Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
1.
BMB Rep ; 54(2): 130-135, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33407994

RESUMEN

Voltage-gated potassium (Kv) channels are involved in many important cellular functions and play pivotal roles in cancer progression. The expression level of Kv2.1 was observed to be higher in the highly metastatic prostate cancer cells (PC-3), specifically in their membrane, than in immortalized prostate cells (WPMY-1 cells) and comparatively less metastatic prostate cancer cells (LNCaP and DU145 cells). However, Kv2.1 expression was significantly decreased when the cells were treated with antioxidants, such as N-acetylcysteine or ascorbic acid, implying that the highly expressed Kv2.1 could detect reactive oxygen species (ROS) in malignant prostate cancer cells. In addition, the blockade of Kv2.1 with stromatoxin-1 or siRNA targeting Kv2.1 significantly inhibited the migration of malignant prostate cancer cells. Our results suggested that Kv2.1 plays an important role as a ROS sensor and that it is a promising therapeutic molecular target in metastasis of prostate cancer. [BMB Reports 2021; 54(2): 130-135].


Asunto(s)
Neoplasias de la Próstata/metabolismo , Canales de Potasio Shab/metabolismo , Línea Celular , Movimiento Celular/efectos de los fármacos , Humanos , Masculino , Células PC-3 , Neoplasias de la Próstata/patología , ARN Mensajero/antagonistas & inhibidores , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/farmacología , Canales de Potasio Shab/antagonistas & inhibidores , Canales de Potasio Shab/genética
2.
BMC Cancer ; 20(1): 1179, 2020 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-33267786

RESUMEN

BACKGROUND: Gastric and colorectal cancers are the most common malignant tumours, leading to a significant number of cancer-related deaths worldwide. Recently, increasing evidence has demonstrated that cancer cells exhibit a differential expression of potassium channels and this can contribute to cancer progression. However, their expression and localisation at the somatic level remains uncertain. In this study, we have investigated the expression levels of KCNB1 and KCNA5 genes encoding ubiquitous Kv2.1 and Kv1.5 potassium channels in gastric and colorectal tumours. METHODS: Gastric and colorectal tumoral and peritumoral tissues were collected to evaluate the expression of KCNB1 and KCNA5 mRNA by quantitative PCR. Moreover, the immunohistochemical staining profile of Kv2.1 and Kv1.5 was assessed on 40 Formalin-Fixed and Paraffin-Embedded (FFPE) gastric carcinoma tissues. Differences in gene expression between tumoral and peritumoral tissues were compared statistically with the Mann-Whitney U test. The association between the clinicopathological features of the GC patients and the expression of both Kv proteins was investigated with χ2 and Fisher's exact tests. RESULTS: The mRNA fold expression of KCNB1 and KCNA5 genes showed a lower mean in the tumoral tissues (0.06 ± 0.17, 0.006 ± 0.009) compared to peritumoral tissues (0.08 ± 0.16, 0.16 ± 0.48, respectively) without reaching the significance rate (p = 0.861, p = 0.152, respectively). Interestingly, Kv2.1 and Kv1.5 immunostaining was detectable and characterised by a large distribution in peritumoral and tumoral epithelial cells. More interestingly, inflammatory cells were also stained. Surprisingly, Kv2.1 and Kv1.5 staining was undoubtedly and predominantly detected in the cytoplasm compartment of tumour cells. Indeed, the expression of Kv2.1 in tumour cells revealed a significant association with the early gastric cancer clinical stage (p = 0.026). CONCLUSION: The data highlight, for the first time, the potential role of Kv1.5 and Kv2.1 in gastrointestinal-related cancers and suggests they may be promising prognostic markers for these tumours.


Asunto(s)
Neoplasias Colorrectales/genética , Canal de Potasio Kv1.5/metabolismo , Canales de Potasio Shab/metabolismo , Neoplasias Gástricas/genética , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos
3.
EBioMedicine ; 61: 103061, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33096484

RESUMEN

BACKGROUND: Diabetic peripheral neuropathy (DPN) is a common complication of diabetes severely afflicting the patients, while there is yet no effective medication against this disease. As Kv2.1 channel functions potently in regulating neurological disorders, the present work was to investigate the regulation of Kv2.1 channel against DPN-like pathology of DPN model mice by using selective Kv2.1 inhibitor SP6616 (ethyl 5-(3-ethoxy-4-methoxyphenyl)-2-(4-hydroxy-3-methoxybenzylidene)-7-methyl-3-oxo-2,3-dihydro-5H-[1,3]thiazolo[3,2-a]pyrimidine-6-carboxylate) as a probe. METHODS: STZ-induced type 1 diabetic mice with DPN (STZ mice) were defined at 12 weeks of age (4 weeks after STZ injection) through behavioral tests, and db/db (BKS Cg-m+/+Leprdb/J) type 2 diabetic mice with DPN (db/db mice) were at 18 weeks of age. SP6616 was administered daily via intraperitoneal injection for 4 weeks. The mechanisms underlying the amelioration of SP6616 on DPN-like pathology were investigated by RT-PCR, western blot and immunohistochemistry technical approaches against diabetic mice, and verified against the STZ mice with Kv2.1 knockdown in dorsal root ganglion (DRG) tissue by injection of adeno associated virus AAV9-Kv2.1-RNAi. Amelioration of SP6616 on the pathological behaviors of diabetic mice was assessed against tactile allodynia, thermal sensitivity and motor nerve conduction velocity (MNCV). FINDINGS: SP6616 treatment effectively ameliorated the threshold of mechanical stimuli, thermal sensitivity and MNCV of diabetic mice. Mechanism research results indicated that SP6616 suppressed Kv2.1 expression, increased the number of intraepidermal nerve fibers (IENFs), improved peripheral nerve structure and vascular function in DRG tissue. In addition, SP6616 improved mitochondrial dysfunction through Kv2.1/CaMKKß/AMPK/PGC-1α pathway, repressed inflammatory response by inhibiting Kv2.1/NF-κB signaling and alleviated apoptosis of DRG neuron through Kv2.1-mediated regulation of Bcl-2 family proteins and Caspase-3 in diabetic mice. INTERPRETATION: Our work has highly supported the beneficial of Kv2.1 inhibition in ameliorating DPN-like pathology and highlighted the potential of SP6616 in the treatment of DPN. FUNDING: Please see funding sources.


