Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Neuropharmacology ; 205: 108910, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-34883135

RESUMEN

The Na-K-2Cl cotransporter NKCC1 and the neuron-specific K-Cl cotransporter KCC2 are considered attractive CNS drug targets because altered neuronal chloride regulation and consequent effects on GABAergic signaling have been implicated in numerous CNS disorders. While KCC2 modulators are not yet clinically available, the loop diuretic bumetanide has been used in clinical studies to treat brain disorders and as a tool for NKCC1 inhibition in preclinical models. Bumetanide is known to have anticonvulsant and neuroprotective effects under some pathophysiological conditions. However, as shown in several species from neonates to adults (mice, rats, dogs, and by extrapolation in humans), at the low clinical doses of bumetanide approved for diuresis, this drug has negligible access into the CNS, reaching levels that are much lower than what is needed to inhibit NKCC1 in cells within the brain parenchyma. Several drug discovery strategies have been used over the last ∼15 years to develop brain-permeant compounds that, ideally, should be selective for NKCC1 to eliminate the diuresis mediated by inhibition of renal NKCC2. The strategies employed to improve the pharmacokinetic and pharmacodynamic properties of NKCC1 blockers include evaluation of other clinically approved loop diuretics; development of lipophilic prodrugs of bumetanide; development of side-chain derivatives of bumetanide; and unbiased high-throughput screening approaches of drug discovery based on large chemical compound libraries. The main outcomes are that (1), non-acidic loop diuretics such as azosemide and torasemide may have advantages as NKCC1 inhibitors vs. bumetanide; (2), bumetanide prodrugs achieve significantly higher brain levels of the parent drug and have lower diuretic activity; (3), the novel bumetanide side-chain derivatives do not exhibit any functionally relevant improvement of CNS accessibility or NKCC1 selectivity vs. bumetanide; (4) novel compounds discovered by high-throughput screening may resolve some of the inherent problems of bumetanide, but as yet this has not been achieved. Thus, further research is needed to optimize the design of brain-permeant NKCC1 inhibitors. Another major challenge is to identify the mechanisms whereby various NKCC1-expressing cellular targets of these drug within (e.g., neurons, oligodendrocytes or astrocytes) and outside the brain parenchyma (e.g., blood-brain barrier, choroid plexus, endocrine and immune system), as well as molecular off-target effects, might contribute to their reported therapeutic and adverse effects.


Asunto(s)
Bumetanida/farmacocinética , Sistema Nervioso Central/efectos de los fármacos , Inhibidores del Simportador de Cloruro Sódico y Cloruro Potásico/farmacocinética , Miembro 2 de la Familia de Transportadores de Soluto 12/efectos de los fármacos , Animales , Humanos
2.
J Clin Neurosci ; 81: 477-484, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33222966

RESUMEN

BACKGROUND/OBJECTIVE: The current study evaluated the analgesic effects of bumetanide as an adjunctive treatment in managing neuropathic pain following spinal cord injury. The peripheral expression level of Na-K-Cl-cotransporter-1 (NKCC1) and K-Cl-cotransporter-2 (KCC2) genes in polymorphonuclear lymphocytes (PMLs) assessed as a possible biomarker indicating central underlying mechanisms. METHODS: This open-label, single-arm, pilot trial of bumetanide (2 mg/day) is an add-on treatment conducted in 14 SCI patients for 19 weeks. The whole duration consisted of three phases: pre-treatment (1 month), titration (3 weeks), and active treatment (4 months). Ultimately, nine patients completed the study. The primary outcome variables were the endpoint pain score measured by the numeric rating scale (NRS), and the short-form McGill Pain Questionnaire. Secondary endpoints included the Short-Form Health Survey that measures the quality of life. Blood samples were collected and used for determining the expression of NKCC1 and KCC2 genes in transcription and translation levels. RESULTS: Bumetanide treatment significantly reduced average pain intensity according to the NRS and the short form of the McGill Pain Questionnaire scores. The baseline expression of KCC2 protein was low between groups and increased significantly following treatment (P < 0.05). Through the current study, pain improvement accompanied by the more significant mean change from the baseline for the overall quality of life. CONCLUSION: These data might be a piece of preliminary evidence for the analgesic effect of bumetanide on neuropathic pain and could support the potential role of the upregulation of KCC2 protein and involvement of GABAergic disinhibition in producing neuropathic pain.


Asunto(s)
Bumetanida/uso terapéutico , Neuralgia/tratamiento farmacológico , Traumatismos de la Médula Espinal/complicaciones , Adulto , Femenino , Humanos , Masculino , Neuralgia/etiología , Proyectos Piloto , Calidad de Vida , Miembro 2 de la Familia de Transportadores de Soluto 12/biosíntesis , Miembro 2 de la Familia de Transportadores de Soluto 12/efectos de los fármacos , Simportadores/efectos de los fármacos , Simportadores/metabolismo
3.
Hear Res ; 361: 152-156, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29459166

RESUMEN

Specific pharmacological blockade of KCNQ (Kv7) channels with XE991 rapidly (within 20 min) and profoundly alters inner ear gravity receptor responses to head motion (Lee et al., 2017). We hypothesized that these effects were attributable to the suppression of K+ secretion following blockade of KCNQ1-KCNE1 channels in vestibular dark cells and marginal cells. To test this hypothesis, K+ secretion was independently inhibited by blocking the Na+-K+-2Cl- cotransporter (NKCC1, Slc12a2) rather than KCNQ1-KCNE1 channels. Acute blockade of NKCC1 with ethacrynic acid (40 mg/kg) eliminated auditory responses (ABRs) within approximately 70 min of injection, but had no effect on vestibular gravity receptor function (VsEPs) over a period of 2 h in the same animals. These findings show that, vestibular gravity receptors are highly resistant to acute disruption of endolymph secretion unlike the auditory system. Based on this we argue that acute suppression of K+ secretion alone does not likely account for the rapid profound effects of XE991 on gravity receptors. Instead the effects of XE991 likely require additional action at KCNQ channels located within the sensory epithelium itself.


Asunto(s)
Ácido Etacrínico/farmacología , Gravitación , Movimientos de la Cabeza , Canales de Potasio KCNQ/metabolismo , Potasio/metabolismo , Inhibidores del Simportador de Cloruro Sódico y Cloruro Potásico/farmacología , Miembro 2 de la Familia de Transportadores de Soluto 12/efectos de los fármacos , Vestíbulo del Laberinto/efectos de los fármacos , Animales , Antracenos/farmacología , Endolinfa/metabolismo , Potenciales Evocados Auditivos del Tronco Encefálico/efectos de los fármacos , Canales de Potasio KCNQ/antagonistas & inhibidores , Ratones Endogámicos C57BL , Bloqueadores de los Canales de Potasio/farmacología , Vías Secretoras , Miembro 2 de la Familia de Transportadores de Soluto 12/metabolismo , Factores de Tiempo , Vestíbulo del Laberinto/citología , Vestíbulo del Laberinto/metabolismo
4.
BMC Neurosci ; 18(1): 42, 2017 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-28490320

RESUMEN

BACKGROUND: Astaxanthin (ATX) is a carotenoid pigment with pleiotropic pharmacological properties that is seen as a possible drug for treating cerebral ischemic injury and subarachnoid hemorrhage. Na+-K+-2Cl- co-transporter-1 (NKCC1), an intrinsic membrane protein expressed by many cell types, is activated by various insults, leading to the formation of cell swelling and brain edema. We previously established that ATX attenuated brain edema and improved neurological outcomes by modulating NKCC1 expression after traumatic brain injury in mice. This paper explored the molecular mechanism of ATX-mediated inhibition of NKCC1 utilizing an in vitro astrocyte stretch injury model. RESULTS: Stretch injury in cultured astrocytes lowered cell viability time-dependently, which was substantially reducing by pretreating with ATX (50 µmol/L). Stretch injury increased Bax level and cleaved caspase-3 activity, and decreased Bcl-2 level and pro-caspase 3 activity, resulting in the apoptosis of astrocytes. Additionally, stretch injury substantially raised the gene and protein expressions of interleukin (IL)-1ß, IL-6, and tumor necrosis factor (TNF)-α and prompted the expression and nuclear translocation of NF-κB. Pretreatment with ATX remarkably prevented the trauma-induced initiation of NF-κB, expressions of pro-inflammatory cytokines, and cell apoptosis. Moreover, stretch injury markedly elevated the gene and protein expression of NKCC1, which was partly blocked by co-treatment with ATX (50 µmol/L) or an NF-κB inhibitor (PDTC, 10 µmol/L). Cleaved caspase-3 activity was partially reduced by PDTC (10 µmol/L) or an NKCC1 inhibitor (bumetanide, 50 µmol/L). CONCLUSIONS: ATX attenuates apoptosis after stretch injury in cultured astrocytes by inhibiting NKCC1 expression, and it acts by reducing the expression of NF-κB-mediated pro-inflammatory factors.


Asunto(s)
Astrocitos/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Miembro 2 de la Familia de Transportadores de Soluto 12/biosíntesis , Animales , Apoptosis/efectos de los fármacos , Astrocitos/patología , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , FN-kappa B/efectos de los fármacos , FN-kappa B/metabolismo , Transducción de Señal , Miembro 2 de la Familia de Transportadores de Soluto 12/efectos de los fármacos , Xantófilas/farmacología
5.
Ann Neurol ; 81(2): 251-265, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28074534

RESUMEN

OBJECTIVE: Rewiring of excitatory glutamatergic neuronal circuits is a major abnormality in epilepsy. Besides the rewiring of excitatory circuits, an abnormal depolarizing γ-aminobutyric acidergic (GABAergic) drive has been hypothesized to participate in the epileptogenic processes. However, a remaining clinically relevant question is whether early post-status epilepticus (SE) evoked chloride dysregulation is important for the remodeling of aberrant glutamatergic neuronal circuits. METHODS: Osmotic minipumps were used to infuse intracerebrally a specific inhibitor of depolarizing GABAergic transmission as well as a functionally blocking antibody toward the pan-neurotrophin receptor p75 (p75NTR ). The compounds were infused between 2 and 5 days after pilocarpine-induced SE. Immunohistochemistry for NKCC1, KCC2, and ectopic recurrent mossy fiber (rMF) sprouting as well as telemetric electroencephalographic and electrophysiological recordings were performed at day 5 and 2 months post-SE. RESULTS: Blockade of NKCC1 after SE with the specific inhibitor bumetanide restored NKCC1 and KCC2 expression, normalized chloride homeostasis, and significantly reduced the glutamatergic rMF sprouting within the dentate gyrus. This mechanism partially involves p75NTR signaling, as bumetanide application reduced SE-induced p75NTR expression and functional blockade of p75NTR decreased rMF sprouting. The early transient (3 days) post-SE infusion of bumetanide reduced rMF sprouting and recurrent seizures in the chronic epileptic phase. INTERPRETATION: Our findings show that early post-SE abnormal depolarizing GABA and p75NTR signaling fosters a long-lasting rearrangement of glutamatergic network that contributes to the epileptogenic process. This finding defines promising and novel targets to constrain reactive glutamatergic network rewiring in adult epilepsy. Ann Neurol 2017;81:251-265.


Asunto(s)
Bumetanida/farmacología , Fibras Musgosas del Hipocampo/efectos de los fármacos , Receptores de Factor de Crecimiento Nervioso/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Inhibidores del Simportador de Cloruro Sódico y Cloruro Potásico/farmacología , Miembro 2 de la Familia de Transportadores de Soluto 12/efectos de los fármacos , Estado Epiléptico/metabolismo , Simportadores/efectos de los fármacos , Ácido gamma-Aminobutírico/efectos de los fármacos , Animales , Bumetanida/administración & dosificación , Masculino , Proteínas del Tejido Nervioso , Ratas , Ratas Wistar , Receptores de Factores de Crecimiento , Inhibidores del Simportador de Cloruro Sódico y Cloruro Potásico/administración & dosificación , Estado Epiléptico/tratamiento farmacológico , Estado Epiléptico/fisiopatología , Cotransportadores de K Cl
6.
J Thorac Cardiovasc Surg ; 153(1): 206-215, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27986254

RESUMEN

OBJECTIVES: Ischemia-reperfusion acute lung injury is characterized by increased vascular permeability, lung edema, and neutrophil sequestration. Ischemia-reperfusion acute lung injury occurs in lung transplantation and other major surgical procedures. Effective regulation of alveolar fluid balance is critical for pulmonary edema. Sodium-potassium-chloride co-transporter regulates alveolar fluid and is associated with inflammation. We hypothesized that sodium-potassium-chloride co-transporter is important in ischemia-reperfusion acute lung injury. Bumetanide, a sodium-potassium-chloride co-transporter inhibitor, is used to treat pulmonary edema clinically. We studied the effect of bumetanide in ischemia-reperfusion acute lung injury. METHODS: Isolated perfusion of mouse lungs in situ was performed. The main pulmonary artery and left atrium were catheterized for lung perfusion and effluent collection for recirculation, respectively, with perfusate consisting of 1 mL blood and 9 mL physiologic solution. Ischemia-reperfusion was induced by 120 minutes of ischemia (no ventilation or perfusion) and reperfused for 60 minutes. Wild-type, SPAK knockout (SPAK-/-), and WNK4 knockin (WNK4D561A/+) mice were divided into control, ischemia-reperfusion, and ischemia-reperfusion + bumetanide groups (n = 6 per group). Bumetanide was administered via perfusate during reperfusion. Measurements were taken of lung wet/dry weight, microvascular permeability, histopathology, cytokine concentrations, and activity of the nuclear factor-κB pathway. RESULTS: In wild-type mice, ischemia-reperfusion caused lung edema (wet/dry weight 6.30 ± 0.36) and hyperpermeability (microvascular permeability, 0.29 ± 0.04), neutrophil sequestration (255.0 ± 55.8 cells/high-power field), increased proinflammatory cytokines, and nuclear factor-κB activation (1.33 ± 0.13). Acute lung injury was more severe in WNK4 mice with more lung edema, permeability, neutrophil sequestration, and nuclear factor-κB activation. Severity of acute lung injury was attenuated in SPAK-/-mice. Bumetanide decreased pulmonary edema (wild-type: wet/dry weight 5.05 ± 0.44, WNK4: wet/dry weight 5.13 ± 0.70), neutrophil sequestration (wild-type: 151.7 ± 27.8 cells/high-power field, WNK4: 135.3 ± 19.1 cells/high-power field), permeability (wild-type: 0.19 ± 0.01, WNK4: 0.21 ± 0.03), cytokines, and nuclear factor-κB activation after ischemia-reperfusion. CONCLUSIONS: Functional reduction of sodium-potassium-chloride co-transporter by genetic or pharmacologic treatment to inhibit sodium-potassium-chloride co-transporter resulted in lower severity of acute lung injury induced by ischemia-reperfusion. Sodium-potassium-chloride co-transporter may present a promising target for therapeutic interventions in a clinical setting.


Asunto(s)
Bumetanida/farmacología , Lesión Pulmonar/prevención & control , Daño por Reperfusión/complicaciones , Miembro 2 de la Familia de Transportadores de Soluto 12/efectos de los fármacos , Enfermedad Aguda , Animales , Permeabilidad Capilar , Modelos Animales de Enfermedad , Lesión Pulmonar/etiología , Lesión Pulmonar/fisiopatología , Masculino , Ratones , Presión Esfenoidal Pulmonar/efectos de los fármacos , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/fisiopatología , Inhibidores del Simportador de Cloruro Sódico y Cloruro Potásico/farmacología , Miembro 2 de la Familia de Transportadores de Soluto 12/metabolismo
7.
Neurotoxicol Teratol ; 60: 82-86, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27826117

RESUMEN

Prolonged ketamine exposure in neonates at anesthetic doses is known to cause long-term impairments of learning and memory. A current theoretical mechanism explains this phenomenon as being neuro-excitotoxicity mediated by compensatory upregulation of N-methyl-d-aspartate receptors (NMDARs), which then initiates widespread neuroapoptosis. Additionally, the excitatory behavior of GABAergic synaptic transmission mediated by GABAA receptors (GABAARs), occurring during the early neuronal development period, is proposed as contributing to the susceptibility of neonatal neurons to ketamine-induced injury. This is due to differential developmental expression patterns of Na+-K+-2Cl- co-transporter (NKCC1) and K+-Cl- co-transporter. Studies have shown that bumetanide, an NKCC1 inhibitor, allows neurons to become inhibitory rather than excitatory early in development. We thus hypothesized that bumetanide co-administration during ketamine treatment would reduce over excitation and protect the neurons from excitotoxicity. In this initial study, the Morris Water Maze test was used to assess the effects of co-administration of ketamine and bumetanide to neonatal Sprague-Dawley rats on long-term learning and memory changes seen later in life. It was revealed that bumetanide, when co-treated with ketamine neonatally, significantly impeded behavioral deficits typically seen in animals exposed to ketamine alone. Therefore, these findings suggest a new mechanism by which neonatal ketamine induced learning impairments can be prevented.


Asunto(s)
Bumetanida/farmacología , Ketamina/efectos adversos , Ketamina/antagonistas & inhibidores , Trastornos de la Memoria/prevención & control , Miembro 2 de la Familia de Transportadores de Soluto 12/efectos de los fármacos , Aprendizaje Espacial/efectos de los fármacos , Animales , Animales Recién Nacidos , Combinación de Medicamentos , Femenino , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Trastornos de la Memoria/inducido químicamente , Ratas
8.
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
9.
Eur J Pharmacol ; 771: 241-6, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26656758

RESUMEN

It has been demonstrated that the antidiuretic hormone vasopressin is able to regulate the expression of Na-K-Cl cotransporters (NKCC1 and NKCC2) in the kidney. The present study investigated the effects of long- and short-term administration of vasopressin on NKCC and the possible signaling mechanism of vasopressin in the mouse distal colon using the siRNA, real-time PCR, western blotting and Ussing chambers method. The results showed the presence of NKCC2 expression in the colon, which was verified with a siRNA technique. The mRNA and protein expression level of NKCC2 significantly increased by about 40% and 90% respectively in response to restricting water intake to 1ml/day/20g for 7 days. In contrast, the NKCC1 expression level was unchanged in the colon. To determine the short-term activation of NKCC2 by vasopressin in vitro, we found that the administration of vasopressin caused a 3-fold increase in mouse colon NKCC2 phosphorylation, which was detected with phosphospecific antibody R5. In addition, the Ussing chamber results showed that NKCC2, cAMP and Ca(2+) signaling pathway may be involved in the vasopressin-induced response. Further, adenylate cyclase inhibitor MDL-12330A and PKA inhibitor H89 and Ca(2+) chelator BAPTA-AM reversed the vasopressin induced NKCC2 phosphorylation level increase by about 35%, 28% and 42% respectively suggesting vasopressin stimulate NKCC2 phosphorylation increase mediated by cAMP-PKA and Ca(2+) signaling in the colon. Collectively, these data suggest that the expression and phosphorylation of NKCC2 are increased in the colon by vasopressin stimulation, in association with enhanced activity of the vasopressin/cAMP and Ca(2+) pathways.


Asunto(s)
Señalización del Calcio/efectos de los fármacos , Colon/metabolismo , Miembro 1 de la Familia de Transportadores de Soluto 12/biosíntesis , Miembro 2 de la Familia de Transportadores de Soluto 12/biosíntesis , Vasopresinas/farmacología , Animales , Quelantes/farmacología , Colon/efectos de los fármacos , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Iminas/farmacología , Isoquinolinas/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación , ARN Interferente Pequeño , Miembro 1 de la Familia de Transportadores de Soluto 12/efectos de los fármacos , Miembro 2 de la Familia de Transportadores de Soluto 12/efectos de los fármacos , Sulfonamidas/farmacología , Privación de Agua
10.
Nat Rev Gastroenterol Hepatol ; 12(8): 446-57, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26122478

RESUMEN

Diarrhoeal disease remains a major health burden worldwide. Secretory diarrhoeas are caused by certain bacterial and viral infections, inflammatory processes, drugs and genetic disorders. Fluid secretion across the intestinal epithelium in secretory diarrhoeas involves multiple ion and solute transporters, as well as activation of cyclic nucleotide and Ca(2+) signalling pathways. In many secretory diarrhoeas, activation of Cl(-) channels in the apical membrane of enterocytes, including the cystic fibrosis transmembrane conductance regulator and Ca(2+)-activated Cl(-) channels, increases fluid secretion, while inhibition of Na(+) transport reduces fluid absorption. Current treatment of diarrhoea includes replacement of fluid and electrolyte losses using oral rehydration solutions, and drugs targeting intestinal motility or fluid secretion. Therapeutics in the development pipeline target intestinal ion channels and transporters, regulatory proteins and cell surface receptors. This Review describes pathogenic mechanisms of secretory diarrhoea, current and emerging therapeutics, and the challenges in developing antidiarrhoeal therapeutics.


Asunto(s)
Diarrea/terapia , Antidiarreicos/uso terapéutico , Infecciones Bacterianas , Canales de Cloruro/antagonistas & inhibidores , Regulador de Conductancia de Transmembrana de Fibrosis Quística/antagonistas & inhibidores , Diarrea/etiología , Gastroenteritis/complicaciones , Motilidad Gastrointestinal/efectos de los fármacos , Humanos , Absorción Intestinal/fisiología , Lisofosfolípidos/uso terapéutico , Terapia Molecular Dirigida/métodos , Bloqueadores de los Canales de Potasio/uso terapéutico , Probióticos/uso terapéutico , Receptores Sensibles al Calcio/agonistas , Intercambiador 3 de Sodio-Hidrógeno , Intercambiadores de Sodio-Hidrógeno/agonistas , Miembro 2 de la Familia de Transportadores de Soluto 12/efectos de los fármacos , Virosis
11.
World J Gastroenterol ; 20(22): 6844-59, 2014 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-24944475

RESUMEN

AIM: To investigate the role of Na(+)/K(+)/2Cl(-) cotransporter 1 (NKCC1) in the regulation of genes involved in cell cycle progression and the clinicopathological significance of its expression in esophageal squamous cell carcinoma (ESCC). METHODS: An immunohistochemical analysis was performed on 68 primary tumor samples obtained from ESCC patients that underwent esophagectomy. NKCC1 expression in human ESCC cell lines was analyzed by Western blotting. Knockdown experiments were conducted using NKCC1 small interfering RNA, and the effects on cell cycle progression were analyzed. The gene expression profiles of cells were analyzed by microarray analysis. RESULTS: Immunohistochemical staining showed that NKCC1 was primarily found in the cytoplasm of carcinoma cells and that its expression was related to the histological degree of differentiation of SCC. NKCC1 was highly expressed in KYSE170 cells. Depletion of NKCC1 in these cells inhibited cell proliferation via G2/M phase arrest. Microarray analysis identified 2527 genes with altered expression levels in NKCC1depleted KYSE170. Pathway analysis showed that the top-ranked canonical pathway was the G2/M DNA damage checkpoint regulation pathway, which involves MAD2L1, DTL, BLM, CDC20, BRCA1, and E2F5. CONCLUSION: These results suggest that the expression of NKCC1 in ESCC may affect the G2/M checkpoint and may be related to the degree of histological differentiation of SCCs. We have provided a deeper understanding of the role of NKCC1 as a mediator and/or a biomarker in ESCC.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma de Células Escamosas/metabolismo , Neoplasias Esofágicas/metabolismo , Puntos de Control de la Fase G2 del Ciclo Celular , Miembro 2 de la Familia de Transportadores de Soluto 12/metabolismo , Anciano , Biomarcadores de Tumor/genética , Western Blotting , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago , Femenino , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Interferencia de ARN , Transducción de Señal , Inhibidores del Simportador de Cloruro Sódico y Cloruro Potásico/farmacología , Miembro 2 de la Familia de Transportadores de Soluto 12/efectos de los fármacos , Miembro 2 de la Familia de Transportadores de Soluto 12/genética , Transfección
12.
Epilepsia ; 55(6): 806-18, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24802699

RESUMEN

Seizures are a common manifestation of acute neurologic insults in neonates and are often resistant to the standard antiepileptic drugs that are efficacious in children and adults. The paucity of evidence-based treatment guidelines, coupled with a rudimentary understanding of disease pathogenesis, has made the current treatment of neonatal seizures empiric and often ineffective, highlighting the need for novel therapies. Key developmental differences in γ-aminobutyric acid (GABA)ergic neurotransmission between the immature and mature brain, and trauma-induced alterations in the function of the cation-chloride cotransporters (CCCs) NKCC1 and KCC2, probably contribute to the poor efficacy of standard antiepileptic drugs used in the treatment of neonatal seizures. Although CCCs are attractive drug targets, bumetanide and other existing CCC inhibitors are suboptimal because of pharmacokinetic constraints and lack of target specificity. Newer approaches including isoform-specific NKCC1 inhibitors with increased central nervous system penetration, and direct and indirect strategies to enhance KCC2-mediated neuronal chloride extrusion, might allow therapeutic modulation of the GABAergic system for neonatal seizure treatment. A PowerPoint slide summarizing this article is available for download in the Supporting Information section here.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Enfermedades del Recién Nacido/tratamiento farmacológico , Convulsiones/tratamiento farmacológico , Simportadores de Cloruro de Sodio-Potasio/efectos de los fármacos , Encéfalo/efectos de los fármacos , Bumetanida/uso terapéutico , Humanos , Recién Nacido , Miembro 2 de la Familia de Transportadores de Soluto 12/efectos de los fármacos , Simportadores/efectos de los fármacos , Cotransportadores de K Cl
13.
Am J Physiol Renal Physiol ; 306(10): F1155-60, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24623142

RESUMEN

Water-handling epithelia are sensitive to the osmotic environment. In this study, the effects of a hypo-osmotic challenge on carbachol (CCh)-induced fluid secretion was investigated using an ex vivo submandibular gland perfusion technique and intracellular pH and Ca(2+) measurements. The osmolality of the perfusion solution was altered to examine the response of the gland to a hypotonic challenge. The flow rate was increased by 34% with a 30% hypotonic solution (225 mosmol/kgH2O), although the Ca(2+) response was unchanged. The lowering of the external Cl(-) by 50% abolished this increase in the 30% hypotonic solution. Furthermore, bumetanide, an inhibitor of the Na(+)-K(+)-2Cl(-) cotransporter (NKCC1), completely inhibited the fluid secretion increase caused by the 30% hypotonic solution, and both the total amount of fluid and the flow rate were identical to those of the isotonic solution. This finding was confirmed by measuring the NKCC1 bumetanide-dependent NH4 (+) transport; Na(+)-K(+)-2Cl(-) transport was upregulated >40% by a 30% hypotonic challenge. Therefore, the increase in CCh-induced fluid secretion in response to hypotonic conditions can be attributed, to a large extent, to the specific activation of the NKCC1.


Asunto(s)
Líquidos Corporales/metabolismo , Presión Osmótica/fisiología , Miembro 2 de la Familia de Transportadores de Soluto 12/metabolismo , Glándula Submandibular/metabolismo , Animales , Bumetanida/farmacología , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Carbacol/farmacología , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Concentración Osmolar , Inhibidores del Simportador de Cloruro Sódico y Cloruro Potásico/farmacología , Miembro 2 de la Familia de Transportadores de Soluto 12/efectos de los fármacos
14.
Anesthesiology ; 119(5): 1096-108, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23788048

RESUMEN

BACKGROUND: It has been shown that γ-aminobutyric acid exerts excitatory actions on the immature brain due to the increased expression of Na(+)-K(+)-2Cl(-) cotransporter isoform 1. The authors sought to clarify whether midazolam, a γ-aminobutyric acid-mimetic hypnotic agent, causes neuronal excitation that can be blocked by bumetanide, a selective inhibitor of Na(+)-K(+)-2Cl(-) cotransporter isoform 1. Furthermore, the authors examined whether bumetanide potentiates the sedative effects of midazolam in neonatal rats. METHODS: The authors measured the effects of midazolam with or without bumetanide on the cytosolic Ca(2+) concentration ([Ca](2+)(i)) in hippocampal slices (n=3 in each condition) from rats at postnatal days 4, 7, and 28 (P4, P7, and P28) using fura-2 microfluorometry. Neuronal activity in the hippocampus and thalamus after intraperitoneal administration of midazolam with or without bumetanide was estimated by immunostaining of phosphorylated cyclic adenosine monophosphate-response element-binding protein (n=12 in each condition). Furthermore, the authors assessed effects of bumetanide on the sedative effect of midazolam by measuring righting reflex latency (n=6 in each condition). RESULTS: Midazolam significantly increased [Ca](2+)(i) in the CA3 area at P4 and P7 but not at P28. Bumetanide inhibited midazolam-induced increase in [Ca](2+)(i). Midazolam significantly up-regulated phosphorylated cyclic adenosine monophosphate-response element-binding protein expression in a bumetanide-sensitive manner in the hippocampus at P7 but not P28. Bumetanide enhanced the sedative effects of midazolam in P4 and P7 but not P28 rats. CONCLUSION: These results suggest that γ-aminobutyric acid A receptor-mediated excitation plays an important role in attenuated sedative effects of midazolam in immature rats.


Asunto(s)
Bumetanida/farmacología , Diuréticos/farmacología , Antagonistas del GABA , Hipnóticos y Sedantes/farmacología , Midazolam/farmacología , Miembro 2 de la Familia de Transportadores de Soluto 12/efectos de los fármacos , Ácido gamma-Aminobutírico/fisiología , Envejecimiento/fisiología , Animales , Animales Recién Nacidos , Región CA3 Hipocampal/metabolismo , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Sinergismo Farmacológico , Ratas , Ratas Sprague-Dawley , Reflejo/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA