Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Bioorg Chem ; 147: 107393, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38691908

RESUMO

Cyclooxygenase-2 plays a vital role in inflammation by catalyzing arachidonic acid conversion toward prostaglandins, making it a prime therapeutic objective. Selective COX-2 inhibitors represent significant progress in anti-inflammatory therapy, offering improved efficacy and fewer side effects. This study describes the synthesis of novel anti-inflammatory compounds from established pharmaceutically marketed agents like fenamates III-V and ibuprofen VI. Through rigorous in vitro testing, compounds 7b-c, and 12a-b demonstrated substantial in vitro selective inhibition, with IC50 values of 0.07 to 0.09 µM, indicating potent pharmacological activity. In vivo assessment, particularly focusing on compound 7c, revealed significant anti-inflammatory effects. Markedly, it demonstrated the highest inhibition of paw thickness (58.62 %) at the 5-hr mark compared to the carrageenan group, indicating its potency in mitigating inflammation. Furthermore, it exhibited a rapid onset of action, with a 54.88 % inhibition observed at the 1-hr mark. Subsequent comprehensive evaluations encompassing analgesic efficacy, histological characteristics, and toxicological properties indicated that compound 7c did not induce gastric ulcers, in contrast to the ulcerogenic tendency associated with mefenamic acid. Moreover, compound 7c underwent additional investigations through in silico methodologies such as molecular modelling, field alignment, and density functional theory. These analyses underscored the therapeutic potential and safety profile of this novel compound, warranting further exploration and development in the realm of pharmaceutical research.


Assuntos
Anti-Inflamatórios não Esteroides , Carragenina , Inibidores de Ciclo-Oxigenase 2 , Ciclo-Oxigenase 2 , Fenamatos , Ibuprofeno , Ibuprofeno/farmacologia , Ibuprofeno/química , Ibuprofeno/síntese química , Ciclo-Oxigenase 2/metabolismo , Animais , Inibidores de Ciclo-Oxigenase 2/farmacologia , Inibidores de Ciclo-Oxigenase 2/síntese química , Inibidores de Ciclo-Oxigenase 2/química , Estrutura Molecular , Anti-Inflamatórios não Esteroides/farmacologia , Anti-Inflamatórios não Esteroides/química , Anti-Inflamatórios não Esteroides/síntese química , Relação Estrutura-Atividade , Fenamatos/farmacologia , Fenamatos/química , Fenamatos/síntese química , Relação Dose-Resposta a Droga , Humanos , Camundongos , Edema/tratamento farmacológico , Edema/induzido quimicamente , Simulação de Acoplamento Molecular , Ratos , Masculino
2.
Cell Mol Life Sci ; 79(1): 65, 2022 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-35013790

RESUMO

Coronavirus disease 2019 (COVID-19), the illness caused by a novel coronavirus now called severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has led to more than 260 million confirmed infections and 5 million deaths to date. While vaccination is a powerful tool to control pandemic spread, medication to relieve COVID-19-associated symptoms and alleviate disease progression especially in high-risk patients is still lacking. In this study, we explore the suitability of the rapid accelerated fibrosarcoma/mitogen-activated protein kinase/extracellular signal-regulated kinase (Raf/MEK/ERK) pathway as a druggable target in the treatment of SARS-CoV-2 infections. We find that SARS-CoV-2 transiently activates Raf/MEK/ERK signaling in the very early infection phase and that ERK1/2 knockdown limits virus replication in cell culture models. We demonstrate that ATR-002, a specific inhibitor of the upstream MEK1/2 kinases which is currently evaluated in clinical trials as an anti-influenza drug, displays strong anti-SARS-CoV-2 activity in cell lines as well as in primary air-liquid-interphase epithelial cell (ALI) cultures, with a safe and selective treatment window. We also observe that ATR-002 treatment impairs the SARS-CoV-2-induced expression of pro-inflammatory cytokines, and thus might prevent COVID-19-associated hyperinflammation, a key player in COVID-19 progression. Thus, our data suggest that the Raf/MEK/ERK signaling cascade may represent a target for therapeutic intervention strategies against SARS-CoV-2 infections and that ATR-002 is a promising candidate for further drug evaluation.


Assuntos
Antivirais/farmacologia , Tratamento Farmacológico da COVID-19 , Fenamatos/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , SARS-CoV-2/efeitos dos fármacos , Células A549 , Adulto , Animais , COVID-19/metabolismo , Linhagem Celular , Células Cultivadas , Chlorocebus aethiops , Citocinas/metabolismo , Humanos , Inflamação/tratamento farmacológico , Inflamação/metabolismo , MAP Quinase Quinase 1/antagonistas & inibidores , MAP Quinase Quinase 1/metabolismo , MAP Quinase Quinase 2/antagonistas & inibidores , MAP Quinase Quinase 2/metabolismo , SARS-CoV-2/fisiologia , Células Vero , Replicação Viral/efeitos dos fármacos
3.
Chem Biodivers ; 20(8): e202300773, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37384873

RESUMO

In this study, twenty new anthranilic acid hydrazones 6-9 (a-e) were synthesized and their structures were characterized by Fourier-transform Infrared (FT-IR), Nuclear Magnetic Resonance (1 H-NMR - 13 C-NMR), and High-resolution Mass Spectroscopy (HR-MS). The inhibitory effects of the compounds against COX-II were evaluated. IC50 values of the compounds were found in the range of >200-0.32 µM and compounds 6e, 8d, 8e, 9b, 9c, and 9e were determined to be the most effective inhibitors. Cytotoxic effects of the most potent compounds were investigated against human hepatoblastoma (Hep-G2) and human healthy embryonic kidney (Hek-293) cell lines. Doxorubicin (IC50 : 8.68±0.16 µM for Hep-G2, 55.29±0.56 µM for Hek-293) was used as standard. 8e is the most active compound, with low IC50 against Hep-G2 (4.80±0.04 µM), high against Hek-293 (159.30±3.12), and high selectivity (33.15). Finally, molecular docking and dynamics studies were performed to understand ligand-protein interactions between the most potent compounds and COX II, Epidermal Growth Factor Receptor (EGFR), and Transforming Growth Factor beta II (TGF-ßII). The docking scores were calculated in the range of -10.609--6.705 kcal/mol for COX-II, -8.652--7.743 kcal/mol for EGFR, and -10.708--8.596 kcal/mol for TGF-ßII.


Assuntos
Antineoplásicos , Fenamatos , Humanos , Simulação de Acoplamento Molecular , Hidrazonas/farmacologia , Hidrazonas/química , Estrutura Molecular , Relação Estrutura-Atividade , Fenamatos/farmacologia , Espectroscopia de Infravermelho com Transformada de Fourier , Células HEK293 , Linhagem Celular Tumoral , Antineoplásicos/farmacologia , Antineoplásicos/química , Receptores ErbB , Anti-Inflamatórios/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais , Proliferação de Células
4.
Org Biomol Chem ; 20(25): 5076-5085, 2022 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-35697330

RESUMO

Human serum albumin (HSA) can bind with numerous drugs, leading to a significant influence on drug pharmacokinetics as well as undesirable drug-drug interactions due to competitive binding. Probing the HSA drug binding site thus offers great opportunities to reveal drug-HSA binding profiles. In the present study, a fluorescent probe (E)-4-(2-(5-(4-(diphenylamino)phenyl)thiophen-2-yl)vinyl)-1-propylpyridin-1-ium (TTPy) has been prepared, which exhibits enhancement of deep-red to near-infrared (NIR) fluorescence upon HSA binding. The competitive binding assay indicated that TTPy can target the HSA binding site of fenamates, a group of non-steroidal anti-inflammatory drugs (NSAIDs), with moderate binding affinity (1.95 × 106 M-1 at 303 K). More interestingly, TTPy enables fluorescent labeling of HSA upon visible light irradiation. This study provides promising ways for HSA drug binding site identification and photochemical protein labeling.


Assuntos
Fenamatos , Albumina Sérica , Sítios de Ligação , Corantes Fluorescentes/química , Humanos , Processos Fotoquímicos , Ligação Proteica , Albumina Sérica/química , Albumina Sérica Humana/metabolismo , Espectrometria de Fluorescência
5.
Plant Physiol ; 180(1): 480-496, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30737267

RESUMO

Many signal perception mechanisms are connected to Ca2+-based second messenger signaling to modulate specific cellular responses. The well-characterized plant hormone auxin elicits a very rapid Ca2+ signal. However, the cellular targets of auxin-induced Ca2+ are largely unknown. Here, we screened a biologically annotated chemical library for inhibitors of auxin-induced Ca2+ entry in plant cell suspensions to better understand the molecular mechanism of auxin-induced Ca2+ and to explore the physiological relevance of Ca2+ in auxin signal transduction. Using this approach, we defined a set of diverse, small molecules that interfere with auxin-induced Ca2+ entry. Based on annotated biological activities of the hit molecules, we found that auxin-induced Ca2+ signaling is, among others, highly sensitive to disruption of membrane proton gradients and the mammalian Ca2+ channel inhibitor bepridil. Whereas protonophores nonselectively inhibited auxin-induced and osmotic stress-induced Ca2+ signals, bepridil specifically inhibited auxin-induced Ca2+ We found evidence that bepridil severely alters vacuolar morphology and antagonized auxin-induced vacuolar remodeling. Further exploration of this plant-tailored collection of inhibitors will lead to a better understanding of auxin-induced Ca2+ entry and its relevance for auxin responses.


Assuntos
Arabidopsis/efeitos dos fármacos , Sinalização do Cálcio/efeitos dos fármacos , Ácidos Indolacéticos/metabolismo , Nicotiana/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Ácido 2,4-Diclorofenoxiacético/farmacologia , Arabidopsis/genética , Proteínas de Bactérias/genética , Bepridil/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , Sinalização do Cálcio/fisiologia , Linhagem Celular , Avaliação Pré-Clínica de Medicamentos/métodos , Fenamatos/farmacologia , Ácidos Indolacéticos/antagonistas & inibidores , Medições Luminescentes , Proteínas Luminescentes/genética , Niclosamida/farmacologia , Células Vegetais/efeitos dos fármacos , Células Vegetais/metabolismo , Reguladores de Crescimento de Plantas/metabolismo , Raízes de Plantas/efeitos dos fármacos , Plantas Geneticamente Modificadas , Nicotiana/genética , Vacúolos/efeitos dos fármacos , Vacúolos/metabolismo
6.
Cell Mol Neurobiol ; 40(8): 1405-1416, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32162200

RESUMO

Voltage-gated sodium channels are crucial mediators of neuronal damage in ischemic and excitotoxicity disease models. Fenamates have been reported to have anti-inflammatory properties following a decrease in prostaglandin synthesis. Several researches showed that fenamates appear to be ion channel modulators and potential neuroprotectants. In this study, the neuroprotective effects of tolfenamic acid, flufenamic acid, and mefenamic acid were tested by glutamate-induced injury in SH-SY5Y cells. Following this, fenamates' effects were examined on both the expression level and the function of hNav1.1 and hNav1.2, which were closely associated with neuroprotection, using Western blot and patch clamp. Finally, the effect of fenamates on the expression of apoptosis-related proteins in SH-SY5Y cells was examined. The results showed that both flufenamic acid and mefenamic acid exhibited neuroprotective effects against glutamate-induced injury in SH-SY5Y cells. They inhibited peak currents of both hNav1.1 and hNav1.2. However, fenamates exhibited decreased inhibitory effects on hNav1.1 when compared to hNav1.2. Correspondingly, the inhibitory effect of fenamates was found to be consistent with the level of neuroprotective effects in vitro. Fenamates inhibited glutamate-induced apoptosis through the modulation of the Bcl-2/Bax-dependent cell death pathways. Taken together, Nav1.2 might play a part in fenamates' neuroprotection mechanism. Nav1.2 and NMDAR might take part in the neuroprotection mechanism of the fenamates. The fenamates inhibited glutamate-induced apoptosis through modulation of the Bcl-2/Bax-dependent cell death pathways.


Assuntos
Fenamatos/farmacologia , Ácido Glutâmico/farmacologia , Receptores de N-Metil-D-Aspartato/metabolismo , ortoaminobenzoatos/farmacologia , Ácido Glutâmico/metabolismo , Humanos , Fármacos Neuroprotetores , Técnicas de Patch-Clamp/métodos , Canais de Sódio Disparados por Voltagem/metabolismo
7.
Eur Biophys J ; 49(7): 591-607, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32940715

RESUMO

GABA and glycine act as inhibitory neurotransmitters in the CNS. Inhibitory neurotransmission is mediated via activation of ionotropic GABAA and glycine receptors. We used a modeling approach to explain the opposite effects of the general anesthetic etomidate (ETM) and fenamate mefenamic acid (MFA) on GABA- and glycine-activated currents recorded in isolated cerebellar Purkinje cells and hippocampal pyramidal neurons, respectively. These drugs potentiated GABAARs but blocked GlyRs. We built a homology model of α1ß GlyR based on the cryo-EM structure of open α1 GlyR, used the α1ß3γ2 GABAAR structure from the PDB, and applied Monte-Carlo energy minimization to optimize models of receptors and ligand-receptor complexes. In silico docking suggests that ETM/MFA bind at the transmembrane ß( +)/α( -) intersubunit interface in GABAAR. Our models predict that the bulky side chain of the highly conserved Arg19' residue at the plus interface side wedges the interface and maintains the conducting receptor state. We hypothesized that MFA/ETM binding at the ß( +)/α( -) interface leads to prolongation of receptor life-time in the open state. Having analyzed different GABAAR and GlyR structures available in the PDB, we found that mutual arrangement of the Arg19' and Gln-26' side chains at the plus and minus interface sides, respectively, plays an important role when the receptor switches from the open to closed state. We show that this process is accompanied by narrowing of the intersubunit interfaces, leading to extrusion of the Arg19' side chain from the interface. Our models allow us to explain the lack of GlyR potentiation in our electrophysiological experiments.


Assuntos
Etomidato/química , Ácido Mefenâmico/química , Neurônios/metabolismo , Proteínas Nucleares/química , Oxirredutases/química , Receptores de GABA-A/química , Anestésicos Gerais/farmacologia , Animais , Sítios de Ligação , Simulação por Computador , Bases de Dados de Proteínas , Eletrofisiologia , Fenamatos/química , Glicina/química , Ligantes , Conformação Molecular , Simulação de Acoplamento Molecular , Método de Monte Carlo , Ligação Proteica , Ratos , Ratos Wistar , Receptores de Glicina/química , Transmissão Sináptica
8.
Org Biomol Chem ; 17(13): 3409-3415, 2019 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-30869715

RESUMO

Rare multiple fluorescence properties including aggregation-induced emission and polymorphism/shape/size-dependent emission were found coexisting in a class of typical non-steroidal anti-inflammatory analgesic drugs, fenamates, which could provide a new approach toward future drug evaluation. Different from the complexity and biological incompatibility of the traditional AIE molecular design, this work opens new avenues to the development of new AIE systems.


Assuntos
Analgésicos/química , Anti-Inflamatórios não Esteroides/química , Fenamatos/química , Corantes Fluorescentes/química , Avaliação de Medicamentos , Fluorescência , Estrutura Molecular , Tamanho da Partícula , Teoria Quântica
9.
J Biol Chem ; 291(29): 15069-81, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27226593

RESUMO

Cyclooxygenase-2 (COX-2) catalyzes the oxygenation of arachidonic acid (AA) and endocannabinoid substrates, placing the enzyme at a unique junction between the eicosanoid and endocannabinoid signaling pathways. COX-2 is a sequence homodimer, but the enzyme displays half-of-site reactivity, such that only one monomer of the dimer is active at a given time. Certain rapid reversible, competitive nonsteroidal anti-inflammatory drugs (NSAIDs) have been shown to inhibit COX-2 in a substrate-selective manner, with the binding of inhibitor to a single monomer sufficient to inhibit the oxygenation of endocannabinoids but not arachidonic acid. The underlying mechanism responsible for substrate-selective inhibition has remained elusive. We utilized structural and biophysical methods to evaluate flufenamic acid, meclofenamic acid, mefenamic acid, and tolfenamic acid for their ability to act as substrate-selective inhibitors. Crystal structures of each drug in complex with human COX-2 revealed that the inhibitor binds within the cyclooxygenase channel in an inverted orientation, with the carboxylate group interacting with Tyr-385 and Ser-530 at the top of the channel. Tryptophan fluorescence quenching, continuous-wave electron spin resonance, and UV-visible spectroscopy demonstrate that flufenamic acid, mefenamic acid, and tolfenamic acid are substrate-selective inhibitors that bind rapidly to COX-2, quench tyrosyl radicals, and reduce higher oxidation states of the heme moiety. Substrate-selective inhibition was attenuated by the addition of the lipid peroxide 15-hydroperoxyeicosatertaenoic acid. Collectively, these studies implicate peroxide tone as an important mechanistic component of substrate-selective inhibition by flufenamic acid, mefenamic acid, and tolfenamic acid.


Assuntos
Inibidores de Ciclo-Oxigenase 2/química , Inibidores de Ciclo-Oxigenase 2/farmacologia , Ciclo-Oxigenase 2/metabolismo , Fenamatos/farmacologia , Substituição de Aminoácidos , Sítios de Ligação , Cristalografia por Raios X , Ciclo-Oxigenase 2/química , Ciclo-Oxigenase 2/genética , Endocanabinoides/metabolismo , Fenamatos/química , Heme/química , Humanos , Técnicas In Vitro , Modelos Moleculares , Mutagênese Sítio-Dirigida , Peróxidos/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidade por Substrato , Triptofano/química
10.
Pharm Res ; 32(10): 3432-46, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26248795

RESUMO

PURPOSE: To examine the expression of fatty acid binding proteins (FABPs) at the human blood-brain barrier (BBB) and to assess their ability to bind lipophilic drugs. METHODS: mRNA and protein expression of FABP subtypes in immortalized human brain endothelial (hCMEC/D3) cells were examined by RT-qPCR and Western blot, respectively. FABPs that were found in hCMEC/D3 cells (hFABPs) were recombinantly expressed and purified from Escherichia coli C41(DE3) cells. Drug binding to these hFABPs was assessed using a fluorescence assay, which measured the ability of a panel of lipophilic drugs to displace the fluorescent probe compound 1-anilinonaphthalene-8-sulfonic acid (ANS). RESULTS: hFABP3, 4 and 5 were expressed in hCMEC/D3 cells at the mRNA and protein level. The competitive ANS displacement assay demonstrated that, in general, glitazones preferentially bound to hFABP5 (Ki: 1.0-28 µM) and fibrates and fenamates preferentially bound to hFABP4 (Ki: 0.100-17 µM). In general, lipophilic drugs appeared to show weaker affinities for hFABP3 relative to hFABP4 and hFABP5. No clear correlation was observed between the molecular structure or physicochemical properties of the drugs and their ability to displace ANS from hFABP3, 4 and 5. CONCLUSIONS: hFABP3, 4 and 5 are expressed at the human BBB and bind differentially to a diverse range of lipophilic drugs. The unique expression and binding patterns of hFABPs at the BBB may therefore influence drug disposition into the brain.


Assuntos
Barreira Hematoencefálica/metabolismo , Proteínas de Ligação a Ácido Graxo/metabolismo , Preparações Farmacêuticas/metabolismo , Ligação Proteica/fisiologia , Isoformas de Proteínas/metabolismo , Naftalenossulfonato de Anilina/metabolismo , Transporte Biológico/fisiologia , Encéfalo/metabolismo , Linhagem Celular , Células Endoteliais/metabolismo , Fenamatos/metabolismo , Ácidos Fíbricos/metabolismo , Corantes Fluorescentes/metabolismo , Humanos , RNA Mensageiro/metabolismo , Tiazolidinedionas/metabolismo
11.
Bioorg Med Chem Lett ; 25(2): 179-83, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25522819

RESUMO

A series of certain novel Schiff bases as fenamate isosteres (VI:a-k) were synthesized to locate analgesic, anti-inflammatory agent with minimal ulcerogenic potential. The structures of the newly synthesized compounds were elucidated on the basis of their elemental analysis as well as IR, and NMR and mass spectroscopic data. All the compounds were evaluated for their anti-inflammatory activity by carrageenan induced paw oedema method. The compounds possessing good anti-inflammatory activity were further tested for analgesic, ulcerogenic, lipid peroxidation potentials and liver toxicity. Compounds (VI-c), (VI-f), (VI-h) and (VI-i) showed the best anti-inflammatory and significant analgesic activities at doses comparable to that of the standard drug Indomethacin. However, compounds (VI-c) and (VI-f) could be considered the most potent anti-inflammatory and analgesic molecules with maximum reduction in gastro-intestinal ulceration with no hepatocyte necrosis or liver degeneration.


Assuntos
Analgésicos/síntese química , Anti-Inflamatórios/síntese química , Antiulcerosos/síntese química , Fenamatos/síntese química , Analgésicos/uso terapêutico , Animais , Anti-Inflamatórios/uso terapêutico , Antiulcerosos/uso terapêutico , Edema/tratamento farmacológico , Edema/patologia , Fenamatos/uso terapêutico , Masculino , Camundongos , Ratos , Ratos Wistar , Bases de Schiff/síntese química , Bases de Schiff/uso terapêutico , Estereoisomerismo
12.
Phys Chem Chem Phys ; 17(12): 7936-48, 2015 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-25720865

RESUMO

In traditional molecular mechanics force fields, intramolecular non-bonded interactions are modelled as intermolecular interactions, and the form of the torsion potential is based on the conformational profiles of small organic molecules. We investigate how a separate model for the intramolecular forces in pharmaceuticals could be more realistic by analysing the low barrier to rotation of the phenyl ring in the fenamates (substituted N-phenyl-aminobenzoic acids), that results in a wide range of observed angles in the numerous fenamate crystal structures. Although the conformational energy changes by significantly less than 10 kJ mol(-1) for a complete rotation of the phenyl ring for fenamic acid, the barrier is only small because of small correlated changes in the other bond and torsion angles. The maxima for conformations where the two aromatic rings approach coplanarity arise from steric repulsion, but the maxima when the two rings are approximately perpendicular arise from a combination of an electronic effect and intramolecular dispersion. Representing the ab initio conformational energy profiles as a cosine series alone is ineffective; however, combining a cos 2ξ term to represent the electronic barrier with an intramolecular atom-atom exp-6 term for all atom pairs separated by three or more bonds (1-4 interactions) provides a very effective representation. Thus we propose a new, physically motivated, generic analytical model of conformational energy, which could be combined with an intermolecular model to form more accurate force-fields for modelling the condensed phases of pharmaceutical-like organic molecules.


Assuntos
Fenamatos/química , Preparações Farmacêuticas/química , Elétrons , Conformação Molecular , Simulação de Dinâmica Molecular , Termodinâmica
13.
Mol Pharmacol ; 85(5): 671-81, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24509840

RESUMO

TWIK-related K(+) 1 (TREK1) potassium channels are members of the two-pore domain potassium channel family and contribute to background potassium conductances in many cell types, where their activity can be regulated by a variety of physiologic and pharmacologic mediators. Fenamates such as FFA (flufenamic acid; 2-{[3-(trifluoromethyl)phenyl]amino}benzoic acid), MFA [mefenamic acid; 2-(2,3-dimethylphenyl)aminobenzoic acid], NFA [niflumic acid; 2-{[3-(trifluoromethyl)phenyl]amino}nicotinic acid], and diclofenac [2-(2-(2,6-dichlorophenylamino)phenyl)acetic acid] and the related experimental drug BL-1249 [(5,6,7,8-tetrahydro-naphthalen-1-yl)-[2-(1H-tetrazol-5-yl)-phenyl]-amine] enhance the activity of TREK1 currents, and we show that BL-1249 is the most potent of these compounds. Alternative translation initiation produces a shorter, N terminus truncated form of TREK1 with a much reduced open probability and a proposed increased permeability to sodium compared with the longer form. We show that both forms of TREK1 can be activated by fenamates and that a number of mutations that affect TREK1 channel gating occlude the action of fenamates but only in the longer form of TREK1. Furthermore, fenamates produce a marked enhancement of current through the shorter, truncated form of TREK1 and reveal a K(+)-selective channel, like the long form. These results provide insight into the mechanism of TREK1 channel activation by fenamates, and, given the role of TREK1 channels in pain, they suggest a novel analgesic mechanism for these compounds.


Assuntos
Fenamatos/farmacologia , Canais de Potássio de Domínios Poros em Tandem/agonistas , Canais de Potássio de Domínios Poros em Tandem/fisiologia , Células HEK293 , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/fisiologia , Mutação/fisiologia , Canais de Potássio de Domínios Poros em Tandem/química , Estrutura Secundária de Proteína
14.
Proc Natl Acad Sci U S A ; 107(47): 20483-8, 2010 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-21059958

RESUMO

Transthyretin (TTR) amyloidosis is a fatal disease for which new therapeutic approaches are urgently needed. We have designed two palindromic ligands, 2,2'-(4,4'-(heptane-1,7-diylbis(oxy))bis(3,5-dichloro-4,1-phenylene)) bis(azanediyl)dibenzoic acid (mds84) and 2,2'-(4,4'-(undecane-1,11-diylbis(oxy))bis(3,5-dichloro-4,1-phenylene)) bis(azanediyl)dibenzoic acid (4ajm15), that are rapidly bound by native wild-type TTR in whole serum and even more avidly by amyloidogenic TTR variants. One to one stoichiometry, demonstrable in solution and by MS, was confirmed by X-ray crystallographic analysis showing simultaneous occupation of both T4 binding sites in each tetrameric TTR molecule by the pair of ligand head groups. Ligand binding by native TTR was irreversible under physiological conditions, and it stabilized the tetrameric assembly and inhibited amyloidogenic aggregation more potently than other known ligands. These superstabilizers are orally bioavailable and exhibit low inhibitory activity against cyclooxygenase (COX). They offer a promising platform for development of drugs to treat and prevent TTR amyloidosis.


Assuntos
Amiloide/biossíntese , Amiloidose/metabolismo , Fenamatos/metabolismo , Ligantes , Pré-Albumina/metabolismo , Amiloide/metabolismo , Amiloidose/tratamento farmacológico , Animais , Varredura Diferencial de Calorimetria , Cromatografia em Gel , Cristalografia por Raios X , Fenamatos/síntese química , Fenamatos/química , Fenamatos/farmacocinética , Fluorometria , Espectrometria de Massas , Camundongos , Modelos Moleculares , Estrutura Molecular , Ultracentrifugação
15.
Ther Deliv ; 14(3): 183-192, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-37158260

RESUMO

Aim: To develop controlled-release tablets based on aminated starch. Materials & methods: Aminated starch was characterized with Fourier transform infrared and x-ray diffraction. Thermogravimetric analysis confirmed the preferential oxidation of crystalline region of starch. Results: The tablets achieved an initial fast release of fenamates, which slows down after 12 h. Drug release was not completed in the simulated intestinal media, which may be due to the stability of imine bond in aminated starch at weakly acidic pH. Drug release was completed in simulated acidic media due to the hydrolysis of imine functionality at strongly acidic pH. Conclusion: Aminated starch with an imine functionality may serve as intestine targeted, controlled drug-delivery system. Mucoadhesive potential of tablets further supports this observation.


Assuntos
Fenamatos , Amido , Amido/química , Sistemas de Liberação de Medicamentos , Liberação Controlada de Fármacos , Difração de Raios X , Comprimidos/química , Espectroscopia de Infravermelho com Transformada de Fourier
16.
Mol Pharmacol ; 82(5): 795-802, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22851714

RESUMO

Niflumic acid, 2-{[3-(trifluoromethyl)phenyl]amino}pyridine-3-carboxylic acid (NFA), a nonsteroidal anti-inflammatory drug that blocks cyclooxygenase (COX), was shown previously to activate [Na(+)](i)-regulated Slo2.1 channels. In this study, we report that other fenamates, including flufenamic acid, mefenamic acid, tolfenamic acid, meclofenamic acid, and a phenyl acetic acid derivative, diclofenac, also are low-potency (EC(50) = 80 µM to 2.1 mM), partial agonists of human Slo2.1 channels heterologously expressed in Xenopus oocytes. Substituent analysis determined that N-phenylanthranilic acid was the minimal pharmacophore for fenamate activation of Slo2.1 channels. The effects of fenamates were biphasic, with an initial rapid activation phase followed by a slow phase of current inhibition. Ibuprofen, a structurally dissimilar COX inhibitor, did not activate Slo2.1. Preincubation of oocytes with ibuprofen did not significantly alter the effects of NFA, suggesting that neither channel activation nor inhibition is associated with COX activity. A point mutation (A278R) in the pore-lining S6 segment of Slo2.1 increased the sensitivity to activation and reduced the inhibition induced by NFA. Together, our results suggest that fenamates bind to two sites on Slo2.1 channels: an extracellular accessible site to activate and a cytoplasmic accessible site in the pore to inhibit currents.


Assuntos
Fenamatos/farmacologia , Canais de Potássio/agonistas , Animais , Inibidores de Ciclo-Oxigenase/farmacologia , Feminino , Fenamatos/química , Humanos , Ibuprofeno/farmacologia , Técnicas In Vitro , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/agonistas , Ácido Niflúmico/farmacologia , Oócitos/efeitos dos fármacos , Oócitos/fisiologia , Técnicas de Patch-Clamp , Mutação Puntual , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/genética , Canais de Potássio Ativados por Sódio , Relação Estrutura-Atividade , Xenopus laevis , ortoaminobenzoatos/farmacologia
17.
J Inorg Biochem ; 228: 111696, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35030390

RESUMO

Through the reaction of copper(II) acetate with nicotinamide (pyridine-3-carboxylic acid amide, niacinamide) and some derivatives of N-phenylanthranilic acid (fenamates), seven new mixed-ligand copper(II) compounds were isolated: [Cu(tolf-O)(tolf-O,O')nia-N)2(EtOH)] (1), [Cu(tolf-O)(tolf-O,O')(nia-N)2(MeOH)] (2), [Cu(meclf-O)(meclf-O,O')(nia-N)2(EtOH)] (3), [Cu(meclf-O)(meclf-O,O')(nia-N)2(MeOH)] (4), [Cu(meclf-O)(meclf-O,O')(nia-N)2(ACN)] (5), [Cu(mef-O)(mef-O,O')(nia-N)2(EtOH)] (6) and [Cu(mef-O)(mef-O,O')(nia-N)2(ACN)] (7) containing a molecule of relevant solvent as ligand in their primary crystal structure (tolf = tolfenamate, meclf = meclofenamate, mef = mefenamate, nia = nicotinamide, EtOH = ethanol, MeOH = methanol, ACN = acetonitrile). The structures of the complexes were determined by single-crystal X-ray analysis. The intermolecular interactions were studied by Hirshfeld surface analysis. The complexes were characterized by IR, UV-vis and EPR spectroscopy and their redox properties were determined by cyclic voltammetry. The interaction of the complexes with bovine serum albumin was studied by fluorescence emission spectroscopy and the albumin-binding constants of the compounds were calculated. The interaction of the complexes with calf-thymus DNA was monitored by diverse techniques (UV-vis spectroscopy, cyclic voltammetry, viscosity measurements) suggesting intercalation as the most possible mode of binding. DNA-competitive studies of the complexes with ethidium bromide were monitored by fluorescence emission spectroscopy. The cytotoxic effects of copper(II) complexes on lung carcinoma cells and healthy cells were determined by the MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] colorimetric technique.


Assuntos
Anti-Inflamatórios não Esteroides/química , Complexos de Coordenação/química , Cobre/química , DNA/química , Niacinamida/química , Soroalbumina Bovina/química , Células A549 , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Antineoplásicos/química , Antineoplásicos/farmacologia , Cristalografia por Raios X/métodos , Espectroscopia de Ressonância de Spin Eletrônica/métodos , Etídio/química , Fenamatos/química , Humanos , Substâncias Intercalantes/química , Oxirredução
18.
Mol Pharmacol ; 80(5): 911-9, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21828194

RESUMO

The brain liver intestine Na(+) channel (BLINaC) is a member of the degenerin/epithelial Na(+) channel gene family of unknown function. Elucidation of the physiological function of BLINaC would benefit greatly from pharmacological tools that specifically affect BLINaC activity. Guided by the close molecular relation of BLINaC to acid-sensing ion channels, we discovered in this study that rat BLINaC (rBLINaC) and mouse BLINaC are inhibited by micromolar concentrations of diarylamidines and nafamostat, similar to acid-sensing ion channels. Inhibition was voltage-dependent, suggesting pore block as the mechanism of inhibition. Furthermore, we identified the fenamate flufenamic acid and related compounds as agonists of rBLINaC. Application of millimolar concentrations of flufenamic acid to rBLINaC induced a robust, Na(+)-selective current, which was blocked partially by amiloride. The identification of an artificial agonist of rBLINaC supports the hypothesis that rBLINaC is opened by an unknown physiological ligand. Inhibition by diarylamidines and activation by fenamates define a unique pharmacological profile for BLINaC, which will be useful to unravel the physiological function of this ion channel.


Assuntos
Encéfalo/metabolismo , Fenamatos/farmacologia , Mucosa Intestinal/metabolismo , Fígado/metabolismo , Bloqueadores dos Canais de Sódio/farmacologia , Canais de Sódio/efeitos dos fármacos , Amilorida/farmacologia , Animais , Benzamidinas , Diminazena/farmacologia , Relação Dose-Resposta a Droga , Guanidinas/farmacologia , Ratos , Xenopus laevis
19.
Cells ; 10(3)2021 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-33809987

RESUMO

Neurodegenerative disorders are desperately lacking treatment options. It is imperative that drug repurposing be considered in the fight against neurodegenerative diseases. Fenamates have been studied for efficacy in treating several neurodegenerative diseases. The purpose of this review is to comprehensively present the past and current research on fenamates in the context of neurodegenerative diseases with a special emphasis on tolfenamic acid and Alzheimer's disease. Furthermore, this review discusses the major molecular pathways modulated by fenamates.


Assuntos
Anti-Inflamatórios não Esteroides/uso terapêutico , Fenamatos/uso terapêutico , Degeneração Neural , Doenças Neurodegenerativas/tratamento farmacológico , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/uso terapêutico , Animais , Anti-Inflamatórios não Esteroides/efeitos adversos , Anti-Inflamatórios não Esteroides/farmacocinética , Fenamatos/efeitos adversos , Fenamatos/farmacocinética , Humanos , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Neurônios/metabolismo , Neurônios/patologia , Fármacos Neuroprotetores/efeitos adversos , Fármacos Neuroprotetores/farmacocinética
20.
J Inorg Biochem ; 218: 111410, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33721718

RESUMO

The interaction of FeCl3 with the fenamate non-steroidal anti-inflammatory drugs has led to the formation and isolation of trinuclear iron(III) complexes, while in the presence of the nitrogen-donors 2,2'-bipyridine or pyridine tetranuclear iron(III) complexes were derived. The five resultant complexes were characterized by diverse techniques (including infrared, electronic and Mössbauer spectroscopy) and their crystal structures were determined by single-crystal X-ray crystallography. These complexes are the first structurally characterized Fe(III)-fenamato complexes. The complexes were evaluated for their ability to scavenge in vitro free radicals such as hydroxyl, 1,1-diphenyl-2-picrylhydrazyl and 2,2΄-azinobis-(3-ethylbenzothiazoline-6-sulfonic acid). The in vitro binding affinity of the complexes to calf-thymus (CT) DNA was examined and their interaction with serum albumins was also investigated. In total, the complexes present promising activity against the radicals tested, and they may bind tightly to CT DNA possibly via intercalation and reversibly to serum albumins.


Assuntos
Complexos de Coordenação/metabolismo , DNA/metabolismo , Fenamatos/metabolismo , Ferro/química , Ferro/metabolismo , Soroalbumina Bovina/metabolismo , Albumina Sérica Humana/metabolismo , Animais , Complexos de Coordenação/química , Cristalografia por Raios X , Fenamatos/química , Humanos , Substâncias Intercalantes/metabolismo , Estrutura Molecular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA