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

Country/Region as subject
Publication year range
1.
Int J Mol Sci ; 24(2)2023 Jan 04.
Article in English | MEDLINE | ID: mdl-36674424

ABSTRACT

The regulation and metabolism of the endocannabinoid system has received extensive attention for their potential neuroprotective effect in neurodegenerative diseases such as Alzheimer's disease (AD), which is characterized by amyloid ß (Aß) -induced cell toxicity, inflammation, and oxidative stress. Using in vitro techniques and two cell lines, the mouse hippocampus-derived HT22 cells and Chinese hamster ovary (CHO) cells expressing human cannabinoid receptor type 1 (CB1), we investigated the ability of endocannabinoids to inhibit Aß aggregation and protect cells against Aß toxicity. The present study provides evidence that endocannabinoids N-arachidonoyl ethanol amide (AEA), noladin and O-arachidonoyl ethanolamine (OAE) inhibit Aß42 aggregation. They were able to provide protection against Aß42 induced cytotoxicity via receptor-mediated and non-receptor-mediated mechanisms in CB1-CHO and HT22 cells, respectively. The aggregation kinetic experiments demonstrate the anti-Aß aggregation activity of some endocannabinoids (AEA, noladin). These data demonstrate the potential role and application of endocannabinoids in AD pathology and treatment.


Subject(s)
Alzheimer Disease , Endocannabinoids , Mice , Animals , Cricetinae , Humans , Endocannabinoids/pharmacology , Endocannabinoids/metabolism , Amyloid beta-Peptides/toxicity , CHO Cells , Cricetulus , Alzheimer Disease/metabolism
2.
Mol Pharm ; 19(8): 2737-2753, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35802484

ABSTRACT

Retinal ganglion cell (RGC) neurodegeneration in glaucoma has potential links with amyloid-ß (Aß) deposition. Targeting the Aß pathway was shown to reduce RGC apoptosis and protect RGCs from degeneration. We report exploratory studies on the amyloid Aß40 aggregation inhibition properties of four cell adhesion peptide (CAP)-gemini surfactants that are intended as building blocks for gene carrier nanoparticles for glaucoma treatment. The CAP-gemini surfactants (18-7N(p1-4)-18) were evaluated as potential Aß40 peptide aggregation inhibitors by a fluorescence kinetic assay and for their binding interactions with Aß40 dimers by molecular docking studies. In vitro Aß40 peptide aggregation inhibition studies showed that the 18-7N(p3)-18 and 18-7N(p1)-18 ligands inhibit Aß40 peptide aggregation and prevent the formation of higher order structures. CDOCKER energies and CDOCKER interaction energies demonstrated that the CAP-gemini surfactants formed more stable complexes in the Aß40 dimer assembly and underwent both polar and nonpolar interactions compared to CAP peptides alone. Also, 18-7N(p3)-18 showed a significantly lower CDOCKER energy compared to that of the unmodified gemini surfactant 18-7NH-18 (p < 0.0001) and 18-7N(p4)-18 (p < 0.001), 18-7N(p1)-18, and 18-7N(p2)-18. Similarly, 18-7N(p3)-18 showed a significantly lower CDOCKER interaction energy compared to that of 18-7NH-18, 18-7N(p4)-18 (p < 0.0001), and 18-7N(p2)-18 (p < 0.001), while 18-7N(p3)-18 and 18-7N(p1)-18 showed similar CDOCKER interaction energies. These studies suggest that a combination of both hydrophobic and electrostatic interactions contributes to the anti-Aß40 aggregation activity of CAP-gemini surfactants. CAP-gemini surfactants showed 10-fold better Aß40 peptide aggregation inhibition compared to previously reported values and could provide a new opportunity for glaucoma treatment as dual-functional gene carriers.


Subject(s)
Glaucoma , Surface-Active Agents , Amyloid beta-Peptides/metabolism , Glaucoma/drug therapy , Humans , Hydrophobic and Hydrophilic Interactions , Molecular Docking Simulation , Peptide Fragments/metabolism , Polymers , Surface-Active Agents/chemistry
3.
Arch Biochem Biophys ; 663: 34-43, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30593770

ABSTRACT

Polyunsaturated fatty acids (PUFAs) are reported to exert beneficial effects in Alzheimer's disease. Some PUFAs are known to reduce amyloid-beta (Aß) toxicity by promoting its degradation and clearance. Studies on the direct interactions of PUFAs with Aß peptides are limited and contradictory. In this study, we report the interactions of fatty acids docosahexaenoic acid (DHA), eicosatetraenoic acid (EPA), α-linolenic acid (ALA), arachidonic acid (ARA), linoleic acid (LNA) and oleic acid (OA) with Aß peptides by carrying out fluorescence based aggregation kinetic experiments, transmission electron microscopy and molecular docking studies. Our investigations demonstrate that all the fatty acids tested exhibit anti-aggregation properties by preventing both Aß40 and Aß42 fibrillogenesis (∼16-84% inhibition). OA and DHA were identified as excellent inhibitors of Aß40 or Aß42 fibrillogenesis respectively (∼84% and 81% inhibition at 25 µM). Molecular docking studies conducted, using the dimer and oligomer models of Aß40 peptide, suggest that these fatty acids interact in the aggregation prone Phe19-Ala21 and the ß-turn region (Asp23-Lys28) whereas a similar study with Aß42 dimer and oligomer models, indicate that the fatty acids were oriented in a hydrophobic region (Gln15, Leu16, Leu17 and Leu34). These results, suggest that DHA, EPA, ALA, ARA, LNA and OA are capable of directly interacting with both Aß40 and Aß42 peptides. These studies will have implications in developing potential therapeutics for Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides/metabolism , Fatty Acids, Unsaturated/metabolism , Peptide Fragments/metabolism , Dimerization , Humans , Kinetics , Microscopy, Electron, Transmission , Molecular Docking Simulation , Protein Binding , Spectrometry, Fluorescence
4.
Harm Reduct J ; 16(1): 17, 2019 02 27.
Article in English | MEDLINE | ID: mdl-30813917

ABSTRACT

PURPOSE: The availability of take home naloxone (THN) was increased for Canadians in 2016, including access to kits via pharmacies. Unlike typical over-the-counter (OTC) and prescription drugs, THN kits may be stored in non-standard conditions, including in vehicles, backpacks, and out of doors. To evaluate whether these non-standard storage conditions affect stability, we investigated the impact of heat and freeze-thaw cycling on naloxone hydrochloride stability. METHODS: To assess the effect of heat, naloxone hydrochloride ampoules were exposed to 80 °C in a temperature-controlled oven for 8 h followed by 16 h at room temperature. To assess the effect of freeze-thaw cycles, naloxone hydrochloride ampoules were exposed to - 20 °C for 16 h followed by 8 h at 4 °C. The impact of these conditions on naloxone hydrochloride stability was evaluated each day for 1 week and after 2 and 4 weeks. The concentration of remaining naloxone hydrochloride was quantified using high-performance liquid chromatography (HPLC). Naloxone hydrochloride ampoules stored at room temperature served as the experimental control. RESULTS: Naloxone hydrochloride ampoules exhibit no changes in drug concentration following exposure to heat or freeze-thaw cycles for up to 28 days compared to ampoules maintained at room temperature (as indicated in the product monograph). CONCLUSIONS: Naloxone hydrochloride remains chemically stable following exposure to heat or freeze-thaw cycles after 28 days. If THN kits are stored in non-standard conditions (for up to 28 days) the active naloxone is likely to remain stable. Despite this, pharmacists should continue to emphasize the importance of appropriate storage of THN kits to ensure optimal efficacy should naloxone administration be required in an emergency situation.


Subject(s)
Naloxone/chemistry , Narcotic Antagonists/chemistry , Calibration , Chromatography, High Pressure Liquid , Drug Packaging , Drug Stability , Freezing , Hot Temperature , Temperature
5.
Anal Chem ; 89(1): 910-915, 2017 01 03.
Article in English | MEDLINE | ID: mdl-27959505

ABSTRACT

96-Well plate has been the traditional method used for screening drug compounds libraries for potential bioactivity. Although this method has been proven successful in testing dose-response analysis, the microliter consumption of expensive reagents and hours of reaction and analysis time call for innovative methods for improvements. This work demonstrates a droplet microfluidic platform that has the potential to significantly reduce the reagent consumption and shorten the reaction and analysis time by utilizing nanoliter-sized droplets as a replacement of wells. This platform is evaluated by applying it to screen drug compounds that inhibit the tau-peptide aggregation, a phenomena related to Alzheimer's disease. In this platform, sample reagents are first dispersed into nanolitre-sized droplets by an immiscible carrier oil and then these droplets are trapped on-demand in the downstream of the microfluidic device. The relative decrease in fluorescence through drug inhibition is characterized using an inverted epifluorescence microscope. Finally, the trapped droplets are released on-demand after each test by manipulating the applied pressures to the channel network which allows continuous processing. The testing results agree well with that obtained from 96-well plates with much lower sample consumption (∼200 times lower than 96-well plate) and reduced reaction time due to increased surface volume ratio (2.5 min vs 2 h).


Subject(s)
Azo Compounds/analysis , Drug Evaluation, Preclinical/methods , Microfluidic Analytical Techniques , Protein Kinase Inhibitors/analysis , Azo Compounds/pharmacology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Humans , Microfluidic Analytical Techniques/instrumentation , Particle Size , Protein Aggregates/drug effects , Protein Kinase Inhibitors/pharmacology
6.
Bioorg Med Chem Lett ; 27(11): 2443-2449, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28400237

ABSTRACT

The 1,2,3,4-tetrahydroacridine derivative tacrine was the first drug approved to treat Alzheimer's disease (AD). It is known to act as a potent cholinesterase inhibitor. However, tacrine was removed from the market due to its hepatotoxicity concerns as it undergoes metabolism to toxic quinonemethide species through the cytochrome P450 enzyme CYP1A2. Despite these challenges, tacrine serves as a useful template in the development of novel multi-targeting anti-AD agents. In this regard, we sought to evaluate the risk of hepatotoxicity in a series of C9 substituted tacrine derivatives that exhibit cholinesterase inhibition properties. The hepatotoxic potential of tacrine derivatives was evaluated using recombinant cytochrome (CYP) P450 CYP1A2 and CYP3A4 enzymes. Molecular docking studies were conducted to predict their binding modes and potential risk of forming hepatotoxic metabolites. Tacrine derivatives compound 1 (N-(3,4-dimethoxybenzyl)-1,2,3,4-tetrahydroacridin-9-amine) and 2 (6-chloro-N-(3,4-dimethoxybenzyl)-1,2,3,4-tetrahydroacridin-9-amine) which possess a C9 3,4-dimethoxybenzylamino substituent exhibited weak binding to CYP1A2 enzyme (1, IC50=33.0µM; 2, IC50=8.5µM) compared to tacrine (CYP1A2 IC50=1.5µM). Modeling studies show that the presence of a bulky 3,4-dimethoxybenzylamino C9 substituent prevents the orientation of the 1,2,3,4-tetrahydroacridine ring close to the heme-iron center of CYP1A2 thereby reducing the risk of forming hepatotoxic species.


Subject(s)
Chemical and Drug Induced Liver Injury/etiology , Cytochrome P-450 CYP1A2 Inhibitors/chemistry , Cytochrome P-450 CYP3A Inhibitors/chemistry , Tacrine/analogs & derivatives , Tacrine/chemistry , Benzoflavones/chemistry , Binding Sites , Cytochrome P-450 CYP1A2/chemistry , Cytochrome P-450 CYP3A/chemistry , Humans , Ketoconazole/chemistry , Molecular Docking Simulation , Risk
7.
Phys Chem Chem Phys ; 19(3): 1953-1962, 2017 Jan 18.
Article in English | MEDLINE | ID: mdl-28009906

ABSTRACT

The effect of variation in the length of surfactant hydrocarbon tail groups was tested in a series of dissymmetric gemini surfactants (N1-alkyl N1,N1,N3,N3-tetramethyl-N3-(6-pyren-6yl)-hexyl)propane-1,3-diammonium dibromide designated as CmC3CnBr, with m = hexyl pyrene, and n = 8, 12, 14, 16, and 18. The aggregation properties of these surfactants have been investigated by means of 1H NMR, fluorescence spectroscopy, surface tension and electrical conductivity measurements. The critical micelle concentration (CMC) was determined using surface tension and confirmed using the specific conductance method. Krafft temperatures and the degree of micelle ionization (α) were obtained from specific conductance measurements. With an increase of the dissymmetry (m/n) ratio, the CMC decreased linearly and an increase in the Krafft temperatures was observed for all of the gemini surfactants. α values for the dissymmetric GS were higher than those of the m-3-m counterparts, which may be attributed to enhanced micelle-micelle interactions that arise from increased hydrophobicity of the hydrocarbon chains. The introduction of the bulky pyrenyl tail group resulted in much lower CMC values compared to their symmetrical counterparts affecting the packing of these surfactants at the air/water interface, which resulted in high-ordered structures (lamellar and inverted micelles). This in turn affected the thermodynamic parameters of the micellization.

8.
Biomacromolecules ; 16(8): 2363-73, 2015 Aug 10.
Article in English | MEDLINE | ID: mdl-26125953

ABSTRACT

Amyloid fibrils, formed by aggregation of improperly folded or intrinsically disordered proteins, are closely related with the pathology of a wide range of neurodegenerative diseases. Hence, there is a great deal of interest in developing molecules that can bind and inhibit amyloid formation. In this regard, we have investigated the effect of two positively charged polysaccharides, chitosan (CHT) and its quarternary derivative N-trimethyl chitosan chloride (TMC), on the aggregation of Aß40 peptide. Our aggregation kinetics and atomic force microscopy (AFM) studies show that both CHT and TMC exhibit a concentration-dependent inhibiting activity on Aß40 fibrillogenesis. Systematic pH-dependent studies demonstrate that the attractive electrostatic interactions between the positively charged moieties in CHT/TMC and the negatively charged residues in Aß40 play a key role in this inhibiting activity. The stronger inhibiting activity of TMC than CHT further suggests the importance of charge density of the polymer chain in interacting with Aß40 and blocking the fibril formation. The possible interactions between CHT/TMC and Aß40 are also revealed at the atomic level by molecular docking simulation, showing that the Aß40 monomer could be primarily stabilized by electrostatic interactions with charged amines of CHT and quaternary amines of TMC, respectively. Binding of CHT/TMC on the central hydrophobic core region of Aß40 peptide may be responsible for blocking the propagation of the nucleus to form fibrillar structures. These results suggest that incorporation of sugar units such as d-glucosamine and N-trimethyl-d-glucosamine into polymer structural template may serve as a new strategy for designing novel antiamyloid molecules.


Subject(s)
Amyloid beta-Peptides/chemistry , Amyloid/chemistry , Chitosan/chemistry , Peptide Fragments/chemistry , Humans , Hydrophobic and Hydrophilic Interactions , Kinetics , Microscopy, Atomic Force , Polymers/chemistry , Polysaccharides/chemistry , Protein Aggregation, Pathological
9.
Bioorg Med Chem Lett ; 25(2): 297-301, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25499431

ABSTRACT

The anti-Alzheimer's agent donepezil is known to bind to the hepatic enzyme CYP3A4, but its relationship with the efflux transporter P-glycoprotein (P-gp) is not as well elucidated. We conducted in vitro inhibition studies of donepezil using human recombinant CYP3A4 and P-gp. These studies show that donepezil is a weak inhibitor of CYP3A4 (IC50=54.68±1.00µM) whereas the reference agent ketoconazole exhibited potent inhibition (CYP3A4 IC50=0.20±0.01µM). P-gp inhibition studies indicate that donepezil exhibits better inhibition relative to CYP3A4 (P-gp EC50=34.85±4.63µM) although it was less potent compared to ketoconazole (P-gp EC50=9.74±1.23µM). At higher concentrations, donepezil exhibited significant inhibition of CYP3A4 (69%, 84% and 87% inhibition at 100, 250 and 500µM, respectively). This indicates its potential to cause drug-drug interactions with other CYP3A4 substrates upon co-administration; however, this scenario is unlikely in vivo due to the low therapeutic concentrations of donepezil. Similarly, donepezil co-administration with P-gp substrates or inhibitors is unlikely to result in beneficial or adverse drug interactions. The molecular docking studies show that the 5,6-dimethoxyindan-1-one moiety of donepezil was oriented closer to the heme center in CYP3A4 whereas in the P-gp binding site, the protonated benzylpiperidine pharmacophore of donepezil played a major role in its binding ability. Energy parameters indicate that donepezil complex with both CYP3A4 and P-gp was less stable (CDOCKER energies=-15.05 and -4.91kcal/mol, respectively) compared to the ketoconazole-CYP3A4 and P-gp complex (CDOCKER energies=-41.89 and -20.03kcal/mol, respectively).


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Alzheimer Disease/drug therapy , Cytochrome P-450 CYP3A/metabolism , Indans/pharmacology , Microsomes, Liver/drug effects , Nootropic Agents/pharmacology , Piperidines/pharmacology , Donepezil , Drug Interactions , Humans , Microsomes, Liver/metabolism , Models, Molecular , Molecular Structure , Structure-Activity Relationship
10.
Bioorg Med Chem Lett ; 25(20): 4505-8, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26351040

ABSTRACT

A group of sixteen iridoids isolated from plants used as anti-inflammatory remedies in Mexican folk medicine were evaluated for their potential to inhibit cyclooxygenase-1 (COX-1) and cyclooxygenase-2 (COX-2) enzymes. From these assays, loganic acid (10) was identified as the most promising compound with both COX-1 (36.0 ± 0.6%) and COX-2 (80.8 ± 4.0%) inhibition at 10 µM. Compound 10 shows a better inhibition against the COX-2 enzyme. Other iridoids tested in the present study showed weak or no inhibition against these enzymes. Furthermore, herein are presented key interactions of iridoid 10 with COX-1 and COX-2 enzymes through molecular docking studies. These studies suggest that 10 exhibits anti-inflammatory activity due to COX inhibition.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Cyclooxygenase Inhibitors/pharmacology , Iridoids/pharmacology , Orobanchaceae/chemistry , Penstemon/chemistry , Vitex/chemistry , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/isolation & purification , Cyclooxygenase Inhibitors/chemistry , Cyclooxygenase Inhibitors/isolation & purification , Dose-Response Relationship, Drug , Humans , Iridoids/chemistry , Iridoids/isolation & purification , Molecular Docking Simulation , Molecular Structure , Structure-Activity Relationship
11.
Bioorg Med Chem ; 23(17): 5681-92, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26211459

ABSTRACT

Steroid sulfatase (STS) catalyzes the hydrolysis of the sulfate ester group in biologically inactive sulfated steroids to give biologically active steroids. Inhibitors of STS are considered to be potential therapeutics for treating hormone-dependent cancers such as ER(+) breast cancer. A series of 4-substituted 17ß-arylsulfonamides of 17ß-aminoestra-1,3,5(10)-trien-3-ol were prepared and examined as STS inhibitors. The presence of a NO2 or Br at the 2-position of the A-ring resulted in a decrease in potency compared to their A-ring-unsubstituted counterparts. However the presence of a nitro group or fluorine atom at the 4-position of the A-ring resulted in an increase in potency and one of these compounds exhibited a Ki(app) value of 1 nM. Modeling studies provided insight into how these compounds interact with active site residues. The anti-proliferative activity of the 3'-Br, 3'-CF3, 4-NO2-3'-Br and 4-NO2-3'-CF3 derivatives were examined using the NCI 60-cell-line panel and found to have mean graph midpoint values of 1.9-3.4 µM.


Subject(s)
Arylsulfatases/chemistry , Enzyme Activation/drug effects , Enzyme Inhibitors/chemistry , Gonadal Steroid Hormones/adverse effects , Steryl-Sulfatase/antagonists & inhibitors , Sulfonamides/chemistry , Arylsulfatases/pharmacology , Humans , Molecular Structure , Sulfonamides/pharmacology
12.
Radiographics ; 34(7): 1793-806, 2014.
Article in English | MEDLINE | ID: mdl-25384279

ABSTRACT

Airway stents are increasingly used to treat symptomatic patients with obstructive tracheobronchial diseases who are not amenable to surgical resection or who have poor performance status, precluding them from resection. The most common conditions that are treated with tracheobronchial stents are primary lung cancer and metastatic disease. However, stents have also been used to treat patients with airway stenosis related to a variety of benign conditions, such as tracheobronchomalacia, relapsing polychondritis, postintubation tracheal stenosis, postoperative anastomotic stenosis, and granulomatous diseases. Additionally, airway stents can be used as a barrier method in the management of esophagorespiratory fistulas. Many types of stents are available from different manufacturers. Principally, they are classified as silicone; covered and uncovered metal; or hybrid, which are made of silicone and reinforced by metal rings. The advantages and disadvantages of each type of airway stent are carefully considered when choosing the most appropriate stent for each patient. Multidetector computed tomography plays an important role in determining the cause and assessing the location and extent of airway obstruction. Moreover, it is very accurate in its depiction of complications after airway stent placement.


Subject(s)
Airway Obstruction/diagnostic imaging , Airway Obstruction/therapy , Multidetector Computed Tomography , Stents , Airway Obstruction/etiology , Bronchial Diseases/complications , Equipment Design , Humans , Lung Diseases/complications , Radiographic Image Interpretation, Computer-Assisted , Tracheal Diseases/complications
13.
Acta Crystallogr Sect E Struct Rep Online ; 70(Pt 5): o554, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24860362

ABSTRACT

The asymmetric unit of the title compound, C15H12ClN3, contains two independent mol-ecules. The quinazoline ring system in each is essentially planar, with maximum deviations of 0.025 (16) and 0.0171 (16) Å. The dihedral angles between quinazoline ring systems and the phenyl rings are 88.25 (8) and 85.28 (16)° in the two independent mol-ecules. In the crystal, alternating independent mol-ecules are linked by N-H⋯N hydrogen bonds, forming chains along [001].

14.
Curr Alzheimer Res ; 21(1): 24-49, 2024.
Article in English | MEDLINE | ID: mdl-38623984

ABSTRACT

Microtubule-Associated Protein Tau (also known as tau) has been shown to accumulate into paired helical filaments and neurofibrillary tangles, which are known hallmarks of Alzheimer's disease (AD) pathology. Decades of research have shown that tau protein undergoes extensive post-translational modifications (PTMs), which can alter the protein's structure, function, and dynamics and impact the various properties such as solubility, aggregation, localization, and homeostasis. There is a vast amount of information describing the impact and role of different PTMs in AD pathology and neuroprotection. However, the complex interplay between these PTMs remains elusive. Therefore, in this review, we aim to comprehend the key post-translational modifications occurring in tau and summarize potential connections to clarify their impact on the physiology and pathophysiology of tau. Further, we describe how different computational modeling methods have helped in understanding the impact of PTMs on the structure and functions of the tau protein. Finally, we highlight the tau PTM-related therapeutics strategies that are explored for the development of AD therapy.


Subject(s)
Alzheimer Disease , Protein Processing, Post-Translational , tau Proteins , tau Proteins/metabolism , Humans , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Protein Processing, Post-Translational/physiology , Animals , Phosphorylation
15.
J Pharmacol Exp Ther ; 345(1): 85-94, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23349335

ABSTRACT

We compared the differential effects of positional isomers of acetylsalicylic acid (o-ASA, m-ASA, and p-ASA) on cyclooxygenase (COX) inhibition, gastric prostaglandin E2 (PGE2), malondialdehyde, tumor necrosis factor-alpha (TNF-α) levels, superoxide dismutase (SOD) activity, human adenocarcinoma colon cancer cell growth inhibition, cell proliferation, apoptosis, and cell-cycle progression. We also evaluated the gastric toxicity exerted by ASA isomers. All ASA isomers inhibit COX enzymes, but only the o-ASA exerted an irreversible inhibitory profile. We did not observe a significant difference between ASA isomers in their ability to decrease the in vivo synthesis of PGE2 and SOD activity. Furthermore, all isomers increased the levels of gastric and TNF-α when administered orally at equimolar doses. We observed a dose-dependent cell growth inhibitory effect; the order of potency was p-ASA > m-ASA ≈ o-ASA. There was a dose-dependent decrease in cell proliferation and an increase in apoptosis, with a concomitant Go/G1 arrest. The ulcerogenic profile of the three ASA isomers showed a significant difference between o-ASA (aspirin) and its two positional isomers when administered orally at equimolar doses (1 mmol/kg); the ulcer index (UI) for o-ASA indicated extensive mucosal injury (UI = 38), whereas m-ASA and p-ASA produced a significantly decreased toxic response (UI = 12 and 8, respectively) under the same experimental conditions. These results suggest that the three positional isomers of ASA exert practically the same biologic profile in vitro and in vivo but showed different safety profiles. The mechanism of gastric ulcer formation exerted by aspirin and its two isomers warrants a more detailed and thorough investigation.


Subject(s)
Antineoplastic Agents/pharmacology , Aspirin/pharmacology , Cell Proliferation/drug effects , Colonic Neoplasms/prevention & control , Cyclooxygenase Inhibitors/pharmacology , Prostaglandin-Endoperoxide Synthases/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Aspirin/chemistry , Aspirin/toxicity , Cell Cycle/drug effects , Chromatography, High Pressure Liquid , Colonic Neoplasms/enzymology , Colonic Neoplasms/pathology , Cyclooxygenase Inhibitors/chemistry , Cyclooxygenase Inhibitors/toxicity , Dose-Response Relationship, Drug , HT29 Cells , Humans , Isomerism , Male , Models, Molecular , Molecular Structure , Rats , Rats, Wistar , Stomach Ulcer/chemically induced , Structure-Activity Relationship
16.
Bioorg Med Chem Lett ; 23(1): 239-43, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23200245

ABSTRACT

The anti-Alzheimer's agent galantamine is known to possess anti-amyloid properties. However the exact mechanisms are not clear. We studied the binding interactions of galantamine with amyloid peptide dimer (Aß(1-40)) through molecular docking and molecular dynamics simulations. Galantamine's binding site within the amyloid peptide dimer was identified by docking experiments and the most stable complex was analyzed by molecular dynamics simulation. These studies show that galantamine was interacting with the central region of the amyloid dimer (Lys16-Ala21) and the C-terminal region (Ile31-Val36) with minimum structural drift of Cα atom in those regions. Strikingly, a significant drift was observed at the turn region from Asp23-Gly29 (Cα atom RMSD=9.2 Å and 11.6 Å at 50 fs and 100 fs respectively). Furthermore, galantamine's binding mode disrupts the key pi-pi stacking interaction between aromatic rings of Phe19 (chain A) and Phe19 (chain B) and intermolecular hydrogen bonds seen in unbound peptide dimer. Noticeably, the azepine tertiary nitrogen of galantamine was in close proximity to backbone CO of Leu34 (distance <3.5 Å) to stabilize the dimer conformation. In summary, the results indicate that galantamine binding to amyloid peptide dimer leads to a significant conformational change at the turn region (Asp23-Gly29) that disrupts interactions between individual ß-strands and promotes a nontoxic conformation of Aß(1-40) to prevent the formation of neurotoxic oligomers.


Subject(s)
Amyloid beta-Peptides/metabolism , Galantamine/metabolism , Peptide Fragments/metabolism , Amyloid beta-Peptides/chemistry , Binding Sites , Dimerization , Galantamine/chemistry , Molecular Docking Simulation , Peptide Fragments/chemistry , Protein Binding , Protein Structure, Tertiary , Thermodynamics
17.
Bioorg Med Chem Lett ; 23(15): 4336-41, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23791077

ABSTRACT

The commonly used beverage and psychostimulant caffeine is known to inhibit human acetylcholinesterase enzyme. This pharmacological activity of caffeine is partly responsible for its cognition enhancing properties. However, the exact mechanisms of its binding to human cholinesterases (acetyl and butyrylcholinesterase; hAChE and hBuChE) are not well known. In this study, we investigated the cholinesterase inhibition by the xanthine derivatives caffeine, pentoxifylline, and propentofylline. Among them, propentofylline was the most potent AChE inhibitor (hAChE IC50=6.40 µM). The hAChE inhibitory potency was of the order: caffeine (hAChE IC50=7.25 µM)50 µM) relative to the reference agent donepezil (hBuChE IC50=13.60 µM). Molecular modeling investigations indicate that caffeine binds primarily in the catalytic site (Ser203, Glu334 and His447) region of hAChE whereas pentoxifylline and propentofylline are able to bind to both the catalytic site and peripheral anionic site due to their increased bulk/size, thereby exhibiting superior AChE inhibition relative to caffeine. In contrast, their lack of hBuChE inhibition is due to a larger binding site and lack of key aromatic amino acids. In summary, our study has important implications in the development of novel caffeine derivatives as selective AChE inhibitors with potential application as cognitive enhancers and to treat various forms of dementia.


Subject(s)
Acetylcholinesterase/chemistry , Cholinesterase Inhibitors/chemistry , Xanthine/chemistry , Acetylcholinesterase/metabolism , Binding Sites , Butyrylcholinesterase/chemistry , Butyrylcholinesterase/metabolism , Caffeine/chemistry , Catalytic Domain , Cholinesterase Inhibitors/metabolism , Humans , Hydrogen Bonding , Molecular Docking Simulation , Pentoxifylline/chemistry , Pentoxifylline/metabolism , Protein Binding , Thermodynamics , Xanthine/metabolism , Xanthines/chemistry , Xanthines/metabolism
18.
ACS Chem Neurosci ; 14(23): 4185-4198, 2023 12 06.
Article in English | MEDLINE | ID: mdl-37972377

ABSTRACT

This study reports the unusual ability of small molecules N-phenylbenzofuran-2-carboxamide (7a) and N-phenylbenzo[b]thiophene-2-carboxamide (7b) to promote and accelerate Aß42 aggregation. In the in vitro aggregation kinetic assays, 7a was able to demonstrate rapid increases in Aß42 fibrillogenesis ranging from 1.5- to 4.7-fold when tested at 1, 5, 10, and 25 µM compared to Aß42-alone control. Similarly, compound 7b also exhibited 2.9- to 4.3-fold increases in Aß42 fibrillogenesis at the concentration range tested. Electron microscopy studies at 1, 5, 10, and 25 µM also demonstrate the ability of compounds 7a and 7b to promote and accelerate Aß42 aggregation with the formation of long, elongated fibril structures. Both 7a and 7b were not toxic to HT22 hippocampal neuronal cells and strikingly were able to prevent Aß42-induced cytotoxicity in HT22 hippocampal neuronal cells (cell viability ∼74%) compared to the Aß42-treated group (cell viability ∼20%). Fluorescence imaging studies using BioTracker 490 green, Hoeschst 33342, and the amyloid binding dye ProteoStat further demonstrate the ability of 7a and 7b to promote Aß42 fibrillogenesis and prevent Aß42-induced cytotoxicity to HT22 hippocampal neuronal cells. Computational modeling studies suggest that both 7a and 7b can interact with the Aß42 oligomer and pentamers and have the potential to modulate the self-assembly pathways. The 8-anilino-1-naphthalenesulfonic acid (ANS) dye binding assay also demonstrates the ability of 7a and 7b to expose the hydrophobic surface of Aß42 to the solvent surface that promotes self-assembly and rapid fibrillogenesis. These studies demonstrate the unique ability of small molecules 7a and 7b to alter the self-assembly and misfolding pathways of Aß42 by promoting the formation of nontoxic aggregates. These findings have direct implications in the discovery and development of novel small-molecule-based chemical and pharmacological tools to study the Aß42 aggregation mechanisms, and in the design of novel antiamyloid therapies to treat Alzheimer's disease.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Humans , Amyloid beta-Peptides/metabolism , Peptide Fragments/metabolism , Alzheimer Disease/metabolism , Neurons/metabolism
19.
Bioorg Med Chem Lett ; 22(14): 4707-12, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22704921

ABSTRACT

We investigated a group of 2-benzylpiperidin-N-benzylpyrimidin-4-amines with various electron-withdrawing or electron-donating groups (EWGs or EDGs, respectively) as multi-targeted Alzheimer's disease (AD) therapeutics. The synthesized derivatives were screened for anti-cholinesterase (AChE and BuChE), anti-Aß-aggregation (AChE- and self-induced) and anti-ß-secretase (BACE-1) activities in an effort to identify lead, multifunctional candidates as part of our multi-targeted approach to treat AD. Biological assessment revealed that the nature of the substituent on the C-4 benzylamine group (e.g., halogen vs methoxy-based) greatly affected the biological profile. In vitro screening identified N(2)-(1-benzylpiperidin-4-yl)-N(4)-(3,4-dimethoxybenzyl)pyrimidine-2,4-diamine (7h) as the lead candidate with a dual ChE (AChE IC(50)=9.9 µM; BuChE IC(50)=11.4 µM), Aß-aggregation (AChE-induced=59.3%; self-induced=17.4% at 100 µM) and BACE-1 (34% inhibition at 10 µM) inhibitory profile along with good cell viability (% neuroblastoma cell viability at 40 µM=81.0%). Molecular modeling studies indicate that a central pyrimidine-2,4-diamine ring serves as a suitable template to develop novel small molecule candidates to target multiple pathological routes in AD.


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Diamines/chemistry , Pyrimidines/chemistry , Alzheimer Disease/drug therapy , Cell Line, Tumor , Cell Survival/drug effects , Diamines/pharmacology , Humans , Models, Molecular , Molecular Structure , Retinoblastoma/pathology , Structure-Activity Relationship
20.
Bioorg Med Chem ; 20(9): 2912-22, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22475926

ABSTRACT

In this study, eighteen new isoxazolo[4,5-d]pyridazin-4(5H)-one derivatives possessing either a 1,3,4-thiadiazole or a 1,2,4-triazole-5-thione moiety were synthesized and tested for anti-inflammatory activity in vitro (COX-1/COX-2, 5-LOX) and in vivo (rat paw edema assay). Compounds 15, 16, 25, 26 and 28-30 showed dual COX-2 (IC(50)'s in the 2.1-10.9 µM range), and 5-LOX (IC(50)'s in the 6.3-63.5 µM range) inhibitory activity. When administered orally to rats, dual COX-2/5-LOX inhibitors showed higher anti-inflammatory activity in vivo (30-45% reduction of the inflammatory response) than the reference drug ibuprofen (18%). Among dual COX-2/5-LOX inhibitors, the most potent compound (28) exhibited the best anti-inflammatory profile by inhibiting both COX-2 (IC(50)=2.1 µM) and 5-LOX (IC(50)=6.3 µM) enzymes. We investigated the binding interactions of compound 28 by an enzyme-ligand molecular modeling (docking) studies, which showed favorable binding interactions in both COX-2 and 5-LOX active sites. Furthermore, the dual acting COX-2/5-LOX compound 28 exhibited a superior gastrointestinal safety profile (ulcer index=0.25) compared to the reference drug ibuprofen (UI=7.0) when administered orally at the same molar dose. These observations suggest that isoxazolo[4,5-d]pyridazin-4(5H)-one analogs represent a new scaffold to design potent, effective, and safe anti-inflammatory agents possessing dual COX-2/5-LOX inhibitory activity.


Subject(s)
Anti-Inflammatory Agents/chemical synthesis , Anti-Inflammatory Agents/pharmacology , Isoxazoles/chemistry , Pyridazines/chemistry , Animals , Anti-Inflammatory Agents/therapeutic use , Arachidonate 5-Lipoxygenase/chemistry , Arachidonate 5-Lipoxygenase/metabolism , Binding Sites , Catalytic Domain , Computer Simulation , Cyclooxygenase 2/chemistry , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/chemistry , Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase 2 Inhibitors/therapeutic use , Edema/drug therapy , Enzyme Activation/drug effects , Humans , Lipoxygenase Inhibitors/chemistry , Lipoxygenase Inhibitors/pharmacology , Lipoxygenase Inhibitors/therapeutic use , Protein Binding/drug effects , Pyridazines/pharmacology , Pyridazines/therapeutic use , Rats
SELECTION OF CITATIONS
SEARCH DETAIL