Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 131
Filtrar
1.
Cell Mol Life Sci ; 80(8): 209, 2023 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-37458846

RESUMO

The corticosteroid hormone, aldosterone, markedly enhances K+ secretion throughout the colon, a mechanism critical to its role in maintaining overall K+ balance. Previous studies demonstrated that basolateral NKCC1 was up-regulated by aldosterone in the distal colon specifically to support K+ secretion-which is distinct from the more well-established role of NKCC1 in supporting luminal Cl- secretion. However, considerable segmental variability exists between proximal and distal colonic ion transport processes, especially concerning their regulation by aldosterone. Furthermore, delineating such region-specific effects has important implications for the management of various gastrointestinal pathologies. Experiments were therefore designed to determine whether aldosterone similarly up-regulates NKCC1 in the proximal colon to support K+ secretion. Using dietary Na+ depletion as a model of secondary hyperaldosteronism in rats, we found that proximal colon NKCC1 expression was indeed enhanced in Na+-depleted (i.e., hyperaldosteronemic) rats. Surprisingly, electrogenic K+ secretion was not detectable by short-circuit current (ISC) measurements in response to either basolateral bumetanide (NKCC1 inhibitor) or luminal Ba2+ (non-selective K+ channel blocker), despite enhanced K+ secretion in Na+-depleted rats, as measured by 86Rb+ fluxes. Expression of BK and IK channels was also found to be unaltered by dietary Na+ depletion. However, bumetanide-sensitive basal and agonist-stimulated Cl- secretion (ISC) were significantly enhanced by Na+ depletion, as was CFTR Cl- channel expression. These data suggest that NKCC1-dependent secretory pathways are differentially regulated by aldosterone in proximal and distal colon. Development of therapeutic strategies in treating pathologies related to aberrant colonic K+/Cl- transport-such as pseudo-obstruction or ulcerative colitis-may benefit from these findings.


Assuntos
Aldosterona , Bumetanida , Animais , Ratos , Aldosterona/farmacologia , Aldosterona/metabolismo , Bumetanida/farmacologia , Bumetanida/metabolismo , Cloretos/metabolismo , Colo , Potássio/metabolismo , Sódio/metabolismo
2.
Mol Pain ; 19: 17448069231159855, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36760008

RESUMO

Previous studies have confirmed the relationship between chloride homeostasis and pain. However, the role of sodium potassium chloride co-transporter isoform 1 (NKCC1) in dorsal horn and dorsal root ganglion neurons (DRGs) in spinal cord injury (SCI)-induced neuropathic pain (NP) remains inconclusive. Therefore, we aimed to explore whether suppression of NKCC1 in the spinal cord and DRGs alleviate the NP of adult rats with thoracic spinal cord contusion. Thirty adult female Sprague-Dawley rats (8 week-old, weighing 250-280 g) were randomly divided into three groups with ten animals in each group (sham, SCI, and bumetanide groups). The paw withdrawal mechanical threshold and paw withdrawal thermal latency were recorded before injury (baseline) and on post-injury days 14, 21, 28, and 35. At the end of experiment, western blotting (WB) analysis, quantitative real-time Polymerase Chain Reaction (PCR) and immunofluorescence were performed to quantify NKCC1 expression. Our results revealed that NKCC1 protein expression in the spinal cord and DRGs was significantly up-regulated in rats with SCI. Intraperitoneal treatment of bumetanide (an NKCC1 inhibitor) reversed the expression of NKCC1 in the dorsal horn and DRGs and ameliorated mechanical ectopic pain and thermal hypersensitivities in the SCI rats. Our study demonstrated the occurrence of NKCC1 overexpression in the spinal cord and DRGs in a rodent model of NP and indicated that changes in the peripheral nervous system also play a major role in promoting pain sensitization after SCI.


Assuntos
Neuralgia , Traumatismos da Medula Espinal , Ratos , Feminino , Animais , Ratos Sprague-Dawley , Bumetanida/metabolismo , Bumetanida/farmacologia , Gânglios Espinais/metabolismo , Corno Dorsal da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/metabolismo , Neuralgia/metabolismo , Medula Espinal/metabolismo , Hiperalgesia/metabolismo
3.
Molecules ; 27(8)2022 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-35458638

RESUMO

Alzheimer's disease (AD) is a neurodegenerative disorder with an increasing need for developing disease-modifying treatments as current therapies only provide marginal symptomatic relief. Recent evidence suggests the γ-aminobutyric acid (GABA) neurotransmitter system undergoes remodeling in AD, disrupting the excitatory/inhibitory (E/I) balance in the brain. Altered expression levels of K-Cl-2 (KCC2) and N-K-Cl-1 (NKCC1), which are cation-chloride cotransporters (CCCs), have been implicated in disrupting GABAergic activity by regulating GABAA receptor signaling polarity in several neurological disorders, but these have not yet been explored in AD. NKCC1 and KCC2 regulate intracellular chloride [Cl-]i by accumulating and extruding Cl-, respectively. Increased NKCC1 expression in mature neurons has been reported in these disease conditions, and bumetanide, an NKCC1 inhibitor, is suggested to show potential therapeutic benefits. This study used primary mouse hippocampal neurons to explore if KCC2 and NKCC1 expression levels are altered following beta-amyloid (Aß1-42) treatment and the potential neuroprotective effects of bumetanide. KCC2 and NKCC1 expression levels were also examined in 18-months-old male C57BL/6 mice following bilateral hippocampal Aß1-42 stereotaxic injection. No change in KCC2 and NKCC1 expression levels were observed in mouse hippocampal neurons treated with 1 nM Aß1-42, but NKCC1 expression increased 30-days post-Aß1-42-injection in the CA1 region of the mouse hippocampus. Primary mouse hippocampal cultures were treated with 1 nM Aß1-42 alone or with various concentrations of bumetanide (1 µM, 10 µM, 100 µM, 1 mM) to investigate the effect of the drug on cell viability. Aß1-42 produced 53.1 ± 1.4% cell death after 5 days, and the addition of bumetanide did not reduce this. However, the drug at all concentrations significantly reduced cell viability, suggesting bumetanide is highly neurotoxic. In summary, these results suggest that chronic exposure to Aß1-42 alters the balance of KCC2 and NKCC1 expression in a region-and layer-specific manner in mouse hippocampal tissue; therefore, this process most likely contributes to altered hippocampal E/I balance in this model. Furthermore, bumetanide induces hippocampal neurotoxicity, thus questioning its suitability for AD therapy. Further investigations are required to examine the effects of Aß1-42 on KCC2 and NKCC1 expression and whether targeting CCCs might offer a therapeutic approach for AD.


Assuntos
Bumetanida , Hipocampo , Membro 2 da Família 12 de Carreador de Soluto , Simportadores , Peptídeos beta-Amiloides , Animais , Bumetanida/metabolismo , Bumetanida/farmacologia , Cloretos/metabolismo , Hipocampo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fragmentos de Peptídeos , Membro 2 da Família 12 de Carreador de Soluto/genética , Membro 2 da Família 12 de Carreador de Soluto/metabolismo , Simportadores/metabolismo
4.
Epilepsia ; 62(1): 269-278, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33140458

RESUMO

OBJECTIVES: The loop diuretic bumetanide has been proposed previously as an adjunct treatment for neonatal seizures because bumetanide is thought to potentiate the action of γ-aminobutyric acid (GABA)ergic drugs such as phenobarbital by preventing abnormal intracellular accumulation of chloride and the subsequent "GABA shift." However, a clinical trial in neonates failed to demonstrate such a synergistic effect of bumetanide, most likely because this drug only poorly penetrates into the brain. This prompted us to develop lipophilic prodrugs of bumetanide, such as the N,N-dimethylaminoethyl ester of bumetanide (DIMAEB), which rapidly enter the brain where they are hydrolyzed by esterases to the parent compound, as demonstrated previously by us in adult rodents. However, it is not known whether esterase activity in neonates is sufficient to hydrolyze ester prodrugs such as DIMAEB. METHODS: In the present study, we examined whether esterases in neonatal serum of healthy term infants are capable of hydrolyzing DIMAEB to bumetanide and whether this activity is different from the serum of adults. Furthermore, to extrapolate the findings to brain tissue, we performed experiments with brain tissue and serum of neonatal and adult rats. RESULTS: Serum from 1- to 2-day-old infants was capable of hydrolyzing DIMAEB to bumetanide at a rate similar to that of serum from adult individuals. Similarly, serum and brain tissue of neonatal rats rapidly hydrolyzed DIMAEB to bumetanide. SIGNIFICANCE: These data provide a prerequisite for further evaluating the potential of bumetanide prodrugs as add-on therapy to phenobarbital and other antiseizure drugs as a new strategy for improving pharmacotherapy of neonatal seizures.


Assuntos
Encéfalo/enzimologia , Bumetanida/metabolismo , Esterases , Ésteres/metabolismo , Pró-Fármacos/metabolismo , Animais , Animais Recém-Nascidos , Encéfalo/metabolismo , Feminino , Humanos , Recém-Nascido , Masculino , Ratos , Soro/enzimologia , Soro/metabolismo
5.
Mol Pharm ; 14(9): 2930-2936, 2017 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-28513167

RESUMO

Monocarboxylate transporter 6 (MCT6; SLC16A5) has been recognized for its role as a xenobiotic transporter, with characterized substrates probenecid, bumetanide, and nateglinide. To date, the impact of commonly ingested dietary compounds on MCT6 function has not been investigated, and therefore, the objective of this study was to evaluate a variety of flavonoids for their potential MCT6-specific interactions. Flavonoids are a large group of polyphenolic phytochemicals found in commonly consumed plant-based products that have been recognized for their dietary health benefits. The uptake of bumetanide in human MCT6 gene-transfected Xenopus laevis oocytes was significantly decreased in the presence of a variety of flavonoids (e.g., quercetin, luteolin, phloretin, and morin), but was not significantly affected by flavonoid glycosides (e.g., naringin, rutin, phlorizin). The IC50 values of quercetin, phloretin, and morin were determined to be 25.3 ± 3.36, 17.3 ± 2.37, and 33.1 ± 3.29 µM, respectively. The mechanism of inhibition of phloretin was reversible and competitive, with a Ki value of 22.8 µM. Furthermore, typical MCT substrates were also investigated for their potential interactions with MCT6. Substrates of MCTs 1, 2, 4, 8, and 10 did not cause any significant decrease in MCT6-mediated bumetanide uptake, suggesting that MCT6 has distinct compound selectivity. In summary, these results suggest that dietary aglycon flavonoids may significantly alter the pharmacokinetics and pharmacodynamics of bumetanide and other MCT6-specific substrates, and may represent potential substrates for MCT6.


Assuntos
Flavonoides/metabolismo , Luteolina/metabolismo , Transportadores de Ácidos Monocarboxílicos/antagonistas & inibidores , Floretina/metabolismo , Quercetina/metabolismo , Animais , Bumetanida/metabolismo , Humanos , Transportadores de Ácidos Monocarboxílicos/metabolismo , Oócitos/metabolismo , Xenopus laevis
6.
Neuroscience ; 318: 34-44, 2016 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-26794590

RESUMO

Population-based studies have demonstrated that children with a history of febrile seizure (FS) perform better than age-matched controls at hippocampus-dependent memory tasks. Here, we report that FSs induce two distinct structural reorganizations in the hippocampus and bidirectionally modify future learning abilities in an age-dependent manner. Compared with age-matched controls, adult mice that had experienced experimental FSs induced by hyperthermia (HT) on postnatal day 14 (P14-HT) performed better in a cognitive task that requires dentate granule cells (DGCs). The enhanced memory performance correlated with an FS-induced persistent increase in the density of large mossy fiber terminals (LMTs) of the DGCs. The memory enhancement was not observed in mice that had experienced HT-induced seizures at P11 which exhibited abnormally located DGCs in addition to the increased LMT density. The ectopic DGCs of the P11-HT mice were abolished by the diuretic bumetanide, and this pharmacological treatment unveiled the masked memory enhancement. Thus, this work provides a novel basis for age-dependent structural plasticity in which FSs influence future brain function.


Assuntos
Febre/complicações , Memória/fisiologia , Neurogênese/fisiologia , Plasticidade Neuronal/fisiologia , Convulsões/fisiopatologia , Envelhecimento , Animais , Bumetanida/metabolismo , Modelos Animais de Doenças , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Camundongos , Convulsões/etiologia , Convulsões/metabolismo
7.
Eur J Pharmacol ; 770: 117-25, 2016 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-26673740

RESUMO

Recently, it has been suggested that bumetanide, an inhibitor of the Na-K-2Cl co-transporter (NKCC1), may be useful in the treatment of central nervous system (CNS) disorders. However, from a physicochemical perspective, bumetanide may not cross the blood-brain barrier to the extent that is necessary for it to be an effective brain NKCC1 inhibitor in vivo. High plasma-protein binding, potentially high brain-tissue binding and putative efflux transporters including organic anion transporter 3 (OAT3) contribute to the poor pharmacokinetic profile of bumetanide. Bidirectional permeability assays are an in vitro method to determine the impact of plasma-protein/brain tissue binding, as well as efflux transport, on the permeability of a compound. We established and validated a cell line stably overexpressing human OAT3 using lentiviral cloning techniques for use in in vitro bidirectional permeability assays. Using efflux transport studies, we show that bumetanide is a transported substrate of human OAT3, exhibiting a transport ratio of ≥1.5, which is attenuated by OAT3 inhibitors. Bidirectional permeability assays were carried out in the presence and absence of either albumin or brain homogenate to elucidate the effect of plasma-protein/brain tissue binding. These tests confirmed the pharmacokinetic limitations for brain delivery of bumetanide. In this experiment, bumetanide is 53% bound to albumin, 77% bound to brain tissue and accumulates in brain cells. Moreover, we conclusively established that bumetanide is a transported substrate of OAT3. Taken together, these bidirectional permeability studies highlight the potential of efflux transporter inhibition as an augmentation strategy for enhanced delivery of bumetanide to the CNS.


Assuntos
Bumetanida/metabolismo , Bumetanida/farmacologia , Membro 2 da Família 12 de Carreador de Soluto/metabolismo , Barreira Hematoencefálica/metabolismo , Bumetanida/farmacocinética , Células HEK293 , Humanos , Transportadores de Ânions Orgânicos Sódio-Independentes/genética , Transportadores de Ânions Orgânicos Sódio-Independentes/metabolismo , Permeabilidade
8.
Eur J Pharmacol ; 746: 167-73, 2015 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-25449033

RESUMO

Bumetanide is increasingly being used for experimental treatment of brain disorders, including neonatal seizures, epilepsy, and autism, because the neuronal Na-K-Cl cotransporter NKCC1, which is inhibited by bumetanide, is implicated in the pathophysiology of such disorders. However, use of bumetanide for treatment of brain disorders is associated with problems, including poor brain penetration and systemic adverse effects such as diuresis, hypokalemic alkalosis, and hearing loss. The poor brain penetration is thought to be related to its high ionization rate and plasma protein binding, which restrict brain entry by passive diffusion, but more recently brain efflux transporters have been involved, too. Multidrug resistance protein 4 (MRP4), organic anion transporter 3 (OAT3) and organic anion transporting polypeptide 2 (OATP2) were suggested to mediate bumetanide brain efflux, but direct proof is lacking. Because MRP4, OAT3, and OATP2 can be inhibited by probenecid, we studied whether this drug alters brain levels of bumetanide in mice. Probenecid (50 mg/kg) significantly increased brain levels of bumetanide up to 3-fold; however, it also increased its plasma levels, so that the brain:plasma ratio (~0.015-0.02) was not altered. Probenecid markedly increased the plasma half-life of bumetanide, indicating reduced elimination of bumetanide most likely by inhibition of OAT-mediated transport of bumetanide in the kidney. However, the diuretic activity of bumetanide was not reduced by probenecid. In conclusion, our study demonstrates that the clinically available drug probenecid can be used to increase brain levels of bumetanide and decrease its elimination, which could have therapeutic potential in the treatment of brain disorders.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Bumetanida/farmacocinética , Moduladores de Transporte de Membrana/farmacologia , Transportadores de Ânions Orgânicos/antagonistas & inibidores , Probenecid/farmacologia , Inibidores de Simportadores de Cloreto de Sódio e Potássio/farmacocinética , Animais , Animais não Endogâmicos , Anticonvulsivantes/sangue , Anticonvulsivantes/metabolismo , Anticonvulsivantes/farmacocinética , Transporte Biológico/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Bumetanida/sangue , Bumetanida/metabolismo , Bumetanida/farmacologia , Linhagem Celular , Diuréticos/sangue , Diuréticos/metabolismo , Diuréticos/farmacocinética , Cães , Interações Medicamentosas , Feminino , Meia-Vida , Humanos , Rim/efeitos dos fármacos , Rim/metabolismo , Camundongos , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Transportadores de Ânions Orgânicos/genética , Transportadores de Ânions Orgânicos/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Inibidores de Simportadores de Cloreto de Sódio e Potássio/sangue , Inibidores de Simportadores de Cloreto de Sódio e Potássio/metabolismo , Inibidores de Simportadores de Cloreto de Sódio e Potássio/farmacologia
9.
J Pharm Pharmacol ; 67(4): 501-10, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25490899

RESUMO

OBJECTIVES: Recent data highlight the potential of bumetanide as a treatment for neonatal seizures and autism, as it facilitates the excitatory to inhibitory switch in gamma-aminobutyric acid signalling. This study examines the extent of blood-brain barrier (BBB) permeation of bumetanide, a key determinant of the efficacy of centrally acting drugs. Furthermore, the impact of efflux transporter organic anion transporter 3 (oat3) inhibition on bumetanide pharmacokinetics was investigated. METHODS: Bumetanide levels in extracellular fluid (ECF) and plasma in the presence and absence of oat3 inhibitor probenecid were monitored using integrated microdialysis. KEY FINDINGS: Following a bumetanide bolus/continuous infusion of 10 mg/kg and 6 mg/kg/h, bumetanide was detected in hippocampal ECF at the estimated concentration of 131 ± 55 ng/ml. Plasma bumetanide levels were ∼20 mg/l at steady state. Coadministration of probenecid resulted in an increase in bumetanide levels in both ECF and plasma, indicating that oat3 inhibition influences the pharmacokinetics of bumetanide primarily in the periphery. CONCLUSION: Although bumetanide reached detectable levels in hippocampal ECF, bumetanide concentration in ECF was low relative to systemic concentration. Oat3 inhibition by probenecid resulted in increased bumetanide concentrations in brain and plasma. As an acute treatment in neonatal seizures, the bumetanide/probenecid combination may hold therapeutic potential.


Assuntos
Adjuvantes Farmacêuticos/farmacologia , Barreira Hematoencefálica/efeitos dos fármacos , Bumetanida/farmacocinética , Líquido Extracelular/metabolismo , Hipocampo/metabolismo , Transportadores de Ânions Orgânicos/antagonistas & inibidores , Probenecid/farmacologia , Animais , Transporte Biológico , Barreira Hematoencefálica/metabolismo , Bumetanida/metabolismo , Bumetanida/farmacologia , Interações Medicamentosas , GABAérgicos/farmacologia , Masculino , Microdiálise , Ratos Sprague-Dawley
10.
Am J Physiol Cell Physiol ; 300(6): C1323-36, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21346157

RESUMO

The intracellular ion homeostasis in cockroach salivary acinar cells during salivation is not satisfactorily understood. This is mainly due to technical problems regarding strong tissue autofluorescence and ineffective ion concentration quantification. For minimizing these problems, we describe the successful application of two-photon (2P) microscopy partly in combination with fluorescence lifetime imaging microscopy (FLIM) to record intracellular Na(+) and Cl(-) concentrations ([Na(+)](i), [Cl(-)](i)) in cockroach salivary acinar cells. Quantitative 2P-FLIM Cl(-) measurements with the dye N-(ethoxycarbonylmethyl)-6-methoxy-quinolinium bromide indicate that the resting [Cl(-)](i) is 1.6 times above the Cl(-) electrochemical equilibrium but is not influenced by pharmacological inhibition of the Na(+)-K(+)-2Cl(-) cotransporter (NKCC) and anion exchanger using bumetanide and 4,4'-diisothiocyanatodihydrostilbene-2,2'-disulfonic acid disodium salt. In contrast, rapid Cl(-) reuptake after extracellular Cl(-) removal is almost totally NKCC mediated both in the absence and presence of dopamine. However, in physiological saline [Cl(-)](i) does not change during dopamine stimulation although dopamine stimulates fluid secretion in these glands. On the other hand, dopamine causes a decrease in the sodium-binding benzofuran isophthalate tetra-ammonium salt (SBFI) fluorescence and an increase in the Sodium Green fluorescence after 2P excitation. This opposite behavior of both dyes suggests a dopamine-induced [Na(+)](i) rise in the acinar cells, which is supported by the determined 2P-action cross sections of SBFI. The [Na(+)](i) rise is Cl(-) dependent and inhibited by bumetanide. The Ca(2+)-ionophore ionomycin also causes a bumetanide-sensitive [Na(+)](i) rise. We propose that a Ca(2+)-mediated NKCC activity in acinar peripheral cells attributable to dopamine stimulation serves for basolateral Na(+) uptake during saliva secretion and that the concomitantly transported Cl(-) is recycled back to the bath.


