Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Microcirculation ; 24(8)2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28857417

RESUMEN

OBJECTIVE: Previously, we found that diet-induced HHcy in mice caused decreased eNOS expression and signaling in mesenteric arteries, but greatly enhanced non-NOS, non-prostacyclin-dependent vasodilation, which involves MEJ communication. To further assess whether HHcy enhances MEJ communication, this study examined endothelium-dependent attenuation of phenylephrine-induced vasoconstriction (myoendothelial feedback) and key molecules involved. METHODS: Myoendothelial feedback was examined in isolated mouse mesenteric arteries, after 6-weeks diet-induced HHcy, using pressure myography. Gap junction (Cx37, Cx40, Cx43), NOS (eNOS, nNOS, iNOS), and potassium channel (IK1) protein expression were measured with immunoblots, and connexin mRNAs with real-time PCR. Contribution of nNOS + iNOS to vasomotor responses was assessed using the drug TRIM. RESULTS: Myoendothelial feedback was significantly (P < .05) enhanced in HHcy arteries compared to control, coincident with significantly greater Cx37 and IK1 protein and Cx37 mRNA. Cx43 protein, but not mRNA, was significantly less in HHcy, and Cx40 was not different. eNOS protein was significantly less in HHcy. nNOS and iNOS were not different. TRIM had little effect on vasomotor function. CONCLUSIONS: Diet-induced HHcy enhanced myoendothelial feedback, and increased Cx37 and IK1 expression may contribute. nNOS or iNOS did not upregulate to compensate for decreased eNOS, and they had little involvement in vasomotor function.


Asunto(s)
Conexinas/metabolismo , Uniones Comunicantes/metabolismo , Regulación de la Expresión Génica , Hiperhomocisteinemia/metabolismo , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/biosíntesis , Arterias Mesentéricas/metabolismo , Animales , Alimentos Formulados/efectos adversos , Uniones Comunicantes/patología , Hiperhomocisteinemia/inducido químicamente , Hiperhomocisteinemia/patología , Hiperhomocisteinemia/fisiopatología , Masculino , Arterias Mesentéricas/patología , Arterias Mesentéricas/fisiopatología , Ratones , Óxido Nítrico Sintasa de Tipo I/biosíntesis , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Óxido Nítrico Sintasa de Tipo III/biosíntesis , Proteína alfa-4 de Unión Comunicante
2.
Biochem Biophys Res Commun ; 470(4): 907-12, 2016 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-26820527

RESUMEN

The intermediate conductance calcium-activated potassium channel (KCa3.1) mediates proliferation of many cell types including fibroblasts, and is a molecular target for intervention in various cell proliferative diseases. Our previous study showed that reduction of KCa3.1 channel expression by lyso-globotriaosylceramide (lyso-Gb3) inhibits differentiation into myofibroblasts and collagen synthesis, which might lead to development of ascending thoracic aortic aneurysm secondary to Fabry disease. However, how lyso-Gb3 downregulates KCa3.1 channel expression is unknown. Therefore, we aimed to investigate the underlying mechanisms of lyso-Gb3-mediated KCa3.1 channel downregulation, focusing on the cAMP signaling pathway. We found that lyso-Gb3 increased the intracellular cAMP concentration by upregulation of adenylyl cyclase 6 and inhibited ERK 1/2 phosphorylation through the protein kinase A (PKA) pathway, leading to the inhibition of KCa3.1 channel synthesis, not the exchange protein directly activated by cAMP (Epac) pathway. Moreover, lyso-Gb3 suppressed expression of class II phosphatidylinositol 3-kinase C2ß (PI3KC2ß) by PKA activation, which reduces the production of phosphatidylinositol 3-phosphate [PI(3)P], and the reduced membrane surface expression of KCa3.1 channel was recovered by increasing the intracellular levels of PI(3)P. Consequently, our findings that lyso-Gb3 inhibited both KCa3.1 channel synthesis and surface expression by increasing intracellular cAMP, and controlled surface expression through changes in PI3KC2ß-mediated PI(3)P production, suggest that modulation of PKA and PI3KC2ß activity to control of KCa3.1 channel expression can be an alternative important target to attenuate ascending thoracic aortic aneurysms in Fabry disease.


Asunto(s)
Membrana Celular/metabolismo , Fosfatidilinositol 3-Quinasas Clase II/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Glucolípidos/administración & dosificación , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/biosíntesis , Fosfatos de Fosfatidilinositol/metabolismo , Esfingolípidos/administración & dosificación , Animales , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/efectos de los fármacos , Ratones , Células 3T3 NIH
3.
Pflugers Arch ; 466(2): 307-17, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23873353

RESUMEN

The present study was designed to investigate the role of advanced glycation end products (AGEs) in intermediate-conductance and small-conductance Ca(2+)-activated potassium channels (KCa3.1 and KCa2.3)-mediated relaxation in rat resistance arteries and the underlying mechanism. The endothelial function of mesenteric arteries was assessed with the use of wire myography. Expression levels of KCa3.1 and KCa2.3 were measured by using Western blot. Reactive oxygen species (ROS) were measured by using dihydroethidium and 2', 7'-dichlorofluorescein diacetate. KCa3.1 and KCa2.3-mediated vasodilatation responses to acetylcholine and NS309 (opener of KCa3.1 and KCa2.3) were impaired by incubation of the third-order mesenteric arteries from normal rats with AGEs (200 µg ml(-1) for 3 h). In cultured human umbilical vein endothelial cells (HUVECs), AGEs increased ROS level and decreased the protein expression of KCa3.1 and KCa2.3. Antioxidant alpha lipoic acid restored the impairment in both vasodilatation function and expression of KCa3.1 and KCa2.3. H2O2 could mimic the effect of AGEs on the protein expression of KCa3.1 and KCa2.3 in cultured HUVECs. These results demonstrate for the first time that AGEs impaired KCa3.1 and KCa2.3-mediated vasodilatation in rat mesenteric arteries via downregulation of both KCa3.1 and KCa2.3, in which the enhanced oxidative stress was involved.


Asunto(s)
Productos Finales de Glicación Avanzada/farmacología , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/efectos de los fármacos , Arterias Mesentéricas/fisiología , Estrés Oxidativo/fisiología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/efectos de los fármacos , Vasodilatación/efectos de los fármacos , Alcanos/farmacología , Animales , Células Endoteliales de la Vena Umbilical Humana , Humanos , Técnicas In Vitro , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/biosíntesis , Masculino , Arterias Mesentéricas/efectos de los fármacos , NG-Nitroarginina Metil Éster/farmacología , Pirazoles/farmacología , Compuestos de Quinolinio/farmacología , Ratas , Ratas Sprague-Dawley , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/biosíntesis
4.
Microcirculation ; 21(8): 747-53, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25047389

RESUMEN

OBJECTIVE: NO and a non-NO/prostacyclin EDH mechanism are major contributors of vascular tone and cerebral blood flow. However, the effect of metabolic syndrome on EDH-mediated responses in cerebral vessels remains unknown and may offer another avenue for therapeutic targeting. The purpose of this study was to investigate EDH-dependent responses in cerebral arteries during metabolic syndrome. METHODS: EDH-dependent dilations were assessed in MCAs isolated from nondiabetic obese and lean Zucker rats in the presence and absence of NS309, an activator of SKCa and IKCa channels. IKCa channel expression and activity were assessed by western blotting and pressure myography, respectively. RESULTS: EDH-mediated dilations were significantly attenuated in the obese compared to the lean Zucker rat MCA. Luminal delivery of 1 µM NS309 enhanced EDH-mediated responses in lean and obese Zucker cerebral vessels. Both dose-dependent dilations to luminal NS309 and IKCa protein expression in pooled cerebral arteries were comparable between the two groups. CONCLUSIONS: Our results suggest that pharmacological targeting of IKCa channels can rescue EDH-mediated dilations in obese Zucker rat MCAs. Compromised EDH-mediated dilations in obesity are not due to impaired IKCa channel expression or activity.


Asunto(s)
Endotelio Vascular , Indoles/farmacología , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/biosíntesis , Obesidad , Oximas/farmacología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/biosíntesis , Vasodilatación/efectos de los fármacos , Animales , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Endotelio Vascular/fisiopatología , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Obesidad/metabolismo , Obesidad/patología , Obesidad/fisiopatología , Ratas , Ratas Zucker
5.
Nephrol Dial Transplant ; 29(2): 313-24, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24166472

RESUMEN

BACKGROUND: Fibroblast activation plays a critical role in diabetic nephropathy (DN). The Ca2+-activated K+ channel KCa3.1 mediates cellular proliferation of many cell types including fibroblasts. KCa3.1 has been reported to be a potential molecular target for pharmacological intervention in a diverse array of clinical conditions. However, the role of KCa3.1 in the activation of myofibroblasts in DN is unknown. These studies assessed the effect of KCa3.1 blockade on renal injury in experimental diabetes. METHODS: As TGF-ß1 plays a central role in the activation of fibroblasts to myofibroblasts in renal interstitial fibrosis, human primary renal interstitial fibroblasts were incubated with TGF-ß1+/- the selective inhibitor of KCa3.1, TRAM34, for 48 h. Two streptozotocin-induced diabetic mouse models were used in this study: wild-type KCa3.1+/+ and KCa3.1-/- mice, and secondly eNOS-/- mice treated with or without a selective inhibitor of KCa3.1 (TRAM34). Then, markers of fibroblast activation and fibrosis were determined. RESULTS: Blockade of KCa3.1 inhibited the upregulation of type I collagen, fibronectin, α-smooth muscle actin, vimentin and fibroblast-specific protein-1 in renal fibroblasts exposed to TGF-ß1 and in kidneys from diabetic mice. TRAM34 reduced TGF-ß1-induced phosphorylation of Smad2/3 and ERK1/2 but not P38 and JNK MAPK in interstitial fibroblasts. CONCLUSIONS: These results suggest that blockade of KCa3.1 attenuates diabetic renal interstitial fibrogenesis through inhibiting activation of fibroblasts and phosphorylation of Smad2/3 and ERK1/2. Therefore, therapeutic interventions to prevent or ameliorate DN through targeted inhibition of KCa3.1 deserve further consideration.


Asunto(s)
Nefropatías Diabéticas/genética , Regulación de la Expresión Génica , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/genética , Corteza Renal/patología , ARN/genética , Animales , Biopsia , Western Blotting , Proliferación Celular , Células Cultivadas , Diabetes Mellitus Experimental , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/patología , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis , Humanos , Inmunohistoquímica , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/antagonistas & inhibidores , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/biosíntesis , Corteza Renal/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Pirazoles/farmacología , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal
6.
Pharmacol Res ; 77: 30-8, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24055799

RESUMEN

Airway smooth muscle (ASM) cell phenotype modulation, characterized by reversible switching between contractile and proliferative phenotypes, is considered to contribute to proliferative diseases such as allergic asthma and chronic obstructive pulmonary disease (COPD). KCa3.1 has been suggested to be involved in regulating ASM cell activation, proliferation, and migration. However, little is known regarding the exact role of KCa3.1 in ASM cell phenotypic modulation. To elucidate the role of KCa3.1 in regulating ASM cell phenotypic modulation, we investigated the effects of KCa3.1 channels on ASM contractile marker protein expression, proliferation and migration of primary human bronchial smooth muscle (BSM) cells. We found that PDGF increased KCa3.1 channel expression in BSM cells with a concomitant marked decrease in the expression of contractile phenotypic marker proteins including smooth muscle myosin heavy chain (SMMHC), smooth muscle α-actin (α-SMA), myocardin and KCa1.1. These changes were significantly attenuated by the KCa3.1 blocker, TRAM-34, or gene silencing of KCa3.1. Pharmacological blockade or gene silencing of KCa3.1 also suppressed PDGF-induced human BSM cell migration and proliferation accompanied by a decrease in intracellular free Ca(2+) levels as a consequence of membrane depolarization, resulting in a reduction in cyclin D1 level and cell cycle arrest at G0-G1 phase. Additionally, PDGF-induced up-regulation of KCa3.1 and down-regulation of BSM contractile marker proteins were regulated by the ERK inhibitor U0126 and the AKT inhibitor LY294002. These findings highlight a novel role for the KCa3.1 channel in human BSM cell phenotypic modulation and provide a potential target for therapeutic intervention for proliferative airway diseases.


Asunto(s)
Canales de Potasio de Conductancia Intermedia Activados por el Calcio/biosíntesis , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/fisiología , Miocitos del Músculo Liso/fisiología , Fenotipo , Regulación hacia Arriba , Actinas/biosíntesis , Bronquios/efectos de los fármacos , Bronquios/metabolismo , Bronquios/fisiología , Butadienos/farmacología , Calcio/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Cromonas/farmacología , Ciclina D1/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Silenciador del Gen , Humanos , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/antagonistas & inhibidores , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/genética , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/biosíntesis , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/genética , Morfolinas/farmacología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Cadenas Pesadas de Miosina/biosíntesis , Nitrilos/farmacología , Proteínas Nucleares/biosíntesis , Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Factor de Crecimiento Derivado de Plaquetas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Pirazoles/farmacología , Transactivadores/biosíntesis , Regulación hacia Arriba/efectos de los fármacos
7.
Am J Respir Cell Mol Biol ; 45(5): 962-8, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21493782

RESUMEN

Migration to draining lymph nodes is a critical requirement for dendritic cells (DCs) to control T-cell-mediated immunity. The calcium-activated potassium channel KCa3.1 has been shown to be involved in regulating cell migration in multiple cell types. In this study, KCa3.1 expression and its functional role in lung DC migration were examined. Fluorescence-labeled antigen was intranasally delivered into mouse lungs to label lung Ag-carrying DCs. Lung CD11c(high)CD11b(low) and CD11c(low)CD11b(high) DCs from PBS-treated and ovalbumin (OVA)-sensitized mice were sorted using MACS and FACS. Indo-1 and DiBAC4(3) were used to measure intracellular Ca(2+) and membrane potential, respectively. The mRNA expression of KCa3.1 was examined using real-time PCR. Expression of KCa3.1 protein and CCR7 was measured using flow cytometry. Migration of two lung DC subsets to lymphatic chemokines was examined using TransWell in the absence or presence of the KCa3.1 blocker TRAM-34. OVA sensitization up-regulated mRNA and protein expression of KCa3.1 in lung DCs, with a greater response by the CD11c(high)CD11b(low) than CD11c(low)CD11b(high) DCs. Although KCa3.1 expression in Ag-carrying DCs was higher than that in non-Ag-carrying DCs in OVA-sensitized mice, the difference was not as prominent. However, Ag-carrying lung DCs expressed significantly higher CCR7 than non-Ag-carrying DCs. CCL19, CCL21, and KCa3.1 activator 1-EBIO induced an increase in intracellular calcium in both DC subsets. In addition, 1-EBIO-induced calcium increase was suppressed by TRAM-34. In vitro blockade of KCa3.1 with TRAM-34 impaired CCL19/CCL21-induced transmigration. In conclusion, KCa3.1 expression in lung DCs is up-regulated by OVA sensitization in both lung DC subsets, and KCa3.1 is involved in lung DC migration to lymphatic chemokines.


Asunto(s)
Movimiento Celular/inmunología , Células Dendríticas/inmunología , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/inmunología , Pulmón/inmunología , Animales , Bencimidazoles/farmacología , Antígenos CD11/inmunología , Calcio/metabolismo , Movimiento Celular/efectos de los fármacos , Quimiocinas/inmunología , Células Dendríticas/efectos de los fármacos , Femenino , Citometría de Flujo , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/agonistas , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/antagonistas & inhibidores , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/biosíntesis , Pulmón/efectos de los fármacos , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/inmunología , Ratones , Ratones Endogámicos BALB C , Ovalbúmina/inmunología , Pirazoles/farmacología , Receptores CCR7/inmunología
8.
J Pharmacol Exp Ther ; 338(2): 528-36, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21602424

RESUMEN

Recently, a new experimental stromal hyperplasia animal model corresponding to clinical benign prostatic hyperplasia (BPH) was established. The main objective of this study was to elucidate the roles of the intermediate-conductance Ca(2+)-activated K(+) channel (K(Ca)3.1) in the implanted urogenital sinus (UGS) of stromal hyperplasia BPH model rats. Using DNA microarray, real-time polymerase chain reaction, Western blot, and/or immunohistochemical analyses, we identified the expression of K(Ca)3.1 and its transcriptional regulators in implanted UGS of BPH model rats and prostate needle-biopsy samples and surgical prostate specimens of BPH patients. We also examined the in vivo effects of a K(Ca)3.1 blocker, 1-[(2-chlorophenyl)diphenylmethyl]-1H-pyrazole (TRAM-34), on the proliferation index of implanted UGS by measurement of UGS weights and proliferating cell nuclear antigen immunostaining. K(Ca)3.1 genes and proteins were highly expressed in implanted UGS rather than in the normal host prostate. In the implanted UGS, the gene expressions of two transcriptional regulators of K(Ca)3.1, repressor element 1-silencing transcription factor and c-Jun, were significantly down- and up-regulated, and the regulations were correlated negatively or positively with K(Ca)3.1 expression, respectively. Positive signals of K(Ca)3.1 proteins were detected exclusively in stromal cells, whereas they were scarcely immunolocalized to basal cells of the epithelium in implanted UGS. In vivo treatment with TRAM-34 significantly suppressed the increase in implanted UGS weights compared with the decrease in stromal cell components. Moreover, significant levels of K(Ca)3.1 expression were observed in human BPH samples. K(Ca)3.1 blockers may be a novel treatment option for patients suffering from BPH.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/antagonistas & inhibidores , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/biosíntesis , Canales de Potasio Calcio-Activados/antagonistas & inhibidores , Canales de Potasio Calcio-Activados/metabolismo , Hiperplasia Prostática/tratamiento farmacológico , Hiperplasia Prostática/metabolismo , Pirazoles/administración & dosificación , Adulto , Anciano , Anciano de 80 o más Años , Animales , Proliferación Celular/efectos de los fármacos , Humanos , Masculino , Persona de Mediana Edad , Bloqueadores de los Canales de Potasio/administración & dosificación , Canales de Potasio Calcio-Activados/genética , Hiperplasia Prostática/patología , Ratas , Adulto Joven
9.
Cell Calcium ; 96: 102384, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33676318

RESUMEN

BACKGROUND: Colorectal cancer (CRC) metastases are the main cause of CRC mortality. Intracellular Ca2+ regulates cell migration and invasion, key factors for metastases. Ca2+ also activates Ca2+-dependent potassium channels which in turn affect Ca2+ driving force. We have previously reported that the expression of the Ca2+ activated potassium channel KCNN4 (SK4) is higher in CRC primary tumors compared to normal tissues. Here, we aimed to investigate the role of SK4 in the physiology of CRC. RESULTS: SK4 protein expression is enhanced in CRC tissues compared to normal colon tissues, with a higher level of KCNN4 in CRC patients with KRAS mutations. At the cellular level, we found that SK4 regulates the membrane potential of HCT116 cells. We also found that its inhibition reduced store operated Ca2+ entry (SOCE) and constitutive Ca2+ entry (CCE), while reducing cell migration. We also found that the activity of SK4 is linked to resistance pathways such as KRAS mutation and the expression of NRF2 and HIF-1α. In addition, the pharmacological inhibition of SK4 reduced intracellular reactive oxygen species (ROS) production, NRF2 expression and HIF1α stabilization. CONCLUSION: Our results suggest that SK4 contributes to colorectal cancer cell migration and invasion by modulating both Ca2+ entry and ROS regulation. Therefore, SK4 could be a potential target to reduce metastasis in KRAS-mutated CRC.


Asunto(s)
Calcio/metabolismo , Movimiento Celular/fisiología , Neoplasias Colorrectales/metabolismo , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/biosíntesis , Mutación/fisiología , Proteínas Proto-Oncogénicas p21(ras) , Movimiento Celular/efectos de los fármacos , Neoplasias Colorrectales/genética , Bases de Datos Genéticas , Células HCT116 , Células HT29 , Humanos , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/antagonistas & inhibidores , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Pirazoles/farmacología
10.
Am J Physiol Cell Physiol ; 299(2): C251-63, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20445171

RESUMEN

KCNN4 channels that provide the driving force for cAMP- and Ca(2+)-induced anion secretion are present in both apical and basolateral membranes of the mammalian colon. However, only a single KCNN4 has been cloned. This study was initiated to identify whether both apical and basolateral KCNN4 channels are encoded by the same or different isoforms. Reverse transcriptase-PCR (RT-PCR), real-time quantitative-PCR (RT-QPCR), and immunofluorescence studies were used to clone and identify tissue-specific expression of KCNN4 isoforms. Three distinct KCNN4 cDNAs that are designated as KCNN4a, KCNN4b, and KCNN4c encoding 425, 424, and 395 amino acid proteins, respectively, were isolated from the rat colon. KCNN4a differs from KCNN4b at both the nucleotide and the amino acid level with distinct 628 bp at the 3'-untranslated region and an additional glutamine at position 415, respectively. KCNN4c differs from KCNN4b by lacking the second exon that encodes a 29 amino acid motif. KCNN4a and KCNN4b/c are identified as smooth muscle- and epithelial cell-specific transcripts, respectively. KCNN4b and KCNN4c transcripts likely encode basolateral (40 kDa) and apical (37 kDa) membrane proteins in the distal colon, respectively. KCNN4c, which lacks the S2 transmembrane segment, requires coexpression of a large conductance K(+) channel beta-subunit for plasma membrane expression. The KCNN4 channel blocker TRAM-34 inhibits KCNN4b- and KCNN4c-mediated (86)Rb (K(+) surrogate) efflux with an apparent inhibitory constant of 0.6 +/- 0.1 and 7.8 +/- 0.4 muM, respectively. We conclude that apical and basolateral KCNN4 K(+) channels that regulate K(+) and anion secretion are encoded by distinct isoforms in colonic epithelial cells.


Asunto(s)
Clonación Molecular/métodos , Colon/metabolismo , Regulación de la Expresión Génica , Variación Genética/fisiología , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/biosíntesis , Secuencia de Aminoácidos , Animales , Colon/fisiología , Femenino , Humanos , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/genética , Mucosa Intestinal/metabolismo , Masculino , Datos de Secuencia Molecular , Especificidad de Órganos , Isoformas de Proteínas/biosíntesis , Isoformas de Proteínas/genética , Ratas , Ratas Sprague-Dawley , Xenopus
11.
J Pharmacol Exp Ther ; 335(2): 284-93, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20671071

RESUMEN

The mechanisms involved in altered endothelial function in obesity-related cardiovascular disease are poorly understood. This study investigates the effect of chronic obesity on endothelium-dependent vasodilation and the relative contribution of nitric oxide (NO), calcium-activated potassium channels (K(Ca)), and myoendothelial gap junctions (MEGJs) in the rat saphenous artery. Obesity was induced by feeding rats a cafeteria-style diet (∼30 kJ as fat) for 16 to 20 weeks, with this model reflecting human dietary obesity etiology. Age- and sex-matched controls received standard chow (∼12 kJ as fat). Endothelium-dependent vasodilation was characterized in saphenous arteries by using pressure myography with pharmacological intervention, Western blotting, immunohistochemistry, and ultrastructural techniques. In saphenous artery from control, acetylcholine (ACh)-mediated endothelium-dependent vasodilation was blocked by NO synthase and soluble guanylate cyclase inhibition, whereas in obese rats, the ACh response was less sensitive to such inhibition. Conversely, the intermediate conductance K(Ca) (IK(Ca)) blocker 1-[(2-chlorophenyl)diphenyl-methyl]-1H pyrazole attenuates ACh-mediated dilation in obese, but not control, vessels. In a similar manner, putative gap junction block with carbenoxolone increased the pEC(50) for ACh in arteries from obese, but not control, rats. IK1 protein and MEGJ expression was up-regulated in the arteries of obese rats, an observation absent in control. Addition of the small conductance K(Ca) blocker apamin had no effect on ACh-mediated dilation in either control or obese rat vessels, consistent with unaltered SK3 expression. Up-regulation of distinct IK(Ca)- and gap junction-mediated pathways at myoendothelial microdomain sites, key mechanisms for endothelial-derived hyperpolarization-type activity, maintains endothelium-dependent vasodilation in diet-induced obese rat saphenous artery. Plasticity of myoendothelial coupling mechanisms represents a significant potential target for therapeutic intervention.


Asunto(s)
Endotelio Vascular/fisiología , Uniones Comunicantes/metabolismo , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/biosíntesis , Músculo Liso Vascular/metabolismo , Obesidad/fisiopatología , Vasodilatación/fisiología , Animales , Western Blotting , Modelos Animales de Enfermedad , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Endotelio Vascular/ultraestructura , Uniones Comunicantes/fisiología , Uniones Comunicantes/ultraestructura , Inmunohistoquímica , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/agonistas , Masculino , Microscopía Electrónica , Músculo Liso Vascular/fisiología , Músculo Liso Vascular/ultraestructura , Miografía , Obesidad/metabolismo , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba , Vasoconstrictores/farmacología , Vasodilatación/efectos de los fármacos
12.
J Pharmacol Exp Ther ; 333(1): 210-7, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20040579

RESUMEN

We tested the hypothesis that changes in arterial blood flow modify the function of endothelial Ca2+-activated K+ channels [calcium-activated K+ channel (K(Ca)), small-conductance calcium-activated K+ channel (SK3), and intermediate calcium-activated K+ channel (IK1)] before arterial structural remodeling. In rats, mesenteric arteries were exposed to increased [+90%, high flow (HF)] or reduced blood flow [-90%, low flow (LF)] and analyzed 24 h later. There were no detectable changes in arterial structure or in expression level of endothelial nitric-oxide synthase, SK3, or IK1. Arterial relaxing responses to acetylcholine and 3-oxime-6,7-dichlore-1H-indole-2,3-dione (NS309; activator of SK3 and IK1) were measured in the absence and presence of endothelium, NO, and prostanoid blockers, and 6,12,19,20,25,26-hexahydro-5,27:13,18:21,24-trietheno-11,7-metheno-7H-dibenzo [b,n] [1,5,12,16]tetraazacyclotricosine-5,13-diium dibromide (UCL 1684; inhibitor of SK3) or 1-[(2-chlorophenyl)diphenylmethyl]-1H-pyrazole (TRAM-34; inhibitor of IK1). In LF arteries, endothelium-dependent relaxation was markedly reduced, due to a reduction in the endothelium-derived hyperpolarizing factor (EDHF) response. In HF arteries, the balance between the NO/prostanoid versus EDHF response was unaltered. However, the contribution of IK1 to the EDHF response was enhanced, as indicated by a larger effect of TRAM-34 and a larger residual NS309-induced relaxation in the presence of UCL 1684. Reduction of blood flow selectively blunts EDHF relaxation in resistance arteries through inhibition of the function of K(Ca) channels. An increase in blood flow leads to a more prominent role of IK1 channels in this relaxation.


Asunto(s)
Endotelio Vascular/metabolismo , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/biosíntesis , Arterias Mesentéricas/metabolismo , Canales de Potasio Calcio-Activados/biosíntesis , Acetilcolina/farmacología , Animales , Factores Biológicos/fisiología , Inhibidores de la Ciclooxigenasa/farmacología , Endotelio Vascular/efectos de los fármacos , Guanilato Ciclasa/farmacología , Indoles/farmacología , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/agonistas , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/antagonistas & inhibidores , Masculino , Arterias Mesentéricas/efectos de los fármacos , Contracción Muscular , Relajación Muscular , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiología , Óxido Nítrico Sintasa/antagonistas & inhibidores , Oximas/farmacología , Canales de Potasio Calcio-Activados/agonistas , Canales de Potasio Calcio-Activados/antagonistas & inhibidores , Ratas , Ratas Endogámicas WKY , Receptores Citoplasmáticos y Nucleares/farmacología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio , Guanilil Ciclasa Soluble , Circulación Esplácnica , Estrés Mecánico
13.
Hypertension ; 75(2): 393-404, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31838908

RESUMEN

Heart failure is associated with sympatho-ßAR (ß-adrenoceptor) activation and cardiac fibrosis. Gal-3 (galectin-3) and KCa3.1 channels that are upregulated in diverse cells of diseased heart are implicated in mediating myocardial inflammation and fibrosis. It remains unclear whether Gal-3 interacts with KCa3.1 leading to cardiac fibrosis in the setting of ßAR activation. We tested the effect of KCa3.1 blocker TRAM-34 on cardiac fibrosis and inflammation in cardiac-restricted ß2-TG (ß2AR overexpressed transgenic) mice and determined KCa3.1 expression in ß2-TG×Gal-3-/- mouse hearts. Mechanisms of KCa3.1 in mediating Gal-3 induced fibroblast activation were studied ex vivo. Expression of Gal-3 and KCa3.1 was elevated in ß2-TG hearts. Gal-3 gene deletion in ß2-TG mice decreased KCa3.1 expression in inflammatory cells but not in fibroblasts. Treatment of ß2-TG mice with TRAM-34 for 1 or 2 months significantly ameliorated cardiac inflammation and fibrosis and reduced Gal-3 level. In cultured fibroblasts, Gal-3 upregulated KCa3.1 expression and channel currents with enhanced membrane potential and Ca2+ entry through TRPV4 (transient receptor potential V4) and TRPC6 (transient receptor potential C6) channels leading to fibroblast activation. In conclusion, ßAR stimulation promotes Gal-3 production that upregulates KCa3.1 channels in noncardiomyocyte cells and activates KCa3.1 channels in fibroblasts leading to hyperpolarization of membrane potential and Ca2+ entry via TRP channels. Gal-3-KCa3.1 signaling mobilizes diverse cells facilitating regional inflammation and fibroblast activation and hence myocardial fibrosis.


Asunto(s)
Cardiomiopatías/genética , Galectina 3/genética , Regulación de la Expresión Génica , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/genética , ARN/genética , Receptores Adrenérgicos beta 2/genética , Animales , Western Blotting , Cardiomiopatías/metabolismo , Cardiomiopatías/patología , Células Cultivadas , Fibrosis/genética , Fibrosis/metabolismo , Fibrosis/patología , Galectina 3/biosíntesis , Inmunohistoquímica , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/biosíntesis , Masculino , Ratones , Ratones Transgénicos , Empalme del ARN , Receptores Adrenérgicos beta 2/biosíntesis , Transducción de Señal
14.
Mol Med Rep ; 20(4): 3406-3414, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31432175

RESUMEN

Ion channels serve important roles in the excitation­contraction coupling of cardiac myocytes. Previous studies have shown that the overexpression or activation of intermediate­conductance calcium­activated potassium channel (SK4, encoded by KCNN4) in embryonic stem cell­derived cardiomyocytes can significantly increase their automaticity. The mechanism underlying this effect is hypothesized to be associated with the activation of hyperpolarization­activated cyclic nucleotide­gated channel 2 (HCN2). The aim of the present study was to explore whether a biological pacemaker could be constructed by overexpressing SK4 alone or in combination with HCN2 in a rat model. Ad­green fluorescent protein (GFP), Ad­KCNN4 and Ad­HCN2 recombinant adenoviruses were injected into the left ventricle of Sprague­Dawley rat hearts. The rats were divided into a GFP group (n=10), an SK4 group (n=10), a HCN2 group (n=10) and an SK4 + HCN2 (SK4/HCN2) group (n=10). The isolated hearts were perfused at 5­7 days following injection, and a complete heart block model was established. Compared with the GFP group, overexpressing SK4 alone did not significantly increase the heart rate after establishment of a complete heart block model [98.1±8.9 vs. 96.7±7.6 beats per min (BPM)], The heart rates in the SK4/HCN2 (139.9±21.9 BPM) and HCN2 groups (111.7±5.5 BPM) were significantly increased compared with the GFP and SK4 groups, and the heart rates in the SK4/HCN2 group were significantly increased compared with the SK4 or HCN2 groups. In the HCN2 (n=8) and the SK4/HCN2 (n=7) groups, the shape of the spontaneous ventricular rhythm was the same as the pacing­induced ectopic rhythm in the transgenically altered site. By contrast, these rhythms were different in the SK4 (n=10) and GFP (n=10) groups. There were no significant differences in action potential duration alternans or ventricular arrhythmia inducibility between the four groups (all P>0.05). Western blotting, reverse transcription­quantitative PCR and immunohistochemistry analyses showed that the expression levels of SK4 and HCN2 were significantly increased at the transgene site. Biological pacemaker activity could be successfully generated by co­overexpression of SK4 and HCN2 without increasing the risk of ventricular arrhythmias. The overexpression of SK4 alone is insufficient to generate biological pacemaker activity. The present study provided evidence that SK4 and HCN2 combined could construct an ectopic pacemaker, laying the groundwork for the development of improved biological pacing mechanisms in the future.


Asunto(s)
Relojes Biológicos/fisiología , Regulación de la Expresión Génica/fisiología , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/biosíntesis , Miocardio/metabolismo , Canales de Potasio/metabolismo , Animales , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/genética , Miocardio/citología , Canales de Potasio/genética , Ratas , Ratas Sprague-Dawley
15.
J Am Heart Assoc ; 8(1): e010418, 2019 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-30563389

RESUMEN

Background Cardiac fibrosis is a core pathological process associated with heart failure. The recruitment and differentiation of primitive fibroblast precursor cells of bone marrow origin play a critical role in pathological interstitial cardiac fibrosis. The KCa3.1 channels are expressed in both ventricular fibroblasts and circulating mononuclear cells in rats and are upregulated by angiotensin II . We hypothesized that KCa3.1 channels mediate the inflammatory microenvironment in the heart, promoting the infiltrated bone marrow-derived circulating mononuclear cells to differentiate into myofibroblasts, leading to myocardial fibrosis. Methods and Results We established a cardiac fibrosis model in rats by infusing angiotensin II to evaluate the impact of the specific KCa3.1 channel blocker TRAM -34 on cardiac fibrosis. At the same time, mouse CD 4+ T cells and rat circulating mononuclear cells were separated to investigate the underlying mechanism of the TRAM -34 anti-cardiac fibrosis effect. TRAM -34 significantly attenuated cardiac fibrosis and the inflammatory reaction and reduced the number of fibroblast precursor cells and myofibroblasts. Inhibition of KCa3.1 channels suppressed angiotensin II -stimulated expression and secretion of interleukin-4 and interleukin-13 in CD 4+ T cells and interleukin-4- or interleukin-13-induced differentiation of monocytes into fibrocytes. Conclusions KCa3.1 channels facilitate myocardial inflammation and the differentiation of bone marrow-derived monocytes into myofibroblasts in cardiac fibrosis caused by angiotensin II infusion.


Asunto(s)
Cardiomiopatías/genética , Regulación de la Expresión Génica , Inflamación/genética , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/genética , Monocitos/patología , Miocardio/metabolismo , Angiotensina II/toxicidad , Animales , Western Blotting , Cardiomiopatías/metabolismo , Cardiomiopatías/patología , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis/genética , Fibrosis/metabolismo , Fibrosis/patología , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/patología , Inmunohistoquímica , Inflamación/metabolismo , Inflamación/patología , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/biosíntesis , Masculino , Monocitos/metabolismo , Miocardio/patología , Miofibroblastos/metabolismo , Miofibroblastos/patología , ARN/genética , Ratas , Ratas Sprague-Dawley
16.
Am J Respir Cell Mol Biol ; 37(5): 525-31, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17585114

RESUMEN

Airway smooth muscle cell hyperplasia contributes to airway remodeling and hyperreactivity characteristic of asthma. Changes to potassium channel activity in proliferating human airway smooth muscle (HASM) cells have been described, but no regulatory role in proliferation has been attributed to them. We sought to investigate the expression of the intermediate conductance calcium-activated potassium channel K(Ca)3.1 in HASM cells and investigate its role in proliferation. Smooth muscle cells derived from human airways were grown in vitro and K(Ca)3.1 channel expression was measured using Western blot, RT-PCR, and patch clamp electrophysiology. Pharmacologic inhibitors of the channel were used in assays of cellular proliferation, and flow cytometry was used to identify cell cycle regulation. HASM cells expressed K(Ca)3.1 channel mRNA, protein, and activity with up-regulation evident after transforming growth factor-beta stimulation. Pharmacologic inhibition of K(Ca)3.1 led to growth arrest in cells stimulated to proliferate with mitogens. These inhibitors did not cause cellular toxicity or induce apoptosis. We have demonstrated, for the first time, the expression of K(Ca)3.1 channels in HASM cells. In addition, we have shown that K(Ca)3.1 channels are important in HASM cell proliferation, making these channels a potential therapeutic target in airway remodeling.


Asunto(s)
Calcio/fisiología , Proliferación Celular , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/fisiología , Músculo Liso/citología , Potasio/metabolismo , Sistema Respiratorio/citología , Células Cultivadas , Humanos , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/biosíntesis , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/genética , Músculo Liso/metabolismo , ARN Mensajero/metabolismo , Sistema Respiratorio/metabolismo , Factor de Crecimiento Transformador beta/fisiología , Regulación hacia Arriba/fisiología
17.
Zhonghua Fu Chan Ke Za Zhi ; 42(2): 111-5, 2007 Feb.
Artículo en Zh | MEDLINE | ID: mdl-17442186

RESUMEN

OBJECTIVE: To study the expression of intermediate-conductance-Ca(2+)-activated K(+) (IKCa1) channels in endometrial cancer and its role in regulating proliferation of endometrial cancer cells. METHODS: Western blot and RT-PCR were used to examine the expression of IKCa1 channels in 13 normal endometrial specimens and 25 endometrial cancer specimens; and RNA interference (RNAi), [(3)H] thymidine incorporation, and inhibitor of IKCa1 channel were used to explore the role of IKCa1 channels in regulation of proliferation of endometrial cancer cells HEC-1A. RESULTS: The expression rate and level of IKCa1 mRNA in endometrial carcinoma (84%, 0.89 +/- 0.52) were higher than in normal endometria (8%, 0.14 +/- 0.12; P < 0.01). The expression rate and level of IKCa1 protein in endometrial carcinomas (80%, 1.18 +/- 0.41) were higher than in normal endometria (15%, 0.71 +/- 0.26; P < 0.01). Clotrimazole, an inhibitor of IKCa1 channels known to suppress the function of the channels, caused a both time- and dose-dependent decrease in cell number of HEC-1A cell. Western blot analysis revealed that the IKCa1 level in whole lysates of the cells transfected with target-IKCa1 small interference RNA (siRNA) was (48.27 +/- 9.07)% of that found in the cells transfected with non-silencing RNA; [(3)H] thymidine incorporation in HEC-1A cells transfected with target-IKCa1 siRNA was also reduced, siRNA inhibited HEC-1A cell proliferation, compared with the cells transfected with non-silencing RNA (P < 0.05). CONCLUSION: The expression of IKCa1 channels may be closely related to the proliferation of endometrial cancer, and down regulation of its expression may suppress its development.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Neoplasias Endometriales/patología , Endometrio/metabolismo , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/biosíntesis , Adulto , Anciano , Línea Celular Tumoral , Clotrimazol/administración & dosificación , Clotrimazol/farmacología , Relación Dosis-Respuesta a Droga , Neoplasias Endometriales/metabolismo , Endometrio/patología , Femenino , Humanos , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/genética , Persona de Mediana Edad , ARN Mensajero/biosíntesis , ARN Interferente Pequeño/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Transfección
18.
Med Sci Monit Basic Res ; 23: 45-57, 2017 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-28280257

RESUMEN

BACKGROUND Accumulating data point to intermediate-conductance calcium-activated potassium channel (IKCa1) as a key player in controlling cell cycle progression and proliferation of human cancer cells. However, the role that IKCa1 plays in the growth of human cervical cancer cells is largely unexplored. MATERIAL AND METHODS In this study, Western blot analysis, immunohistochemical staining, and RT-PCR were first used for IKCa1protein and gene expression assays in cervical cancer tissues and HeLa cells. Then, IKCa1 channel blocker and siRNA were employed to inhibit the functionality of IKCa1 and downregulate gene expression in HeLa cells, respectively. After these treatments, we examined the level of cell proliferation by MTT method and measured IKCa1 currents by conventional whole-cell patch clamp technique. Cell apoptosis was assessed using the Annexin V-FITC/Propidium Iodide (PI) double-staining apoptosis detection kit. RESULTS We demonstrated that IKCa1 mRNA and protein are preferentially expressed in cervical cancer tissues and HeLa cells. We also showed that the IKCa1 channel blocker, clotrimazole, and IKCa1 channel siRNA can be used to suppress cervical cancer cell proliferation and decrease IKCa1 channel current. IKCa1 downregulation by specific siRNAs induced a significant increase in the proportion of apoptotic cells in HeLa cells. CONCLUSIONS IKCa1 is overexpressed in cervical cancer tissues, and IKCa1 upregulation in cervical cancer cell linea enhances cell proliferation, partly by reducing the proportion of apoptotic cells.


Asunto(s)
Canales de Potasio de Conductancia Intermedia Activados por el Calcio/metabolismo , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/patología , Adulto , Bloqueadores de los Canales de Calcio/farmacología , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Clotrimazol/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica , Células HeLa , Humanos , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/biosíntesis , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/genética , Persona de Mediana Edad , Técnicas de Placa-Clamp , ARN Mensajero/genética , ARN Mensajero/metabolismo , Neoplasias del Cuello Uterino/genética
19.
Oncotarget ; 7(28): 43924-43938, 2016 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-27270322

RESUMEN

Approximately 50-75% of patients with serous ovarian carcinoma (SOC) experience recurrence within 18 months after first-line treatment. Current clinical indicators are inadequate for predicting the risk of recurrence. In this study, we used 7 publicly available microarray datasets to identify gene signatures related to recurrence in optimally debulked SOC patients, and validated their expressions in an independent clinic cohort of 127 patients using immunohistochemistry (IHC). We identified a two-gene signature including KCNN4 and S100A14 which was related to recurrence in optimally debulked SOC patients. Their mRNA expression levels were positively correlated and regulated by DNA copy number alterations (CNA) (KCNN4: p=1.918e-05) and DNA promotermethylation (KCNN4: p=0.0179; S100A14: p=2.787e-13). Recurrence prediction models built in the TCGA dataset based on KCNN4 and S100A14 individually and in combination showed good prediction performance in the other 6 datasets (AUC:0.5442-0.9524). The independent cohort supported the expression difference between SOC recurrences. Also, a KCNN4 and S100A14-centered protein interaction subnetwork was built from the STRING database, and the shortest regulation path between them, called the KCNN4-UBA52-KLF4-S100A14 axis, was identified. This discovery might facilitate individualized treatment of SOC.


Asunto(s)
Biomarcadores de Tumor/análisis , Cistadenocarcinoma Seroso/patología , Recurrencia Local de Neoplasia/patología , Neoplasias Glandulares y Epiteliales/patología , Neoplasias Ováricas/patología , Adulto , Anciano , Anciano de 80 o más Años , Proteínas de Unión al Calcio/análisis , Proteínas de Unión al Calcio/biosíntesis , Carcinoma Epitelial de Ovario , Cistadenocarcinoma Seroso/mortalidad , Femenino , Perfilación de la Expresión Génica , Humanos , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/análisis , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/biosíntesis , Estimación de Kaplan-Meier , Factor 4 Similar a Kruppel , Persona de Mediana Edad , Recurrencia Local de Neoplasia/mortalidad , Neoplasias Glandulares y Epiteliales/mortalidad , Neoplasias Ováricas/mortalidad , Pronóstico , Transcriptoma
20.
PLoS One ; 10(12): e0145259, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26689552

RESUMEN

The KCa3.1 K+ channel has been proposed as a novel target for pulmonary diseases such as asthma and pulmonary fibrosis. It is expressed in epithelia but its expression and function in primary human bronchial epithelial cells (HBECs) has not been described. Due to its proposed roles in the regulation of cell proliferation, migration, and epithelial fluid secretion, inhibiting this channel might have either beneficial or adverse effects on HBEC function. The aim of this study was to assess whether primary HBECs express the KCa3.1 channel and its role in HBEC function. Primary HBECs from the airways of healthy and asthmatic subjects, SV-transformed BEAS-2B cells and the neoplastic H292 epithelial cell line were studied. Primary HBECs, BEAS-2B and H292 cells expressed KCa3.1 mRNA and protein, and robust KCa3.1 ion currents. KCa3.1 protein expression was increased in asthmatic compared to healthy airway epithelium in situ, and KCa3.1 currents were larger in asthmatic compared to healthy HBECs cultured in vitro. Selective KCa3.1 blockers (TRAM-34, ICA-17043) had no effect on epithelial cell proliferation, wound closure, ciliary beat frequency, or mucus secretion. However, several features of TGFß1-dependent epithelial-mesenchymal transition (EMT) were inhibited by KCa3.1 blockade. Treatment with KCa3.1 blockers is likely to be safe with respect to airway epithelial biology, and may potentially inhibit airway remodelling through the inhibition of EMT.


Asunto(s)
Bronquios/metabolismo , Células Epiteliales/metabolismo , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/biosíntesis , Mucosa Respiratoria/metabolismo , Asma/metabolismo , Línea Celular Tumoral , Transición Epitelial-Mesenquimal , Regulación de la Expresión Génica , Humanos , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/antagonistas & inhibidores , Factor de Crecimiento Transformador beta1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA