Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 113
Filter
1.
Am J Ther ; 30(5): e447-e453, 2023.
Article in English | MEDLINE | ID: mdl-37713689

ABSTRACT

BACKGROUND: Deposition of wild-type or mutant transthyretin (TTR) amyloid fibrils in the myocardium causes TTR amyloid cardiomyopathy (ATTR-CM). Targeted therapeutics for ATTR-CM include TTR stabilizers (tafamidis and diflunisal) and oligonucleotide drugs (revusiran, patisiran, and inotersen). TTR stabilizers prevent dissociation of transthyretin tetramers. Transthyretin monomers can misfold and form amyloid fibrils. TTR stabilizers thereby limit amyloid fibrils development and deposition. Oligonucleotide drugs inhibit hepatic synthesis of transthyretin, which decreases transthyretin protein levels and thus the amyloid fibril substrate. AREAS OF UNCERTAINTY: To study the safety and efficacy of targeted therapeutics in patients with ATTR-CM, we performed a pooled analysis. A random-effects model with the Mantel-Haenszel method was used to pool the data. DATA SOURCES: A literature search was performed using PubMed, Cochrane CENTRAL, and Embase databases using the search terms "cardiac amyloidosis" AND "tafamidis" OR "patisiran" OR "inotersen" OR "revusiran" OR "diflunisal." THERAPEUTIC ADVANCES: We identified 6 studies that compared targeted therapeutics with placebo. One study was stopped prematurely because of increased mortality in the targeted therapeutics arm. Pooled analysis included 1238 patients, of which 738 patients received targeted therapeutics and 500 patients received placebo. When compared with placebo, targeted therapeutics significantly reduced all-cause mortality [OR 0.39, 95% confidence interval (CI): 0.16-0.97, P = 0.04]. Only 2 studies reported the effect on cardiovascular-related hospitalizations. There was a trend toward an improvement in global longitudinal strain (mean difference -0.69, 95% CI: -1.44 to 0.05, P = 0.07). When compared with placebo, there was no increase in serious adverse events with targeted therapeutics (OR 1.06, 95% CI: 0.78-1.44, P = 0.72). CONCLUSION: Evidence from the pooled analysis revealed targeted therapeutics improve survival and are well-tolerated. These findings suggest a potential role for targeted therapeutics in the treatment of patients with ATTR-CM.


Subject(s)
Amyloid Neuropathies, Familial , Cardiomyopathies , Diflunisal , Humans , Amyloid Neuropathies, Familial/drug therapy , Prealbumin/metabolism , Prealbumin/therapeutic use , Diflunisal/pharmacology , Diflunisal/therapeutic use , Oligonucleotides/pharmacology , Oligonucleotides/therapeutic use , Cardiomyopathies/drug therapy
2.
ChemMedChem ; 18(4): e202200599, 2023 02 14.
Article in English | MEDLINE | ID: mdl-36533570

ABSTRACT

Here we report the encapsulation of an osteosarcoma stem cell (OSC) potent gallium(III)-diflunisal complex 1 into polymeric nanoparticles, and its delivery into osteosarcoma cells. At the optimum feed (20 %, 1 NP20 ), nanoparticle encapsulation of 1 enhances potency towards bulk osteosarcoma cells and OSCs (cultured in monolayer and three-dimensional systems). Strikingly, the nanoparticle formulation exhibits up to 5645-fold greater potency towards OSCs than frontline anti-osteosarcoma drugs, doxorubicin and cisplatin. The nanoparticle formulation evokes a similar mechanism of action as the payload, which bodes well for future translation. Specifically, the nanoparticle formulation induces nuclear DNA damage, cyclooxygenase-2 downregulation, and caspase-dependent apoptosis. To the best of our knowledge, this is the first study to demonstrate that polymeric nanoparticles can be used to effectively deliver an OSC-active metal complex into osteosarcoma cells.


Subject(s)
Bone Neoplasms , Diflunisal , Gallium , Nanoparticles , Osteosarcoma , Humans , Diflunisal/pharmacology , Micelles , Gallium/pharmacology , Cell Line, Tumor , Osteosarcoma/drug therapy , Polymers/pharmacology , Neoplastic Stem Cells
3.
Int J Nanomedicine ; 16: 1457-1472, 2021.
Article in English | MEDLINE | ID: mdl-33654396

ABSTRACT

PURPOSE: Rheumatoid arthritis is an autoimmune disorder that directly affects joints. However, other body organs including heart, eyes, skin, blood vessels and lungs may also be affected. The purpose of this study was to design and evaluate a nanoemulgel formulation of diflunisal (DIF) and solubility enhanced diflunisal (DIF-IC) for enhanced topical anti-inflammatory activity. METHODOLOGY: Nanoemulsion formulations of both DIF and DIF-IC were prepared and incorporated in three different gelling agents, namely carboxymethylcellulose sodium (CMC-Na), sodium alginate (Na-ALG) and xanthan gum (XG). All the formulations were evaluated in term of particle size, pH, conductivity, viscosity, zeta potential and in vitro drug release. The formulation 2 (NE2) of both DIF and DIF-IC which expressed optimum release and satisfactory physicochemical properties was incorporated with gelling agents to produce final nanoemulgel formulations. The optimized nanoemulgel formulation was subjected to three different in vivo anti-inflammatory models including carrageenan-induced paw edema model, histamine-induced paw edema model and formalin-induced paw edema model. RESULTS: DIF-IC-loaded nanoemulgel formulations yielded significantly enhanced in vitro skin permeation than DIF-loaded nanoemulgel. The nanoemulgel formulation of DIF-IC formulated with XG produced improved in vivo anti-inflammatory activity. CONCLUSION: It was recommended that DIF-IC-based nanoemulgel formulation prepared with XG could be a better option for effective topical treatment of inflammatory conditions.


Subject(s)
Diflunisal/administration & dosage , Drug Delivery Systems , Emulsions/chemistry , Nanogels/chemistry , Polyethylene Glycols/chemistry , Polyethyleneimine/chemistry , Administration, Topical , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Carrageenan , Diflunisal/chemistry , Diflunisal/pharmacology , Diflunisal/therapeutic use , Disease Models, Animal , Drug Compounding , Drug Liberation , Edema/drug therapy , Edema/pathology , Electric Conductivity , Hydrogen-Ion Concentration , Male , Particle Size , Permeability , Phase Transition , Rats , Skin/drug effects , Skin Absorption/drug effects , Solubility , Surface-Active Agents/chemistry , Viscosity
4.
J Med Chem ; 64(1): 797-811, 2021 01 14.
Article in English | MEDLINE | ID: mdl-33369426

ABSTRACT

In the kynurenine pathway for tryptophan degradation, an unstable metabolic intermediate, α-amino-ß-carboxymuconate-ε-semialdehyde (ACMS), can nonenzymatically cyclize to form quinolinic acid, the precursor for de novo biosynthesis of nicotinamide adenine dinucleotide (NAD+). In a competing reaction, ACMS is decarboxylated by ACMS decarboxylase (ACMSD) for further metabolism and energy production. Therefore, the inhibition of ACMSD increases NAD+ levels. In this study, an Food and Drug Administration (FDA)-approved drug, diflunisal, was found to competitively inhibit ACMSD. The complex structure of ACMSD with diflunisal revealed a previously unknown ligand-binding mode and was consistent with the results of inhibition assays, as well as a structure-activity relationship (SAR) study. Moreover, two synthesized diflunisal derivatives showed half-maximal inhibitory concentration (IC50) values 1 order of magnitude better than diflunisal at 1.32 ± 0.07 µM (22) and 3.10 ± 0.11 µM (20), respectively. The results suggest that diflunisal derivatives have the potential to modulate NAD+ levels. The ligand-binding mode revealed here provides a new direction for developing inhibitors of ACMSD.


Subject(s)
Carboxy-Lyases/metabolism , Diflunisal/metabolism , Enzyme Inhibitors/metabolism , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/metabolism , Binding Sites , Biosynthetic Pathways/drug effects , Carboxy-Lyases/antagonists & inhibitors , Catalytic Domain , Crystallography, X-Ray , Diflunisal/analogs & derivatives , Diflunisal/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Inhibitory Concentration 50 , Kynurenine/metabolism , Molecular Docking Simulation , NAD/metabolism , Pseudomonas fluorescens/enzymology , Structure-Activity Relationship , Tryptophan/metabolism
5.
Amyloid ; 28(1): 24-29, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32811187

ABSTRACT

Transthyretin (TTR) tetramer dissociation is rate limiting for aggregation and subunit exchange. Slowing of TTR tetramer dissociation via kinetic stabiliser binding slows cardiomyopathy progression. Quadruplicate subunit exchange comparisons of the drug candidate AG10, and the drugs tolcapone, diflunisal, and tafamidis were carried out at 1, 5, 10, 20 and 30 µM concentrations in 4 distinct pooled wild type TTR (TTRwt) human plasma samples. These experiments reveal that the concentration dependence of the efficacy of each compound at inhibiting TTR dissociation was primarily determined by the ratio between the stabiliser's dissociation constants from TTR and albumin, which competes with TTR to bind kinetic stabilisers. The best stabilisers, tafamidis (80 mg QD), AG10 (800 mg BID), and tolcapone (3 x 100 mg over 12 h), exhibit very similar kinetic stabilisation at the plasma concentrations resulting from these doses. At a 10 µM plasma concentration, AG10 is slightly more potent as a kinetic stabiliser vs. tolcapone and tafamidis (which are similar), which are substantially more potent than diflunisal. Dissociation of TTR can be limited to 10% of its normal rate at concentrations of 5.7 µM AG10, 10.3 µM tolcapone, 12.0 µM tafamidis, and 188 µM diflunisal. The potency similarities revealed by our study suggest that differences in safety, adsorption and metabolism, pharmacokinetics, and tissue distribution become important for kinetic stabiliser clinical use decisions.


Subject(s)
Amyloid Neuropathies, Familial/drug therapy , Amyloid/genetics , Cardiomyopathies/drug therapy , Prealbumin/genetics , Amyloid/antagonists & inhibitors , Amyloid/blood , Amyloid/chemistry , Amyloid Neuropathies, Familial/blood , Amyloid Neuropathies, Familial/genetics , Amyloid Neuropathies, Familial/pathology , Benzoates/pharmacology , Benzoxazoles/pharmacology , Cardiomyopathies/blood , Cardiomyopathies/genetics , Cardiomyopathies/pathology , Diflunisal/pharmacology , Humans , Kinetics , Prealbumin/chemistry , Protein Aggregates/drug effects , Protein Binding/drug effects , Protein Multimerization/drug effects , Protein Subunits/antagonists & inhibitors , Protein Subunits/blood , Protein Subunits/chemistry , Protein Subunits/genetics , Pyrazoles/pharmacology , Tolcapone/pharmacology
6.
Bioorg Med Chem ; 28(23): 115794, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33091848

ABSTRACT

In the past few years, attempts have been made to use decision criteria beyond Lipinski's guidelines (Rule of five) to guide drug discovery projects more effectively. Several variables and formulations have been proposed and investigated within the framework of multiparameter optimization methods to guide drug discovery. In this context, the combination of Ligand Efficiency Indices (LEI) has been predominantly used to map and monitor the drug discovery process in a retrospective fashion. Here we provide an example of the use of a novel application of the LEI methodology for prospective lead optimization by using the transthyretin (TTR) fibrillogenesis inhibitor iododiflunisal (IDIF) as example. Using this approach, a number of compounds with theoretical efficiencies higher than the reference compound IDIF were identified. From this group, ten compounds were selected, synthesized and biologically tested. Half of the compounds (5, 6, 7, 8 and 10) showed potencies in terms of IC50 inhibition of TTR aggregation equal or higher than the lead compound. These optimized compounds mapped within the region of more efficient candidates in the corresponding experimental nBEI-NSEI plot, matching their position in the theoretical optimization plane that was used for the prediction. Due to their upstream (North-Eastern) position in the progression lines of NPOL = 3 or 4 of the nBEI-NSEI plot, three of them (5, 6 and 8) are more interesting candidates than iododiflunisal because they have been optimized in the three crucial LEI variables of potency, size and polarity at the same time. This is the first example of the effectiveness of using the combined LEIs within the decision process to validate the application of the LEI formulation for the prospective optimization of lead compounds.


Subject(s)
Ligands , Prealbumin/metabolism , Diflunisal/analogs & derivatives , Diflunisal/pharmacology , Humans , Kinetics , Mutagenesis, Site-Directed , Prealbumin/antagonists & inhibitors , Prealbumin/genetics , Protein Binding , Protein Multimerization/drug effects , Structure-Activity Relationship
7.
J Vis Exp ; (155)2020 01 14.
Article in English | MEDLINE | ID: mdl-32009635

ABSTRACT

Chitosan-poly(vinyl alcohol) hydrogels can be produced by the freeze-thawing method without using toxic crosslinking agents. The applications of these systems are limited by their characteristics (e.g., porosity, flexibility, swelling capacity, drug loading and drug release capacity), which depend on the freezing conditions and the kind and ratio of polymers. This protocol describes how to prepare hydrogels from chitosan and poly(vinyl alcohol) at 50/50 w/w % of polymer composition and varying the freezing temperature (-4 °C, -20 °C, -80 °C) and freeze-thawing cycles (4, 5, 6 freezing cycles). FT-IR spectra, SEM micrograph and porosimetry data of hydrogels were obtained. Also, the swelling capacity and drug loading and release of diflunisal were assessed. Results from SEM micrographs and porosimetry show that the pore size decreases, while the porosity increases at lower temperatures. The swelling percentage was higher at the minor freezing temperature. The release of diflunisal from the hydrogels has been studied. All the networks maintain the drug release for 30 h and it has been observed that a simple diffusion mechanism regulates the diflunisal release according to Korsmeyer-Peppas and Higuchi models.


Subject(s)
Chitosan/chemistry , Hydrogels/chemistry , Polyvinyl Alcohol/chemistry , Diflunisal/pharmacology , Freezing
8.
EMBO Rep ; 20(10): e47788, 2019 10 04.
Article in English | MEDLINE | ID: mdl-31418171

ABSTRACT

Extracellular HMGB1 triggers inflammation following infection or injury and supports tumorigenesis in inflammation-related malignancies. HMGB1 has several redox states: reduced HMGB1 recruits inflammatory cells to injured tissues forming a heterocomplex with CXCL12 and signaling via its receptor CXCR4; disulfide-containing HMGB1 binds to TLR4 and promotes inflammatory responses. Here we show that diflunisal, an aspirin-like nonsteroidal anti-inflammatory drug (NSAID) that has been in clinical use for decades, specifically inhibits in vitro and in vivo the chemotactic activity of HMGB1 at nanomolar concentrations, at least in part by binding directly to both HMGB1 and CXCL12 and disrupting their heterocomplex. Importantly, diflunisal does not inhibit TLR4-dependent responses. Our findings clarify the mode of action of diflunisal and open the way to the rational design of functionally specific anti-inflammatory drugs.


Subject(s)
Chemokine CXCL12/metabolism , Diflunisal/pharmacology , HMGB1 Protein/metabolism , Leukocytes/metabolism , 3T3 Cells , Animals , Chemotaxis/drug effects , Diflunisal/chemistry , Disulfides/metabolism , Glycyrrhizic Acid/pharmacology , Humans , Inflammation/pathology , Leukocytes/drug effects , Macrophages/drug effects , Macrophages/metabolism , Magnetic Resonance Spectroscopy , Mice
9.
Biol Pharm Bull ; 41(7): 979-984, 2018.
Article in English | MEDLINE | ID: mdl-29962408

ABSTRACT

Hereditary transthyretin (TTR)-related amyloidosis is caused by mutations in the TTR gene. The mutations destabilize the tetramer and/or monomer of TTR, and thus the stabilization of TTR is a key strategy for the treatment of TTR-related amyloidosis. In this review, we summarized the natural products and synthetic compounds that have been shown to inhibit the amyloidogenesis of TTR. The stabilizers and/or the amyloid fibril disrupters isolated from natural sources may become lead compounds for the treatment of TTR-related amyloidosis.


Subject(s)
Amyloid/antagonists & inhibitors , Amyloidosis/drug therapy , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Biological Products/pharmacology , Prealbumin/metabolism , Amyloid/metabolism , Amyloidosis/genetics , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Benzoxazoles/pharmacology , Benzoxazoles/therapeutic use , Biological Products/therapeutic use , Diflunisal/pharmacology , Diflunisal/therapeutic use , Humans , Mutation , Prealbumin/genetics
10.
J Med Chem ; 60(17): 7434-7446, 2017 09 14.
Article in English | MEDLINE | ID: mdl-28771355

ABSTRACT

Noncovalent binding of biopharmaceuticals to human serum albumin protects against enzymatic degradation and renal clearance. Herein, we investigated the effect of mono- or divalent small-molecule albumin binders for half-life extension of peptides. For proof-of-principle, the clinically relevant glucagon-like peptide 1 (GLP-1) was functionalized with diflunisal, indomethacin, or both. In vitro, all GLP-1 analogues had subnanomolar GLP-1 receptor potency. Surface plasmon resonance revealed that both small molecules were able to confer albumin affinity to GLP-1 and indicated that affinity is increased for divalent analogues. In lean mice, the divalent GLP-1 analogues were superior to monovalent analogues with respect to control of glucose homeostasis and suppression of food intake. Importantly, divalent GLP-1 analogues showed efficacy comparable to liraglutide, an antidiabetic GLP-1 analogue that carries a long-chain fatty acid. Finally, pharmacokinetic investigations of a divalent GLP-1 analogue demonstrated a promising gain in circulatory half-life and absorption time compared to its monovalent equivalent.


Subject(s)
Albumins/metabolism , Diflunisal/analogs & derivatives , Drug Design , Glucagon-Like Peptide 1/analogs & derivatives , Hypoglycemic Agents/chemistry , Indomethacin/analogs & derivatives , Animals , Blood Glucose/analysis , Blood Glucose/metabolism , Diflunisal/metabolism , Diflunisal/pharmacokinetics , Diflunisal/pharmacology , Eating/drug effects , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide 1/pharmacokinetics , Glucagon-Like Peptide 1/pharmacology , Glucagon-Like Peptide-1 Receptor/metabolism , Half-Life , Hypoglycemic Agents/metabolism , Hypoglycemic Agents/pharmacokinetics , Hypoglycemic Agents/pharmacology , Indomethacin/metabolism , Indomethacin/pharmacokinetics , Indomethacin/pharmacology , Mice, Inbred C57BL
11.
PLoS One ; 12(8): e0183046, 2017.
Article in English | MEDLINE | ID: mdl-28817613

ABSTRACT

The motor protein prestin is a member of the SLC26 family of anion antiporters and is essential to the electromotility of cochlear outer hair cells and for hearing. The only direct inhibitor of electromotility and the associated charge transfer is salicylate, possibly through direct interaction with an anion-binding site on prestin. In a screen to identify other inhibitors of prestin activity, we explored the effect of the non-steroid anti-inflammatory drug diflunisal, which is a derivative of salicylate. We recorded prestin activity by whole-cell patch clamping HEK cells transiently expressing prestin and mouse outer hair cells. We monitored the impact of diflunisal on the prestin-dependent non-linear capacitance and electromotility. We found that diflunisal triggers two prestin-associated effects: a chloride independent increase in the surface area and the specific capacitance of the membrane, and a chloride dependent inhibition of the charge transfer and the electromotility in outer hair cells. We conclude that diflunisal affects the cell membrane organization and inhibits prestin-associated charge transfer and electromotility at physiological chloride concentrations. The inhibitory effects on hair cell function are noteworthy given the proposed use of diflunisal to treat neurodegenerative diseases.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Chlorides/metabolism , Diflunisal/pharmacology , Molecular Motor Proteins/antagonists & inhibitors , Animals , Cell Membrane/metabolism , Cell Membrane/physiology , Cells, Cultured , HEK293 Cells , Hair Cells, Auditory, Outer/drug effects , Hair Cells, Auditory, Outer/metabolism , Hair Cells, Auditory, Outer/physiology , Humans , Membrane Potentials , Mice , Mice, Inbred C57BL , Molecular Motor Proteins/metabolism
12.
Mycopathologia ; 182(11-12): 1025-1036, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28795317

ABSTRACT

Indoor mold due to water damage causes serious human respiratory disorders, and the remediation to homes, schools, and businesses is a major expense. Prevention of mold infestation of building materials would reduce health problems and building remediation costs. Nonsteroidal anti-inflammatory drugs (NSAIDs) inhibit yeasts and a limited number of filamentous fungi. The purpose of this research was to determine the possible inhibitory activity of nonsteroidal anti-inflammatory drugs (NSAIDs) on germination, fungal growth, and reproduction of Chaetomium globosum and other important filamentous fungi that occur in water-damaged buildings. Several NSAIDs were found to inhibit C. globosum germination, growth, and reproduction. The most effective NSAIDs inhibiting C. globosum were ibuprofen, diflunisal, and diclofenac. Fusarium oxysporum, Fusarium solani, Aspergillus niger, and Stachybotrys atra were also tested on the various media with similar results obtained. However, F. oxysporum and A. niger exhibited a higher level of resistance to aspirin and NaSAL when compared to the C. globosum isolates. The inhibition exhibited by NSAIDs was variable depending on growth media and stage of fungal development. These compounds have a great potential of inhibiting fungal growth on building materials such as gypsum board. Formulations of sprays or building materials with NSAID-like chemical treatments may hold promise in reducing mold in homes and buildings.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antifungal Agents/pharmacology , Cell Proliferation/drug effects , Chaetomium/growth & development , Germination/drug effects , Acetaminophen/pharmacology , Aspergillus/drug effects , Aspergillus/growth & development , Aspirin/pharmacology , Chaetomium/drug effects , Diclofenac/pharmacology , Diflunisal/pharmacology , Fusarium/drug effects , Fusarium/growth & development , Humans , Ibuprofen/pharmacology , Lung Diseases, Fungal/prevention & control , Microbial Sensitivity Tests , Mycoses/prevention & control , Stachybotrys/drug effects , Stachybotrys/growth & development
13.
FEBS Lett ; 591(15): 2311-2322, 2017 08.
Article in English | MEDLINE | ID: mdl-28656718

ABSTRACT

The ß-clamp is the processivity-promoting factor for most of the enzymes in prokaryotic DNA replication; hence, it is a crucial drug target. In the present study, we investigated the ß-clamp from Helicobacter pylori, aiming to seek potential drug molecules against this gastric-cancer-causing bacterium. An in silico screening of Food and Drug Administration (FDA) approved drugs against the H. pylori ß-clamp, followed by its in vitro inhibition using a surface competition approach, yielded the drug diflunisal as a positive initial hit. Diflunisal inhibits the growth of H. pylori in the micromolar range. We determined the structure of diflunisal in complex with the ß-clamp to show that the drug binds at subsite I, which is a protein-protein interaction site. Successful identification of FDA-approved molecules against H. pylori may lead to better and faster drug development.


Subject(s)
Anti-Bacterial Agents/pharmacology , DNA Polymerase III/antagonists & inhibitors , DNA Polymerase III/chemistry , Diflunisal/pharmacology , Helicobacter pylori/drug effects , Anti-Bacterial Agents/chemistry , Binding Sites , Crystallography, X-Ray , DNA Ligases/metabolism , DNA Polymerase III/metabolism , Diflunisal/chemistry , Drug Approval , Drug Evaluation, Preclinical/methods , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Helicobacter pylori/enzymology , Inhibitory Concentration 50 , Molecular Docking Simulation , Protein Conformation , United States , United States Food and Drug Administration
14.
CNS Neurosci Ther ; 23(7): 605-619, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28570028

ABSTRACT

BACKGROUND: The absence of transthyretin (TTR) in AD mice decreases brain Aß clearance and reduces the low-density lipoprotein receptor-related protein 1 (LRP1). It is possible that neuroprotection by TTR is dependent on its tetramer structural stability, as studies using TTR mutants showed that unstable L55P TTR has low affinity for Aß, and TTR tetrameric stabilizers such as iododiflunisal ameliorate AD features in vivo. METHODS: We firstly investigated TTR folding status in human plasma measuring the resistance to urea denaturation. The importance of TTR stability on Aß internalization was studied in human cerebral microvascular endothelial (hCMEC/D3) and hepatoma cells (HepG2), by flow cytometry. To investigate the fate of Aß at the blood-brain barrier, Aß efflux from hCMEC/D3 cells seeded on transwells was measured using ELISA. Further, to assess Aß colocalization with lysosomes, Lysotracker was used. Moreover, levels of LRP1 were assessed in the liver and plasma of mice with different TTR backgrounds or treated with iododiflunisal. RESULTS: We showed that TTR stability is decreased in AD and that WT TTR and drug-stabilized L55P TTR are able to increase uptake of Aß. Furthermore, measurement of Aß efflux showed that stable or stabilized TTR increased Aß efflux from the basolateral to the apical side. Moreover, HepG2 cells incubated with Aß in the presence of WT TTR, but not L55P TTR, showed an increased number of lysosomes. Further, in the presence of WT TTR, Aß peptide colocalized with lysosomes, indicating that only stable TTR assists Aß internalization, leading to its degradation. Finally, we demonstrated that only stable TTR can increase LRP1 levels. CONCLUSION: TTR stabilization exerts a positive effect on Aß clearance and LRP1 levels, suggesting that TTR protective role in AD is dependent on its stability. These results provide relevant information for the design of TTR-based therapeutic strategies for AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Prealbumin/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Blood-Brain Barrier/metabolism , Cell Line , Diflunisal/analogs & derivatives , Diflunisal/pharmacology , Escherichia coli , Humans , Liver/drug effects , Liver/metabolism , Low Density Lipoprotein Receptor-Related Protein-1 , Lysosomes/metabolism , Mice, Transgenic , Prealbumin/chemistry , Presenilin-1/genetics , Presenilin-1/metabolism , Protein Denaturation , Protein Multimerization , Protein Stability , Receptors, LDL/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Urea/metabolism
15.
J Med Chem ; 60(13): 5749-5758, 2017 07 13.
Article in English | MEDLINE | ID: mdl-28587455

ABSTRACT

Several strategies against Alzheimer disease (AD) are directed to target Aß-peptides. The ability of transthyretin (TTR) to bind Aß-peptides and the positive effect exerted by some TTR stabilizers for modulating the TTR-Aß interaction have been previously studied. Herein, key structural features of the interaction between TTR and the Aß(12-28) peptide (3), the essential recognition element of Aß, have been unravelled by STD-NMR spectroscopy methods in solution. Molecular aspects related to the role of the TTR stabilizer iododiflunisal (IDIF, 5) on the TTR-Aß complex have been also examined. The NMR results, assisted by molecular modeling protocols, have provided a structural model for the TTR-Aß interaction, as well as for the ternary complex formed in the presence of IDIF. This basic structural information could be relevant for providing light on the mechanisms involved in the ameliorating effects of AD symptoms observed in AD/TTR± animal models after IDIF treatment and eventually for designing new molecules toward AD therapeutic drugs.


Subject(s)
Amyloid beta-Peptides/metabolism , Diflunisal/analogs & derivatives , Prealbumin/metabolism , Protein Interaction Maps/drug effects , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/chemistry , Crystallography, X-Ray , Diflunisal/chemistry , Diflunisal/pharmacology , Humans , Magnetic Resonance Spectroscopy , Molecular Docking Simulation , Prealbumin/chemistry
16.
J Microencapsul ; 33(5): 475-86, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27499126

ABSTRACT

Diflunisal (DIF) is non-steroidal anti-inflammatory drug used in the treatment of rheumatoid arthritis, osteoarthritis. The current engrossment was aimed at formulation and assessment of DIF-loaded solid lipid nanoparticles (SLNs) for topical/dermal application. SLNs formulated by hot homogenisation method based on microemulsification technique were spherical with a mean size of 124.0 ± 2.07 nm; PDI 0.294 ± 0.15. The cumulative amount permeated/area was 109.99 ± 0.008 µg/cm(2), along with permeation flux (6.30 ± 0.09 µg/cm(2)/h) and skin retention (11.74 ± 0.155 µg/cm(2)) across mice skin. The SLNs of DIF showed significant decrease in fluid volume, granuloma tissue weight, leukocyte count/mm(3) after application of SLN formulation in mice air pouch model. Similarly, in mice ear oedema and rat paw oedema model, there was 2.30 and 1.29 time increase in percentage inhibition of oedema after SLN formulation application, respectively, as compared with conventional cream. Hence, the SLNs of DIF may prove to be a potential nanocarrier to effectively treat the local inflammatory conditions associated with arthritis.


Subject(s)
Arthritis, Experimental/drug therapy , Diflunisal , Drug Carriers , Nanoparticles/chemistry , Skin Absorption , Animals , Diflunisal/chemistry , Diflunisal/pharmacology , Drug Carriers/chemistry , Drug Carriers/pharmacology , Female , Mice , Rats
17.
Antimicrob Agents Chemother ; 60(9): 5322-30, 2016 09.
Article in English | MEDLINE | ID: mdl-27324764

ABSTRACT

Staphylococcus aureus osteomyelitis is a common and debilitating invasive infection of bone. Treatment of osteomyelitis is confounded by widespread antimicrobial resistance and the propensity of bacteria to trigger pathological changes in bone remodeling that limit antimicrobial penetration to the infectious focus. Adjunctive therapies that limit pathogen-induced bone destruction could therefore limit morbidity and enhance traditional antimicrobial therapies. In this study, we evaluate the efficacy of the U.S. Food and Drug Administration-approved, nonsteroidal anti-inflammatory (NSAID) compound diflunisal in limiting S. aureus cytotoxicity toward skeletal cells and in preventing bone destruction during staphylococcal osteomyelitis. Diflunisal is known to inhibit S. aureus virulence factor production by the accessory gene regulator (agr) locus, and we have previously demonstrated that the Agr system plays a substantial role in pathological bone remodeling during staphylococcal osteomyelitis. Consistent with these observations, we find that diflunisal potently inhibits osteoblast cytotoxicity caused by S. aureus secreted toxins independently of effects on bacterial growth. Compared to commonly used NSAIDs, diflunisal is uniquely potent in the inhibition of skeletal cell death in vitro Moreover, local delivery of diflunisal by means of a drug-eluting, bioresorbable foam significantly limits bone destruction during S. aureus osteomyelitis in vivo Collectively, these data demonstrate that diflunisal potently inhibits skeletal cell death and bone destruction associated with S. aureus infection and may therefore be a useful adjunctive therapy for osteomyelitis.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bone Density Conservation Agents/pharmacology , Delayed-Action Preparations/pharmacology , Diflunisal/pharmacology , Drug Repositioning , Osteomyelitis/drug therapy , Staphylococcal Infections/drug therapy , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Survival/drug effects , Female , Gene Expression , Humans , Mice , Mice, Inbred C57BL , Osteoblasts/cytology , Osteoblasts/drug effects , Osteomyelitis/microbiology , Osteomyelitis/pathology , Primary Cell Culture , Staphylococcal Infections/microbiology , Staphylococcal Infections/pathology , Staphylococcus aureus/drug effects , Staphylococcus aureus/genetics , Staphylococcus aureus/growth & development , Staphylococcus aureus/pathogenicity , Trans-Activators/antagonists & inhibitors , Trans-Activators/genetics , Trans-Activators/metabolism , Treatment Outcome
18.
Elife ; 52016 05 31.
Article in English | MEDLINE | ID: mdl-27244239

ABSTRACT

Salicylate and acetylsalicylic acid are potent and widely used anti-inflammatory drugs. They are thought to exert their therapeutic effects through multiple mechanisms, including the inhibition of cyclo-oxygenases, modulation of NF-κB activity, and direct activation of AMPK. However, the full spectrum of their activities is incompletely understood. Here we show that salicylate specifically inhibits CBP and p300 lysine acetyltransferase activity in vitro by direct competition with acetyl-Coenzyme A at the catalytic site. We used a chemical structure-similarity search to identify another anti-inflammatory drug, diflunisal, that inhibits p300 more potently than salicylate. At concentrations attainable in human plasma after oral administration, both salicylate and diflunisal blocked the acetylation of lysine residues on histone and non-histone proteins in cells. Finally, we found that diflunisal suppressed the growth of p300-dependent leukemia cell lines expressing AML1-ETO fusion protein in vitro and in vivo. These results highlight a novel epigenetic regulatory mechanism of action for salicylate and derivative drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Diflunisal/pharmacology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute/drug therapy , Salicylic Acid/pharmacology , p300-CBP Transcription Factors/antagonists & inhibitors , Acetyl Coenzyme A/antagonists & inhibitors , Acetyl Coenzyme A/metabolism , Acetylation/drug effects , Animals , Antineoplastic Agents/chemistry , Binding, Competitive , Catalytic Domain , Cell Line, Tumor , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Diflunisal/chemistry , Enzyme Inhibitors/chemistry , HEK293 Cells , Humans , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Leukocytes/drug effects , Leukocytes/enzymology , Leukocytes/pathology , Mice , Mice, SCID , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Protein Binding , RUNX1 Translocation Partner 1 Protein/genetics , RUNX1 Translocation Partner 1 Protein/metabolism , Salicylic Acid/chemistry , Signal Transduction , Structure-Activity Relationship , Xenograft Model Antitumor Assays , p300-CBP Transcription Factors/genetics , p300-CBP Transcription Factors/metabolism
19.
PLoS One ; 11(4): e0153529, 2016.
Article in English | MEDLINE | ID: mdl-27093678

ABSTRACT

Amyloid formation of the human plasma protein transthyretin (TTR) is associated with several human disorders, including familial amyloidotic polyneuropathy (FAP) and senile systemic amyloidosis. Dissociation of TTR's native tetrameric assembly is the rate-limiting step in the conversion into amyloid, and this feature presents an avenue for intervention because binding of an appropriate ligand to the thyroxin hormone binding sites of TTR stabilizes the native tetrameric assembly and impairs conversion into amyloid. The desired features for an effective TTR stabilizer include high affinity for TTR, high selectivity in the presence of other proteins, no adverse side effects at the effective concentrations, and a long half-life in the body. In this study we show that the commonly used flame retardant tetrabromobisphenol A (TBBPA) efficiently stabilizes the tetrameric structure of TTR. The X-ray crystal structure shows TBBPA binding in the thyroxine binding pocket with bromines occupying two of the three halogen binding sites. Interestingly, TBBPA binds TTR with an extremely high selectivity in human plasma, and the effect is equal to the recently approved drug tafamidis and better than diflunisal, both of which have shown therapeutic effects against FAP. TBBPA consequently present an interesting scaffold for drug design. Its absorption, metabolism, and potential side-effects are discussed.


Subject(s)
Excipients/chemistry , Polybrominated Biphenyls/chemistry , Prealbumin/chemistry , Amyloid/metabolism , Amyloidosis/metabolism , Benzoxazoles/pharmacology , Binding Sites/physiology , Cell Line, Tumor , Crystallography, X-Ray/methods , Diflunisal/pharmacology , Drug Design , Half-Life , Humans , Ligands , Polybrominated Biphenyls/metabolism , Prealbumin/metabolism , Protein Binding/physiology , Thyroxine/pharmacology
20.
Anticancer Agents Med Chem ; 16(10): 1266-74, 2016.
Article in English | MEDLINE | ID: mdl-26320814

ABSTRACT

A series of diflunisal 4-thiazolidinones were synthesized. Some selected compounds were determined at one dose towards the full panel of 60 human cancer cell lines by National Cancer Institute. 2',4'-Difluoro-4-hydroxy-N-[4-oxo-2-(thiophen-2-yl)-1,3-thiazolidin-3-yl]biphenyl- 3-carboxamide (4a) demonstrated the most marked effect on K-562 cancer cell line with 58.59 % growth inhibition at 10 µM. Compound 4a was evaluated in vitro using the MTT colorimetric method against human leukemia cell line K-562 and mouse embryonic fibroblasts cell line NIH- 3T3 at different doses for cell viability and growth inhibition. Compound 4a exhibited anticancer activity with IC50 value of 5.2 µM against K-562 cells and did not display cytotoxicity towards NIH-3T3 cells compared with diflunisal. In addition, this compound could be an interesting prototype as an antiproliferative agent.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Diflunisal/chemistry , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Chemistry Techniques, Synthetic , Diflunisal/pharmacology , Drug Screening Assays, Antitumor , Humans , K562 Cells/drug effects , Mice , NIH 3T3 Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...