Assuntos
Cloretos/metabolismo , Baratas/citologia , Baratas/metabolismo , Microscopia de Fluorescência/métodos , Sódio/metabolismo , Animais , Bumetanida/metabolismo , Dopamina/metabolismo , Fluorescência , Corantes Fluorescentes/metabolismo , Compostos de Quinolínio/metabolismo , Glândulas Salivares/citologia , Inibidores de Simportadores de Cloreto de Sódio e Potássio/metabolismo , Simportadores de Cloreto de Sódio-Potássio/metabolismo
11.
Am J Physiol Lung Cell Mol Physiol ; 298(2): L210-31, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19965983

RESUMO

The serous acini of airway submucosal glands are important for fluid secretion in the lung. Serous cells are also sites of expression of the cystic fibrosis transmembrane conductance regulator (CFTR) Cl(-) channel. However, the mechanisms of serous cell fluid secretion remain poorly defined. In this study, serous acinar cells were isolated from porcine bronchi and studied using optical techniques previously used to examine fluid secretion in rat parotid and murine nasal acinar cells. When stimulated with the cholinergic agonist carbachol, porcine serous cells shrank by approximately 20% (observed via DIC microscopy) after a profound elevation of intracellular [Ca(2+)] ([Ca(2+)](i); measured by simultaneous fura 2 fluorescence imaging). Upon removal of agonist and relaxation of [Ca(2+)](i) to resting levels, cells swelled back to resting volume. Similar results were observed during stimulation with histamine and ATP, and elevation of [Ca(2+)](i) was found to be necessary and sufficient to activate shrinkage. Cell volume changes were associated with changes in [Cl(-)](i) (measured using SPQ fluorescence), suggesting that shrinkage and swelling are caused by loss and gain of intracellular solute content, respectively, likely reflecting changes in the secretory state of the cells. Shrinkage was inhibited by niflumic acid but not by GlyH-101, suggesting Ca(2+)-activated secretion is mediated by alternative non-CFTR Cl(-) channels, possibly including Ano1 (TMEM16A), expressed on the apical membrane of porcine serous cells. Optimal cell swelling/solute uptake required activity of the Na(+)K(+)2Cl(-) cotransporter and Na(+)/H(+) exchanger, both of which are expressed on the basolateral membrane of serous acini and likely contribute to sustaining transepithelial secretion.


Assuntos
Líquidos Corporais/metabolismo , Brônquios/citologia , Cálcio/metabolismo , Glândulas Exócrinas/metabolismo , Animais , Anoctamina-1 , Brônquios/metabolismo , Bumetanida/metabolismo , Carbacol/metabolismo , Tamanho Celular , Células Cultivadas , Canais de Cloreto/metabolismo , Cloretos/metabolismo , Agonistas Colinérgicos/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/antagonistas & inibidores , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Perfilação da Expressão Gênica , Humanos , Camundongos , Dados de Sequência Molecular , Ácido Niflúmico/metabolismo , Ratos , Inibidores de Simportadores de Cloreto de Sódio e Potássio/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Simportadores de Cloreto de Sódio-Potássio/metabolismo , Membro 2 da Família 12 de Carreador de Soluto , Suínos
12.
Development ; 136(16): 2837-48, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19633174

RESUMO

Endolymph is the specialised extracellular fluid present inside the inner ear. In mammals, disruptions to endolymph homeostasis can result in either collapse or distension of the endolymphatic compartment in the cochlea, with concomitant hearing loss. The zebrafish little ears (lte) mutant shows a collapse of the otic vesicle in the larva, apparently owing to a loss of endolymphatic fluid in the ear, together with an over-inflation of the swim bladder. Mutant larvae display signs of abnormal vestibular function by circling and swimming upside down. The two available alleles of lte are homozygous lethal: mutant larvae fail to thrive beyond 6 days post-fertilisation. Patterning of the otic vesicle is apparently normal. However, the expression of several genes thought to play a role in endolymph production is downregulated, including the sodium-potassium-chloride cotransporter gene nkcc1 (slc12a2) and several Na(+)/K(+)-ATPase channel subunit genes. We show here that lte mutations correspond to lesions in nkcc1. Each allele has a point mutation that disrupts splicing, leading to frame shifts in the coding region that predict the generation of truncated products. Endolymph collapse in the lte/nkcc1 mutant shows distinct parallels to that seen in mouse Nkcc1 mutants, validating zebrafish as a model for the study of endolymph disorders. The collapse in ear volume can be ameliorated in the to27d allele of lte by injection of a morpholino that blocks splicing at an ectopic site introduced by the mutation. This exemplifies the use of morpholinos as potential therapeutic agents for genetic disease.


Assuntos
Sacos Aéreos/metabolismo , Orelha Interna/metabolismo , Endolinfa/metabolismo , Isoformas de Proteínas/metabolismo , Simportadores de Cloreto de Sódio-Potássio/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra , Sacos Aéreos/anatomia & histologia , Processamento Alternativo , Animais , Sequência de Bases , Padronização Corporal/fisiologia , Bumetanida/metabolismo , Orelha Interna/anatomia & histologia , Embrião não Mamífero/anatomia & histologia , Embrião não Mamífero/fisiologia , Furosemida/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Homeostase , Hibridização In Situ , Camundongos , Dados de Sequência Molecular , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/metabolismo , Fenótipo , Isoformas de Proteínas/genética , Inibidores de Simportadores de Cloreto de Sódio e Potássio/metabolismo , Simportadores de Cloreto de Sódio-Potássio/genética , Membro 2 da Família 12 de Carreador de Soluto , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética
13.
FASEB J ; 23(4): 1168-76, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19103648

RESUMO

Plasma membrane chloride (Cl(-)) pathways play an important role in neuronal physiology. Here, we investigated the role of NKCC1 cotransporters (a secondary active Cl(-) uptake mechanism) in Cl(-) handling in cultured rat dorsal root ganglion neurons (DRGNs) and motor neurons (MNs) derived from fetal stage embryonic day 14. Gramicidin-perforated patch-clamp recordings revealed that DRGNs accumulate intracellular Cl(-) through a bumetanide- and Na(+)-sensitive mechanism, indicative of the functional expression of NKCC1. Western blotting confirmed the expression of NKCC1 in both DRGNs and MNs, but immunocytochemistry experiments showed a restricted expression in dendrites of MNs, which contrasts with a homogeneous expression in DRGNs. Both MNs and DRGNs could be readily loaded with or depleted of Cl(-) during GABA(A) receptor activation at depolarizing or hyperpolarizing membrane potentials. After loading, the rate of recovery to the resting Cl(-) concentration (i.e., [Cl(-)](i) decrease) was similar in both cell types and was unaffected by lowering the extracellular Na(+) concentration. In contrast, the recovery on depletion (i.e., [Cl(-)](i) increase) was significantly faster in DRGNs in control conditions but not in low extracellular Na(+). The experimental observations could be reproduced by a mathematical model for intracellular Cl(-) kinetics, in which DRGNs show higher NKCC1 activity and smaller Cl(-)-handling volume than MNs. On the basis of these results, we conclude that embryonic DRGNs show a higher somatic functional expression of NKCC1 than embryonic MNs. The high NKCC1 activity in DRGNs is important for maintaining high [Cl(-)](i), whereas lower NKCC1 activity in MNs allows large [Cl(-)](i) variations during neuronal activity.


Assuntos
Cloretos/metabolismo , Gânglios Espinais/metabolismo , Neurônios Motores/metabolismo , Receptores de GABA-A/metabolismo , Simportadores de Cloreto de Sódio-Potássio/fisiologia , Animais , Antibacterianos/metabolismo , Antibacterianos/farmacologia , Bumetanida/metabolismo , Bumetanida/farmacologia , Células Cultivadas , Eletrofisiologia , Embrião de Mamíferos , Gânglios Espinais/citologia , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/embriologia , Gramicidina/metabolismo , Gramicidina/farmacologia , Imuno-Histoquímica , Cinética , Modelos Estatísticos , Neurônios Motores/citologia , Neurônios Motores/efeitos dos fármacos , Neurônios Motores/fisiologia , Técnicas de Patch-Clamp , Ratos , Ratos Endogâmicos , Receptores de GABA/metabolismo , Receptores de GABA-A/fisiologia , Inibidores de Simportadores de Cloreto de Sódio e Potássio , Simportadores de Cloreto de Sódio-Potássio/metabolismo , Ácido gama-Aminobutírico/metabolismo
14.
Neurochem Res ; 33(8): 1574-81, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18404376

RESUMO

The Na(+)/H(+) exchanger has been the only unequivocally demonstrated H(+)-transport mechanism in the synaptosomal preparation. We had previously suggested that a Cl(-)-H(+) symporter (in its acidifying mode) is involved in cytosolic pH regulation in the synaptosomal preparation. Supporting this suggestion, we now show that: (1) when synaptosomes are transferred from PSS to either gluconate or sulfate solutions, the Fura-2 ratio remains stable instead of increasing as it does in 50 mM K solution. This indicates that these anions do not promote a plasma membrane depolarization. (2) Based in the recovery rate from the cytosolic alkalinization, the anionic selectivity of the Cl(-)-H(+) symporter is NO(3)(-) > Br(-) > Cl(-) >> I(-) = isethionate = sulfate = methanesulfonate = gluconate. (3) PCMB 10 muM inhibits the gluconate-dependent alkalinization by 30 +/- 6%. (4) Neither Niflumic acid, 9AC, Bumetanide nor CCCP inhibits the recovery from the cytosolic alkalinization.


Assuntos
Ânions/metabolismo , Antiporters/genética , Antiporters/metabolismo , Encéfalo/metabolismo , Sinaptossomos/metabolismo , Sequência de Aminoácidos , Animais , Sulfonatos de Arila/metabolismo , Bumetanida/metabolismo , Cálcio/metabolismo , Carbonil Cianeto m-Clorofenil Hidrazona/metabolismo , Corantes Fluorescentes/metabolismo , Gluconatos/metabolismo , Concentração de Íons de Hidrogênio , Ionóforos/metabolismo , Ácido Niflúmico/metabolismo , Potássio/metabolismo , Ratos , Inibidores de Simportadores de Cloreto de Sódio e Potássio/metabolismo , Sulfatos/metabolismo
15.
J Exp Biol ; 207(Pt 21): 3707-16, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15371478

RESUMO

We examined the effects of bathing saline Na+/K+ ratio, bumetanide and hydrochlorothiazide on fluid and ion transport by serotonin-stimulated Malpighian tubules of Rhodnius prolixus. Previous pharmacological and electrophysiological studies indicate that a bumetanide-sensitive Na+/K+/2Cl- cotransporter is the primary route for basolateral ion entry into the cell during fluid secretion. The goal of this study was to resolve the apparent conflict between relatively high secretion rates by tubules bathed in K+-free saline and the evidence that Na+/K+/2Cl- cotransporters described in other systems have an absolute requirement for all three ions for translocation. Our measurements of fluid secretion rate, ion fluxes and electrophysiological responses to serotonin show that fluid secretion in K+-free saline is bumetanide sensitive and hydrochlorothiazide insensitive. Dose-response curves of secretion rate versus bumetanide concentration were identical for tubules bathed in K+-free and control saline with IC50 values of 2.6 x 10(-6) mmol l(-1) and 2.9 x 10(-6) mmol l(-1), respectively. Double-reciprocal plots of K+ flux versus bathing saline K+ concentration showed that increasing Na+ concentration in the bathing fluid increased Kt but had no effect on Jmax, consistent with competitive inhibition of K+ transport by Na+. We propose that the competition between Na+ and K+ for transport by the bumetanide-sensitive transporter is part of an autonomous mechanism by which Malpighian tubules regulate haemolymph K+ concentration.


Assuntos
Bumetanida/metabolismo , Túbulos de Malpighi/metabolismo , Potássio/metabolismo , Rhodnius/fisiologia , Simportadores de Cloreto de Sódio-Potássio/metabolismo , Sódio/metabolismo , Análise de Variância , Animais , Bumetanida/farmacologia , Hidroclorotiazida/metabolismo , Transporte de Íons/efeitos dos fármacos , Túbulos de Malpighi/efeitos dos fármacos , Potenciais da Membrana , Microeletrodos , Potássio/sangue , Rhodnius/metabolismo , Serotonina/farmacologia
16.
Pharmacotherapy ; 24(5): 630-7, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15162897

RESUMO

Food significantly affects the pharmacokinetics of oral loop diuretics in healthy individuals, but studies have not been performed in patients with edema. Because of this omission, food's effect on pharmacokinetics has been overlooked and may decrease the pharmacodynamic response in patients who rely on diuretics. Despite this potential interaction, reference manuals do not provide warnings about the effects of food on loop diuretic absorption. We reviewed the published human studies investigating the effects of food on loop diuretics. Peak plasma concentrations and urinary recovery were significantly decreased when taken with food, but only one study showed a corresponding decrease in total urine output, which is likely related to the diuretic threshold effect. These healthy subjects probably were always above the diuretic threshold under both fed and fasting conditions and thus could not augment their urine output. Based on these data in healthy subjects, the special implications for patients who routinely take diuretics are discussed. Therefore, food is more likely to have a clinical effect on the diuretic threshold given its effect in healthy subjects and the special considerations for patients with edema. Additional studies are needed to help answer these questions. Until such data are available, the most conservative, effective clinical approach is to administer oral loop diuretics without food.


Assuntos
Bumetanida , Diurese/efeitos dos fármacos , Diuréticos , Alimentos , Furosemida , Administração Oral , Área Sob a Curva , Bumetanida/metabolismo , Bumetanida/farmacocinética , Bumetanida/farmacologia , Ensaios Clínicos como Assunto , Diuréticos/metabolismo , Diuréticos/farmacocinética , Diuréticos/farmacologia , Furosemida/metabolismo , Furosemida/farmacocinética , Furosemida/farmacologia , Humanos , Absorção Intestinal , Sulfonamidas/metabolismo , Sulfonamidas/farmacocinética , Sulfonamidas/farmacologia , Torasemida
17.
Exp Physiol ; 87(4): 437-45, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12392107

RESUMO

We used the short-circuit current (I(sc)) and patch-clamp techniques to investigate the effects of methoxsalen (MTX) on the electrogenic Cl- secretion of the mouse jejunum. MTX stimulated a sustained increase in Isc that was dose dependent. Bumetanide inhibited MTX-stimulated Isc in a dose-dependent manner consistent with activation of Cl- secretion. MTX failed to stimulate I(sc) following maximal activation of the cAMP pathway by forskolin, but did increase Isc after a submaximal dose of forskolin. Glibenclamide, a blocker of the cystic fibrosis transmembrane conductance regulator (CFTR), reduced the MTX-stimulated increase of Isc by 59 +/- 6%. The cAMP-dependent K+ channel blocker 293B did not alter the MTX-activated I(sc); however, clotrimazole, an intermediate Ca2(+)-activated K+ channel (IK(Ca)) blocker, reduced the MTX-stimulated I(sc). MTX did not alter Na(+)-glucose cotransport across the mouse jejunum. In cell-attached membrane patches, MTX increased the open probability of the basolateral IK(Ca) channel of isolated crypts. These data suggest that the CFTR and IK(Ca) channels participate in the MTX-activated, sustained Cl- secretory response of the mouse jejunum.


Assuntos
Cloro/metabolismo , Mucosa Intestinal/metabolismo , Jejuno/metabolismo , Metoxaleno/farmacologia , Canais de Potássio/metabolismo , Animais , Bário/metabolismo , Bário/farmacologia , Bumetanida/administração & dosagem , Bumetanida/metabolismo , Colforsina/metabolismo , Colforsina/farmacologia , Relação Dose-Resposta a Droga , Glibureto/metabolismo , Glibureto/farmacologia , Técnicas In Vitro , Mucosa Intestinal/efeitos dos fármacos , Transporte de Íons/efeitos dos fármacos , Transporte de Íons/fisiologia , Jejuno/efeitos dos fármacos , Masculino , Metoxaleno/metabolismo , Camundongos , Técnicas de Patch-Clamp , Canais de Potássio/efeitos dos fármacos , Valores de Referência , Sensibilidade e Especificidade
18.
Am J Physiol Cell Physiol ; 280(3): C491-9, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11171568

RESUMO

Although Na(+)-K(+)-2Cl(-) cotransport has been demonstrated in cultured bovine corneal endothelial cells, its presence and role in the native tissue have been disputed. Using RT-PCR we have now identified a partial clone of the cotransporter protein in freshly dissected as well as in cultured corneal endothelial and epithelial cells. The deduced amino acid sequence of this protein segment is 99% identical to that of the bovine isoform (bNKCC1). [(3)H]bumetanide binding shows that the cotransporter sites are located in the basolateral membrane region at a density of 1.6 pmol/mg of protein, close to that in lung epithelium. Immunocytochemistry confirms the basolateral location of the cotransporter. We calculate the turnover rate of the cotransporter to be 83 s(-1). Transendothelial fluid transport, determined from deepithelialized rabbit corneal thickness measurements, is partially inhibited (30%) by bumetanide in a dose-dependent manner. Our results demonstrate that Na(+)-K(+)-2Cl(-) cotransporters are present in the basolateral domain of freshly dissected bovine corneal endothelial cells and contribute to fluid transport across corneal endothelial preparations.


Assuntos
Líquidos Corporais/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/fisiologia , Endotélio Corneano/metabolismo , Sequência de Aminoácidos/genética , Animais , Sequência de Bases/genética , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/fisiologia , Bumetanida/metabolismo , Bumetanida/farmacologia , Bovinos , Células Cultivadas , Clonagem Molecular , Córnea/anatomia & histologia , Córnea/efeitos dos fármacos , Córnea/metabolismo , Imuno-Histoquímica , Dados de Sequência Molecular , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Simportadores de Cloreto de Sódio-Potássio , Distribuição Tecidual
19.
Am J Physiol Cell Physiol ; 279(6): C1710-21, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11078685

RESUMO

In this study, we examined the Na(+)-K(+)-Cl(-) cotransporter activity and expression in rat cortical astrocyte differentiation. Astrocyte differentiation was induced by dibutyryl cAMP (DBcAMP, 0. 25 mM) for 7 days, and cells changed from a polygonal to process-bearing morphology. Basal activity of the cotransporter was significantly increased in DBcAMP-treated astrocytes (P < 0.05). Expression of an approximately 161-kDa cotransporter protein was increased by 91% in the DBcAMP-treated astrocytes. Moreover, the specific [(3)H]bumetanide binding was increased by 67% in the DBcAMP-treated astrocytes. Inhibition of protein synthesis by cyclohexamide (2-3 microgram/ml) significantly attenuated the DBcAMP-mediated upregulation of the cotransporter activity and expression. The Na(+)-K(+)-Cl(-) cotransporter in astrocytes has been suggested to play a role in K(+) uptake. In 75 mM extracellular K(+) concentration, the cotransporter-mediated K(+) influx was stimulated by 147% in nontreated cells and 79% in DBcAMP-treated cells (P < 0.05). To study whether this high K(+)-induced stimulation of the cotransporter is attributed to membrane depolarization and Ca(2+) influx, the role of the L-type voltage-dependent Ca(2+) channel was investigated. The high-K(+)-mediated stimulation of the cotransporter activity was abolished in the presence of either 0.5 or 1.0 microM of the L-type channel blocker nifedipine or Ca(2+)-free HEPES buffer. A rise in intracellular free Ca(2+) in astrocytes was observed in high K(+). These results provide the first evidence that the Na(+)-K(+)-Cl(-) cotransporter protein expression can be regulated selectively when intracellular cAMP is elevated. The study also demonstrates that the cotransporter in astrocytes is stimulated by high K(+) in a Ca(2+)-dependent manner.


Assuntos
Astrócitos/metabolismo , Bucladesina/farmacologia , Proteínas de Transporte/metabolismo , Potássio/farmacocinética , Animais , Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/fisiologia , Bumetanida/metabolismo , Bumetanida/farmacologia , Cálcio/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/metabolismo , Proteínas de Transporte/análise , Proteínas de Transporte/biossíntese , Células Cultivadas , Córtex Cerebral/citologia , Diuréticos/metabolismo , Diuréticos/farmacologia , Imunofluorescência , Nifedipino/farmacologia , Ratos , Ratos Sprague-Dawley , Simportadores de Cloreto de Sódio-Potássio , Trítio
20.
Hepatology ; 31(6): 1296-304, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10827155

RESUMO

We have cloned two complementary DNAs (cDNAs), RL-Mtx-1 and RL-Mtx-2, corresponding to the bile acid- sensitive methotrexate carrier from rat liver by direct full-length rapid amplification of cDNA ends polymerase chain reaction (RACE-PCR) using degenerated primers that were deduced from published sequences of tumor cell methotrexate transporters. When expressed in Xenopus laevis oocytes and cosM6 cells, both clones mediate methotrexate and bumetanide transport. RL-Mtx-1 consists of 2,445 bp with an open reading frame of 1,536 bp. The corresponding protein with 512 amino acids has a molecular weight of 58 kd. RL-Mtx-2 (2,654 bp) differs by an additional insert of 203 bp. This insert is located in frame at position 1,196 of the RL-Mtx-1 and contains the typical splice junction sites at the 5' and 3' end, indicating that the RL-Mtx-2 messenger RNA (mRNA) is generated by alternative splicing. The insert contains a stop codon that shortens the RL-Mtx-2 protein to 330 amino acids (38 kd). Both cDNAs contain the binding site sequence for the dioxin/nuclear translocator responsive element (Ah/Arnt-receptor) in conjunction with a barbiturate recognition sequence (Barbie box). Preliminary results show that the Barbie box acts as a negative regulatory element. The two liver cDNA clones show homologies to the published sequences of folate and the reduced folate carriers, but no homology is found to the transport systems for organic anions like the Ntcp1, oatp1, OAT-K1, and OAT1. Expression of the mRNA for the methotrexate carrier is found in liver, kidney, heart, brain, spleen, lung, and skeletal muscle, but not in the testis as revealed by Northern blot analysis. The highest abundance of the mRNA is found in the kidney.


Assuntos
Transportadores de Cassetes de Ligação de ATP , Ácidos e Sais Biliares/farmacologia , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Clonagem Molecular , Fígado/metabolismo , Proteínas de Membrana Transportadoras , Proteínas de Neoplasias , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Sequência de Aminoácidos/genética , Animais , Sequência de Bases/genética , Bumetanida/metabolismo , Células COS/metabolismo , Células Cultivadas , DNA Complementar/genética , Feminino , Fígado/citologia , Masculino , Metotrexato/metabolismo , Antígenos de Histocompatibilidade Menor , Dados de Sequência Molecular , Oócitos/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Proteína Carregadora de Folato Reduzido , Distribuição Tecidual , Xenopus laevis
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA