenosine Triphosphatases/chemistry , DNA-Binding Proteins/chemistry , Drug Design , Enzyme Inhibitors/chemistry , Histones/chemistry , Isoxazoles/chemistry , Peptide Fragments/chemistry , Protein Processing, Post-Translational , ATPases Associated with Diverse Cellular Activities , Adenosine Triphosphatases/antagonists & inhibitors , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Binding Sites , Biotinylation , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Histones/antagonists & inhibitors , Histones/metabolism , Humans , Isoxazoles/chemical synthesis , Isoxazoles/pharmacology , Kinetics , Ligands , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/metabolism , Pliability , Protein Conformation , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sulfonamides/chemical synthesis , Sulfonamides/chemistry , Sulfonamides/pharmacology , meta-Aminobenzoates/chemical synthesis , meta-Aminobenzoates/chemistry , meta-Aminobenzoates/pharmacology
14.
Arch Pharm Res ; 37(2): 232-8, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23733585

ABSTRACT

Neu2000 (NEU; 2-hydroxy-5-(2,3,5,6-tetrafluoro-4-trifluoromethyl-benzylamino)-benzoic acid), a recently developed derivative of acetylsalicylic acid and sulfasalazine, potently protects against neuronal cell death following ischemic brain injury by antagonizing NMDA receptor-mediated neuronal toxicity and oxidative stress. However, it has yet to be determined whether NEU can attenuate hypoxia-induced impairment of neuronal electrical activity. In this study, we carried out extracellular recordings of hippocampal slices in order to investigate the effects of NEU on the electrical activity of neurons exposed to a hypoxic insult (oxygen and glucose deprivation). NEU prominently suppressed hypoxia-induced impairment of neuronal activity in a concentration-dependent manner. NEU, at a low dose (1 µM), competently depressed the hypoxia-induced convulsive activity in a manner similar to trolox. Furthermore, high concentrations of NEU (50 µM) markedly abolished all hypoxia-mediated impairment of neuronal activity and accelerated the slow recovery of neuronal activity more efficiently than ifenprodil and APV. These results suggest that NEU attenuates hypoxia-induced impairment of neuronal activity more potently than the antioxidant, trolox, and the NMDA receptor antagonists, ifenprodil and APV. We propose that NEU is a striking pharmacological candidate for neuroprotection against hypoxia because of its defensive action on hypoxia-mediated impairment of electrical neurotransmission as well as its neuroprotective action against neuronal cell death induced by exposure to pathological hypoxic conditions.


Subject(s)
Antioxidants/pharmacology , CA3 Region, Hippocampal/drug effects , Fluorobenzenes/pharmacology , Hypoxia/physiopathology , Neuroprotective Agents/pharmacology , Salicylates/pharmacology , Synaptic Transmission/drug effects , meta-Aminobenzoates/pharmacology , Animals , CA3 Region, Hippocampal/metabolism , CA3 Region, Hippocampal/physiopathology , Dose-Response Relationship, Drug , Glucose/metabolism , In Vitro Techniques , Microelectrodes , Neurons/drug effects , Oxygen/metabolism , Rats , Rats, Sprague-Dawley
15.
J Pharmacol Sci ; 122(2): 149-52, 2013.
Article in English | MEDLINE | ID: mdl-23698159

ABSTRACT

Neu2000, acting as an antioxidant with N-methyl-d-aspartate-receptor antagonism, demonstrates excellent protection against ischemic insults in rodents. In this study, we report that Neu2000 also dramatically enhances the activity of kainate (KA) receptors. Neu2000 non-competitively and reversibly potentiated KA-evoked responses in a voltage-independent manner, mainly by increasing the open probability of KA receptor channels.


Subject(s)
Antioxidants/pharmacology , Cerebral Cortex , Fluorobenzenes/pharmacology , Neurons/metabolism , Receptors, Kainic Acid/metabolism , Salicylates/pharmacology , meta-Aminobenzoates/pharmacology , Animals , Evoked Potentials/drug effects , Mice , Mice, Inbred ICR , Receptors, Kainic Acid/physiology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Stimulation, Chemical
16.
Toxicol In Vitro ; 27(2): 788-97, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23268106

ABSTRACT

Neu2000 [2-hydroxy-5-(2,3,5,6-tetrafluoro-4 trifluoromethylbenzylamino) benzoic acid] is a dual-acting neuroprotective agent that functions both as a noncompetitive N-methyl-d-aspartate (NMDA) receptor antagonist and a free radical scavenger. In the present study, we investigated the scavenging activity of Neu2000 on various classes of reactive oxygen species and reactive nitrogen species (ROS/RNS) as well as its efficacy for reducing free radicals and oxidative stress/damage induced in spinal cord mitochondrial preparations. Neu2000 exerted scavenging activity against superoxide, nitric oxide, and hydroxyl radicals, and efficiently scavenged peroxynitrite. In the mitochondrial studies, Neu2000 markedly inhibited ROS/RNS and hydrogen peroxide levels following antimycin treatment. In addition, Neu2000 effectively scavenged hydroxyl radicals generated by iron(III)-ascorbate, reduced protein carbonyl formation mediated by hydroxyl radicals and peroxynitrite, and prevented glutathione oxidation caused by tert-butyl hydroperoxide in isolated mitochondria. Interestingly, incubation of isolated mitochondria with Neu2000 followed by centrifugation and removal of the supernatant also resulted in a concentration-dependent decrease in lipid peroxidation. This observation suggests that Neu2000 enters mitochondria to target free radicals or indirectly affects mitochondrial function in a manner that promotes antioxidant activity. The results of the present study demonstrate that Neu2000 possesses potent in vitro antioxidant activity due, most likely, to its active phenoxy group.


Subject(s)
Antioxidants/pharmacology , Fluorobenzenes/pharmacology , Mitochondria/drug effects , Neuroprotective Agents/pharmacology , Salicylates/pharmacology , meta-Aminobenzoates/pharmacology , Animals , Female , Free Radicals/metabolism , Glutathione/metabolism , In Vitro Techniques , Iron/metabolism , Mitochondria/metabolism , Nitric Oxide/metabolism , Oxidative Stress/drug effects , Protein Carbonylation/drug effects , Rats , Rats, Long-Evans , Spinal Cord/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...