Asunto(s)
Neuropatías Diabéticas/metabolismo , Pirimidinas/farmacología , Canales de Potasio Shab/antagonistas & inhibidores , Tiazoles/farmacología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Biomarcadores , Calcio/metabolismo , Diabetes Mellitus Experimental , Neuropatías Diabéticas/tratamiento farmacológico , Neuropatías Diabéticas/etiología , Neuropatías Diabéticas/patología , Modelos Animales de Enfermedad , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Ganglios Espinales/patología , Regulación de la Expresión Génica/efectos de los fármacos , Inmunohistoquímica , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/genética , Mitocondrias/metabolismo , Neuritas/efectos de los fármacos , Neuritas/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Unión Proteica , Pirimidinas/química , Canales de Potasio Shab/genética , Canales de Potasio Shab/metabolismo , Transducción de Señal , Tiazoles/química
4.
Sci Rep ; 10(1): 5771, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-32238829

RESUMEN

Household air pollution has adverse effects on cardiovascular health. One of the major sources of household air pollutants is the combustion of cooking oils during cooking. Trans, trans-2,4-decadienal (tt-DDE) is a type of dienaldehyde that is present in a wide range of food and food products. It is a byproduct of the peroxidation of linoleic acid following the heating of oil during cooking. The mechanisms of the associations between household air pollution and cardiac arrhythmias are currently unclear. The purpose of this study was to determine effects of tt-DDE on the ion currents in H9c2 cells. The IK and ICa,L in H9c2 cells treated with and without tt-DDE were measured using the whole-cell patch clamp method. Expressions of Kv2.1 and Cav1.2 in H9c2 cells treated with and without tt-DDE were measured by western blot analysis. After the H9c2 cells had been exposed to tt-DDE, the IK and ICa,L were significantly decreased. The expression of Kv2.1, unlike that of Cav1.2, was also significantly decreased in these cells. These changes in IK and ICa,L that were induced by tt-DDE may help to explain the association between cardiac arrhythmogenesis and cooking-oil fumes.


Asunto(s)
Contaminación del Aire Interior/efectos adversos , Aldehídos/efectos adversos , Miocitos Cardíacos/efectos de los fármacos , Aceites/efectos adversos , Contaminantes Atmosféricos/efectos adversos , Animales , Arritmias Cardíacas/etiología , Arritmias Cardíacas/metabolismo , Canales de Calcio Tipo L/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Culinaria , Humanos , Transporte Iónico/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Ratas , Canales de Potasio Shab/metabolismo
5.
FASEB J ; 33(12): 14680-14689, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31682765

RESUMEN

Voltage-gated potassium (K+) channel subfamily B member 1 (KCNB1, Kv2.1) and integrin-α5 form macromolecular complexes-named integrin-α5-KCNB1 complexes (IKCs)-in the human brain, but their function was poorly understood. Here we report that membrane depolarization triggered IKC intracellular signals mediated by small GTPases of the Ras subfamily and protein kinase B (Akt) to advance the development of filopodia and lamellipodia in Chinese hamster ovary cells, stimulate their motility, and enhance neurite outgrowth in mouse neuroblastoma Neuro2a cells. Five KCNB1 mutants (L211P, R312H G379R, G381R, and F416L) linked to severe infancy or early-onset epileptic encephalopathy exhibited markedly defective conduction. However, although L211P, G379R, and G381R normally engaged Ras/Akt and stimulated cell migration, R312H and F416L failed to activate Ras/Akt signaling and did not enhance cell migration. Taken together, these data suggest that IKCs modulate cellular plasticity via Ras and Akt signaling. As such, defective IKCs may cause epilepsy through mechanisms other than dysregulated excitability such as, for example, abnormal neuronal development and resulting synaptic connectivity.-Yu, W., Shin, M. R., Sesti, F. Complexes formed with integrin-α5 and KCNB1 potassium channel wild type or epilepsy-susceptibility variants modulate cellular plasticity via Ras and Akt signaling.


Asunto(s)
Epilepsia/genética , Integrina alfa5/metabolismo , Mutación , Plasticidad Neuronal , Canales de Potasio Shab/metabolismo , Animales , Células CHO , Línea Celular Tumoral , Movimiento Celular , Cricetinae , Cricetulus , Ratones , Neuronas/metabolismo , Neuronas/fisiología , Unión Proteica , Proteínas Proto-Oncogénicas c-akt/metabolismo , Canales de Potasio Shab/genética , Transducción de Señal , Proteínas ras/metabolismo
6.
FASEB J ; 33(9): 9762-9774, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31162977

RESUMEN

The KCNE2 single transmembrane-spanning voltage-gated potassium (Kv) channel ß subunit is ubiquitously expressed and essential for normal function of a variety of cell types, often via regulation of the KCNQ1 Kv channel. A polymorphism upstream of KCNE2 is associated with reduced lung function in human populations, but the pulmonary consequences of KCNE2 gene disruption are unknown. Here, germline deletion of mouse Kcne2 reduced pulmonary expression of potassium channel α subunits Kcnq1 and Kcnb1 but did not alter expression of other Kcne genes. Kcne2 colocalized and coimmunoprecipitated with Kcnq1 in mouse lungs, suggesting the formation of pulmonary Kcnq1-Kcne2 potassium channel complexes. Kcne2 deletion reduced blood O2, increased CO2, increased pulmonary apoptosis, and increased inflammatory mediators TNF-α, IL-6, and leukocytes in bronchoalveolar lavage (BAL) fluids. Consistent with increased pulmonary vascular leakage, Kcne2 deletion increased plasma, BAL albumin, and the BAL:plasma albumin concentration ratio. Kcne2-/- mouse lungs exhibited baseline induction of the reperfusion injury salvage kinase pathway but were less able to respond via this pathway to imposed pulmonary ischemia/reperfusion injury (IRI). We conclude that KCNE2 regulates KCNQ1 in the lungs and is required for normal lung function and resistance to pulmonary IRI. Our data support a causal relationship between KCNE2 gene disruption and lung dysfunction.-Zhou, L., Köhncke, C., Hu, Z., Roepke, T. K., Abbott, G. W. The KCNE2 potassium channel ß subunit is required for normal lung function and resilience to ischemia and reperfusion injury.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Lesión Pulmonar/metabolismo , Canales de Potasio con Entrada de Voltaje/metabolismo , Daño por Reperfusión/metabolismo , Animales , Citocinas/genética , Citocinas/metabolismo , Femenino , Mutación de Línea Germinal , Inflamación/metabolismo , Canal de Potasio KCNQ1/genética , Canal de Potasio KCNQ1/metabolismo , Ratones , Ratones Noqueados , Fosforilación , Canales de Potasio con Entrada de Voltaje/genética , Daño por Reperfusión/genética , Canales de Potasio Shab/genética , Canales de Potasio Shab/metabolismo
7.
Glia ; 66(10): 2200-2208, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30151916

RESUMEN

For long times astrocytes had been regarded as supporting cells, passively filling the spaces between neuronal cell bodies and their extensions. Now it is known that astrocytes are actively involved in a variety of important biological functions such as regulating cerebral blood flow, supporting neuronal metabolism, controlling the extracellular potassium concentration, and clearing neurotransmitters from the extracellular space. In line with this multitude of tasks astrocytes display conspicuous functional and regional heterogeneity. Using three complementary labeling methods nine classes of astrocytes have been differentiated, which were termed protoplasmic, fibrous, velate, radial, and perivascular astrocytes in addition to Bergmann, marginal, and ependymal glial cells. To complete this list retinal Müller cells and a largely forgotten astrocytic cell type, the "feathered cell" of Fañanas need to be added. So far, Fañanas cells could be only recognized with the tedious gold-sublimate procedure. Consequently, data indicating a potential biological function are completely missing. In a parallel investigation we used a battery of antibodies against potassium channels and related proteins to identify potential marker proteins for the immunocytochemical visualization of distinct cell types in the cerebellar cortex. Here we present novel marker proteins, the Kv2.2 potassium channel and calsenilin, to visualize Fañanas cells in the cerebellar Purkinje cell layer. Such markers will allow to identify Fañanas cell subsequent to patching and electrophysiological characterization. This may pave the path to obtain new functional data, which may be helpful to understand the role of these enigmatic cells in normal biological function and disease.


Asunto(s)
Cerebelo/citología , Cerebelo/metabolismo , Técnica del Anticuerpo Fluorescente , Neuroglía/citología , Neuroglía/metabolismo , Animales , Anticuerpos , Técnica del Anticuerpo Fluorescente/métodos , Expresión Génica , Proteínas de Interacción con los Canales Kv/metabolismo , Masculino , Microscopía Confocal , Ratas Wistar , Canales de Potasio Shab/metabolismo , Coloración y Etiquetado
8.
Clin Exp Pharmacol Physiol ; 45(12): 1257-1264, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30003581

RESUMEN

The Kv2.1 channel plays an important role in the regulation against pancreatic ß-cell dysfunctions. Therefore, it is regarded as a promising target for drug discovery against type 2 diabetes. In the present study, we found that the small molecule 4-ethoxy-N-{[6-(2-thienyl)-7H-[1,2,4]triazolo[3,4-b][1,3,4]thiadiazin-3-yl]methyl}aniline (ETA), a novel Kv2.1 inhibitor, may be capable of promoting glucose-stimulated insulin secretion and protecting from apoptosis in pancreatic INS-832/13 cells. The assay of ETA on type 2 diabetic mice induced by high-fat diet (HFD)/streptozocin (STZ) confirmed its potency in ameliorating glucose homeostasis. ETA administration reduced fasting blood glucose and glycated haemoglobin levels, improved oral glucose tolerance, and increased serum insulin levels in HFD/STZ mice. Mechanism study demonstrated that ETA protected INS-832/13 cells involving the regulation against protein kinase B and extracellular-regulated protein kinase 1/2 signalling pathways. Our study has confirmed the underlying regulation of Kv2.1 against ß-cell function and also addressed the potential of ETA as a lead compound in the treatment of type 2 diabetes mellitus.


Asunto(s)
Hiperglucemia/tratamiento farmacológico , Hiperglucemia/patología , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/patología , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio Shab/antagonistas & inhibidores , Tiadiazinas/farmacología , Animales , Apoptosis/efectos de los fármacos , Transporte Biológico/efectos de los fármacos , Calcio/metabolismo , Glucosa/metabolismo , Homeostasis/efectos de los fármacos , Hiperglucemia/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Bloqueadores de los Canales de Potasio/uso terapéutico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Canales de Potasio Shab/metabolismo , Transducción de Señal/efectos de los fármacos , Tiadiazinas/uso terapéutico
9.
J Neurochem ; 144(4): 483-497, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29265365

RESUMEN

Intracellular protein trafficking is tightly regulated, and improper trafficking might be the fundamental provocateur for human diseases including neurodegeneration. In neurons, protein trafficking to and from the plasma membrane affects synaptic plasticity. Voltage-gated potassium channel 2.1 (Kv2.1) is a predominant delayed rectifier potassium (K+ ) current, and electrical activity patterns of dopamine (DA) neurons within the substantia nigra are generated and modulated by the orchestrated function of different ion channels. The pathological hallmark of Parkinson's disease (PD) is the progressive loss of these DA neurons, resulting in the degeneration of striatal dopaminergic terminals. However, whether trafficking of Kv2.1 channels contributes to PD remains unclear. In this study, we demonstrated that MPTP/MPP+ increases the surface expression of the Kv2.1 channel and causes nigrostriatal degeneration by using a subchronic MPTP mouse model. The inhibition of the Kv2.1 channel by using a specific blocker, guangxitoxin-1E, protected nigrostriatal projections against MPTP/MPP+ insult and thus facilitated the recovery of motor coordination. These findings highlight the importance of trafficking of Kv2.1 channels in the pathogenesis of PD.


Asunto(s)
Intoxicación por MPTP/metabolismo , Neostriado/patología , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Canales de Potasio Shab/metabolismo , Sustancia Negra/patología , Animales , Proteínas de Artrópodos/farmacología , Biotinilación , Línea Celular Tumoral , Neuronas Dopaminérgicas/metabolismo , Intoxicación por MPTP/patología , Intoxicación por MPTP/prevención & control , Ratones , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Degeneración Nerviosa/inducido químicamente , Fármacos Neuroprotectores/farmacología , Equilibrio Postural/efectos de los fármacos , Canales de Potasio Shab/antagonistas & inhibidores , Venenos de Araña/farmacología
10.
J Biol Chem ; 292(37): 15512-15524, 2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28768770

RESUMEN

NMDA receptors (NMDARs) are Ca2+-permeant, ligand-gated ion channels activated by the excitatory neurotransmitter glutamate and have well-characterized roles in the nervous system. The expression and function of NMDARs in pancreatic ß-cells, by contrast, are poorly understood. Here, we report a novel function of NMDARs in ß-cells. Using a combination of biochemistry, electrophysiology, and imaging techniques, we now show that NMDARs have a key role in mediating the effect of leptin to modulate ß-cell electrical activity by promoting AMP-activated protein kinase (AMPK)-dependent trafficking of KATP and Kv2.1 channels to the plasma membrane. Blocking NMDAR activity inhibited the ability of leptin to activate AMPK, induce KATP and Kv2.1 channel trafficking, and promote membrane hyperpolarization. Conversely, activation of NMDARs mimicked the effect of leptin, causing Ca2+ influx, AMPK activation, and increased trafficking of KATP and Kv2.1 channels to the plasma membrane, and triggered membrane hyperpolarization. Moreover, leptin potentiated NMDAR currents and triggered NMDAR-dependent Ca2+ influx. Importantly, NMDAR-mediated signaling was observed in rat insulinoma 832/13 cells and in human ß-cells, indicating that this pathway is conserved across species. The ability of NMDARs to regulate potassium channel surface expression and thus, ß-cell excitability provides mechanistic insight into the recently reported insulinotropic effects of NMDAR antagonists and therefore highlights the therapeutic potential of these drugs in managing type 2 diabetes.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Canales KATP/metabolismo , Leptina/metabolismo , Receptores de N-Metil-D-Aspartato/agonistas , Canales de Potasio Shab/metabolismo , Transducción de Señal , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Proteínas Quinasas Activadas por AMP/metabolismo , Adulto , Animales , Biotinilación , Señalización del Calcio/efectos de los fármacos , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Células Cultivadas , Humanos , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/efectos de los fármacos , Ligandos , Moduladores del Transporte de Membrana/farmacología , Transporte de Proteínas/efectos de los fármacos , Ratas , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/metabolismo , Transducción de Señal/efectos de los fármacos , Propiedades de Superficie
11.
Cell Death Dis ; 8(4): e2737, 2017 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-28383553

RESUMEN

Oxidative modification of the voltage-gated potassium (K+) channel KCNB1 promotes apoptosis in the neurons of cortex and hippocampus through a signaling pathway mediated by Src tyrosine kinases. How oxidation of the channel is transduced into Src recruitment and activation, however, was not known. Here we show that the apoptotic signal originates from integrins, which form macromolecular complexes with KCNB1 channels. The initial stimulus is transduced to Fyn and possibly other Src family members by focal adhesion kinase (FAK). Thus KCNB1 and integrin alpha chain V (integrin-α5) coimmunoprecipitated in the mouse brain and these interactions were retained upon channel's oxidation. Pharmacological inhibition of integrin signaling or FAK suppressed apoptosis induced by oxidation of KCNB1, as well as FAK and Src/Fyn activation. Most importantly, the activation of the integrin-FAK-Src/Fyn cascade was negligible in the presence of non-oxidizable C73A KCNB1 mutant channels, even though they normally interacted with integrin-α5. This leads us to conclude that the transition between the non-oxidized and oxidized state of KCNB1 activates integrin signaling. KCNB1 oxidation may favor integrin clustering, thereby facilitating the recruitment and activation of FAK and Src/Fyn kinases.


Asunto(s)
Apoptosis/fisiología , Encéfalo/metabolismo , Integrina alfa5/metabolismo , Canales de Potasio Shab/metabolismo , Transducción de Señal/fisiología , Sustitución de Aminoácidos , Animales , Células CHO , Cricetinae , Cricetulus , Quinasa 1 de Adhesión Focal/genética , Quinasa 1 de Adhesión Focal/metabolismo , Integrina alfa5/genética , Ratones , Mutación Missense , Proteínas Proto-Oncogénicas c-fyn/genética , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Canales de Potasio Shab/genética , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
12.
Sci Rep ; 7: 41646, 2017 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-28139741

RESUMEN

Heterotetramer voltage-gated K+ (KV) channels KV2.1/KV6.4 display a gating charge-voltage (QV) distribution composed by two separate components. We use state dependent chemical accessibility to cysteines substituted in either KV2.1 or KV6.4 to assess the voltage sensor movements of each subunit. By comparing the voltage dependences of chemical modification and gating charge displacement, here we show that each gating charge component corresponds to a specific subunit forming the heterotetramer. The voltage sensors from KV6.4 subunits move at more negative potentials than the voltage sensors belonging to KV2.1 subunits. These results indicate that the voltage sensors from the tetrameric channels move independently. In addition, our data shows that 75% of the total charge is attributed to KV2.1, while 25% to KV6.4. Thus, the most parsimonious model for KV2.1/KV6.4 channels' stoichiometry is 3:1.


Asunto(s)
Activación del Canal Iónico , Multimerización de Proteína , Canales de Potasio Shab/química , Canales de Potasio Shab/metabolismo , Secuencia de Aminoácidos , Línea Celular , Células Cultivadas , Humanos , Potenciales de la Membrana , Subunidades de Proteína , Canales de Potasio Shab/genética
13.
Sci Rep ; 7: 40155, 2017 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-28065942

RESUMEN

The electric activity of lower motor neurons (MNs) appears to play a role in determining cell-vulnerability in MN diseases. MN excitability is modulated by cholinergic inputs through C-type synaptic boutons, which display an endoplasmic reticulum-related subsurface cistern (SSC) adjacent to the postsynaptic membrane. Besides cholinergic molecules, a constellation of proteins involved in different signal-transduction pathways are clustered at C-type synaptic sites (M2 muscarinic receptors, Kv2.1 potassium channels, Ca2+ activated K+ [SK] channels, and sigma-1 receptors [S1R]), but their collective functional significance so far remains unknown. We have previously suggested that neuregulin-1 (NRG1)/ErbBs-based retrograde signalling occurs at this synapse. To better understand signalling through C-boutons, we performed an analysis of the distribution of C-bouton-associated signalling proteins. We show that within SSC, S1R, Kv2.1 and NRG1 are clustered in highly specific, non-overlapping, microdomains, whereas ErbB2 and ErbB4 are present in the adjacent presynaptic compartment. This organization may define highly ordered and spatially restricted sites for different signal-transduction pathways. SSC associated proteins are disrupted in axotomised MNs together with the activation of microglia, which display a positive chemotactism to C-bouton sites. This indicates that C-bouton associated molecules are also involved in neuroinflammatory signalling in diseased MNs, emerging as new potential therapeutic targets.


Asunto(s)
Neuronas Motoras/metabolismo , Neurregulina-1/metabolismo , Traumatismos de los Nervios Periféricos/metabolismo , Terminales Presinápticos/metabolismo , Receptor ErbB-2/metabolismo , Receptor ErbB-4/metabolismo , Animales , Células Cultivadas , Retículo Endoplásmico/metabolismo , Ratones , Neuronas Motoras/ultraestructura , Terminales Presinápticos/ultraestructura , Receptores sigma/metabolismo , Canales de Potasio Shab/metabolismo , Transducción de Señal , Receptor Sigma-1
14.
Cell Death Dis ; 7: e2216, 2016 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-27148689

RESUMEN

Kv2.1 as a voltage-gated potassium (Kv) channel subunit has a pivotal role in the regulation of glucose-stimulated insulin secretion (GSIS) and pancreatic ß-cell apoptosis, and is believed to be a promising target for anti-diabetic drug discovery, although the mechanism underlying the Kv2.1-mediated ß-cell apoptosis is obscure. Here, the small molecular compound, ethyl 5-(3-ethoxy-4-methoxyphenyl)-2-(4-hydroxy-3-methoxybenzylidene)-7-methyl-3-oxo-2,3-dihydro-5H-[1,3]thiazolo[3,2-a]pyrimidine-6-carboxylate (SP6616) was discovered to be a new Kv2.1 inhibitor. It was effective in both promoting GSIS and protecting ß cells from apoptosis. Evaluation of SP6616 on either high-fat diet combined with streptozocin-induced type 2 diabetic mice or db/db mice further verified its efficacy in the amelioration of ß-cell dysfunction and glucose homeostasis. SP6616 treatment efficiently increased serum insulin level, restored ß-cell mass, decreased fasting blood glucose and glycated hemoglobin levels, and improved oral glucose tolerance. Mechanism study indicated that the promotion of SP6616 on ß-cell survival was tightly linked to its regulation against both protein kinases C (PKC)/extracellular-regulated protein kinases 1/2 (Erk1/2) and calmodulin(CaM)/phosphatidylinositol 3-kinase(PI3K)/serine/threonine-specific protein kinase (Akt) signaling pathways. To our knowledge, this may be the first report on the underlying pathway responsible for the Kv2.1-mediated ß-cell protection. In addition, our study has also highlighted the potential of SP6616 in the treatment of type 2 diabetes.


Asunto(s)
Diabetes Mellitus Experimental/tratamiento farmacológico , Hipoglucemiantes/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Bloqueadores de los Canales de Potasio/farmacología , Pirimidinas/farmacología , Canales de Potasio Shab/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Tiazoles/farmacología , Animales , Células CHO , Calmodulina/antagonistas & inhibidores , Calmodulina/genética , Calmodulina/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Cricetulus , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patología , Descubrimiento de Drogas , Regulación de la Expresión Génica , Prueba de Tolerancia a la Glucosa , Hipoglucemiantes/química , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Masculino , Ratones , Ratones Transgénicos , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosfatidilinositol 3-Quinasa/genética , Fosfatidilinositol 3-Quinasa/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Bloqueadores de los Canales de Potasio/química , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/genética , Proteína Quinasa C/metabolismo , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirimidinas/química , Canales de Potasio Shab/genética , Canales de Potasio Shab/metabolismo , Transducción de Señal , Bibliotecas de Moléculas Pequeñas/química , Estreptozocina , Tiazoles/química
15.
Physiol Rep ; 4(6)2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27033450

RESUMEN

Delayed rectifier voltage-gated K(+)(Kv) channels play an important role in the regulation of the electrophysiological properties of neurons. In mouse dorsal root ganglion (DRG) neurons, a large fraction of the delayed rectifier current is carried by both homotetrameric Kv2 channels and heterotetrameric channels consisting of Kv2 and silent Kv (KvS) subunits (i.e., Kv5-Kv6 and Kv8-Kv9). However, little is known about the contribution of Kv2-mediated currents during the postnatal development ofDRGneurons. Here, we report that the Stromatoxin-1 (ScTx)-sensitive fraction of the total outward K(+)current (IK) from mouseDRGneurons gradually decreased (~13%,P < 0.05) during the first month of postnatal development. Because ScTx inhibits both Kv2.1- and Kv2.2-mediated currents, this gradual decrease may reflect a decrease in currents containing either subunit. However, the fraction of Kv2.1 antibody-sensitive current that only reflects the Kv2.1-mediated currents remained constant during that same period. These results suggested that the fractional contribution of Kv2.2-mediated currents relative toIKdecreased with postnatal age. SemiquantitativeRT-PCRanalysis indicated that this decrease can be attributed to developmental changes in Kv2.2 expression as themRNAlevels of the Kv2.2 subunit decreased gradually between 1 and 4 weeks of age. In addition, we observed age-dependent fluctuations in themRNAlevels of the Kv6.3, Kv8.1, Kv9.1, and Kv9.3 subunits. These results support an important role of both Kv2 and KvS subunits in the postnatal maturation ofDRGneurons.


Asunto(s)
Ganglios Espinales/metabolismo , Neuronas/metabolismo , Potasio/metabolismo , Canales de Potasio Shab/metabolismo , Factores de Edad , Animales , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica , Activación del Canal Iónico , Masculino , Potenciales de la Membrana , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Péptidos/farmacología , Bloqueadores de los Canales de Potasio/farmacología , ARN Mensajero/metabolismo , Canales de Potasio Shab/antagonistas & inhibidores , Canales de Potasio Shab/genética , Venenos de Araña/farmacología
16.
Cell Death Dis ; 7: e2100, 2016 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-26890139

RESUMEN

Altered neuronal excitability is emerging as an important feature in Alzheimer's disease (AD). Kv2.1 potassium channels are important modulators of neuronal excitability and synaptic activity. We investigated Kv2.1 currents and its relation to the intrinsic synaptic activity of hippocampal neurons from 3xTg-AD (triple transgenic mouse model of Alzheimer's disease) mice, a widely employed preclinical AD model. Synaptic activity was also investigated by analyzing spontaneous [Ca(2+)]i spikes. Compared with wild-type (Non-Tg (non-transgenic mouse model)) cultures, 3xTg-AD neurons showed enhanced spike frequency and decreased intensity. Compared with Non-Tg cultures, 3xTg-AD hippocampal neurons revealed reduced Kv2.1-dependent Ik current densities as well as normalized conductances. 3xTg-AD cultures also exhibited an overall decrease in the number of functional Kv2.1 channels. Immunofluorescence assay revealed an increase in Kv2.1 channel oligomerization, a condition associated with blockade of channel function. In Non-Tg neurons, pharmacological blockade of Kv2.1 channels reproduced the altered pattern found in the 3xTg-AD cultures. Moreover, compared with untreated sister cultures, pharmacological inhibition of Kv2.1 in 3xTg-AD neurons did not produce any significant modification in Ik current densities. Reactive oxygen species (ROS) promote Kv2.1 oligomerization, thereby acting as negative modulator of the channel activity. Glutamate receptor activation produced higher ROS levels in hippocampal 3xTg-AD cultures compared with Non-Tg neurons. Antioxidant treatment with N-Acetyl-Cysteine was found to rescue Kv2.1-dependent currents and decreased spontaneous hyperexcitability in 3xTg-AD neurons. Analogous results regarding spontaneous synaptic activity were observed in neuronal cultures treated with the antioxidant 6-hydroxy-2,5,7,8-tetramethylchroman-2-carboxylic acid (Trolox). Our study indicates that AD-related mutations may promote enhanced ROS generation, oxidative-dependent oligomerization, and loss of function of Kv2.1 channels. These processes can be part on the increased neuronal excitability of these neurons. These steps may set a deleterious vicious circle that eventually helps to promote excitotoxic damage found in the AD brain.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Hipocampo/metabolismo , Neuronas/metabolismo , Canales de Potasio Shab/metabolismo , Enfermedad de Alzheimer/patología , Animales , Calcio/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Hipocampo/efectos de los fármacos , Hipocampo/patología , Masculino , Ratones , Neuronas/efectos de los fármacos , Neuronas/patología , Especies Reactivas de Oxígeno/metabolismo , Canales de Potasio Shab/antagonistas & inhibidores , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo
17.
Acta Neuropathol Commun ; 4: 15, 2016 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-26891847

RESUMEN

INTRODUCTION: Increasing evidence implicates the role of the cell types surrounding motor neurons, such as interneurons and glial cells, in non-cell autonomous neurodegeneration of amyotrophic lateral sclerosis (ALS). C-boutons, the large cholinergic synapses that innervate spinal α-motor neurons to control their excitability, are progressively lost from motor neurons in both human ALS and mutant Cu/Zn superoxide dismutase 1 (SOD1)-ALS mice. Neuregulin-1 (NRG1), a trophic factor implicated in neural development, transmission, and synaptic plasticity, has been reported to localize in the synapse of C-boutons. However, the roles of NRG1 in maintenance of motor neuron health and activity, as well as the functional consequences of its alteration in motor neuron disease, are not fully understood. RESULTS: NRG1 was localized to the post-synaptic face of C-boutons and its expression was significantly lost in SOD1-ALS mice and human ALS patients. Losses of NRG1 expression and C-boutons occurred almost contemporaneously in SOD1-ALS mice. In addition, expressions of ErbB3 and ErbB4, receptors for NRG1, were reduced in the motor neurons of SOD1-ALS mice. Furthermore, viral-mediated delivery of type III-NRG1 to the spinal cord restored the number of C-boutons and extended the survival time of SOD1-ALS mice. CONCLUSIONS: These results suggest that maintenance of NRG1-ErbB4/3 axis by supplementation of NRG1 confers neuroprotection in motor neuron disease, partly through the maintenance of C-boutons of spinal motor neurons.


Asunto(s)
Esclerosis Amiotrófica Lateral , Neuronas Motoras/patología , Neurregulina-1/metabolismo , Neuroprotección/fisiología , Terminales Presinápticos/metabolismo , Médula Espinal/patología , Adulto , Factores de Edad , Anciano , Anciano de 80 o más Años , Esclerosis Amiotrófica Lateral/tratamiento farmacológico , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Animales , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica/genética , Humanos , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Persona de Mediana Edad , Neuronas Motoras/metabolismo , Mutación/genética , Proteínas del Tejido Nervioso/metabolismo , Cambios Post Mortem , Receptor ErbB-3/metabolismo , Canales de Potasio Shab/metabolismo , Superóxido Dismutasa/genética , Superóxido Dismutasa-1 , Proteínas de Transporte Vesicular de Acetilcolina/metabolismo
18.
Pharmacol Rep ; 68(2): 457-61, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26922553

RESUMEN

BACKGROUND: The aim of the present study was to assess the effects of perifosine-a third generation alkylphospholipid analog with anti-tumor properties-on the activity of Kv2.1 channels. METHODS: The whole-cell patch clamp technique was applied to follow the modulatory effect of perifosine on Kv2.1 channels expressed in HEK293 cells. RESULTS: Obtained data provide evidence that perifosine application decreases the whole cell Kv2.1 currents in a concentration-independent manner. Perifosine induces a hyperpolarizing shift in the voltage dependence of Kv2.1 channels inactivation without altering the voltage dependence of channels activation. The kinetics of Kv2.1 closed-state inactivation was accelerated by perifosine, with no significant effects on the recovery rate from inactivation. CONCLUSIONS: Taken together, these results show that perifosine modified the Kv2.1 inactivation gating resulting in a decrease of the current amplitude. These data will help to elucidate the mechanism of action of this promising anti-cancer drug on ion channels and their possible implications.


Asunto(s)
Antineoplásicos/farmacología , Activación del Canal Iónico/efectos de los fármacos , Fosforilcolina/análogos & derivados , Canales de Potasio con Entrada de Voltaje/metabolismo , Canales de Potasio Shab/metabolismo , Línea Celular , Células HEK293 , Humanos , Cinética , Potenciales de la Membrana/efectos de los fármacos , Técnicas de Placa-Clamp/métodos , Fosforilcolina/farmacología , Potasio/metabolismo
19.
J Biol Chem ; 290(50): 29676-90, 2015 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-26453299

RESUMEN

In pancreatic ß-cells, voltage-gated potassium 2.1 (Kv2.1) channels are the dominant delayed rectifier potassium channels responsible for action potential repolarization. Here, we report that leptin, a hormone secreted by adipocytes known to inhibit insulin secretion, causes a transient increase in surface expression of Kv2.1 channels in rodent and human ß-cells. The effect of leptin on Kv2.1 surface expression is mediated by the AMP-activated protein kinase (AMPK). Activation of AMPK mimics whereas inhibition of AMPK occludes the effect of leptin. Inhibition of Ca(2+)/calmodulin-dependent protein kinase kinase ß, a known upstream kinase of AMPK, also blocks the effect of leptin. In addition, the cAMP-dependent protein kinase (PKA) is involved in Kv2.1 channel trafficking regulation. Inhibition of PKA prevents leptin or AMPK activators from increasing Kv2.1 channel density, whereas stimulation of PKA is sufficient to promote Kv2.1 channel surface expression. The increased Kv2.1 surface expression by leptin is dependent on actin depolymerization, and pharmacologically induced actin depolymerization is sufficient to enhance Kv2.1 surface expression. The signaling and cellular mechanisms underlying Kv2.1 channel trafficking regulation by leptin mirror those reported recently for ATP-sensitive potassium (KATP) channels, which are critical for coupling glucose stimulation with membrane depolarization. We show that the leptin-induced increase in surface KATP channels results in more hyperpolarized membrane potentials than control cells at stimulating glucose concentrations, and the increase in Kv2.1 channels leads to a more rapid repolarization of membrane potential in cells firing action potentials. This study supports a model in which leptin exerts concerted trafficking regulation of KATP and Kv2.1 channels to coordinately inhibit insulin secretion.


Asunto(s)
Islotes Pancreáticos/efectos de los fármacos , Leptina/farmacología , Canales de Potasio/metabolismo , Transporte de Proteínas/efectos de los fármacos , Canales de Potasio Shab/metabolismo , Adenilato Quinasa/metabolismo , Animales , Calcio/metabolismo , Línea Celular , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Humanos , Insulina/metabolismo , Secreción de Insulina , Islotes Pancreáticos/citología , Islotes Pancreáticos/metabolismo , Sodio/metabolismo
20.
Mol Hum Reprod ; 21(11): 865-83, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26405173

RESUMEN

Human parturition is associated with many pro-inflammatory mediators which are regulated by the nuclear factor-kappaB (NF-κB) family of transcription factors. In the present study, we employed a ChIP-on-chip approach to define genomic loci within chromatin of PHM1-31 myometrial cells that were occupied by RelA-containing NF-κB dimers in response to a TNF stimulation of 1 h. In TNF-stimulated PHM1-31 cells, anti-RelA serum enriched 13 300 chromatin regions; importantly, 11 110 regions were also enriched by anti-RelA antibodies in the absence of TNF. DNA sequences in these regions, from both unstimulated or TNF-stimulated PHM1-31 cultures, were associated with genic regions including IκBα, COX-2, IL6RN, Jun and KCNMB3. TNF-induced binding events at a consensus κB site numbered 1667; these were represented by 112 different instances of the consensus κB motif. Of the 1667 consensus κB motif occurrences, 770 (46.2%) were identified within intronic regions. In unstimulated PHM1-31 cells, anti-RelA-serum-enriched regions were associated with sequences corresponding to open reading frames of ion channel subunit genes including CACNB3 and KCNB1. Moreover, in unstimulated cells, the consensus κB site was identified 2116 times, being defined by 103 different sequence instances of this motif. Of these 2116 consensus κB motifs, 1089 (51.5%) were identified within intronic regions. Parallel expression array analyses in PHM1-31 cultures demonstrated that TNF stimulated a >2-fold induction in 51 genes and a fold repression of >1.5 in 18 others. We identified 14 anti-RelA-serum-enriched genomic regions that correlated with 17 TNF-inducible genes, such as COX2, Egr-1, Jun, IκBα and IL6, as well as five regions associated with TNF-mediated gene repression, including Col1A2.


Asunto(s)
Miocitos del Músculo Liso/metabolismo , Miometrio/citología , FN-kappa B/metabolismo , Regiones Promotoras Genéticas/genética , Factor de Transcripción ReIA/metabolismo , Canales de Calcio/genética , Canales de Calcio/metabolismo , Femenino , Humanos , Miocitos del Músculo Liso/efectos de los fármacos , FN-kappa B/genética , Embarazo , Multimerización de Proteína , Canales de Potasio Shab/genética , Canales de Potasio Shab/metabolismo , Factor de Transcripción ReIA/genética , Activación Transcripcional/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA