Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
Am J Physiol Renal Physiol ; 326(6): F957-F970, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38601986

RESUMEN

Stretch-activated two-pore domain K+ (K2P) channels play important roles in many visceral organs, including the urinary bladder. The TWIK-related K+ channel TREK-1 is the predominantly expressed K2P channel in the urinary bladder of humans and rodents. Downregulation of TREK-1 channels was observed in the urinary bladder of patients with detrusor overactivity, suggesting their involvement in the pathogenesis of voiding dysfunction. This study aimed to characterize the long-term effects of TREK-1 on bladder function with global and smooth muscle-specific TREK-1 knockout (KO) mice. Bladder morphology, bladder smooth muscle (BSM) contractility, and voiding patterns were evaluated up to 12 mo of age. Both sexes were included in this study to probe the potential sex differences. Smooth muscle-specific TREK-1 KO mice were used to distinguish the effects of TREK-1 downregulation in BSM from the neural pathways involved in the control of bladder contraction and relaxation. TREK-1 KO mice developed enlarged urinary bladders (by 60.0% for males and by 45.1% for females at 6 mo; P < 0.001 compared with the age-matched control group) and had a significantly increased bladder capacity (by 137.7% at 12 mo; P < 0.0001) and compliance (by 73.4% at 12 mo; P < 0.0001). Bladder strips isolated from TREK-1 KO mice exhibited decreased contractility (peak force after KCl at 6 mo was 1.6 ± 0.7 N/g compared with 3.4 ± 2.0 N/g in the control group; P = 0.0005). The lack of TREK-1 channels exclusively in BSM did not replicate the bladder phenotype observed in TREK-1 KO mice, suggesting a strong neurogenic origin of TREK-1-related bladder dysfunction.NEW & NOTEWORTHY This study compared voiding function and bladder phenotypes in global and smooth muscle-specific TREK-1 KO mice. We found significant age-related changes in bladder contractility, suggesting that the lack of TREK-1 channel activity might contribute to age-related changes in bladder smooth muscle physiology.


Asunto(s)
Hipertrofia , Ratones Noqueados , Contracción Muscular , Músculo Liso , Canales de Potasio de Dominio Poro en Tándem , Vejiga Urinaria , Animales , Canales de Potasio de Dominio Poro en Tándem/genética , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Canales de Potasio de Dominio Poro en Tándem/deficiencia , Vejiga Urinaria/fisiopatología , Vejiga Urinaria/metabolismo , Vejiga Urinaria/patología , Músculo Liso/metabolismo , Músculo Liso/fisiopatología , Músculo Liso/patología , Masculino , Femenino , Envejecimiento/metabolismo , Ratones , Ratones Endogámicos C57BL , Factores de Edad , Micción
2.
BMC Urol ; 19(1): 40, 2019 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-31113422

RESUMEN

BACKGROUND: Previously published results from our laboratory identified a mechano-gated two-pore domain potassium channel, TREK-1, as a main mechanosensor in the smooth muscle of the human urinary bladder. One of the limitations of in vitro experiments on isolated human detrusor included inability to evaluate in vivo effects of TREK-1 on voiding function, as the channel is also expressed in the nervous system, and may modulate micturition via neural pathways. Therefore, in the present study, we aimed to assess the role of TREK-1 channel in bladder function and voiding patterns in vivo by using TREK-1 knockout (KO) mice. METHODS: Adult C57BL/6 J wild-type (WT, N = 32) and TREK-1 KO (N = 33) mice were used in this study. The overall phenotype and bladder function were evaluated by gene and protein expression of TREK-1 channel, in vitro contractile experiments using detrusor strips in response to stretch and pharmacological stimuli, and cystometry in unanesthetized animals. RESULTS: TREK-1 KO animals had an elevated basal muscle tone and enhanced spontaneous activity in the detrusor without detectable changes in bladder morphology/histology. Stretch applied to isolated detrusor strips increased the amplitude of spontaneous contractions by 109% in the TREK-1 KO group in contrast to a 61% increase in WT mice (p ≤ 0.05 to respective baseline for each group). The detrusor strips from TREK-1 KO mice also generated more contractile force in response to electric field stimulation and high potassium concentration in comparison to WT group (p ≤ 0.05 for both tests). However, cystometric recordings from TREK-1 KO mice revealed a significant increase in the duration of the intermicturition interval, enhanced bladder capacity and increased number of non-voiding contractions in comparison to WT mice. CONCLUSIONS: Our results provide evidence that global down-regulation of TREK-1 channels has dual effects on detrusor contractility and micturition patterns in vivo. The observed differences are likely due to expression of TREK-1 channel not only in detrusor myocytes but also in afferent and efferent neural pathways involved in regulation of micturition which may underly the "mixed" voiding phenotype in TREK-1 KO mice.


Asunto(s)
Contracción Muscular/fisiología , Canales de Potasio de Dominio Poro en Tándem/deficiencia , Vejiga Urinaria/fisiología , Micción/fisiología , Animales , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
3.
Neuropharmacology ; 144: 29-36, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30332606

RESUMEN

Alcohol excitation of the ventral tegmental area (VTA) is important in neurobiological processes related to the development of alcoholism. The ionotropic receptors on VTA neurons that mediate ethanol-induced excitation have not been identified. Quinidine blocks ethanol excitation of VTA neurons, and blockade of two-pore potassium channels is among the actions of quinidine. Therefore two-pore potassium channels in the VTA may be potential targets for the action of ethanol. Here, we explored whether ethanol activation of VTA neurons is mediated by the two-pore potassium channel KCNK13. Extracellular recordings of the response of VTA neurons to ethanol were performed in combination with knockdown of Kcnk13 using a short hairpin RNA (shRNA) in C57BL/6 J mice. Real-time PCR and immunohistochemistry were used to examine expression of this channel in the VTA. Finally, the role of KCNK13 in binge-like drinking was examined in the drinking in the dark test after knockdown of the channel. Kcnk13 expression in the VTA was increased by acute ethanol exposure. Ethanol-induced excitation of VTA neurons was selectively reduced by shRNA targeting Kcnk13. Importantly, knockdown of Kcnk13 in the VTA resulted in increased alcohol drinking. These results are consistent with the idea that ethanol stimulates VTA neurons at least in part by inhibiting KCNK13, a specific two-pore potassium channel, and that KCNK13 can control both VTA neuronal activity and binge drinking. KCNK13 is a novel alcohol-sensitive molecular target and may be amenable to the development of pharmacotherapies for alcoholism treatment.


Asunto(s)
Potenciales de Acción/efectos de los fármacos , Consumo Excesivo de Bebidas Alcohólicas/metabolismo , Depresores del Sistema Nervioso Central/farmacología , Etanol/farmacología , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Área Tegmental Ventral/efectos de los fármacos , Potenciales de Acción/fisiología , Animales , Consumo Excesivo de Bebidas Alcohólicas/patología , Dopamina/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Canales de Potasio de Dominio Poro en Tándem/deficiencia , Canales de Potasio de Dominio Poro en Tándem/genética , ARN Mensajero/metabolismo , Técnicas de Cultivo de Tejidos , Área Tegmental Ventral/metabolismo , Área Tegmental Ventral/patología
4.
Immunity ; 49(1): 56-65.e4, 2018 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-29958799

RESUMEN

Potassium (K+) efflux across the plasma membrane is thought to be an essential mechanism for ATP-induced NLRP3 inflammasome activation, yet the identity of the efflux channel has remained elusive. Here we identified the two-pore domain K+ channel (K2P) TWIK2 as the K+ efflux channel triggering NLRP3 inflammasome activation. Deletion of Kcnk6 (encoding TWIK2) prevented NLRP3 activation in macrophages and suppressed sepsis-induced lung inflammation. Adoptive transfer of Kcnk6-/- macrophages into mouse airways after macrophage depletion also prevented inflammatory lung injury. The K+ efflux channel TWIK2 in macrophages has a fundamental role in activating the NLRP3 inflammasome and consequently mediates inflammation, pointing to TWIK2 as a potential target for anti-inflammatory therapies.


Asunto(s)
Inflamasomas/metabolismo , Inflamación/fisiopatología , Macrófagos/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Adenosina Trifosfato/metabolismo , Adenosina Trifosfato/farmacología , Animales , Caspasa 1/deficiencia , Caspasa 1/metabolismo , Línea Celular , Inflamasomas/efectos de los fármacos , Interleucina-1beta/metabolismo , Lipopolisacáridos/farmacología , Lesión Pulmonar/metabolismo , Lesión Pulmonar/fisiopatología , Macrófagos/trasplante , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/deficiencia , Canales de Potasio/efectos de los fármacos , Canales de Potasio/metabolismo , Canales de Potasio de Dominio Poro en Tándem/antagonistas & inhibidores , Canales de Potasio de Dominio Poro en Tándem/deficiencia , Quinina/farmacología , ARN Interferente Pequeño/farmacología , Receptores Purinérgicos P2X7/deficiencia , Receptores Purinérgicos P2X7/metabolismo , Sepsis/metabolismo , Sepsis/fisiopatología , Transducción de Señal/efectos de los fármacos
5.
Neuron ; 97(2): 299-312.e6, 2018 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-29290552

RESUMEN

Microglia exhibit two modes of motility: they constantly extend and retract their processes to survey the brain, but they also send out targeted processes to envelop sites of tissue damage. We now show that these motility modes differ mechanistically. We identify the two-pore domain channel THIK-1 as the main K+ channel expressed in microglia in situ. THIK-1 is tonically active, and its activity is potentiated by P2Y12 receptors. Inhibiting THIK-1 function pharmacologically or by gene knockout depolarizes microglia, which decreases microglial ramification and thus reduces surveillance, whereas blocking P2Y12 receptors does not affect membrane potential, ramification, or surveillance. In contrast, process outgrowth to damaged tissue requires P2Y12 receptor activation but is unaffected by blocking THIK-1. Block of THIK-1 function also inhibits release of the pro-inflammatory cytokine interleukin-1ß from activated microglia, consistent with K+ loss being needed for inflammasome assembly. Thus, microglial immune surveillance and cytokine release require THIK-1 channel activity.


Asunto(s)
Interleucina-1beta/fisiología , Microglía/fisiología , Canales de Potasio de Dominio Poro en Tándem/fisiología , Adenosina Trifosfato/farmacología , Animales , Movimiento Celular , Polaridad Celular , Forma de la Célula , Extensiones de la Superficie Celular/fisiología , Quimiotaxis/fisiología , Inflamasomas/metabolismo , Potenciales de la Membrana , Ratones , Ratones Noqueados , Microglía/efectos de los fármacos , Potasio/fisiología , Canales de Potasio de Dominio Poro en Tándem/antagonistas & inhibidores , Canales de Potasio de Dominio Poro en Tándem/deficiencia , Ratas , Ratas Sprague-Dawley , Receptores Purinérgicos P2Y12/fisiología , Transcriptoma
6.
Endocrinology ; 158(12): 4129-4138, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29069360

RESUMEN

Primary aldosteronism (PA) is a common form of endocrine hypertension that is characterized by the excessive production of aldosterone relative to suppressed plasma renin levels. PA is usually caused by either a unilateral aldosterone-producing adenoma or bilateral adrenal hyperplasia. Somatic mutations have been identified in several genes that encode ion pumps and channels that may explain the aldosterone excess in over half of aldosterone-producing adenomas, whereas the pathophysiology of bilateral adrenal hyperplasia is largely unknown. A number of mouse models of hyperaldosteronism have been described that recreate some features of the human disorder, although none replicate the genetic basis of human PA. Animal models that reproduce the genotype-phenotype associations of human PA are required to establish the functional mechanisms that underlie the endocrine autonomy and deregulated cell growth of the affected adrenal and for preclinical studies of novel therapeutics. Herein, we discuss the differences in adrenal physiology across species and describe the genetically modified mouse models of PA that have been developed to date.


Asunto(s)
Glándulas Suprarrenales/fisiología , Glándulas Suprarrenales/fisiopatología , Modelos Animales de Enfermedad , Hiperaldosteronismo/fisiopatología , Proteína de la Poliposis Adenomatosa del Colon/deficiencia , Proteína de la Poliposis Adenomatosa del Colon/genética , Glándulas Suprarrenales/metabolismo , Animales , Criptocromos/deficiencia , Criptocromos/genética , Humanos , Hiperaldosteronismo/genética , Hiperaldosteronismo/metabolismo , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/deficiencia , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/genética , Ratones Noqueados , Ratones Transgénicos , Canales de Potasio/deficiencia , Canales de Potasio/genética , Canales de Potasio de Dominio Poro en Tándem/deficiencia , Canales de Potasio de Dominio Poro en Tándem/genética , Especificidad de la Especie
7.
Am J Physiol Lung Cell Mol Physiol ; 313(6): L1030-L1046, 2017 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-28839101

RESUMEN

We previously proposed a role for the two-pore domain potassium (K2P) channel TREK-1 in hyperoxia (HO)-induced lung injury. To determine whether redundancy among the three TREK isoforms (TREK-1, TREK-2, and TRAAK) could protect from HO-induced injury, we now examined the effect of deletion of all three TREK isoforms in a clinically relevant scenario of prolonged HO exposure and mechanical ventilation (MV). We exposed WT and TREK-1/TREK-2/TRAAK-deficient [triple knockout (KO)] mice to either room air, 72-h HO, MV [high and low tidal volume (TV)], or a combination of HO + MV and measured quasistatic lung compliance, bronchoalveolar lavage (BAL) protein concentration, histologic lung injury scores (LIS), cellular apoptosis, and cytokine levels. We determined surfactant gene and protein expression and attempted to prevent HO-induced lung injury by prophylactically administering an exogenous surfactant (Curosurf). HO treatment increased lung injury in triple KO but not WT mice, including an elevated LIS, BAL protein concentration, and markers of apoptosis, decreased lung compliance, and a more proinflammatory cytokine phenotype. MV alone had no effect on lung injury markers. Exposure to HO + MV (low TV) further decreased lung compliance in triple KO but not WT mice, and HO + MV (high TV) was lethal for triple KO mice. In triple KO mice, the HO-induced lung injury was associated with decreased surfactant protein (SP) A and SPC but not SPB and SPD expression. However, these changes could not be explained by alterations in the transcription factors nuclear factor-1 (NF-1), NKX2.1/thyroid transcription factor-1 (TTF-1) or c-jun, or lamellar body levels. Prophylactic Curosurf administration did not improve lung injury scores or compliance in triple KO mice.


Asunto(s)
Hiperoxia/metabolismo , Lesión Pulmonar/metabolismo , Canales de Potasio de Dominio Poro en Tándem/deficiencia , Canales de Potasio/deficiencia , Proteínas Asociadas a Surfactante Pulmonar/biosíntesis , Animales , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Hiperoxia/genética , Hiperoxia/patología , Lipopolisacáridos/toxicidad , Lesión Pulmonar/genética , Lesión Pulmonar/patología , Ratones , Ratones Noqueados , Proteínas Asociadas a Surfactante Pulmonar/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
8.
Respir Physiol Neurobiol ; 244: 17-25, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28673876

RESUMEN

Despite intensive research, the exact function of TASK potassium channels in central and peripheral chemoreception is still under debate. In this study, we investigated the respiration of unrestrained TASK-3 (TASK-3-/-) and TASK-1/TASK-3 double knockout (TASK-1/3-/-) adult male mice in vivo using a plethysmographic device. Ventilation parameters of TASK-3-/- mice were normal under control condition (21% O2) and upon hypoxia and hypercapnia they displayed the physiological increase of ventilation. TASK-1/3-/- mice showed increased ventilation under control conditions. This increase of ventilation was caused by increased tidal volumes (VT), a phenomenon similarly observed in TASK-1-/- mice. Under acute hypoxia, TASK-1/3-/- mice displayed the physiological increase of the minute volume. Interestingly, this increase was not related to an increase of the respiratory frequency (fR), as observed in wild-type mice, but was caused by a strong increase of VT. This particular respiratory phenotype is reminiscent of the respiratory phenotype of carotid body-denervated rodents in the compensated state. Acute hypercapnia (5% CO2) stimulated ventilation in TASK-1/3-/- and wild-type mice to a similar extent; however, at higher CO2 concentrations (>5% CO2) the stimulation of ventilation was more pronounced in TASK-1/3-/- mice. At hyperoxia (100% O2), TASK-1-/-, TASK-3-/- and wild-type mice showed the physiological small decrease of ventilation. In sharp contrast, TASK-1/3-/- mice exhibited an abnormal increase of ventilation under hyperoxia. In summary, these measurements showed a grossly normal respiration of TASK-3-/- mice and a respiratory phenotype of TASK-1/3-/- mice that was characterized by a markedly enhanced tidal volume, similar to the one observed in TASK-1-/- mice. The abnormal hyperoxia response, exclusively found in TASK-1/3-/- double mutant mice, indicates that both TASK-1 and TASK-3 are essential for the hyperoxia-induced hypoventilation. The peculiar respiratory phenotype of TASK-1/3 knockout mice is reminiscent of the respiration of animals with long-term carotid body dysfunction. Taken together, TASK-1 and TASK-3 appear to serve specific and distinct roles in the complex processes underlying chemoreception and respiratory control.


Asunto(s)
Hiperoxia/metabolismo , Proteínas del Tejido Nervioso/deficiencia , Canales de Potasio de Dominio Poro en Tándem/deficiencia , Canales de Potasio/deficiencia , Respiración , Animales , Dióxido de Carbono/metabolismo , Femenino , Hipercapnia/metabolismo , Hipoxia/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Fenotipo , Pletismografía Total , Canales de Potasio/genética , Canales de Potasio de Dominio Poro en Tándem/genética , Volumen de Ventilación Pulmonar/fisiología
9.
J Neurochem ; 141(2): 236-246, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28192611

RESUMEN

Spinal cord injury (SCI) involves complex pathological process which can be complicated by secondary injury. TREK-1 is a member of the two-pore domain potassium (K2P) channel family, which can be modulated by a number of physiological and pathological stimuli. Recent studies suggest that TREK-1 plays an active role in depression, pain and neuroprotection. However, its role in the pathological process after SCI remains unclear. In this study, we tested the expression and function of TREK-1 in spinal cord of mice after traumatic SCI. TREK-1 was widely expressed in mice spinal cord, including astrocytes and neurons. Deficiency of TREK-1 significantly exacerbated focal inflammatory responses as indicated by the increased accumulation of microglia/macrophage as well as pro-inflammatory factor interleukin-1 beta (IL-1ß) and tumor necrosis factor alpha expression. Meanwhile, TREK-1 knockout mice showed enhanced reactive astrogliosis, chondroitin sulphate proteoglycans (CSPGs) production and decreased glutamate transporter-1 expression compared to the wide-type mice after SCI. Furthermore, TREK-1 deficiency promoted neurons and oligodendrocytes apoptosis, aggravated demyelination, cavity formation and retarded motor recovery. In summary, our findings provide the first in vivo evidence suggesting that TREK-1 may thereby constitute a promising therapeutic target to treat acute SCI.


Asunto(s)
Enfermedades Desmielinizantes/metabolismo , Enfermedades Desmielinizantes/patología , Canales de Potasio de Dominio Poro en Tándem/deficiencia , Recuperación de la Función/fisiología , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/patología , Animales , Apoptosis/fisiología , Femenino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Distribución Aleatoria
10.
Respir Physiol Neurobiol ; 245: 13-28, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-27838333

RESUMEN

TASK-1 potassium channels have been implicated in central and peripheral chemoreception; however, the precise contribution of TASK-1 for the control of respiration is still under debate. Here, we investigated the respiration of unrestrained adult and neonatal TASK-1 knockout mice (TASK-1-/-) using a plethysmographic device. Respiration in adult female TASK-1-/- mice under control (21% O2), hypoxia and hypercapnia was unaffected. Under acute hypoxia male TASK-1-/- mice exhibited a reduced increase of the respiratory frequency (fR) compared to wildtypes. However, the tidal volume (VT) of male TASK-1-/- mice was strongly enhanced. The volatile anesthetic isoflurane induced in male TASK-1-/- and male wild type mice (TASK-1+/+) a similar respiratory depression. Neonatal TASK-1-/- mice demonstrated a 30-40% decrease of the minute volume, caused by a reduction of the fR under control condition (21% O2). Under hypoxia, neonatal TASK-1-/- mice more frequently stopped breathing (apnea>3s) suggesting an increased hypoxia-sensitivity. As reported before, this increased hypoxia sensitivity had no influence on the survival rate of neonatal TASK-1-/- mice. In adult and neonatal mice, TASK-1 gene deletion induced a significant prolongation of the relaxation time (RT), which is a parameter for expiration kinetics. Additionally, screening for mutations in the human TASK-1 gene in 155 cases of sudden infant death syndrome (SIDS) was inconclusive. In conclusion, these data are suggestive for an increased hypoxia-sensitivity of neonatal TASK-1-/- mice, however, without causing an increase in neonatal lethality. In adult female TASK-1-/- mice respiration was unaffected, whereas adult male TASK-1-/- mice showed a modified breathing pattern. These results are suggestive for sex-specific mechanisms for compensating the inactivation of TASK-1 in mice.


Asunto(s)
Proteínas del Tejido Nervioso/deficiencia , Canales de Potasio de Dominio Poro en Tándem/deficiencia , Respiración , Caracteres Sexuales , Envejecimiento/metabolismo , Anestésicos por Inhalación/farmacología , Animales , Animales Recién Nacidos , Estudios de Cohortes , Femenino , Humanos , Hipercapnia/fisiopatología , Lactante , Isoflurano/farmacología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Pletismografía Total , Canales de Potasio de Dominio Poro en Tándem/genética , Respiración/efectos de los fármacos , Muerte Súbita del Lactante/genética , Volumen de Ventilación Pulmonar/fisiología
11.
J Am Heart Assoc ; 5(4): e002865, 2016 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-27098968

RESUMEN

BACKGROUND: Two-pore K(+) channels have emerged as potential targets to selectively regulate cardiac cell membrane excitability; however, lack of specific inhibitors and relevant animal models has impeded the effort to understand the role of 2-pore K(+) channels in the heart and their potential as a therapeutic target. The objective of this study was to determine the role of mechanosensitive 2-pore K(+) channel family member TREK-1 in control of cardiac excitability. METHODS AND RESULTS: Cardiac-specific TREK-1-deficient mice (αMHC-Kcnk(f/f)) were generated and found to have a prevalent sinoatrial phenotype characterized by bradycardia with frequent episodes of sinus pause following stress. Action potential measurements from isolated αMHC-Kcnk2(f/f) sinoatrial node cells demonstrated decreased background K(+) current and abnormal sinoatrial cell membrane excitability. To identify novel pathways for regulating TREK-1 activity and sinoatrial node excitability, mice expressing a truncated allele of the TREK-1-associated cytoskeletal protein ßIV-spectrin (qv(4J) mice) were analyzed and found to display defects in cell electrophysiology as well as loss of normal TREK-1 membrane localization. Finally, the ßIV-spectrin/TREK-1 complex was found to be downregulated in the right atrium from a canine model of sinoatrial node dysfunction and in human cardiac disease. CONCLUSIONS: These findings identify a TREK-1-dependent pathway essential for normal sinoatrial node cell excitability that serves as a potential target for selectively regulating sinoatrial node cell function.


Asunto(s)
Canales de Potasio de Dominio Poro en Tándem/fisiología , Nodo Sinoatrial/fisiología , Potenciales de Acción/fisiología , Animales , Ecocardiografía , Ratones , Ratones Noqueados , Canales de Potasio de Dominio Poro en Tándem/deficiencia
12.
J Mol Cell Cardiol ; 97: 24-35, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27103460

RESUMEN

The two-pore domain potassium (K(+)) channel TWIK-1 (or K2P1.1) contributes to background K(+) conductance in diverse cell types. TWIK-1, encoded by the KCNK1 gene, is present in the human heart with robust expression in the atria, however its physiological significance is unknown. To evaluate the cardiac effects of TWIK-1 deficiency, we studied zebrafish embryos after knockdown of the two KCNK1 orthologues, kcnk1a and kcnk1b. Knockdown of kcnk1a or kcnk1b individually caused bradycardia and atrial dilation (p<0.001 vs. controls), while ventricular stroke volume was preserved. Combined knockdown of both kcnk1a and kcnk1b resulted in a more severe phenotype, which was partially reversed by co-injection of wild-type human KCNK1 mRNA, but not by a dominant negative variant of human KCNK1 mRNA. To determine whether genetic variants in KCNK1 might cause atrial fibrillation (AF), we sequenced protein-coding regions in two independent cohorts of patients (373 subjects) and identified three non-synonymous variants, p.R171H, p.I198M and p.G236S, that were all located in highly conserved amino acid residues. In transfected mammalian cells, zebrafish and wild-type human TWIK-1 channels had a similar cellular distribution with predominant localization in the endosomal compartment. Two-electrode voltage-clamp experiments using Xenopus oocytes showed that both zebrafish and wild-type human TWIK-1 channels produced K(+) currents that are sensitive to external K(+) concentration as well as acidic pH. There were no effects of the three KCNK1 variants on cellular localization, current amplitude or reversal potential at pH7.4 or pH6. Our data indicate that TWIK-1 has a highly conserved role in cardiac function and is required for normal heart rate and atrial morphology. Despite the functional importance of TWIK-1 in the atrium, genetic variation in KCNK1 is not a common primary cause of human AF.


Asunto(s)
Remodelación Atrial/genética , Estudios de Asociación Genética , Atrios Cardíacos/metabolismo , Frecuencia Cardíaca/genética , Canales de Potasio de Dominio Poro en Tándem/genética , Adulto , Anciano , Animales , Fibrilación Atrial/genética , Fibrilación Atrial/metabolismo , Fibrilación Atrial/patología , Fibrilación Atrial/fisiopatología , Femenino , Expresión Génica , Técnicas de Inactivación de Genes , Variación Genética , Atrios Cardíacos/anatomía & histología , Atrios Cardíacos/patología , Humanos , Masculino , Persona de Mediana Edad , Mutación , Linaje , Canales de Potasio de Dominio Poro en Tándem/deficiencia , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Transporte de Proteínas , Factores de Riesgo , Pez Cebra
13.
PLoS One ; 10(5): e0126781, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26001192

RESUMEN

BACKGROUND: TREK-1 deficient alveolar epithelial cells (AECs) secrete less IL-6, more MCP-1, and contain less F-actin. Whether these alterations in cytokine secretion and F-actin content are related remains unknown. We now hypothesized that cytokine secretion from TREK-1-deficient AECs was regulated by cytoskeletal rearrangements. METHODS: We determined F-actin and α-tubulin contents of control, TREK-1-deficient and TREK-1-overexpressing human A549 cells by confocal microscopy and western blotting, and measured IL-6 and MCP-1 levels using real-time PCR and ELISA. RESULTS: Cytochalasin D decreased the F-actin content of control cells. Jasplakinolide increased the F-actin content of TREK-1 deficient cells, similar to the effect of TREK-1 overexpression in control cells. Treatment of control and TREK-1 deficient cells with TNF-α, a strong stimulus for IL-6 and MCP-1 secretion, had no effect on F-actin structures. The combination of TNF-α+cytochalasin D or TNF-α+jasplakinolide had no additional effect on the F-actin content or architecture when compared to cytochalasin D or jasplakinolide alone. Although TREK-1 deficient AECs contained less F-actin at baseline, quantified biochemically, they contained more α-tubulin. Exposure to nocodazole disrupted α-tubulin filaments in control and TREK-1 deficient cells, but left the overall amount of α-tubulin unchanged. Although TNF-α had no effect on the F-actin or α-tubulin contents, it increased IL-6 and MCP-1 production and secretion from control and TREK-1 deficient cells. IL-6 and MCP-1 secretions from control and TREK-1 deficient cells after TNF-α+jasplakinolide or TNF-α+nocodazole treatment was similar to the effect of TNF-α alone. Interestingly, cytochalasin D decreased TNF-α-induced IL-6 but not MCP-1 secretion from control but not TREK-1 deficient cells. CONCLUSION: Although cytochalasin D, jasplakinolide and nocodazole altered the F-actin and α-tubulin structures of control and TREK-1 deficient AEC, the changes in cytokine secretion from TREK-1 deficient cells cannot be explained by cytoskeletal rearrangements in these cells.


Asunto(s)
Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Actinas/metabolismo , Western Blotting , Línea Celular , Quimiocina CCL2/metabolismo , Citocalasina D/farmacología , Depsipéptidos/farmacología , Ensayo de Inmunoadsorción Enzimática , Células Epiteliales , Humanos , Interleucina-6/metabolismo , Microscopía Confocal , Nocodazol/farmacología , Canales de Potasio de Dominio Poro en Tándem/deficiencia , Canales de Potasio de Dominio Poro en Tándem/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Tubulina (Proteína)/metabolismo
14.
Mol Brain ; 7: 80, 2014 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-25406588

RESUMEN

BACKGROUND: Two-pore domain K(+) (K2P) channels have been shown to modulate neuronal excitability. However, physiological function of TWIK-1, the first identified member of the mammalian K2P channel family, in neuronal cells is largely unknown. RESULTS: We found that TWIK-1 proteins were expressed and localized mainly in the soma and proximal dendrites of dentate gyrus granule cells (DGGCs) rather than in distal dendrites or mossy fibers. Gene silencing demonstrates that the outwardly rectifying K(+) current density was reduced in TWIK-1-deficient granule cells. TWIK-1 deficiency caused a depolarizing shift in the resting membrane potential (RMP) of DGGCs and enhanced their firing rate in response to depolarizing current injections. Through perforant path stimulation, TWIK-1-deficient granule cells showed altered signal input-output properties with larger EPSP amplitude values and increased spiking compared to control DGGCs. In addition, supra-maximal perforant path stimulation evoked a graded burst discharge in 44% of TWIK-1-deficient cells, which implies impairment of EPSP-spike coupling. CONCLUSIONS: These results showed that TWIK-1 is functionally expressed in DGGCs and contributes to the intrinsic excitability of these cells. The TWIK-1 channel is involved in establishing the RMP of DGGCs; it attenuates sub-threshold depolarization of the cells during neuronal activity, and contributes to EPSP-spike coupling in perforant path-to-granule cell synaptic transmission.


Asunto(s)
Potenciales de Acción , Giro Dentado/citología , Giro Dentado/metabolismo , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Animales , Potenciales Postsinápticos Excitadores , Técnicas de Silenciamiento del Gen , Masculino , Ratones Endogámicos C57BL , Canales de Potasio/metabolismo , Canales de Potasio de Dominio Poro en Tándem/deficiencia , Sinapsis/metabolismo
15.
Arch Dermatol Res ; 306(10): 885-902, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25318378

RESUMEN

TASK-3 (KCNK9 or K2P9.1) channels are thought to promote proliferation and/or survival of malignantly transformed cells, most likely by increasing their hypoxia tolerance. Based on our previous results that suggested mitochondrial expression of TASK-3 channels, we hypothesized that TASK-3 channels have roles in maintaining mitochondrial activity. In the present work we studied the effect of reduced TASK-3 expression on the mitochondrial function and survival of WM35 and A2058 melanoma cells. TASK-3 knockdown cells had depolarized mitochondrial membrane potential and contained a reduced amount of mitochondrial DNA. Compared to their scrambled shRNA-transfected counterparts, they demonstrated diminished responsiveness to the application of the mitochondrial uncoupler [(3-chlorophenyl)hydrazono]malononitrile (CCCP). These observations indicate impaired mitochondrial function. Further, TASK-3 knockdown cells presented reduced viability, decreased total DNA content, altered cell morphology, and reduced surface area. In contrast to non- and scrambled shRNA-transfected melanoma cell lines, which did not present noteworthy apoptotic activity, almost 50 % of the TASK-3 knockdown cells exhibited strong Annexin-V-specific immunofluorescence signal. Sequestration of cytochrome c from the mitochondria to the cytosol, increased caspase 3 activity, and translocation of the apoptosis-inducing factor from mitochondria to cell nuclei were also demonstrated in TASK-3 knockdown cells. Interference with TASK-3 channel expression, therefore, induces caspase-dependent and -independent apoptosis of melanoma cells, most likely via causing mitochondrial depolarization. Consequently, TASK-3 channels may be legitimate targets of future melanoma therapies.


Asunto(s)
Apoptosis , Melanoma/metabolismo , Potencial de la Membrana Mitocondrial , Mitocondrias/metabolismo , Canales de Potasio de Dominio Poro en Tándem/deficiencia , Interferencia de ARN , Neoplasias Cutáneas/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Línea Celular Tumoral , Proliferación Celular , Regulación hacia Abajo , Metabolismo Energético , Regulación Neoplásica de la Expresión Génica , Humanos , Melanoma/genética , Melanoma/patología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Canales de Potasio de Dominio Poro en Tándem/genética , Transducción de Señal , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Factores de Tiempo , Transfección , Desacopladores/farmacología
16.
Hypertension ; 64(6): 1260-5, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25245387

RESUMEN

TWIK-2 (KCNK6) is a member of the 2-pore domain (K2P) family of potassium channels, which are highly expressed in the vascular system. We tested the hypothesis that TWIK-2 deficiency leads to pulmonary hypertension. TWIK-2 knockout mice and their wildtype littermates at 8 weeks of age had similar mean right ventricular systolic pressures (24±3 and 21±3 mm Hg, respectively.) Significantly, by 20 weeks of age, the mean right ventricular systolic pressures in TWIK-2 knockout mice increased to 35±3 mm Hg (P≤0.036), whereas mean right ventricular systolic pressures in wildtype littermates remained at 22±3 mm Hg. Elevated mean right ventricular systolic pressures in the TWIK-2 knockout mice was accompanied by pulmonary vascular remodeling as determined by a 25% increase in the cross-sectional area of the vessels occupied by the vessel wall. Additionally, secondary branches of the pulmonary artery from 20-week-old TWIK-2 knockout mice showed an enhanced contractile response to U46619 (10(-6) moles/L), a thromboxane A2 mimetic, which was completely abolished with the Rho-kinase inhibitor, Y27632 (10(-6) and 10(-5) moles/L). Treatment of TWIK-2 knockout mice with the Rho-kinase inhibitor, fasudil, in the drinking water for 12 weeks, abolished the development of pulmonary hypertension and attenuated the vessel remodeling. We concluded that mice deficient in the TWIK-2 channel develop pulmonary hypertension between 8 and 20 weeks of age through a mechanism involving Rho-kinase. Our results suggest that downregulation of TWIK-2 in the pulmonary vasculature may be an underlying mechanism in the development of pulmonary hypertension.


Asunto(s)
ADN/genética , Regulación de la Expresión Génica , Hipertensión Pulmonar/genética , Canales de Potasio de Dominio Poro en Tándem/deficiencia , Arteria Pulmonar/metabolismo , Quinasas Asociadas a rho/genética , Animales , Presión Sanguínea , Modelos Animales de Enfermedad , Genotipo , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/fisiopatología , Masculino , Ratones , Ratones Noqueados , Canales de Potasio de Dominio Poro en Tándem/genética , Arteria Pulmonar/fisiopatología , Función Ventricular Derecha , Quinasas Asociadas a rho/biosíntesis
17.
Crit Care Med ; 42(11): e692-701, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25126877

RESUMEN

OBJECTIVES: We previously reported the expression of the two-pore-domain K channel TREK-1 in lung epithelial cells and proposed a role for this channel in the regulation of alveolar epithelial cytokine secretion. In this study, we focused on investigating the role of TREK-1 in vivo in the development of hyperoxia-induced lung injury. DESIGN: Laboratory animal experiments. SETTING: University research laboratory. SUBJECTS: Wild-type and TREK-1-deficient mice. INTERVENTIONS: Mice were anesthetized and exposed to 1) room air, no mechanical ventilation, 2) 95% hyperoxia for 24 hours, and 3) 95% hyperoxia for 24 hours followed by mechanical ventilation for 4 hours. MEASUREMENTS AND MAIN RESULTS: Hyperoxia exposure accentuated lung injury in TREK-1-deficient mice but not controls, resulting in increase in lung injury scores, bronchoalveolar lavage fluid cell numbers, and cellular apoptosis and a decrease in quasi-static lung compliance. Exposure to a combination of hyperoxia and injurious mechanical ventilation resulted in further morphological lung damage and increased lung injury scores and bronchoalveolar lavage fluid cell numbers in control but not TREK-1-deficient mice. At baseline and after hyperoxia exposure, bronchoalveolar lavage cytokine levels were unchanged in TREK-1-deficient mice compared with controls. Exposure to hyperoxia and mechanical ventilation resulted in an increase in bronchoalveolar lavage interleukin-6, monocyte chemotactic protein-1, and tumor necrosis factor-α levels in both mouse types, but the increase in interleukin-6 and monocyte chemotactic protein-1 levels was less prominent in TREK-1-deficient mice than in controls. Lung tissue macrophage inflammatory protein-2, keratinocyte-derived cytokine, and interleukin-1ß gene expression was not altered by hyperoxia in TREK-1-deficient mice compared with controls. Furthermore, we show for the first time TREK-1 expression on alveolar macrophages and unimpaired tumor necrosis factor-α secretion from TREK-1-deficient macrophages. CONCLUSIONS: TREK-1 deficiency resulted in increased sensitivity of lungs to hyperoxia, but this effect is less prominent if overwhelming injury is induced by the combination of hyperoxia and injurious mechanical ventilation. TREK-1 may constitute a new potential target for the development of novel treatment strategies against hyperoxia-induced lung injury.


Asunto(s)
Lesión Pulmonar Aguda/patología , Citocinas/metabolismo , Hiperoxia/complicaciones , Canales de Potasio de Dominio Poro en Tándem/deficiencia , Lesión Pulmonar Aguda/etiología , Lesión Pulmonar Aguda/terapia , Animales , Western Blotting , Líquido del Lavado Bronquioalveolar/química , Citocinas/genética , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Macrófagos Alveolares/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal/métodos , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Distribución Aleatoria , Reacción en Cadena en Tiempo Real de la Polimerasa , Valores de Referencia , Respiración Artificial , Medición de Riesgo , Índice de Severidad de la Enfermedad
18.
Am J Physiol Regul Integr Comp Physiol ; 305(1): R60-7, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23637138

RESUMEN

K2P6.1 or TWIK-2, a two-pore domain K channel, is an important regulator of cardiovascular function. K2P6.1 is highly expressed in vascular smooth muscle and endothelium. Mice (8-12 wk) lacking functional K2P6.1 (K2P6.1(-/-)) are hypertensive and have enhanced vascular contractility. It is not known whether the lack of functional K2P6.1 in endothelium has a role in the vascular dysfunction in K2P6.1(-/-) mice. We tested the hypothesis: K2P6.1(-/-) mice have impaired endothelium-dependent relaxations. K2P6.1(-/-) mice were ∼35 mmHg more hypertensive than WT mice at both 8-12 wk (young adult) and 20-24 wk (mature mice, P < 0.01; n = 8-10). Endothelium-dependent relaxations of the thoracic aorta were evaluated by isometric myography after contraction with phenylephrine (10(-6) M). Maximal ACh-dependent relaxations were increased from 65 ± 1% to 73 ± 1% in the aorta from young adult (P < 0.01; n = 6) and from 45 ± 1% to 74 ± 1% in the aorta from mature (P < 0.001; n = 5) K2P6.1(-/-) mice compared with K2P6.1(+/+) littermates. However, in the aorta from young adult and mature K2P6.1(+/+) mice, 10(-5) M indomethacin, a cyclooxygenase inhibitor, increased maximal ACh relaxations to knockout levels. Enhanced relaxation was also seen with ATP, a P2Y purinergic agonist, and A23187, a nonreceptor-based agonist in mature K2P6.1(-/-) mice. Mature adult aorta from K2P6.1(-/-) showed an attenuated ACh-mediated contraction in the presence of nitro-l-arginine methyl ester (l-NAME) and without precontraction of 0.97 mN vs. 7.5 mN in K2P6.1(-/-) and K2P6.1(+/+) (P < 0.001; n = 5). In summary, K2P6.1(-/-) mice, which are hypertensive, have enhanced endothelium-dependent relaxations in the aorta due to the suppression of an indomethacin-sensitive constrictor component.


Asunto(s)
Aorta Torácica/fisiología , Endotelio Vascular/fisiología , Canales de Potasio de Dominio Poro en Tándem/deficiencia , Canales de Potasio de Dominio Poro en Tándem/fisiología , Vasodilatación/fisiología , Animales , Calcimicina/farmacología , Modelos Animales de Enfermedad , Hipertensión/etiología , Hipertensión/fisiopatología , Indometacina/farmacología , Masculino , Ratones , Ratones Noqueados , NG-Nitroarginina Metil Éster/farmacología , Fenilefrina/farmacología , Canales de Potasio de Dominio Poro en Tándem/genética , Vasoconstricción/efectos de los fármacos , Vasoconstricción/fisiología , Vasodilatación/efectos de los fármacos
19.
Am J Physiol Lung Cell Mol Physiol ; 304(4): L276-86, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23275623

RESUMEN

We recently proposed a role for the two-pore-domain K(+) (K2P) channel Trek-1 in the regulation of cytokine release from mouse alveolar epithelial cells (AECs) by demonstrating decreased interleukin-6 (IL-6) secretion from Trek-1-deficient cells, but the underlying mechanisms remained unknown. This study was designed to investigate the mechanisms by which Trek-1 decreases IL-6 secretion. We hypothesized that Trek-1 regulates tumor necrosis factor-α (TNF-α)-induced IL-6 release via NF-κB-, p38-, and PKC-dependent pathways. We found that Trek-1 deficiency decreased IL-6 secretion from mouse and human AECs at both transcriptional and translational levels. While NF-κB/p65 phosphorylation was unchanged, p38 phosphorylation was decreased in Trek-1-deficient cells, and pharmacological inhibition of p38 decreased IL-6 secretion in control but not Trek-1-deficient cells. Similarly, pharmacological inhibition of PKC also decreased IL-6 release, and we found decreased phosphorylation of the isoforms PKC/PKDµ (Ser(744/748)), PKCθ, PKCδ, PKCα/ßII, and PKCζ/λ, but not PKC/PKDµ (Ser(916)) in Trek-1-deficient AECs. Phosphorylation of PKCθ, a Ca(2+)-independent isoform, was intact in control cells but impaired in Trek-1-deficient cells. Furthermore, TNF-α did not elevate the intracellular Ca(2+) concentration in control or Trek-1-deficient cells, and removal of extracellular Ca(2+) did not impair IL-6 release. In summary, we report the expression of Trek-1 in human AECs and propose that Trek-1 deficiency may alter both IL-6 translation and transcription in AECs without affecting Ca(2+) signaling. The results of this study identify Trek-1 as a new potential target for the development of novel treatment strategies against acute lung injury.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Interleucina-6/metabolismo , Canales de Potasio de Dominio Poro en Tándem/fisiología , Lesión Pulmonar Aguda/fisiopatología , Animales , Calcio/metabolismo , Humanos , Ratones , Canales de Potasio de Dominio Poro en Tándem/deficiencia , Proteína Quinasa C/fisiología , Transducción de Señal/fisiología , Factor de Necrosis Tumoral alfa/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA