Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Epilepsia ; 62(6): 1460-1471, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33955541

RESUMEN

OBJECTIVES: Bumetanide was suggested as an adjunct to phenobarbital for suppression of neonatal seizures. This suggestion was based on the idea that bumetanide, by reducing intraneuronal chloride accumulation through inhibition of the Na-K-2Cl cotransporter NKCC1, may attenuate or abolish depolarizing γ-aminobutyric acid (GABA) responses caused by birth asphyxia. However, a first proof-of-concept clinical trial failed. This could have had several reasons, including bumetanide's poor brain penetration, the wide cellular NKCC1 expression pattern in the brain, and problems with the general concept of NKCC1's role in neonatal seizures. We recently replicated the clinical failure of bumetanide to potentiate phenobarbital's effect in a novel rat model of birth asphyxia. In this study, a clinically relevant dose (0.3 mg/kg) of bumetanide was used that does not lead to NKCC1-inhibitory brain levels. The aim of the present experiments was to examine whether a much higher dose (10 mg/kg) of bumetanide is capable of potentiating phenobarbital in this rat model. Furthermore, the effects of the two lipophilic bumetanide derivatives, the ester prodrug N,N-dimethylaminoethylester of bumetanide (DIMAEB) and the benzylamine derivative bumepamine, were examined at equimolar doses. METHODS: Intermittent asphyxia was induced for 30 min by exposing male and female P11 rat pups to three 7 + 3 min cycles of 9% and 5% O2 at constant 20% CO2 . All control pups exhibited neonatal seizures after the asphyxia. RESULTS: Even at 10 mg/kg, bumetanide did not potentiate the effect of a submaximal dose (15 mg/kg) of phenobarbital on seizure incidence, whereas a significant suppression of neonatal seizures was determined for combinations of phenobarbital with DIMAEB or, more effectively, bumepamine, which, however, does not inhibit NKCC1. Of interest, the bumepamine/phenobarbital combination prevented the neurodegenerative consequences of asphyxia and seizures in the hippocampus. SIGNIFICANCE: Both bumepamine and DIMAEB are promising tools that may help to develop more effective lead compounds for clinical trials.


Asunto(s)
Anticonvulsivantes/farmacología , Asfixia Neonatal/complicaciones , Asfixia Neonatal/tratamiento farmacológico , Bencilaminas/uso terapéutico , Bumetanida/uso terapéutico , Hipocampo/patología , Degeneración Nerviosa/patología , Fenobarbital/farmacología , Convulsiones/tratamiento farmacológico , Convulsiones/etiología , Animales , Animales Recién Nacidos , Anticonvulsivantes/farmacocinética , Bencilaminas/farmacocinética , Encéfalo/metabolismo , Bumetanida/análogos & derivados , Bumetanida/farmacocinética , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Femenino , Masculino , Fenobarbital/farmacocinética , Embarazo , Ratas , Miembro 2 de la Familia de Transportadores de Soluto 12/biosíntesis
2.
Bioorg Chem ; 100: 103878, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32361486

RESUMEN

Herein, we describe the design and synthesis of new benzenesulfonamide derivatives as selective COX-2 inhibitors based on bumetanide scaffold. Benzenesulfonamides bearing both the pyrazole 6b and the triazoles 9a, 9c were good inhibitors of COX-2 with IC50 values of 0.32, 0.28 and 0.17 µM, respectively. These benzenesulfonamides 6b, 9a and 9c exhibited a higher selectivity index than the reference drug celecoxib. Molecular modeling study showed that incorporation of bumetanide led to a unique binding mode that is most likely the reason for the observed significant COX-2 selectivity. The anti-inflammatory activity of synthesized compounds revealed that triazoles 9a and 9c demonstrated higher efficacy than celecoxib upon using in vivo carrageenan-induced rat paw edema model. Most of the prepared compounds possess low ulcerogenic potential when administered orally. Therefore, these compounds have a great potential to be developed as safe therapeutics for inflammation, pain, and other diseases where COX-2 plays important role in their pathophysiology.


Asunto(s)
Antiinflamatorios/química , Antiinflamatorios/farmacología , Bumetanida/análogos & derivados , Bumetanida/farmacología , Inhibidores de la Ciclooxigenasa 2/química , Inhibidores de la Ciclooxigenasa 2/farmacología , Animales , Antiinflamatorios/síntesis química , Antiinflamatorios/uso terapéutico , Bumetanida/síntesis química , Bumetanida/uso terapéutico , Dominio Catalítico/efectos de los fármacos , Ciclooxigenasa 2/química , Ciclooxigenasa 2/metabolismo , Inhibidores de la Ciclooxigenasa 2/síntesis química , Inhibidores de la Ciclooxigenasa 2/uso terapéutico , Diseño de Fármacos , Edema/tratamiento farmacológico , Edema/metabolismo , Masculino , Ratones , Simulación del Acoplamiento Molecular , Ratas , Sulfonamidas/síntesis química , Sulfonamidas/química , Sulfonamidas/farmacología , Sulfonamidas/uso terapéutico , Bencenosulfonamidas
3.
Neuropharmacology ; 162: 107754, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31476353

RESUMEN

Temporal lobe epilepsy (TLE) is the most common type of focal epilepsies, affecting approximately 35 million people worldwide. Despite the introduction of numerous novel antiepileptic drugs during the last decades, the proportion of patients with therapy-resistant TLE is still high. As an impaired cellular chloride homeostasis appears involved in disease pathophysiology, bumetanide, an antagonist to Na-K-Cl cotransporters, gained interest as potential therapeutic option. However, bumetanide induces a strong diuretic effect and displays poor penetration across the blood-brain barrier (BBB). To reduce these unwanted effects, we modified the already described BUM690 by exchanging the allyl-into a trifluoro-ethyl group to yield BUM532. Furthermore, we exchanged the nitrogen for oxygen in the trifluoro-ethyl group to yield BUM97. In the intrahippocampal kainic acid mouse model of TLE BUM532 ±â€¯phenobarbital (PB), bumetanide ±â€¯PB and PB alone significantly reduced hippocampal paroxysmal discharges (HPDs) but not spike trains. By contrast, treatment with BUM97 suppressed HPDs as well as spike trains dose-dependently, more pronounced compared to the other tested compounds and exerted a synergistic anticonvulsant effect with PB. Moreover, at higher doses BUM97 achieved long-lasting reduction of spike trains. In pentylenetetrazole-induced acute seizures only BUM532 combined with a sub-effective dose of PB increased the seizure threshold. No diuretic effects were observed at any dose of the three derivatives. Our data demonstrate the successful optimization of the pharmacological profile of bumetanide and the potential of the improved derivative BUM97 for the treatment of therapy-resistant TLE, in particular in combinatorial drug regimens with a GABA mimetic.


Asunto(s)
Anticonvulsivantes/farmacología , Barrera Hematoencefálica/metabolismo , Encéfalo/efectos de los fármacos , Bumetanida/análogos & derivados , Epilepsia Refractaria/tratamiento farmacológico , Epilepsia del Lóbulo Temporal/tratamiento farmacológico , Inhibidores del Simportador de Cloruro Sódico y Cloruro Potásico/farmacología , Animales , Encéfalo/metabolismo , Encéfalo/fisiopatología , Bumetanida/farmacología , Convulsivantes , Modelos Animales de Enfermedad , Diuréticos/efectos adversos , Relación Dosis-Respuesta a Droga , Epilepsia Refractaria/inducido químicamente , Epilepsia Refractaria/fisiopatología , Electroencefalografía , Epilepsia del Lóbulo Temporal/inducido químicamente , Epilepsia del Lóbulo Temporal/fisiopatología , Agonistas de Aminoácidos Excitadores/toxicidad , Ácido Kaínico/toxicidad , Ratones , Pentilenotetrazol , Fenobarbital/farmacología , Convulsiones/fisiopatología , Convulsiones/prevención & control , Miembro 2 de la Familia de Transportadores de Soluto 12/genética , Miembro 2 de la Familia de Transportadores de Soluto 12/metabolismo
4.
Int J Mol Sci ; 20(8)2019 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-31013775

RESUMEN

AqB013 and AqB050 compounds inhibit aquaporin 1 (AQP1), a dual water and ion channel implicated in tumour angiogenesis. We tested AqB013 and AqB050 either as monotherapy or in combination on tube formation of murine endothelial cells (2H-11 and 3B-11) and human umbilical vascular endothelial cells (HUVECs). The mechanism underlying their anti-tubulogenic effect was explored by examining cell viability, induction of apoptosis and migration using 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) assay, Annexin V/propidium iodide apoptosis assay and scratch wound assay. Tube formation of all the cell lines was inhibited by AqB013, AqB050 and the combination of the two compounds. The inhibition of 2H-11 and 3B-11 was frequently accompanied by impaired migration, whereas that of HUVEC treated with AqB050 and the combination was associated with reduced cell viability due to apoptosis. AqB013 and AqB050 exhibited an anti-tubulogenic effect through inhibition of AQP1-mediated cell migration and induction of apoptosis. Together with previously reported anti-tumour cell effect of AqB013 and AqB050, our findings support further evaluation of these compounds as potential cancer therapeutics.


Asunto(s)
Apoptosis/efectos de los fármacos , Acuaporina 1/antagonistas & inhibidores , Bumetanida/farmacología , Movimiento Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Bumetanida/análogos & derivados , Supervivencia Celular/efectos de los fármacos , Células Endoteliales/metabolismo , Humanos
5.
Neuropharmacology ; 143: 186-204, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30248303

RESUMEN

Based on the potential role of Na-K-Cl cotransporters (NKCCs) in epileptic seizures, the loop diuretic bumetanide, which blocks the NKCC1 isoforms NKCC1 and NKCC2, has been tested as an adjunct with phenobarbital to suppress seizures. However, because of its physicochemical properties, bumetanide only poorly penetrates through the blood-brain barrier. Thus, concentrations needed to inhibit NKCC1 in hippocampal and neocortical neurons are not reached when using doses (0.1-0.5 mg/kg) in the range of those approved for use as a diuretic in humans. This prompted us to search for a bumetanide derivative that more easily penetrates into the brain. Here we show that bumepamine, a lipophilic benzylamine derivative of bumetanide, exhibits much higher brain penetration than bumetanide and is more potent than the parent drug to potentiate phenobarbital's anticonvulsant effect in two rodent models of chronic difficult-to-treat epilepsy, amygdala kindling in rats and the pilocarpine model in mice. However, bumepamine suppressed NKCC1-dependent giant depolarizing potentials (GDPs) in neonatal rat hippocampal slices much less effectively than bumetanide and did not inhibit GABA-induced Ca2+ transients in the slices, indicating that bumepamine does not inhibit NKCC1. This was substantiated by an oocyte assay, in which bumepamine did not block NKCC1a and NKCC1b after either extra- or intracellular application, whereas bumetanide potently blocked both variants of NKCC1. Experiments with equilibrium dialysis showed high unspecific tissue binding of bumetanide in the brain, which, in addition to its poor brain penetration, further reduces functionally relevant brain concentrations of this drug. These data show that CNS effects of bumetanide previously thought to be mediated by NKCC1 inhibition can also be achieved by a close derivative that does not share this mechanism. Bumepamine has several advantages over bumetanide for CNS targeting, including lower diuretic potency, much higher brain permeability, and higher efficacy to potentiate the anti-seizure effect of phenobarbital.


Asunto(s)
Anticonvulsivantes/farmacología , Bencilaminas/farmacología , Bumetanida/farmacología , Fenobarbital/farmacología , Animales , Anticonvulsivantes/síntesis química , Anticonvulsivantes/química , Anticonvulsivantes/farmacocinética , Bencilaminas/síntesis química , Bencilaminas/química , Bencilaminas/farmacocinética , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Bumetanida/análogos & derivados , Bumetanida/química , Bumetanida/farmacocinética , Evaluación Preclínica de Medicamentos , Sinergismo Farmacológico , Epilepsia/tratamiento farmacológico , Epilepsia/metabolismo , Femenino , Ratones , Oocitos , Fenobarbital/farmacocinética , Ratas Wistar , Convulsiones/tratamiento farmacológico , Convulsiones/metabolismo , Inhibidores del Simportador de Cloruro Sódico y Cloruro Potásico/química , Inhibidores del Simportador de Cloruro Sódico y Cloruro Potásico/farmacocinética , Inhibidores del Simportador de Cloruro Sódico y Cloruro Potásico/farmacología , Miembro 2 de la Familia de Transportadores de Soluto 12/metabolismo , Técnicas de Cultivo de Tejidos , Xenopus laevis
6.
Neuropharmacology ; 117: 182-194, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28192112

RESUMEN

There is accumulating evidence that bumetanide, which has been used over decades as a potent loop diuretic, also exerts effects on brain disorders, including autism, neonatal seizures, and epilepsy, which are not related to its effects on the kidney but rather mediated by inhibition of the neuronal Na-K-Cl cotransporter isoform NKCC1. However, following systemic administration, brain levels of bumetanide are typically below those needed to inhibit NKCC1, which critically limits its clinical use for treating brain disorders. Recently, active efflux transport at the blood-brain barrier (BBB) has been suggested as a process involved in the low brain:plasma ratio of bumetanide, but it is presently not clear which transporters are involved. Understanding the processes explaining the poor brain penetration of bumetanide is needed for developing strategies to improve the brain delivery of this drug. In the present study, we administered probenecid and more selective inhibitors of active transport carriers at the BBB directly into the brain of mice to minimize the contribution of peripheral effects on the brain penetration of bumetanide. Furthermore, in vitro experiments with mouse organic anion transporter 3 (Oat3)-overexpressing Chinese hamster ovary cells were performed to study the interaction of bumetanide, bumetanide derivatives, and several known inhibitors of Oats on Oat3-mediated transport. The in vivo experiments demonstrated that the uptake and efflux of bumetanide at the BBB is much more complex than previously thought. It seems that both restricted passive diffusion and active efflux transport, mediated by Oat3 but also organic anion-transporting polypeptide (Oatp) Oatp1a4 and multidrug resistance protein 4 explain the extremely low brain concentrations that are achieved after systemic administration of bumetanide, limiting the use of this drug for targeting abnormal expression of neuronal NKCC1 in brain diseases.


Asunto(s)
Barrera Hematoencefálica/fisiología , Encéfalo/metabolismo , Bumetanida/farmacocinética , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/fisiología , Transportadores de Anión Orgánico Sodio-Independiente/fisiología , Proteínas de Transporte de Catión Orgánico/fisiología , Animales , Transporte Biológico/efectos de los fármacos , Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/efectos de los fármacos , Bumetanida/análogos & derivados , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Difusión , Femenino , Moduladores del Transporte de Membrana/farmacología , Ratones , Transportadores de Anión Orgánico/antagonistas & inhibidores , Transportadores de Anión Orgánico/genética , Transportadores de Anión Orgánico Sodio-Independiente/antagonistas & inhibidores , Transportadores de Anión Orgánico Sodio-Independiente/metabolismo , Probenecid/farmacología
7.
Epilepsy Behav ; 59: 42-9, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27088517

RESUMEN

The Na(+)-K(+)-Cl(-) cotransporter NKCC1 plays a major role in the regulation of intraneuronal Cl(-) concentration. Abnormal functionality of NKCC1 has been implicated in several brain disorders, including epilepsy. Bumetanide is the only available selective NKCC1 inhibitor, but also inhibits NKCC2, which can cause severe adverse effects during treatment of brain disorders. A NKCC1-selective bumetanide derivative would therefore be a desirable option. In the present study, we used the Xenopus oocyte heterologous expression system to compare the effects of bumetanide and several derivatives on the two major human splice variants of NKCCs, hNKCC1A and hNKCC2A. The derivatives were selected from a series of ~5000 3-amino-5-sulfamoylbenzoic acid derivatives, covering a wide range of structural modifications and diuretic potencies. To our knowledge, such structure-function relationships have not been performed before for NKCC1. Half maximal inhibitory concentrations (IC50s) of bumetanide were 0.68 (hNKCC1A) and 4.0µM (hNKCC2A), respectively, indicating that this drug is 6-times more potent to inhibit hNKCC1A than hNKCC2A. Side chain substitutions in the bumetanide molecule variably affected the potency to inhibit hNKCC1A. This allowed defining the minimal structural requirements necessary for ligand interaction. Unexpectedly, only a few of the bumetanide derivatives examined were more potent than bumetanide to inhibit hNKCC1A, and most of them also inhibited hNKCC2A, with a highly significant correlation between IC50s for the two NKCC isoforms. These data indicate that the structural requirements for inhibition of NKCC1 and NKCC2 are similar, which complicates development of bumetanide-related compounds with high selectivity for NKCC1.


Asunto(s)
Anticonvulsivantes/farmacología , Anticonvulsivantes/uso terapéutico , Bumetanida/análogos & derivados , Bumetanida/farmacología , Diuréticos/farmacología , Epilepsia/tratamiento farmacológico , Miembro 2 de la Familia de Transportadores de Soluto 12/efectos de los fármacos , Animales , Humanos , Oocitos , Miembro 2 de la Familia de Transportadores de Soluto 12/genética , Relación Estructura-Actividad , Xenopus
8.
Mol Pharmacol ; 89(1): 133-40, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26467039

RESUMEN

Aquaporins (AQPs) in the major intrinsic family of proteins mediate fluxes of water and other small solutes across cell membranes. AQP1 is a water channel, and under permissive conditions, a nonselective cation channel gated by cGMP. In addition to mediating fluid transport, AQP1 expression facilitates rapid cell migration in cell types including colon cancers and glioblastoma. Work here defines new pharmacological derivatives of bumetanide that selectively inhibit the ion channel, but not the water channel, activity of AQP1. Human AQP1 was analyzed in the Xenopus laevis oocyte expression system by two-electrode voltage clamp and optical osmotic swelling assays. The aquaporin ligand bumetanide derivative AqB011 was the most potent blocker of the AQP1 ion conductance (IC50 of 14 µM), with no effect on water channel activity (at up to 200 µM). The order of potency for inhibition of the ionic conductance was AqB011 > AqB007 >> AqB006 ≥ AqB001. Migration of human colon cancer (HT29) cells was assessed with a wound-closure assay in the presence of a mitotic inhibitor. AqB011 and AqB007 significantly reduced migration rates of AQP1-positive HT29 cells without affecting viability. The order of potency for AQP1 ion channel block matched the order for inhibition of cell migration, as well as in silico modeling of the predicted order of energetically favored binding. Docking models suggest that AqB011 and AqB007 interact with the intracellular loop D domain, a region involved in AQP channel gating. Inhibition of AQP1 ionic conductance could be a useful adjunct therapeutic approach for reducing metastasis in cancers that upregulate AQP1 expression.


Asunto(s)
Acuaporina 1/antagonistas & inhibidores , Bumetanida/análogos & derivados , Bumetanida/farmacología , Movimiento Celular/efectos de los fármacos , Activación del Canal Iónico/efectos de los fármacos , Animales , Acuaporina 1/química , Movimiento Celular/fisiología , Relación Dosis-Respuesta a Droga , Femenino , Células HT29 , Humanos , Activación del Canal Iónico/fisiología , Estructura Secundaria de Proteína , Xenopus laevis
9.
Am J Physiol ; 265(5 Pt 1): G942-54, 1993 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-8238524

RESUMEN

The loop diuretic bumetanide is an organic monocarboxylic organic anion assumed to be transported into hepatocytes by a transport system for bile acids. The structural requirements of 22 bumetanide analogues were analyzed for an interaction with bile acid uptake into isolated rat hepatocytes. Whereas bumetanide inhibited the hepatocellular uptake of [14C]cholate to the same degree as its own uptake, derivatization altered affinity and specificity and yielded compounds that selectively inhibited either cholate or taurocholate uptake or uptake of both. No correlation was found between the diuretic potency of bumetanide derivatives, reflecting the affinity to the Na(+)-K(+)-Cl- cotransporter, and their affinity to hepatic bile salt transport. Computer-aided model building combined with the calculation of potential energy maps showed a strictly amphipathic charge separation in bumetanide analogues as in bile acids. Ranking bumetanide compounds by their mean inhibitory concentration values, inhibition constants, and their type of competition, we conclude that at least three binding domains in the proteins are essential for recognition by bile acid transporters, namely two hydrophobic and an anionic side, and that for the anionic binding region a carbonyl atom in the ligands as an electron donor group is sufficient for ligand interaction.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Bumetanida/análogos & derivados , Bumetanida/farmacología , Hígado/metabolismo , Animales , Transporte Biológico/efectos de los fármacos , Bumetanida/química , Células Cultivadas , Cinética , Hígado/efectos de los fármacos , Masculino , Modelos Estructurales , Conformación Molecular , Estructura Molecular , Ratas , Ratas Wistar , Relación Estructura-Actividad
10.
Am J Physiol ; 253(2 Pt 1): C243-52, 1987 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-3618761

RESUMEN

(Na + K + Cl) cotransport is the major mechanism of salt transport across the apical membrane of the epithelial cells of the thick ascending limb of Henle's loop of mammalian kidney and the site of action of "loop" diuretics such as furosemide and bumetanide. We have identified a 150-kDa protein in membranes from dog kidney cortex that is photolabeled by a radiolabeled, benzophenone analogue of bumetanide, [3H]4-benzoyl-5-sulfamoyl-3-(3-thenyloxy)benzoic acid ([3H]BSTBA). Several pieces of evidence strongly suggest that this 150-kDa protein is at least part of the (Na + K + Cl) cotransport system. 1) Photoincorporation of [3H]BSTBA into this protein is completely blocked by inclusion of 10 microM unlabeled bumetanide in the photolysis medium. 2) Photoincorporation of [3H]BSTBA into this protein shows a saturable dependence on [3H]BSTBA concentration, with a K 1/2 (approximately 0.1 microM) very similar to that for reversible [3H]BSTBA binding to kidney membranes. 3) Photolabeling of this protein by [3H]BSTBA requires the simultaneous presence of Na, K, and Cl in the photolysis medium. 4) When crude membranes from dog kidney cortex are centrifuged on sucrose density gradients, saturable [3H]bumetanide binding and photoincorporation of [3H]BSTBA in the 150-kDa region show a very similar distribution among the 15 gradient fractions collected. [3H]BSTBA is also photoincorporated into at least two lower molecular mass proteins, the largest of which is approximately 50 kDa.(ABSTRACT TRUNCATED AT 250 WORDS)


Asunto(s)
Bumetanida , Proteínas Portadoras/metabolismo , Diuréticos , Corteza Renal/metabolismo , Animales , Bumetanida/análogos & derivados , Perros , Peso Molecular , Concentración Osmolar , Simportadores de Cloruro de Sodio-Potasio , Rayos Ultravioleta
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...