Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 168
Filter
Add more filters

Publication year range
1.
Cell Commun Signal ; 22(1): 233, 2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38641599

ABSTRACT

BACKGROUND: Multiple neurodegenerative diseases are induced by the formation and deposition of protein aggregates. In particular, the microtubule-associated protein Tau leads to the development of so-called tauopathies characterized by the aggregation of hyperphosphorylated Tau within neurons. We recently showed that the constitutive activity of the serotonin receptor 7 (5-HT7R) is required for Tau hyperphosphorylation and aggregation through activation of the cyclin-dependent kinase 5 (CDK5). We also demonstrated physical interaction between 5-HT7R and CDK5 at the plasma membrane suggesting that the 5-HT7R/CDK5 complex is an integral part of the signaling network involved in Tau-mediated pathology. METHODS: Using biochemical, microscopic, molecular biological, computational and AI-based approaches, we investigated structural requirements for the formation of 5-HT7R/CDK5 complex. RESULTS: We demonstrated that 5-HT7R domains responsible for coupling to Gs proteins are not involved in receptor interaction with CDK5. We also created a structural model of the 5-HT7R/CDK5 complex and refined the interaction interface. The model predicted two conserved phenylalanine residues, F278 and F281, within the third intracellular loop of 5-HT7R to be potentially important for complex formation. While site-directed mutagenesis of these residues did not influence Gs protein-mediated receptor signaling, replacement of both phenylalanines by alanine residues significantly reduced 5-HT7R/CDK5 interaction and receptor-mediated CDK5 activation, leading to reduced Tau hyperphosphorylation and aggregation. Molecular dynamics simulations of 5-HT7R/CDK5 complex for wild-type and receptor mutants confirmed binding interface stability of the initial model. CONCLUSIONS: Our results provide a structural basis for the development of novel drugs targeting the 5-HT7R/CDK5 interaction interface for the selective treatment of Tau-related disorders, including frontotemporal dementia and Alzheimer's disease.


Subject(s)
Cyclin-Dependent Kinase 5 , Enzyme Activation , Receptors, Serotonin , Humans , Alzheimer Disease/metabolism , Cyclin-Dependent Kinase 5/chemistry , Cyclin-Dependent Kinase 5/genetics , Cyclin-Dependent Kinase 5/metabolism , Phosphorylation , Receptors, Serotonin/chemistry , Receptors, Serotonin/genetics , Receptors, Serotonin/metabolism , Signal Transduction
2.
Angew Chem Int Ed Engl ; 63(3): e202312942, 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38062619

ABSTRACT

The development of a small-molecule probe designed to selectively target neurons would enhance the exploration of intricate neuronal structures and functions. Among such probes, NeuO stands out as the pioneer and has gained significant traction in the field of research. Nevertheless, neither the mechanism behind neuron-selectivity nor the cellular localization has been determined. Here, we introduce NeuM, a derivative of NeuO, designed to target neuronal cell membranes. Furthermore, we elucidate the mechanism behind the selective neuronal membrane trafficking that distinguishes neurons. In an aqueous buffer, NeuM autonomously assembles into micellar structures, leading to the quenching of its fluorescence (Φ=0.001). Upon exposure to neurons, NeuM micelles were selectively internalized into neuronal endosomes via clathrin-mediated endocytosis. Through the endocytic recycling pathway, NeuM micelles integrate into neuronal membrane, dispersing fluorescent NeuM molecules in the membrane (Φ=0.61). Molecular dynamics simulations demonstrated that NeuM, in comparison to NeuO, possesses optimal lipophilicity and molecular length, facilitating its stable incorporation into phospholipid layers. The stable integration of NeuM within neuronal membrane allows the prolonged monitoring of neurons, as well as the visualization of intricate neuronal structures.


Subject(s)
Clathrin , Micelles , Clathrin/metabolism , Endocytosis/physiology , Endosomes/metabolism , Neurons/metabolism
3.
Int J Mol Sci ; 24(4)2023 Feb 16.
Article in English | MEDLINE | ID: mdl-36835378

ABSTRACT

In traditional herbal medicine, the Polyscias fruticosa has been frequently used for the treatment of ischemia and inflammation. Oxidative stress mediated by elevated glutamate levels cause neuronal cell death in ischemia and various neurodegenerative diseases. However, so far, the neuroprotective effects of this plant extract against glutamate-mediated cell death have not been investigated in cell models. The current study investigates the neuroprotective effects of ethanol extracts of Polyscias fruticosa (EEPF) and elucidates the underlying molecular mechanisms of EEPFs relevant to neuroprotection against glutamate-mediated cell death. The oxidative stress-mediated cell death was induced by 5 mM glutamate treatment in HT22 cells. The cell viability was measured by a tetrazolium-based EZ-Cytox reagent and Calcein-AM fluorescent dye. Intracellular Ca2+ and ROS levels were measured by fluorescent dyes, fluo-3 AM and 2',7'-dichlorodihydrofluorescein diacetate (DCF-DA), respectively. Protein expressions of p-AKT, BDNF, p-CREB, Bax, Bcl-2, and apoptosis-inducing factor (AIF) were determined by western blot analysis. The apoptotic cell death was measured by flow cytometry. The in vivo efficacy of EEPF was evaluated using the Mongolian gerbil mouse by surgery-induced brain ischemia. EEPF treatment showed a neuroprotective effect against glutamate-induced cell death. The EEPF co-treatment reduced the intracellular Ca2+ and ROS and apoptotic cell death. Furthermore, it recovered the p-AKT, p-CREB, BDNF, and Bcl-2 levels decreased by glutamate. The EEPF co-treatment suppressed the activation of apoptotic Bax, the nuclear translocation of AIF, and mitogen-activated protein kinase (MAPK) pathway proteins (ERK1/2, p38, JNK). Further, EEPF treatment significantly rescued the degenerative neurons in the ischemia-induced Mongolian gerbil in vivo model. EEPF exhibited neuroprotective properties that suppress glutamate-mediated neurotoxicity. The underlying mechanism of EEPF is increasing the level of p-AKT, p-CREB, BDNF, and Bcl-2 associated with cell survival. It has therapeutic potential for the treatment of glutamate-mediated neuropathology.


Subject(s)
Ethanol , Magnoliopsida , Neurons , Neuroprotective Agents , Plant Extracts , Animals , bcl-2-Associated X Protein/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Cell Line , Glutamic Acid/metabolism , Hippocampus/metabolism , Neurons/drug effects , Neuroprotective Agents/pharmacology , Oxidative Stress , Plant Extracts/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Magnoliopsida/chemistry
4.
J Enzyme Inhib Med Chem ; 36(1): 856-868, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33771089

ABSTRACT

The present study describes evaluation of epigenetic regulation by a small molecule as the therapeutic potential for treatment of Huntington's disease (HD). We identified 5-allyloxy-2-(pyrrolidin-1-yl)quinoline (APQ) as a novel SETDB1/ESET inhibitor using a combined in silico and in vitro cell based screening system. APQ reduced SETDB1 activity and H3K9me3 levels in a HD cell line model. In particular, not only APQ reduced H3K9me3 levels in the striatum but it also improved motor function and neuropathological symptoms such as neuronal size and activity in HD transgenic (YAC128) mice with minimal toxicity. Using H3K9me3-ChIP and genome-wide sequencing, we also confirmed that APQ modulates H3K9me3-landscaped epigenomes in YAC128 mice. These data provide that APQ, a novel small molecule SETDB1 inhibitor, coordinates H3K9me-dependent heterochromatin remodelling and can be an epigenetic drug for treating HD, leading with hope in clinical trials of HD.


Subject(s)
Disease Models, Animal , Enzyme Inhibitors/pharmacology , Heterochromatin/drug effects , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Huntington Disease/drug therapy , Neurons/drug effects , Animals , Behavior, Animal/drug effects , Biosensing Techniques , Cell Survival/drug effects , Cells, Cultured , Enzyme Inhibitors/chemistry , Fluorescence Resonance Energy Transfer , Heterochromatin/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Huntington Disease/metabolism , Huntington Disease/pathology , Mice , Mice, Transgenic , Molecular Structure , Neurons/metabolism , Neurons/pathology
5.
Molecules ; 26(11)2021 May 31.
Article in English | MEDLINE | ID: mdl-34073016

ABSTRACT

The pathogenesis of several neurodegenerative diseases such as Alzheimer's or Huntington's disease has been associated with metabolic dysfunctions caused by imbalances in the brain and cerebral spinal fluid levels of neuroactive metabolites. Kynurenine monooxygenase (KMO) is considered an ideal therapeutic target for the regulation of neuroactive tryptophan metabolites. Despite significant efforts, the known KMO inhibitors lack blood-brain barrier (BBB) permeability and upon the mimicking of the substrate binding mode, are subject to produce reactive oxygen species as a side reaction. The computational drug design is further complicated by the absence of complete crystal structure information for human KMO (hKMO). In the current work, we performed virtual screening of readily available compounds using several protein-ligand complex pharmacophores. Each of the pharmacophores accounts for one of three distinct reported KMO protein-inhibitor binding conformations. As a result, six novel KMO inhibitors were discovered based on an in vitro fluorescence assay. Compounds VS1 and VS6 were predicted to be BBB permeable and avoid the hydrogen peroxide production dilemma, making them valuable, novel hit compounds for further drug property optimization and advancement in the drug design pipeline.


Subject(s)
Kynurenine 3-Monooxygenase/antagonists & inhibitors , Kynurenine 3-Monooxygenase/metabolism , Blood-Brain Barrier/metabolism , Brain/metabolism , Computational Biology/methods , Drug Design , Drug Evaluation, Preclinical/methods , Enzyme Inhibitors/pharmacology , Humans , Kynurenine/metabolism , Kynurenine 3-Monooxygenase/chemistry , Molecular Docking Simulation/methods , Neurodegenerative Diseases/drug therapy , Protein Conformation
6.
Bioorg Med Chem Lett ; 30(4): 126882, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31889666

ABSTRACT

Development of highly effective, safe, and fast-acting anti-depressants is urgently required for the treatment of major depressive disorder. It has been suggested that targeting 5-HT2A and 5-HT2C in addition to inhibition of serotonin reuptake may be beneficial in generating anti-depressant agents with better pharmacology and less adverse effects. We have developed phthalazinone-based compounds that potently bind to 5-HT2A, 5-HT2C, and the serotonin transporter. The representative compounds 11j and 11l displayed strong binding affinities against these targets, and showed favorable toxicity profiles as determined by hERG binding and CYP inhibition assays. Furthermore, these compounds presented promising anti-depressant effects comparable to fluoxetine and also synergistic effects with fluoxetine in forced swimming test, which implicates these compounds can be developed to help the treatment of major depressive disorder.


Subject(s)
Antidepressive Agents/chemistry , Azoles/chemistry , Receptor, Serotonin, 5-HT2A/chemistry , Receptor, Serotonin, 5-HT2C/chemistry , Serotonin Plasma Membrane Transport Proteins/chemistry , Animals , Antidepressive Agents/metabolism , Antidepressive Agents/pharmacology , Behavior, Animal/drug effects , Drug Design , Fluoxetine/chemistry , Fluoxetine/pharmacology , Humans , Inhibitory Concentration 50 , Mice , Receptor, Serotonin, 5-HT2A/metabolism , Receptor, Serotonin, 5-HT2C/metabolism , Serotonin 5-HT2 Receptor Antagonists/chemistry , Serotonin 5-HT2 Receptor Antagonists/metabolism , Serotonin 5-HT2 Receptor Antagonists/pharmacology , Serotonin Plasma Membrane Transport Proteins/metabolism , Selective Serotonin Reuptake Inhibitors/chemistry , Selective Serotonin Reuptake Inhibitors/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology , Structure-Activity Relationship
7.
J Enzyme Inhib Med Chem ; 35(1): 1568-1580, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32752896

ABSTRACT

Herein, two new series of N-substituted indole-based analogues were rationally designed, synthesized via microwave heating technology, and evaluated as noteworthy MAO-B potential inhibitors. Compared to the reported indazole-based hits VI and VII, compounds 4b and 4e exhibited higher inhibitory activities over MAO-B with IC50 values of 1.65 and 0.78 µM, respectively. When compared to the modest selectivity index of rasagiline (II, a well-known MAO-B inhibitor, SI > 50), both 4b and 4e also showed better selectivity indices (SI > 60 and 120, respectively). A further kinetic evaluation of the most potent derivative (4e) displayed a competitive mode of inhibition (inhibition constant (K i)/MAO-B = 94.52 nM). Reasonable explanations of the elicited biological activities were presented via SAR study and molecular docking simulation. Accordingly, the remarkable MAO-B inhibitory activity of 4e (N-(1-(3-fluorobenzoyl)-1H-indol-5-yl)pyrazine-2-carboxamide), with its selectivity and competitive inhibition, advocates its potential role as a promising lead worthy of further optimization.


Subject(s)
Drug Discovery , Indoles/pharmacology , Monoamine Oxidase Inhibitors/pharmacology , Monoamine Oxidase/metabolism , Dose-Response Relationship, Drug , Humans , Indoles/chemistry , Kinetics , Models, Molecular , Molecular Structure , Monoamine Oxidase Inhibitors/chemical synthesis , Monoamine Oxidase Inhibitors/chemistry , Structure-Activity Relationship
8.
Bioorg Med Chem Lett ; 29(10): 1168-1172, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30928197

ABSTRACT

Highly effective and safe drugs for the treatment of neuropathic pain are urgently required and it was shown that blocking T-type calcium channels can be a promising strategy for drug development for neuropathic pain. We have developed pyrrolidine-based T-type calcium channel inhibitors by structural hybridization and subsequent assessment of in vitro activities against Cav3.1 and Cav3.2 channels. Profiling of in vitro ADME properties of compounds was also carried out. The representative compound 17h showed comparable in vivo efficacy to gabapentin in the SNL model, which indicates T-type calcium channel inhibitors can be developed as effective therapeutics for neuropathic pain.


Subject(s)
Analgesics/chemistry , Calcium Channel Blockers/chemistry , Calcium Channels, T-Type/metabolism , Neuralgia/drug therapy , Pyrrolidines/chemistry , Analgesics/pharmacology , Animals , Calcium Channel Blockers/pharmacology , Calcium Signaling/drug effects , Disease Models, Animal , Gabapentin/metabolism , Ganglia, Spinal/drug effects , Humans , Ligation , Microsomes, Liver/drug effects , Molecular Structure , Pyrrolidines/pharmacology , Rats , Structure-Activity Relationship
9.
Bioorg Chem ; 92: 103261, 2019 11.
Article in English | MEDLINE | ID: mdl-31542718

ABSTRACT

Inhibition of IKK-ß (inhibitor of nuclear factor kappa-B kinase subunit beta) has been broadly documentedas a promising approach for treatment of acute and chronic inflammatory diseases, cancer, and autoimmune diseases. Recently, we have identified a novel class of thiazolidine-2,4-diones as structurally novel modulators for IKK-ß. Herein, we report a hit optimization study via analog synthesis strategy aiming to acquire more potent derivative(s), probe the structure activity relationship (SAR), and get reasonable explanations for the elicited IKK-ß inhibitory activities though an in silico docking simulation study. Accordingly, a new series of eighteen thiazolidine-2,4-dione derivatives was rationally designed, synthesized, identified with different spectroscopic techniques and biologically evaluated as noteworthy IKK-ß potential modulators. Successfully, new IKK-ß potent modulators were obtained, including the most potent analog up-to-date 7m with IC50 value of 260 nM. A detailed structure activity relationship (SAR) was discussed and a mechanistic study for 7m was carried out indicating its irreversible inhibition mode with IKK-ß (Kinact value = 0.01 (min-1). Furthermore, the conducted in silico simulation study provided new insights for the binding modes of this novel class of modulators with IKK-ß.


Subject(s)
Drug Design , I-kappa B Kinase/antagonists & inhibitors , Molecular Docking Simulation , Protein Kinase Inhibitors/pharmacology , Thiazolidinediones/pharmacology , Dose-Response Relationship, Drug , Humans , I-kappa B Kinase/metabolism , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship , Thiazolidinediones/chemical synthesis , Thiazolidinediones/chemistry
10.
Molecules ; 24(22)2019 Nov 12.
Article in English | MEDLINE | ID: mdl-31726716

ABSTRACT

In this study, we investigate the atomistic details of Keap1-Nrf2 inhibitors by in-depth modeling techniques, including molecular dynamics (MD) simulations, and the path-based free energy method of umbrella sampling (US). The protein-protein interaction (PPI) of Keap1-Nrf2 is implicated in several neurodegenerative diseases like cancer, diabetes, and cardiomyopathy. A better understanding of the five sub-pocket binding sites for Nrf2 (ETGE and DLG motifs) inside the Kelch domain would expedite the inhibitor design process. We selected four protein-ligand complexes with distinct co-crystal ligands and binding occupancies inside the Nrf2 binding site. We performed 100 ns of MD simulation for each complex and analyzed the trajectories. From the results, it is evident that one ligand (1VV) has flipped inside the binding pocket, whereas the remaining three were stable. We found that Coulombic (Arg483, Arg415, Ser363, Ser508, and Ser602) and Lennard-Jones (Tyr525, Tyr334, and Tyr572) interactions played a significant role in complex stability. The obtained binding free energy values from US simulations were consistent with the potencies of simulated ligands. US simulation highlight the importance of basic and aromatic residues in the binding pocket. A detailed description of the dissociation process brings valuable insight into the interaction of the four selected protein-ligand complexes, which could help in the future to design more potent PPI inhibitors.


Subject(s)
Drug Discovery , Kelch-Like ECH-Associated Protein 1/chemistry , Molecular Docking Simulation , Molecular Dynamics Simulation , NF-E2-Related Factor 2/chemistry , Protein Binding/drug effects , Binding Sites , Drug Discovery/methods , Hydrogen Bonding , Kelch-Like ECH-Associated Protein 1/metabolism , Ligands , Molecular Structure , NF-E2-Related Factor 2/metabolism , Quantitative Structure-Activity Relationship , Structure-Activity Relationship
11.
Molecules ; 24(18)2019 Sep 05.
Article in English | MEDLINE | ID: mdl-31491978

ABSTRACT

Here, we describe the synthesis of disubstituted pyrimidine derivatives and their biological evaluation as selective 5-HT2C agonists. To improve selectivity for 5-HT2C over other subtypes, we synthesized two series of disubstituted pyrimidines with fluorophenylalkoxy groups at either the 5-position or 4-position and varying cyclic amines at the 2-position. The in vitro cell-based assay and binding assay identified compounds 10a and 10f as potent 5-HT2C agonists. Further studies on selectivity to 5-HT subtypes and drug-like properties indicated that 2,4-disubstituted pyrimidine 10a showed a highly agonistic effect on the 5-HT2C receptor, with excellent selectivity, as well as exceptional drug-like properties, including high plasma and microsomal stability, along with low CYP inhibition. Thus, pyrimidine 10a could be considered a viable lead compound as a 5-HT2C selective agonist.


Subject(s)
Pyrimidines/chemical synthesis , Pyrimidines/pharmacology , Receptor, Serotonin, 5-HT2C/chemistry , Serotonin 5-HT2 Receptor Agonists/chemical synthesis , Serotonin 5-HT2 Receptor Agonists/pharmacology , Chemistry Techniques, Synthetic , Dose-Response Relationship, Drug , Drug Stability , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Structure , Protein Binding , Pyrimidines/chemistry , Receptor, Serotonin, 5-HT2C/metabolism , Serotonin 5-HT2 Receptor Agonists/chemistry , Structure-Activity Relationship
12.
Bioorg Med Chem ; 26(21): 5596-5611, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30385226

ABSTRACT

Two new series of 5-subtituted and 5,6-disubstituted pyrrolo[2,3-d]pyrimidine octamides (4a-o and 6a-g) and their corresponding free amines 5a-m and 7a-g have been synthesized and biologically evaluated for their antiproliferative activity against three human cancer cell lines. The 5,6-disubstituted octamides 6d-g as well as the amine derivative 7b have shown the best anticancer activity with single digit micromolar GI50 values over the tested cancer cells, and low cytotoxic effects (GI50 > 10.0 µM) against HFF-1 normal cell. A structure activity relationship (SAR) study has been established and disclosed that terminal octamide moiety at C2 as well as disubstitution with fluorobenzyl piperazines at C5 and C6 of pyrrolo[2,3-d]pyrimidine are the key structural features prerequisite for best antiproliferative activity. Moreover, the most active member 6f was tested for its antiproliferative activity over a panel of 60 cancer cell lines at NCI, and exhibited distinct broad spectrum anticancer activity with submicromolar GI50 and TGI values over multiple cancer cells. Kinase profile of compound 6f over 53 oncogenic kinases at 10 µM concentration showed its highly selective inhibitory activity towards FGFR4, Tie2 and TrkA kinases. The observed activity of 6f against TrkA (IC50 = 2.25 µM), FGFR4 (IC50 = 6.71 µM) and Tie2 (IC50 = 6.84 µM) was explained by molecular docking study, which also proposed that 6f may be a type III kinase inhibitor, binding to an allosteric site rather than kinase hinge region. Overall, compound 6f may serve as a promising anticancer lead compound that could be further optimized for development of potent anticancer agents.


Subject(s)
Antineoplastic Agents/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Pyrroles/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Humans , Molecular Docking Simulation , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacokinetics , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Pyrimidines/pharmacokinetics , Pyrroles/chemical synthesis , Pyrroles/chemistry , Pyrroles/pharmacokinetics , Receptor, Fibroblast Growth Factor, Type 4/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 4/chemistry , Receptor, TIE-2/antagonists & inhibitors , Receptor, TIE-2/chemistry , Receptor, trkA/antagonists & inhibitors , Receptor, trkA/chemistry
13.
Bioorg Med Chem ; 26(15): 4382-4389, 2018 08 15.
Article in English | MEDLINE | ID: mdl-30054191

ABSTRACT

In this study, we designed and synthesized several novel "Y"-shaped biaryl PPARδ agonists. Structure-activity relationship (SAR) studies demonstrated that compound 3a was the most active agonist with an EC50 of 2.6 nM. We also synthesized and evaluated enantiospecific R and S isomers of compound 3a to confirm that R isomer (EC50 = 0.7 nM) shows much more potent activity than S isomer (EC50 = 6.1 nM). Molecular docking studies between the PPAR ligand binding domain and enantiospecific R and S isomers of compound 3a were performed. In vitro absorption, distribution, metabolism, excretion, and toxicity (ADMET) and in vivo PK profiles show that compound 3a possesses superior drug-like properties including good bioavailability. Our overall results clearly demonstrate that this orally administrable PPARδ agonist 3a is a viable drug candidate for the treatment of various PPARδ-related disorders.


Subject(s)
Acetates/chemistry , Biphenyl Compounds/chemical synthesis , PPAR delta/agonists , Acetates/chemical synthesis , Acetates/pharmacokinetics , Administration, Oral , Animals , Binding Sites , Biphenyl Compounds/chemistry , Biphenyl Compounds/pharmacology , Crystallography, X-Ray , Drug Design , Half-Life , Humans , Inhibitory Concentration 50 , Mice , Microsomes/metabolism , Molecular Docking Simulation , PPAR delta/metabolism , Protein Structure, Tertiary , Rats , Stereoisomerism , Structure-Activity Relationship , Thiazoles/chemistry , Thiazoles/metabolism
14.
Bioorg Med Chem ; 26(1): 232-244, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29198609

ABSTRACT

Benzyloxyphenyl moiety is a common structure of highly potent, selective and reversible inhibitors of monoamine oxidase B (MAO-B), safinamide and sembragiline. We synthesized 4-(benzyloxy)phenyl and biphenyl-4-yl derivatives including halogen substituents on the terminal aryl unit. In addition, we modified the carbon linker between amine group and the biaryl linked unit. Among synthesized compounds, 12c exhibited the most potent and selective MAO-B inhibitory effect (hMAO-B IC50: 8.9 nM; >10,000-fold selectivity over MAO-A) as a competitive inhibitor. In addition, 12c showed greater MAO-B inhibitory activity and selectivity compared to well-known MAO-B inhibitors such as selegiline, safinamide and sembragiline. In the MPTP-induced mouse model of Parkinson's disease (PD), 12c significantly protected the tyrosine hydroxylase (TH)-immunopositive DAergic neurons and attenuated the PD-associated behavioral deficits. This study suggests characteristic structures as a MAO-B inhibitor that may provide a good insight for the development of therapeutic agents for PD.


Subject(s)
Benzene Derivatives/pharmacology , Monoamine Oxidase Inhibitors/pharmacology , Monoamine Oxidase/metabolism , Parkinson Disease/drug therapy , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/administration & dosage , Animals , Benzene Derivatives/chemical synthesis , Benzene Derivatives/chemistry , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Male , Mice , Mice, Inbred C57BL , Molecular Structure , Monoamine Oxidase Inhibitors/chemical synthesis , Monoamine Oxidase Inhibitors/chemistry , Parkinson Disease/metabolism , Parkinson Disease/pathology , Structure-Activity Relationship
15.
J Enzyme Inhib Med Chem ; 33(1): 1460-1471, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30231778

ABSTRACT

The treatment of neuropathic pain is one of the urgent unmet medical needs and T-type calcium channels are promising therapeutic targets for neuropathic pain. Several potent T-type channel inhibitors showed promising in vivo efficacy in neuropathic pain animal models and are being investigated in clinical trials. Herein we report development of novel pyrrolidine-based T-type calcium channel inhibitors by pharmacophore mapping and structural hybridisation followed by evaluation of their Cav3.1 and Cav3.2 channel inhibitory activities. Among potent inhibitors against both Cav3.1 and Cav3.2 channels, a promising compound 20n based on in vitro ADME properties displayed satisfactory plasma and brain exposure in rats according to in vivo pharmacokinetic studies. We further demonstrated that 20n effectively improved the symptoms of neuropathic pain in both SNL and STZ neuropathic pain animal models, suggesting modulation of T-type calcium channels can be a promising therapeutic strategy for the treatment of neuropathic pain.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Channels, T-Type/metabolism , Neuralgia/drug therapy , Pyrrolidines/pharmacology , Animals , Calcium Channel Blockers/chemical synthesis , Calcium Channel Blockers/chemistry , Disease Models, Animal , HEK293 Cells , Humans , Ligation , Male , Mice , Mice, Knockout , Molecular Structure , Neuralgia/chemically induced , Neuralgia/metabolism , Pyrrolidines/chemical synthesis , Pyrrolidines/chemistry , Rats , Rats, Sprague-Dawley , Spinal Nerves/surgery , Streptozocin
16.
Acta Neuropathol ; 134(5): 729-748, 2017 11.
Article in English | MEDLINE | ID: mdl-28593442

ABSTRACT

Huntington's disease (HD) is an autosomal-dominant inherited neurological disorder caused by expanded CAG repeats in exon 1 of the Huntingtin (HTT) gene. Altered histone modifications and epigenetic mechanisms are closely associated with HD suggesting that transcriptional repression may play a pathogenic role. Epigenetic compounds have significant therapeutic effects in cellular and animal models of HD, but they have not been successful in clinical trials. Herein, we report that dSETDB1/ESET, a histone methyltransferase (HMT), is a mediator of mutant HTT-induced degeneration in a fly HD model. We found that nogalamycin, an anthracycline antibiotic and a chromatin remodeling drug, reduces trimethylated histone H3K9 (H3K9me3) levels and pericentromeric heterochromatin condensation by reducing the expression of Setdb1/Eset. H3K9me3-specific ChIP-on-ChIP analysis identified that the H3K9me3-enriched epigenome signatures of multiple neuronal pathways including Egr1, Fos, Ezh1, and Arc are deregulated in HD transgenic (R6/2) mice. Nogalamycin modulated the expression of the H3K9me3-landscaped epigenome in medium spiny neurons and reduced mutant HTT nuclear inclusion formation. Moreover, nogalamycin slowed neuropathological progression, preserved motor function, and extended the life span of R6/2 mice. Together, our results indicate that modulation of SETDB1/ESET and H3K9me3-dependent heterochromatin plasticity is responsible for the neuroprotective effects of nogalamycin in HD and that small compounds targeting dysfunctional histone modification and epigenetic modification by SETDB1/ESET may be a rational therapeutic strategy in HD.


Subject(s)
Chromatin Assembly and Disassembly/physiology , Heterochromatin/metabolism , Huntington Disease/metabolism , Animals , Chromatin Immunoprecipitation , Disease Models, Animal , Disease Progression , Gene Expression Regulation , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Huntington Disease/mortality , Huntington Disease/pathology , Mice , Survival Rate
17.
J Comput Aided Mol Des ; 31(10): 929-941, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28913661

ABSTRACT

Cyclophilin D (CypD) is a mitochondria-specific cyclophilin that is known to play a pivotal role in the formation of the mitochondrial permeability transition pore (mPTP).The formation and opening of the mPTP disrupt mitochondrial homeostasis, cause mitochondrial dysfunction and eventually lead to cell death. Several recent studies have found that CypD promotes the formation of the mPTP upon binding to ß amyloid (Aß) peptides inside brain mitochondria, suggesting that neuronal CypD has a potential to be a promising therapeutic target for Alzheimer's disease (AD). In this study, we generated an energy-based pharmacophore model by using the crystal structure of CypD-cyclosporine A (CsA) complex and performed virtual screening of ChemDiv database, which yielded forty-five potential hit compounds with novel scaffolds. We further tested those compounds using mitochondrial functional assays in neuronal cells and identified fifteen compounds with excellent protective effects against Aß-induced mitochondrial dysfunction. To validate whether these effects derived from binding to CypD, we performed surface plasmon resonance (SPR)-based direct binding assays with selected compounds and discovered compound 29 was found to have the equilibrium dissociation constants (KD) value of 88.2 nM. This binding affinity value and biological activity correspond well with our predicted binding mode. We believe that this study offers new insights into the rational design of small molecule CypD inhibitors, and provides a promising lead for future therapeutic development.


Subject(s)
Cyclophilins/antagonists & inhibitors , Cyclosporine/chemistry , Mitochondria/drug effects , Neuroprotective Agents/chemistry , Amyloid beta-Peptides/chemistry , Animals , Binding Sites , Cell Survival , Peptidyl-Prolyl Isomerase F , Cyclophilins/metabolism , Cyclosporine/pharmacology , Databases, Pharmaceutical , HT29 Cells , Humans , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Molecular Docking Simulation , Neuroprotective Agents/pharmacology , Protein Binding , Protein Conformation , Structure-Activity Relationship
18.
J Comput Aided Mol Des ; 31(10): 877-889, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28879500

ABSTRACT

ERG-associated protein with the SET domain (ESET/SET domain bifurcated 1/SETDB1/KMT1E) is a histone lysine methyltransferase (HKMT) and it preferentially tri-methylates lysine 9 of histone H3 (H3K9me3). SETDB1/ESET leads to heterochromatin condensation and epigenetic gene silencing. These functional changes are reported to correlate with Huntington's disease (HD) progression and mood-related disorders which make SETDB1/ESET a viable drug target. In this context, the present investigation was performed to identify novel peptide-competitive small molecule inhibitors of the SETDB1/ESET by a combined in silico-in vitro approach. A ligand-based pharmacophore model was built and employed for the virtual screening of ChemDiv and Asinex database. Also, a human SETDB1/ESET homology model was constructed to supplement the data further. Biological evaluation of the selected 21 candidates singled out 5 compounds exhibiting a notable reduction of the H3K9me3 level via inhibitory potential of SETDB1/ESET activity in SETDB1/ESET-inducible cell line and HD striatal cells. Later on, we identified two compounds as final hits that appear to have neuronal effects without cytotoxicity based on the result from MTT assay. These compounds hold the calibre to become the future lead compounds and can provide structural insights into more SETDB1/ESET-focused drug discovery research. Moreover, these SETDB1/ESET inhibitors may be applicable for the preclinical study to ameliorate neurodegenerative disorders via epigenetic regulation.


Subject(s)
Benzene Derivatives/chemistry , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Protein Methyltransferases/antagonists & inhibitors , Benzene Derivatives/pharmacology , Binding Sites , Cell Line , Cell Survival , Computer Simulation , Databases, Pharmaceutical , Humans , Ligands , Methylation , Molecular Docking Simulation , Neurons/cytology , Neurons/drug effects , Protein Binding , Protein Methyltransferases/metabolism , Structure-Activity Relationship
19.
Molecules ; 22(9)2017 Aug 26.
Article in English | MEDLINE | ID: mdl-28846591

ABSTRACT

A series of pyrimidine derivatives 4a-i were synthesized and evaluated for their binding affinities towards 5-HT2C receptors. With regard to designed molecules 4a-i, the influence of the size of alkyl ether and the absolute configuration of a stereogenic center on the 5-HT2C binding affinity and selectivity was studied. The most promising diasteromeric mixtures 4d and 4e were selected in the initial radioligand binding assay and they were further synthesized as optically active forms starting from optically active alcohols 5d and 5e, prepared by an enzymatic kinetic resolution. Pyrimidine analogue (R,R)-4e displayed an excellent 5-HT2C binding affinity with good selectivity values against a broad range of other 5-HT receptor subtypes.


Subject(s)
Pyrimidines/chemical synthesis , Receptor, Serotonin, 5-HT2C/metabolism , Serotonin 5-HT2 Receptor Agonists/chemical synthesis , Animals , CHO Cells , Cricetulus , Models, Molecular , Molecular Structure , Pyrimidines/chemistry , Pyrimidines/pharmacology , Serotonin 5-HT2 Receptor Agonists/chemistry , Serotonin 5-HT2 Receptor Agonists/pharmacology , Structure-Activity Relationship
20.
Mol Pharmacol ; 90(5): 530-539, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27573669

ABSTRACT

α-Pinene is a major monoterpene of the pine tree essential oils. It has been reported that α-pinene shows anxiolytic and hypnotic effects upon inhaled administration. However, hypnotic effect by oral supplementation and the molecular mechanism of α-pinene have not been determined yet. By combining in vivo sleep behavior, ex vivo electrophysiological recording from brain slices, and in silico molecular modeling, we demonstrate that (-)-α-pinene shows sleep enhancing property through a direct binding to GABAA-benzodiazepine (BZD) receptors by acting as a partial modulator at the BZD binding site. The effect of (-)-α-pinene on sleep-wake profiles was evaluated by recording electroencephalogram and electromyogram. The molecular mechanism of (-)-α-pinene was investigated by electrophysiology and molecular docking study. (-)-α-pinene significantly increased the duration of non-rapid eye movement sleep (NREMS) and reduced the sleep latency by oral administration without affecting duration of rapid eye movement sleep and delta activity. (-)-α-pinene potentiated the GABAA receptor-mediated synaptic response by increasing the decay time constant of sIPSCs in hippocampal CA1 pyramidal neurons. These effects of (-)-α-pinene on sleep and inhibitory synaptic response were mimicked by zolpidem, acting as a modulator for GABAA-BZD receptors, and fully antagonized by flumazenil, an antagonist for GABAA-BZD receptor. (-)-α-pinene was found to bind to aromatic residues of α1- and -γ2 subunits of GABAA-BZD receptors in the molecular model. We conclude that (-)-α-pinene enhances the quantity of NREMS without affecting the intensity of NREMS by prolonging GABAergic synaptic transmission, acting as a partial modulator of GABAA-BZD receptors and directly binding to the BZD binding site of GABAA receptor.


Subject(s)
Benzodiazepines/metabolism , Eye Movements/drug effects , Monoterpenes/pharmacology , Pinus/chemistry , Plant Oils/pharmacology , Receptors, GABA-A/metabolism , Sleep/drug effects , Animals , Bicyclic Monoterpenes , Binding Sites , Flumazenil/chemistry , Flumazenil/pharmacology , Inhibitory Postsynaptic Potentials/drug effects , Male , Mice, Inbred C57BL , Mice, Inbred ICR , Models, Molecular , Monoterpenes/chemistry , Pentobarbital , Pyridines/chemistry , Pyridines/pharmacology , Sleep, REM/drug effects , Time Factors , Wakefulness/drug effects , Zolpidem
SELECTION OF CITATIONS
SEARCH DETAIL