Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 91
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Pharm ; 21(5): 2501-2511, 2024 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-38574292

RESUMEN

The molecular structures of nonsteroidal anti-inflammatory drugs (NSAIDs) vary, but most contain a carboxylic acid functional group (RCOOH). This functional group is known to be related to the mechanism of cyclooxygenase inhibition and also causes side effects, such as gastrointestinal bleeding. This study proposes a new role for RCOOH in NSAIDs: facilitating the interaction at the binding site II of serum albumins. We used bovine serum albumin (BSA) as a model to investigate the interactions with ligands at site II. Using dansyl-proline (DP) as a fluorescent site II marker, we demonstrated that only negatively charged NSAIDs such as ibuprofen (IBP), naproxen (NPX), diflunisal (DFS), and ketoprofen (KTP) can efficiently displace DP from the albumin binding site. We confirmed the importance of RCOO by neutralizing IBP and NPX through esterification, which reduced the displacement of DP. The competition was also monitored by stopped-flow experiments. While IBP and NPX displaced DP in less than 1 s, the ester derivatives were ineffective. We also observed a higher affinity of negatively charged NSAIDs using DFS as a probe and ultrafiltration experiments. Molecular docking simulations showed an essential salt bridge between the positively charged residues Arg409 and Lys413 with RCOO-, consistent with the experimental findings. We performed a ligand dissociation pathway and corresponding energy analysis by applying molecular dynamics. The dissociation of NPX showed a higher free energy barrier than its ester. Apart from BSA, we conducted some experimental studies with human serum albumin, and similar results were obtained, suggesting a general effect for other mammalian serum albumins. Our findings support that the RCOOH moiety affects not only the mechanism of action and side effects but also the pharmacokinetics of NSAIDs.


Asunto(s)
Antiinflamatorios no Esteroideos , Ácidos Carboxílicos , Ibuprofeno , Simulación del Acoplamiento Molecular , Naproxeno , Albúmina Sérica Bovina , Albúmina Sérica Bovina/química , Albúmina Sérica Bovina/metabolismo , Antiinflamatorios no Esteroideos/química , Sitios de Unión , Animales , Ácidos Carboxílicos/química , Bovinos , Ibuprofeno/química , Naproxeno/química , Unión Proteica , Cetoprofeno/química , Diflunisal/química , Humanos , Ligandos
2.
J Phys Chem B ; 127(45): 9710-9723, 2023 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-37917720

RESUMEN

Understanding the interaction between the drug:carrier complex and protein is essential for the development of a new drug-delivery system. However, the majority of reports are based on an understanding of interactions between the drug and protein. Here, we present our findings on the interaction of the anti-inflammatory drug diflunisal with the drug carrier cyclodextrin (CD) and the protein lysozyme, utilizing steady-state and time-resolved fluorescence spectroscopy. Our findings reveal a different pattern of molecular interaction between the inclusion complex of ß-CD (ß-CD) or hydroxypropyl-ß-CD (HP-ß-CD) (as the host) and diflunisal (as the guest) in the presence of protein lysozyme. The quantum yield for the 1:2 guest:host complex is twice that of the 1:1 guest:host complex, indicating a more stable hydrophobic microenvironment created in the 1:2 complex. Consequently, the nonradiative decay pathway is significantly reduced. The interaction is characterized by ultrafast solvation dynamics and time-resolved fluorescence resonance energy transfer. The solvation dynamics of the lysozyme becomes 10% faster under the condition of binding with the drug, indicating a negligible change in the polar environment after binding. In addition, the fluorescence lifetime of diflunisal (acceptor) is increased by 50% in the presence of the lysozyme (donor), which indicates that the drug molecule is bound to the binding pocket on the surface of the protein, and the average distance between active tryptophan in the hydrophobic region and diflunisal is calculated to be approximately 50 Å. Excitation and emission matrix spectroscopy reveals that the tryptophan emission increases 3-5 times in the presence of both diflunisal and CD. This indicates that the tryptophan of lysozyme may be present in a more hydrophobic environment in the presence of both diflunisal and CD. Our observations on the interaction of diflunisal with ß-CD and lysozyme are well supported by molecular dynamics simulation. Results from this study may have an impact on the development of a better drug-delivery system in the future. It also reveals a fundamental molecular mechanism of interaction of the drug-carrier complex with the protein.


Asunto(s)
Ciclodextrinas , Diflunisal , Diflunisal/química , Ciclodextrinas/química , Triptófano , Muramidasa , Espectrometría de Fluorescencia , 2-Hidroxipropil-beta-Ciclodextrina/química , Preparaciones Farmacéuticas
3.
Int J Mol Sci ; 23(23)2022 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-36499692

RESUMEN

The unexpected dissolution behaviour of amorphous diflunisal-chitosan solid dispersions (kneading method) with respect to the crystalline co-evaporated systems is the starting point of this research. This work is an in-depth study of the diflunisal release behaviour from either chitosan or carboxymethylchitosan dispersions. The microstructure is not usually considered when designing this type of products; however, it is essential to understand the process of solvent penetration and subsequent drug release through a polymeric system, as has been evidenced in this study. In accordance with the kinetic data analysed, it is possible to conclude that the porous structure, conditioned by the sample preparation method, can be considered the main factor involved in diflunisal release. The low mean pore size (1-2 µm), low porosity, and high tortuosity of the amorphous kneaded products are responsible for the slow drug release in comparison with the crystalline coevaporated systems, which exhibit larger pore size (8-10 µm) and lower tortuosity. Nevertheless, all diflunisal-carboxymethylchitosan products show similar porous microstructure and overlapping dissolution profiles. The drug release mechanisms obtained can also be related to the porous structure. Fickian diffusion was the main mechanism involved in drug release from chitosan, whereas an important contribution of erosion was detected for carboxymethylchitosan systems, probably due to its high solubility.


Asunto(s)
Quitosano , Diflunisal , Liberación de Fármacos , Quitosano/química , Solubilidad , Diflunisal/química , Polímeros/química
4.
Int J Nanomedicine ; 16: 1457-1472, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33654396

RESUMEN

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.


Asunto(s)
Diflunisal/administración & dosificación , Sistemas de Liberación de Medicamentos , Emulsiones/química , Nanogeles/química , Polietilenglicoles/química , Polietileneimina/química , Administración Tópica , Animales , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Carragenina , Diflunisal/química , Diflunisal/farmacología , Diflunisal/uso terapéutico , Modelos Animales de Enfermedad , Composición de Medicamentos , Liberación de Fármacos , Edema/tratamiento farmacológico , Edema/patología , Conductividad Eléctrica , Concentración de Iones de Hidrógeno , Masculino , Tamaño de la Partícula , Permeabilidad , Transición de Fase , Ratas , Piel/efectos de los fármacos , Absorción Cutánea/efectos de los fármacos , Solubilidad , Tensoactivos/química , Viscosidad
5.
Int J Mol Sci ; 21(19)2020 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-32998442

RESUMEN

Transthyretin (TTR) is a homotetrameric protein involved in human amyloidosis, including familial amyloid polyneuropathy (FAP). Discovering small-molecule stabilizers of the TTR tetramer is a therapeutic strategy for these diseases. Tafamidis, the only approved drug for FAP treatment, is not effective for all patients. Herein, we discovered that benzbromarone (BBM), a uricosuric drug, is an effective TTR stabilizer and inhibitor against TTR amyloid fibril formation. BBM rendered TTR more resistant to urea denaturation, similarly to iododiflunisal (IDIF), a very potent TTR stabilizer. BBM competes with thyroxine for binding in the TTR central channel, with an IC50 similar to IDIF and tafamidis. Results obtained by isothermal titration calorimetry (ITC) demonstrated that BBM binds TTR with an affinity similar to IDIF, tolcapone and tafamidis, confirming BBM as a potent binder of TTR. The crystal structure of the BBM-TTR complex shows two molecules binding deeply in the thyroxine binding channel, forming strong intermonomer hydrogen bonds and increasing the stability of the TTR tetramer. Finally, kinetic analysis of the ability of BBM to inhibit TTR fibrillogenesis at acidic pH and comparison with other stabilizers revealed that benzbromarone is a potent inhibitor of TTR amyloidogenesis, adding a new interesting scaffold for drug design of TTR stabilizers.


Asunto(s)
Benzbromarona/química , Reposicionamiento de Medicamentos , Fármacos Neuroprotectores/química , Prealbúmina/química , Tiroxina/química , Amiloide/antagonistas & inhibidores , Benzbromarona/metabolismo , Benzoxazoles/química , Benzoxazoles/metabolismo , Sitios de Unión , Unión Competitiva , Cristalografía por Rayos X , Diflunisal/análogos & derivados , Diflunisal/química , Diflunisal/metabolismo , Expresión Génica , Humanos , Enlace de Hidrógeno , Cinética , Simulación del Acoplamiento Molecular , Fármacos Neuroprotectores/metabolismo , Prealbúmina/agonistas , Prealbúmina/genética , Prealbúmina/metabolismo , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Estabilidad Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Termodinámica , Tiroxina/metabolismo , Tolcapona/química , Tolcapona/metabolismo
6.
Spectrochim Acta A Mol Biomol Spectrosc ; 234: 118265, 2020 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-32203686

RESUMEN

Both pretty low solubility and high membrane permeability of diflunisal (DIF) would affect significantly its oral bioavailability as a typical non-steroidal anti-inflammatory substance. Meanwhile, pyrazinamide (PZA), known as one kind of important anti-tuberculosis drugs, has also several certain side effects. These deficiencies affect the large-scale clinical use of such drugs. Solid-state pharmaceutical co-crystallization is of contemporary interest since it offers an easy and efficient way to produce prospective materials with tunable improved properties. In the current work, a novel solid phase drug-drug co-crystal involving DIF and PZA with molar ratio 1:1 was prepared through the mechanical grinding approach, and vibrational spectroscopic techniques including terahertz time-domain spectroscopy (THz-TDS) and Raman spectroscopy were performed to identify DIF, PZA and their pharmaceutical drug-drug co-crystal. The absorption peaks observed in the THz spectra of the co-crystal were at 0.35, 0.65, 1.17, 1.31 and 1.42 THz respectively, which are obviously different from parent materials. Similarly, Raman spectra could also be used to characterize the difference shown between the co-crystal and parent compounds. Structures and vibrational patterns of three kinds of possible co-crystal theoretical forms (form I, II and III) between DIF and PZA have been simulated by performing density functional theory (DFT) calculations. Theoretical results and THz/Raman vibrational spectra of DIF-PZA co-crystal show that the DIF links to PZA via the carboxylic acid-pyridine hetero-synthon association establishing the theoretical form I, which is a much-higher degree of agreement with experimental results than those of other two co-crystal forms. These results provide us a unique method for characterizing the composition of co-crystal structures, and also provide a wealth of drug-drug co-crystal structural information for improving physicochemical properties and pharmacological activities of specific drugs at the molecular-level.


Asunto(s)
Teoría Funcional de la Densidad , Diflunisal/química , Pirazinamida/química , Espectrometría Raman , Espectroscopía de Terahertz , Cristalización , Conformación Molecular , Vibración
7.
Pak J Pharm Sci ; 33(5(Supplementary)): 2307-2315, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33832905

RESUMEN

Low aqueous solubility and bioavailability is the limiting factor to achieve desired therapeutic efficacy for variety of new and existing drug moieties. The goal of the present study was to explore the effects of ß-cyclodextrin (ßCD) and hydroxypropyl-ß-cyclodextrin (HPßCD) on the solubility and dissolution profile of diflunisal (DIF) prepared by using two different methods (physical mixing and solvent evaporation) at DIF-cyclodextrins weight ratios of 1:1, 1:2 and 1:4. The phase solubility studies demonstrated that DIF solubility increased proportionally with an increase in ßCD and HPßCD concentration. The inclusion complexes were subjected to characterization of scanning electron microscopy (SEM), fourier transform infrared spectroscopy (FTIR), differential scanning calorimetry (DSC) and X-ray diffractometry (XRD). Solvent evaporation yielded higher DIF solubility than physical mixing method. HPßCD-DIF inclusion complexes yielded higher dissolution profile than ßCD complexes when prepared under same experimental design. FTIR, DSC and XRD confirmed the successful inclusion of DIF into cyclodextrin (ßCD/HPßCD) by both preparation methods with enhanced water solubility and drug release in comparison with pure drug.


Asunto(s)
2-Hidroxipropil-beta-Ciclodextrina/química , Antiinflamatorios no Esteroideos/química , Diflunisal/química , Excipientes/química , beta-Ciclodextrinas/química , Composición de Medicamentos , Liberación de Fármacos , Solubilidad , Solventes/química
8.
Sci Rep ; 9(1): 13672, 2019 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-31541162

RESUMEN

It is well settled that the amyloidogenic properties of the plasma protein transporter transthyretin (TTR) can be modulated by compounds that stabilize its native tetrameric conformation. TTR is also present in cerebrospinal fluid where it can bind to Aß-peptides and prevent Aß aggregation. We have previously shown that treatment of Alzheimer's Disease (AD) model mice with iododiflunisal (IDIF), a TTR tetramer stabilizing compound, prevents AD pathologies. This evidence positioned IDIF as a new lead drug for AD. In dissecting the mechanism of action of IDIF, we disclose here different labeling strategies for the preparation of 131I-labeled IDIF and 131I- and 124I-labeled TTR, which have been further used for the preparation of IDIF-TTR complexes labeled either on the compound or the protein. The biodistribution of all labeled species after intravenous administration has been investigated in mice using ex vivo and in vivo techniques. Our results confirm the capacity of TTR to cross the blood brain barrier (BBB) and suggest that the formation of TTR-IDIF complexes enhances BBB permeability of both IDIF and TTR. The increased TTR and IDIF brain concentrations may result in higher Aß-peptide sequestration capacity with the subsequent inhibition of AD symptoms as we have previously observed in mice.


Asunto(s)
Encéfalo/diagnóstico por imagen , Diflunisal/análogos & derivados , Radioisótopos de Yodo/química , Prealbúmina/química , Prealbúmina/farmacocinética , Administración Intravenosa , Péptidos beta-Amiloides/metabolismo , Animales , Autorradiografía , Barrera Hematoencefálica/química , Encéfalo/metabolismo , Diflunisal/administración & dosificación , Diflunisal/química , Diflunisal/farmacocinética , Ratones , Tomografía de Emisión de Positrones , Prealbúmina/administración & dosificación , Distribución Tisular
9.
EMBO Rep ; 20(10): e47788, 2019 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-31418171

RESUMEN

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.


Asunto(s)
Quimiocina CXCL12/metabolismo , Diflunisal/farmacología , Proteína HMGB1/metabolismo , Leucocitos/metabolismo , Células 3T3 , Animales , Quimiotaxis/efectos de los fármacos , Diflunisal/química , Disulfuros/metabolismo , Ácido Glicirrínico/farmacología , Humanos , Inflamación/patología , Leucocitos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Espectroscopía de Resonancia Magnética , Ratones
10.
Int J Pharm ; 563: 293-303, 2019 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-30951860

RESUMEN

The current study aimed to develop an effective transdermal nanovesicular carrier of diflunisal that provides enhanced delivery through the skin. Two types of nanovesicles, ethosomes and transfersomes, were investigated and compared to conventional liposomes. Ethosomes with variable ethanol contents (10, 30 and 50%) and transfersomes using different edge activators, including sodium deoxycholate, sodium cholate and sodium taurocholate, were prepared and characterized. The obtained vesicles revealed good entrapment efficiencies (46.73-65.99%), nanometric vesicle sizes (453.10-796.80 nm) and negative zeta potential values (-45.40 to -86.90 mV). Ethosomes with 30% ethanol and sodium deoxycholate-containing transfersomes were incorporated into hydrogels to evaluate their in vitro release and permeation patterns. Nanovesicular hydrogels exhibited more sustained diflunisal release than did corresponding dispersions. Compared to liposomal hydrogel, both carriers proved the superiority of diflunisal permeation and flux across the skin. Confocal laser scanning microscopy showed improved penetration of rhodamine-loaded nanovesicles through skin layers with a wider distribution and higher fluorescence intensity. Compared to liposomes, selected nanovesicles exhibited remarkable antinociceptive and anti-inflammatory effects manifested by significant reduction in number of writhings and significantly higher inhibition of paw oedema. Hence, the developed nanovesicles could be considered promising carriers for transdermal delivery of diflunisal for pain and inflammation management.


Asunto(s)
Analgésicos/administración & dosificación , Antiinflamatorios/administración & dosificación , Diflunisal/administración & dosificación , Hidrogeles/administración & dosificación , Absorción Cutánea/efectos de los fármacos , Ácido Acético , Administración Cutánea , Analgésicos/química , Animales , Antiinflamatorios/química , Carragenina , Diflunisal/química , Liberación de Fármacos , Edema/inducido químicamente , Edema/tratamiento farmacológico , Hidrogeles/química , Liposomas , Masculino , Ratones , Dolor/inducido químicamente , Dolor/tratamiento farmacológico , Ratas Wistar , Piel/metabolismo
11.
Dalton Trans ; 47(39): 13761-13765, 2018 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-30225483

RESUMEN

We report a novel series of cobalt(iii)-polypridyl complexes, 4-6, that can selectively release diflunisal, a nonsteroidal anti-inflammatory drug, under reducing conditions. Remarkably, the 1,10-phenanthroline-bearing complex 5 displays selective potency towards hard-to-kill cancer stem cells (CSCs) (IC50 = 2.1 ± 0.1 µM) over bulk cancer (IC50 = 3.9 ± 0.2 µM) and normal cells (IC50 = 21.2 ± 1.3 µM). This complex induces CSC apoptosis by DNA damage and cyclooxygenase-2 inhibition.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Cobalto/química , Complejos de Coordinación/química , Complejos de Coordinación/farmacología , Diflunisal/química , Células Madre Neoplásicas/efectos de los fármacos , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Humanos , Concentración 50 Inhibidora , Células Madre Neoplásicas/patología
12.
J Phys Chem B ; 122(40): 9409-9418, 2018 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-30222347

RESUMEN

In the present study, we attempt to characterize fluorinated ligand-serum albumin interaction in solution by a set of one-dimensional 19F ligand-based experiments. In this regard, a model system diflunisal (DFL)-human serum albumin (HSA) has been chosen to benchmark the utility of 19F relaxation and diffusion-based experiments in deciphering ligand-protein interactions. Further, we extend the application of a similar set of 19F experiments to unravel the molecular interaction in an unexplored system of 2,6-difluorobenzoic acid (DFBA)-bovine serum albumin (BSA). Interaction analysis of DFBA-SA is of particular interest because DFBA is not only a stable metabolite of a number of pesticides but also used as the starting reagent of many fluorinated drugs. Observation of 19F-1H & 1H-1H saturation transfer difference effects confirmed binding of the ligands to SA. Further, these ligand-protein complexes were probed in terms of the dissociation constant ( KD), number of binding sites ( n), bound fraction of the ligand ( Pb), the complex lifetime (τres), and exchange rate ( Kex). Although Carr-Purcell-Meiboom-Gill (CPMG)-based transverse relaxation and diffusion analysis quantified the former three quantities, the latter two were determined by the constant time fast pulsing CPMG method. Additionally, 19F competition binding experiments performed with well-characterized BSA site markers and DFBA indicated nonspecific binding of DFBA to BSA, whereas similar measurements in the case of HSA with DFL and DFBA revealed superior binding interaction of DFL with SA.


Asunto(s)
Benzoatos/metabolismo , Diflunisal/metabolismo , Albúmina Sérica Humana/metabolismo , Benzoatos/química , Sitios de Unión , Difusión , Diflunisal/química , Flúor/química , Humanos , Ligandos , Resonancia Magnética Nuclear Biomolecular/métodos , Unión Proteica , Albúmina Sérica Humana/química
13.
Molecules ; 23(8)2018 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-30082676

RESUMEN

Cyclooxygenase enzymes play a vital role in inflammatory pathways in the human body. Apart from their relation with inflammation, the additional involvement of COX-2 enzyme with cancer activity was recently discovered. In some cancer types the level of COX-2 enzyme is increased indicating that this enzyme could be a suitable target for cancer therapy. Based on these findings, we have synthesized some new diflunisal thiosemicarbazides and 1,2,4-triazoles and tested them against androgen-independent prostate adenocarcinoma (PC-3), colon carcinoma (HCT-116), human breast cancer (T47D), breast carcinoma (MCF7) and human embryonic kidney (HEK-293) cell lines. Specifically, the diflunisal and thiosemicarbazide functionality are combined during the synthesis of original compounds anticipating a potency enhancement. Compounds 6, 10, 15 and 16 did not show cytotoxic effects for the HEK293 cell line. Among them, compounds 15 and 16 demonstrated anticancer activity for the breast cancer cell line T47D, whereas compounds 6 and 10 which are thiosemicarbazide derivatives displayed anti-tumourigenic activity against the PC-3 cell line, consistent with the literature. However, no activity was observed for the HCT-116 cancer cell line with the tested thiosemicarbazide derivatives. Only compound 16 displayed activity against the HCT-116 cell line. Therefore, it was speculated that the diflunisal and thiosemicarbazide functionalities potentiate anticancer activity on prostate cancer and the thiosemicarbazide functionality decreases the anticancer activity of diflunisal on colon cancer cell lines. In order to gain insight into the anticancer activity and COX-2 inhibition, molecular docking studies were carried out for COX-1 and COX-2 enzymes utilizing the newly synthesized compounds 15, and 16. Both 15 and 16 showed high selectivity and affinity toward COX-2 isozyme over COX-1, which is in agreement with the experimental results.


Asunto(s)
Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Inhibidores de la Ciclooxigenasa/farmacología , Diflunisal/química , Simulación del Acoplamiento Molecular , Antineoplásicos/química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Inhibidores de la Ciclooxigenasa 2/química , Inhibidores de la Ciclooxigenasa 2/farmacología , Inhibidores de la Ciclooxigenasa/síntesis química , Inhibidores de la Ciclooxigenasa/química , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Células HCT116 , Células HEK293 , Humanos , Células MCF-7 , Masculino , Semicarbacidas/química , Relación Estructura-Actividad
14.
Int J Pharm ; 533(1): 206-224, 2017 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-28943207

RESUMEN

Diflunisal (DIF) is used for treatment of rheumatoid arthritis, osteoarthritis etc. DIF-phospholipid complex (DIF-PL complex) was prepared by solvent-evaporation method and characterized by molecular docking studies, SEM, FTIR, DSC, PXRD studies. Further, the DIF-PL complex was incorporated into supramolecular nano-engineered lipidic carriers (SNLCs) for transdermal delivery. The optimization exercise was done using Face centered cubic design (FCCD) after screening of variables by L8 Taguchi orthogonal array design. The optimized SNLC formulation depicted average particle size (188.1nm), degree of entrapment (86.77±3.33%), permeation flux (5.47±0.48µg/cm2/h) and skin retention (17.72±0.68µg/cm2). The dermatokinetic studies revealed the higher concentration of DIF in dermis. The Confocal laser scanning microscopy (CSLM) studies revealed penetration of SNLCs into the deeper layers of skin. The results of mice ear edema depicted significant inhibition of ear edema (76.37±12.52%; p<0.05). In CFA induced rheumatoid arthritis model, the inhibition of paw edema was significantly higher (73.85±14.5%). The levels of TNF-α were reduced in synovial fluid (146.74±1.69pg/mL) and serum (132.43±2.70pg/mL). Furthermore, the licking and biting time was reduced in formalin induced hyperalgesia model. Hence, it can be concluded that dual formulation strategy based SNLCs were promising in treatment of pain and inflammation associated with rheumatoid arthritis.


Asunto(s)
Antiinflamatorios no Esteroideos/administración & dosificación , Artritis Experimental/tratamiento farmacológico , Artritis Reumatoide/tratamiento farmacológico , Diflunisal/administración & dosificación , Lípidos/administración & dosificación , Nanopartículas/administración & dosificación , Administración Cutánea , Animales , Articulación del Tobillo/efectos de los fármacos , Articulación del Tobillo/patología , Antiinflamatorios no Esteroideos/química , Artritis Experimental/patología , Artritis Reumatoide/patología , Diflunisal/química , Diseño de Fármacos , Edema/inducido químicamente , Edema/tratamiento farmacológico , Femenino , Formaldehído , Geles , Hiperalgesia/inducido químicamente , Hiperalgesia/tratamiento farmacológico , Lípidos/química , Ratones , Nanopartículas/química , Ratas Wistar , Piel/efectos de los fármacos , Piel/metabolismo , Absorción Cutánea , Xilenos
15.
J Med Chem ; 60(13): 5749-5758, 2017 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-28587455

RESUMEN

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.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Diflunisal/análogos & derivados , Prealbúmina/metabolismo , Mapas de Interacción de Proteínas/efectos de los fármacos , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/química , Cristalografía por Rayos X , Diflunisal/química , Diflunisal/farmacología , Humanos , Espectroscopía de Resonancia Magnética , Simulación del Acoplamiento Molecular , Prealbúmina/química
16.
FEBS Lett ; 591(15): 2311-2322, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28656718

RESUMEN

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.


Asunto(s)
Antibacterianos/farmacología , ADN Polimerasa III/antagonistas & inhibidores , ADN Polimerasa III/química , Diflunisal/farmacología , Helicobacter pylori/efectos de los fármacos , Antibacterianos/química , Sitios de Unión , Cristalografía por Rayos X , ADN Ligasas/metabolismo , ADN Polimerasa III/metabolismo , Diflunisal/química , Aprobación de Drogas , Evaluación Preclínica de Medicamentos/métodos , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Helicobacter pylori/enzimología , Concentración 50 Inhibidora , Simulación del Acoplamiento Molecular , Conformación Proteica , Estados Unidos , United States Food and Drug Administration
17.
J Inorg Biochem ; 170: 85-97, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28231454

RESUMEN

From the reaction of ZnCl2 with the non-steroidal anti-inflammatory drug diflunisal (Hdifl), complex [Zn(difl-O)2(MeOH)4], 1 was formed, while in the presence of a N,N'-donor heterocyclic ligand 2,2'-bipyridylamine (bipyam), 2,2'-bipyridine (bipy), 1,10-phenanthroline (phen) and 2,2'-dipyridylketone oxime (Hpko), the complexes [Zn(difl-O,O')2(bipyam)], 2, [Zn(difl-O,O')2(bipy)], 3, [Zn(difl-O,O')2(phen)], 4 and [Zn(difl-O)2(Hpko)2], 5 were isolated, respectively. The complexes were characterized by physicochemical and spectroscopic techniques and the crystal structures of complexes 2, 3 and 5 were determined by X-ray crystallography. The ability of the complexes to scavenge 1,1-diphenyl-picrylhydrazyl, 2,2'-azinobis(3-ethylbenzothiazoline-6-sulfonic acid) and hydroxyl radicals and to inhibit soybean lipoxygenase was studied and the complexes were more active than free Hdifl. The interaction of the complexes with serum albumins was monitored by fluorescence emission spectroscopy and the corresponding binding constants were calculated. UV-vis spectroscopy, viscosity measurements and fluorescence emission spectroscopy for the competitive studies of the complexes with ethidium bromide were employed to investigate the interaction of the complexes with calf-thymus DNA and revealed intercalation as the most possible DNA-binding mode. Computational techniques were used to identify possible binding sites of albumins and DNA, and determine the druggability of human and bovine serum albumins with the five novel complexes. The majority of the complexes are stronger binders than the free Hdifl. This is the first study incorporating experimental and computational results to explore the binding activity of metal-NSAID complexes with DNA and serum albumins, suggesting their application as potential metallodrugs.


Asunto(s)
Antioxidantes , ADN/química , Diflunisal , Albúmina Sérica Bovina/química , Zinc/química , Animales , Antioxidantes/síntesis química , Antioxidantes/química , Bovinos , Diflunisal/síntesis química , Diflunisal/química , Humanos , Estructura Molecular
18.
J Microencapsul ; 33(5): 475-86, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27499126

RESUMEN

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.


Asunto(s)
Artritis Experimental/tratamiento farmacológico , Diflunisal , Portadores de Fármacos , Nanopartículas/química , Absorción Cutánea , Animales , Diflunisal/química , Diflunisal/farmacología , Portadores de Fármacos/química , Portadores de Fármacos/farmacología , Femenino , Ratones , Ratas
19.
J Inorg Biochem ; 162: 9-21, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27318424

RESUMEN

The reaction of NiCl2 with the non-steroidal anti-inflammatory drug diflunisal (Hdifl) resulted in the formation of complex [Ni(difl-O)2(MeOH)4], 1. The co-existence of a N,N'-donor heterocyclic ligand 2,2'-dipyridylketone oxime (Hpko), 1,10-phenanthroline (phen), 2,2'-bipyridine (bipy) and 2,2'-bipyridylamine (bipyam) led to the formation of complexes [Ni(difl-O)2(Hpko-N,N')2], 2, [Ni(difl)2(phen)(MeOH)2], 3, [Ni(difl)2(bipy)(MeOH)2], 4 and [Ni(difl-O,O')2(bipyam)], 5, respectively. The complexes were characterized by physicochemical and spectroscopic techniques and the crystal structures of complexes 1 and 2 were determined by X-ray crystallography. The ability of the complexes to scavenge in vitro 1,1-diphenyl-picrylhydrazyl, 2,2'-azinobis(3-ethylbenzothiazoline-6-sulfonic acid) and hydroxyl radicals was investigated; the complexes were more active scavengers than free Hdifl. The interaction of the complexes with serum albumins was investigated by fluorescence emission spectroscopy and the binding constants of the compounds to the albumins were calculated. UV spectroscopy, cyclic voltammetry and viscosity measurements as well as fluorescence emission spectroscopy for the competitive studies of the complexes with ethidium bromide were employed so as to monitor the interaction of the compounds with calf-thymus DNA and revealed intercalation as the most possible mode of binding.


Asunto(s)
Antiinflamatorios no Esteroideos/química , Complejos de Coordinación/química , Diflunisal/química , Depuradores de Radicales Libres/química , Níquel/química , 2,2'-Dipiridil/análogos & derivados , 2,2'-Dipiridil/química , Aminopiridinas/química , Animales , Benzotiazoles/antagonistas & inhibidores , Benzotiazoles/química , Unión Competitiva , Compuestos de Bifenilo/antagonistas & inhibidores , Compuestos de Bifenilo/química , Bovinos , Complejos de Coordinación/síntesis química , Cristalografía por Rayos X , ADN/química , Etidio/química , Depuradores de Radicales Libres/síntesis química , Radical Hidroxilo/antagonistas & inhibidores , Radical Hidroxilo/química , Modelos Moleculares , Fenantrolinas/química , Picratos/antagonistas & inhibidores , Picratos/química , Unión Proteica , Albúmina Sérica/química , Ácidos Sulfónicos/antagonistas & inhibidores , Ácidos Sulfónicos/química
20.
Elife ; 52016 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-27244239

RESUMEN

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.


Asunto(s)
Antineoplásicos/farmacología , Diflunisal/farmacología , Inhibidores Enzimáticos/farmacología , Regulación Leucémica de la Expresión Génica , Leucemia Mieloide Aguda/tratamiento farmacológico , Ácido Salicílico/farmacología , Factores de Transcripción p300-CBP/antagonistas & inhibidores , Acetilcoenzima A/antagonistas & inhibidores , Acetilcoenzima A/metabolismo , Acetilación/efectos de los fármacos , Animales , Antineoplásicos/química , Unión Competitiva , Dominio Catalítico , Línea Celular Tumoral , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Diflunisal/química , Inhibidores Enzimáticos/química , Células HEK293 , Humanos , Leucemia Mieloide Aguda/enzimología , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Leucocitos/efectos de los fármacos , Leucocitos/enzimología , Leucocitos/patología , Ratones , Ratones SCID , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Unión Proteica , Proteína 1 Compañera de Translocación de RUNX1/genética , Proteína 1 Compañera de Translocación de RUNX1/metabolismo , Ácido Salicílico/química , Transducción de Señal , Relación Estructura-Actividad , Ensayos Antitumor por Modelo de Xenoinjerto , Factores de Transcripción p300-CBP/genética , Factores de Transcripción p300-CBP/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...