Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 207
Filtrar
1.
Life Sci ; 289: 120203, 2022 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-34875252

RESUMEN

OBJECTIVE: To assess the functional role of Hyperpolarization-activated cyclic nucleotide-gated gated channel (HCN) subtypes in the aging bladder phenotype characterized by diminished bladder volume sensation (BVS) with or without the detrusor instability (DI). METHODS: Expression of HCN subtypes was examined by quantitative RT-PCR and Western blot in aged male Fisher 344 rats (n = 15) and young rats (n = 15). Nocturnal urination and awake cystometry (CMG) were assessed in presence and absence of a steady state HCN channel blockade achieved with daily oral gavage of vehicle or Ivabradine (HCN blocker) 6 mg/kg for 7 days. RESULTS: The association of BVS with the age-related downregulation (~30%) of cAMP sensitive HCN1, HCN2 subtypes, and (~50%) upregulation of cAMP insensitive HCN3 subtype is evinced by the doubling in the mean urine volume of nocturnal voids (0.82 ± 0.22 mL vs 0.41 ± 0.12 mL; n = 10; p < 0.05) predicting an age-related rise in the micturition volume threshold (p < 0.0001) in CMG, which is raised further by Ivabradine treatment (p < 0.0005). Ivabradine also doubled non-voiding contractions (NVC) and maximum voiding pressure (MVP) in young and aged rats, respectively (p < 0.0001) to abolish the age-related, innate two -fold elevation in NVC not accompanied with MVP rise in untreated aged rats (p < 0.005). CONCLUSION: The age-related HCN downregulation is mechanistically linked to the exhibition of aging bladder phenotype with the manifestation of DI following steady state blockade of HCN channels in Ivabradine treated young rats. The amplification of MVP in aged rats mediated by FDA approved Ivabradine hints at potential repurposing opportunity in detrusor underactivity.


Asunto(s)
Envejecimiento/metabolismo , Regulación de la Expresión Génica , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/biosíntesis , Canales de Potasio/biosíntesis , Vejiga Urinaria de Baja Actividad/metabolismo , Vejiga Urinaria/metabolismo , Envejecimiento/patología , Animales , Masculino , Ratas , Ratas Endogámicas F344 , Vejiga Urinaria/patología , Vejiga Urinaria de Baja Actividad/patología
2.
Mol Neurobiol ; 58(7): 3575-3587, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33772465

RESUMEN

Peripheral neuropathy is the most frequent dose-limiting adverse effect of oxaliplatin. Acute pain symptoms that are induced or exacerbated by cold occur in almost all patients immediately following the first infusions. Evidence has shown that oxaliplatin causes ion channel expression modulations in dorsal root ganglia neurons, which are thought to contribute to peripheral hypersensitivity. Most dysregulated genes encode ion channels involved in cold and mechanical perception, noteworthy members of a sub-group of potassium channels of the K2P family, TREK and TRAAK. Downregulation of these K2P channels has been identified as an important tuner of acute oxaliplatin-induced hypersensitivity. We investigated the molecular mechanisms underlying this peripheral dysregulation in a murine model of neuropathic pain triggered by a single oxaliplatin administration. We found that oxaliplatin-mediated TREK-TRAAK downregulation, as well as downregulation of other K+ channels of the K2P and Kv families, involves a transcription factor known as the neuron-restrictive silencer factor (NRSF) and its epigenetic co-repressors histone deacetylases (HDACs). NRSF knockdown was able to prevent most of these K+ channel mRNA downregulation in mice dorsal root ganglion neurons as well as oxaliplatin-induced acute cold and mechanical hypersensitivity. Interestingly, pharmacological inhibition of class I HDAC reproduces the antinociceptive effects of NRSF knockdown and leads to an increased K+ channel expression in oxaliplatin-treated mice.


Asunto(s)
Regulación hacia Abajo/fisiología , Epigénesis Genética/fisiología , Hiperalgesia/metabolismo , Oxaliplatino/toxicidad , Canales de Potasio de Dominio Poro en Tándem/biosíntesis , Transcripción Genética/fisiología , Animales , Antineoplásicos/toxicidad , Regulación hacia Abajo/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Hiperalgesia/inducido químicamente , Hiperalgesia/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Canales de Potasio/biosíntesis , Canales de Potasio/genética , Canales de Potasio de Dominio Poro en Tándem/genética , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/biosíntesis , Proteínas Represoras/genética , Transcripción Genética/efectos de los fármacos
3.
J Gen Physiol ; 150(8): 1189-1201, 2018 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-29941431

RESUMEN

The human ether-a-go-go-related gene (hERG) encodes a voltage-gated potassium channel that controls repolarization of cardiac action potentials. Accumulating evidence suggests that most disease-related hERG mutations reduce the function of the channel by disrupting protein biogenesis of the channel in the endoplasmic reticulum (ER). However, the molecular mechanism underlying the biogenesis of ERG K+ channels is largely unknown. By forward genetic screening, we identified an ER-located chaperone CNX-1, the worm homologue of mammalian chaperone Calnexin, as a critical regulator for the protein biogenesis of UNC-103, the ERG-type K+ channel in Caenorhabditis elegans Loss-of-function mutations of cnx-1 decreased the protein level and current density of the UNC-103 K+ channel and suppressed the behavioral defects caused by a gain-of-function mutation in unc-103 Moreover, CNX-1 facilitated tetrameric assembly of UNC-103 channel subunits in a liposome-assisted cell-free translation system. Further studies showed that CNX-1 act in parallel to DNJ-1, another ER-located chaperone known to regulate maturation of UNC-103 channels, on controlling the protein biogenesis of UNC-103. Importantly, Calnexin interacted with hERG proteins in the ER in HEK293T cells. Deletion of calnexin reduced the expression and current densities of endogenous hERG K+ channels in SH-SY5Y cells. Collectively, we reveal an evolutionarily conserved chaperone CNX-1/Calnexin controlling the biogenesis of ERG-type K+ channels.


Asunto(s)
Proteínas de Caenorhabditis elegans/fisiología , Proteínas de Unión al Calcio/fisiología , Retículo Endoplásmico/metabolismo , Canales de Potasio Éter-A-Go-Go/biosíntesis , Proteínas del Choque Térmico HSP40/fisiología , Canales de Potasio/biosíntesis , Secuencia de Aminoácidos , Animales , Animales Modificados Genéticamente , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/genética , Proteínas de Unión al Calcio/genética , Calnexina/metabolismo , Células HEK293 , Proteínas del Choque Térmico HSP40/genética , Humanos , Ratones
4.
Biochemistry ; 55(30): 4212-9, 2016 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-27384110

RESUMEN

Cell free protein synthesis (CFPS) has emerged as a promising methodology for protein expression. While polypeptide production is very reliable and efficient using CFPS, the correct cotranslational folding of membrane proteins during CFPS is still a challenge. In this contribution, we describe a two-step protocol in which the integral membrane protein is initially expressed by CFPS as a precipitate followed by an in vitro folding procedure using lipid vesicles for converting the protein precipitate to the correctly folded protein. We demonstrate the feasibility of using this approach for the K(+) channels KcsA and MVP and the amino acid transporter LeuT. We determine the crystal structure of the KcsA channel obtained by CFPS and in vitro folding to show the structural similarity to the cellular expressed KcsA channel and to establish the feasibility of using this two-step approach for membrane protein production for structural studies. Our studies show that the correct folding of these membrane proteins with complex topologies can take place in vitro without the involvement of the cellular machinery for membrane protein biogenesis. This indicates that the folding instructions for these complex membrane proteins are contained entirely within the protein sequence.


Asunto(s)
Proteínas Bacterianas/biosíntesis , Proteínas Bacterianas/química , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/química , Canales de Potasio/biosíntesis , Canales de Potasio/química , Proteínas Bacterianas/genética , Sistema Libre de Células , Cristalografía por Rayos X , Técnicas In Vitro , Membrana Dobles de Lípidos/química , Proteínas de la Membrana/genética , Modelos Moleculares , Canales de Potasio/genética , Conformación Proteica , Pliegue de Proteína , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/genética
5.
J Cancer Res Ther ; 12(1): 248-53, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27072246

RESUMEN

AIMS: Cisplatin, a platinum-based drug, is an important weapon against many types of cancer. It is well-known that cisplatin induces apoptosis. Potassium channel plays very important role in several signaling pathways. To investigate the possibility that potassium channels also have a role in the cellular response to cisplatin, we examined the effect of cisplatin on the activity of potassium channels on CT26 cell, the colon carcinoma cell line. MATERIALS AND METHODS: The cells were cultured in DMEM, supplemented with 10< heat-inactivated fetal bovine serum. At mid-log phase, cultures were harvested, washed twice in phosphate-buffered saline, and resuspended in culture medium before use. Cells were voltage-clamped using the whole-cell patch clamp technique. Membrane current data were collected and amplified. STATISTICAL ANALYSIS: Differences between two groups were assessed by paired t-test and one sample t-test to compare the relative values. One-way ANOVA was used for all experiment with more than two groups. RESULTS: Potassium currents were detected in CT26 cells and the currents were reduced by the application of tetraethylammonium (TEA) chloride, iberiotoxin, a big conductance calcium-activated potassium channel blocker and barium. The potassium currents were enhanced to 192< by the application of cisplatin (0.5 mM). Moreover, the increase of potassium currents by cisplatin was further inhibited by the application of TEA confirming the action of cisplatin on potassium channels. In addition, relative current induced by cisplatin in CT26 cells was bit larger than in normal IEC-6 cells. CONCLUSION: Potassium currents were detected in CT26 cells and the currents were reduced by the application of tetraethylammonium (TEA) chloride, iberiotoxin, a big conductance calcium-activated potassium channel blocker and barium. The potassium currents were enhanced to 192< by the application of cisplatin (0.5 mM). Moreover, the increase of potassium currents by cisplatin was further inhibited by the application of TEA confirming the action of cisplatin on potassium channels. In addition, relative current induced by cisplatin in CT26 cells was bit larger than in normal IEC-6 cells.


Asunto(s)
Carcinoma/tratamiento farmacológico , Cisplatino/administración & dosificación , Neoplasias del Colon/tratamiento farmacológico , Canales de Potasio/biosíntesis , Apoptosis/efectos de los fármacos , Calcio/metabolismo , Carcinoma/genética , Carcinoma/patología , Línea Celular Tumoral , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Humanos , Técnicas de Placa-Clamp , Péptidos/administración & dosificación , Potasio/metabolismo , Canales de Potasio/genética , Transducción de Señal/efectos de los fármacos
6.
J Biol Chem ; 290(30): 18575-83, 2015 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-26100633

RESUMEN

Although recent studies have shown the sodium-activated potassium channel SLACK (KCNT1) can contribute to neuronal excitability, there remains little information on the physiological role of the closely related SLICK (KCNT2) channel. Activation of SLICK channels may be important during pathological states such as ischemia, in which an increase in intracellular sodium and chloride can perturb membrane potential and ion homeostasis. We have identified two NFκB-binding sites within the promoter region of the human SLICK (KCNT2) and orthologous rat Slick (Kcnt2) genes, suggesting that conditions in which NFκB transcriptional activity is elevated promote expression of this channel. NFκB binding to the rat Slick promoter was confirmed in vivo by ChIP analyses, and NFκB was found differentially bound to the two sites. We verified NFκB transcriptional regulation of SLICK/Slick by mutational analyses and studying gene expression by luciferase assay in P19 cells, where NFκB is constitutively active. For the rat gene, activation of the Slick promoter was found to be additive in single NFκB mutations and synergistic in double mutations. Unexpectedly, for the human gene, NFκB exhibited cooperativity in activating the SLICK promoter. The human SLICK promoter constructs were then tested under hypoxic conditions in PC-12 cells, where NFκB is not active. Only under hypoxic conditions could luciferase activity be detected; the double NFκB mutant construct failed to exhibit activity. Transcriptional regulation of Slick by NFκB was verified in primary neurons. The Slick transcript decreased 24 h after NFκB inhibition. Our data show SLICK expression is predominantly under the control of NFκB. Because neuronal NFκB activation occurs during stressful stimuli such as hypoxia and injury, our findings suggest that SLICK is a neuroprotective gene.


Asunto(s)
FN-kappa B/metabolismo , Neuronas/metabolismo , Canales de Potasio/metabolismo , Transcripción Genética , Animales , Hipoxia de la Célula/genética , Ganglios Espinales/metabolismo , Regulación de la Expresión Génica , Humanos , FN-kappa B/genética , Células PC12 , Canales de Potasio/biosíntesis , Canales de Potasio/genética , Canales de potasio activados por Sodio , Regiones Promotoras Genéticas , Ratas , Transducción de Señal , Sodio/metabolismo
7.
Stem Cells Transl Med ; 4(5): 476-82, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25769653

RESUMEN

Stem cell therapy requires a nontoxic and high-throughput method to achieve a pure cell population to prevent teratomas that can occur if even one cell in the implant has not been transformed. A promising method to detect and separate cells expressing a particular gene is RNA beacon technology. However, developing a successful, specific beacon to a particular transfected gene can take months to develop and in some cases is impossible. Here, we report on an off-the-shelf universal beacon that decreases the time and cost of applying beacon technology to select any living cell population transfected with an exogenous gene.


Asunto(s)
Colorantes Fluorescentes/química , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/aislamiento & purificación , Células Madre Mesenquimatosas/citología , Canales de Potasio/aislamiento & purificación , ARN Mensajero/aislamiento & purificación , Animales , Rastreo Celular/métodos , Perros , Humanos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/biosíntesis , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Sondas Moleculares/genética , Canales de Potasio/biosíntesis , Canales de Potasio/genética , ARN Mensajero/biosíntesis
8.
Biomed Res Int ; 2014: 380398, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25045667

RESUMEN

Prostate cancer is the most common noncutaneous cancer among men in the United States. A genetic contribution to prostate cancer risk has been documented, but knowledge of the molecular mechanisms involved in prostate cancer initiation is still not well understood. Loss of heterozygosity (LOH) of chromosomal regions is crucial in tumor progression. In human prostate cancer, several chromosomal regions demonstrating a high frequency of LOH have been previously identified. KCTD11 (REN) is a tumor suppressor gene mapping on human chromosome 17p13.2, whose expression is frequently lost in human medulloblastoma and in several other cancer types. KCTD11 acts as a negative regulator of the Hedgehog (Hh) signaling. Here, we demonstrated that KCTD11 LOH is a common genetic lesion in human prostate adenocarcinoma. Indeed, nuclear KCTD11 protein expression is strongly reduced in primary prostate cancer, and this event correlated with overexpression of proteins acting into the Hedgehog pathway. Low levels of KCTD11 mRNA have been also observed in prostatic cancer cells, and ectopic overexpression of KCTD11 led to growth arrest. Our study demonstrates and supports that KCTD11, as well as negatively regulated downstream effectors belonging to Hh signaling, plays a role in prostate cancer pathogenesis. This could be suitable to characterize new diagnostic and therapeutic markers.


Asunto(s)
Adenocarcinoma/genética , Genes Supresores de Tumor , Canales de Potasio/biosíntesis , Neoplasias de la Próstata/genética , Adenocarcinoma/patología , Proteínas de Ciclo Celular , Deleción Cromosómica , Regulación Neoplásica de la Expresión Génica , Humanos , Pérdida de Heterocigocidad , Masculino , Canales de Potasio/genética , Neoplasias de la Próstata/patología , Transducción de Señal/genética , Transferasas
9.
Proc Natl Acad Sci U S A ; 111(12): 4620-5, 2014 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-24616516

RESUMEN

Many ion channels, both selective and nonselective, have reentrant pore loops that contribute to the architecture of the permeation pathway. It is a fundamental feature of these diverse channels, regardless of whether they are gated by changes of membrane potential or by neurotransmitters, and is critical to function of the channel. Misfolding of the pore loop leads to loss of trafficking and expression of these channels on the cell surface. Mature tetrameric potassium channels contain an α-helix within the pore loop. We systematically mutated the "pore helix" residues of the channel Kv1.3 and assessed the ability of the monomer to fold into a tertiary reentrant loop. Our results show that pore loop residues form a canonical α-helix in the monomer early in biogenesis and that disruption of tertiary folding is caused by hydrophilic substitutions only along one face of this α-helix. These results provide insight into the determinants of the reentrant pore conformation, which is essential for ion channel function.


Asunto(s)
Canales de Potasio/biosíntesis , Secuencia de Aminoácidos , Animales , Electroforesis en Gel de Poliacrilamida , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Canales de Potasio/química , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido
10.
J Cardiovasc Pharmacol ; 63(6): 533-43, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24566462

RESUMEN

One of the main strategies for cancer therapy is to use tyrosine kinase inhibitors for inhibiting tumor proliferation. Increasing evidence has demonstrated the potential risks of cardiac arrhythmias (such as prolonged QT interval) of these drugs. We report here that a widely used selective inhibitor of Src tyrosine kinases, 4-amino-5-(4-chlorophenyl)-7-(t-butyl)pyrazolo[3,4-d]pyrimidine (PP2), can inhibit and prevent ß-adrenergic stimulation of cardiac pacemaker activity. First, in dissected rat sinus node, PP2 inhibited and prevented the isoproterenol-induced increase of spontaneous beating rate. Second, in isolated rat sinus node myocytes, PP2 suppressed the hyperpolarization-activated "funny" current (traditionally called cardiac pacemaker current, I(f)) by negatively shifting the activation curve and decelerating activation kinetics. Third, in isolated rat sinus node myocytes, PP2 decreased the Src kinase activity, the cell surface expression, and tyrosine phosphorylation of hyperpolarization-activated, cyclic nucleotide-modulated channel 4 (HCN4) channel proteins. Finally, in human embryonic kidney 293 cells overexpressing recombinant human HCN4 channels, PP2 reversed the enhancement of HCN4 channels by isoproterenol and inhibited 573x, a cyclic adenosine momophosphate-insensitive human HCN4 mutant. These results demonstrated that inhibition of Src kinase activity in heart by PP2 decreased and prevented ß-adrenergic stimulation of cardiac pacemaker activity. These effects are mediated, at least partially, by a cAMP-independent attenuation of channel activity and cell surface expression of HCN4, the main channel protein that controls the heart rate.


Asunto(s)
Agonistas Adrenérgicos beta/farmacología , Pirimidinas/farmacología , Nodo Sinoatrial/efectos de los fármacos , Familia-src Quinasas/antagonistas & inhibidores , Animales , Células Cultivadas , Células HEK293 , Humanos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/biosíntesis , Proteínas Musculares/biosíntesis , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/enzimología , Canales de Potasio/biosíntesis , Ratas , Ratas Sprague-Dawley , Nodo Sinoatrial/enzimología , Familia-src Quinasas/metabolismo
11.
Neuroreport ; 24(3): 131-6, 2013 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-23370493

RESUMEN

TRESK gene recombinant adenovirus (10 IU/ml), which has been constructed successfully in our previous study, was implemented through an intrathecal injection. The fact that the method can effectively upregulate the expression of TRESK mRNA in the dorsal root ganglia of spared nerve injury in rats was verified. We also investigated the role of TRESK gene recombinant adenovirus in attenuating tactile allodynia and thermal hyperalgesia in spared nerve injury rats. Spared nerve injury to the sciatic nerve induced persistent tactile allodynia, but had no effect on thermal hyperalgesia. Intrathecal injection of TRESK gene recombinant adenovirus (25 µl) into the region of lumbar enlargement in advance reduced tactile allodynia. Moreover, intrathecal injection of TRESK gene recombinant adenovirus (25 µl) significantly alleviated the activation of astrocytes in spinal cord induced by spared nerve injury. The current study shows that an intrathecal injection of the TRESK gene recombinant adenovirus attenuated the activity of astrocytes in spinal cord, which contributed to relieving neuropathic pain in spared nerve injury rats. According to the result reported in our previous study, attenuating the expression of TRESK in dorsal root ganglia was involved in the development of neuropathic pain. On the basis of these results, we theorized that the therapeutic utility of upregulation of TRESK in dorsal root ganglia was effective in relieving neuropathic pain syndromes induced by peripheral nerve injury.


Asunto(s)
Adenoviridae/genética , Canales de Potasio/uso terapéutico , Ciática/tratamiento farmacológico , Animales , Astrocitos/metabolismo , Astrocitos/patología , Modelos Animales de Enfermedad , Ganglios Espinales/metabolismo , Vectores Genéticos , Proteína Ácida Fibrilar de la Glía/metabolismo , Hiperalgesia/tratamiento farmacológico , Inyecciones Espinales , Masculino , Dimensión del Dolor , Canales de Potasio/biosíntesis , Canales de Potasio/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Ciática/patología , Médula Espinal/metabolismo
12.
J Mol Cell Cardiol ; 53(4): 532-41, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22824041

RESUMEN

In contrast with pathological hypertrophy, exercise-induced physiological hypertrophy is not associated with electrical abnormalities or increased arrhythmia risk. Recent studies have shown that increased cardiac-specific expression of phosphoinositide-3-kinase-α (PI3Kα), the key mediator of physiological hypertrophy, results in transcriptional upregulation of ion channel subunits in parallel with the increase in myocyte size (cellular hypertrophy) and the maintenance of myocardial excitability. The experiments here were undertaken to test the hypothesis that Akt1, which underlies PI3Kα-induced cellular hypertrophy, mediates the effects of augmented PI3Kα signaling on the transcriptional regulation of cardiac ion channels. In contrast to wild-type animals, chronic exercise (swim) training of mice (Akt1(-/-)) lacking Akt1 did not result in ventricular myocyte hypertrophy. Ventricular K(+) current amplitudes and the expression of K(+) channel subunits, however, were increased markedly in Akt1(-/-) animals with exercise training. Expression of the transcripts encoding inward (Na(+) and Ca(2+)) channel subunits were also increased in Akt1(-/-) ventricles following swim training. Additional experiments in a transgenic mouse model of inducible cardiac-specific expression of constitutively active PI3Kα (icaPI3Kα) revealed that short-term activation of PI3Kα signaling in the myocardium also led to the transcriptional upregulation of ion channel subunits. Inhibition of cardiac Akt activation with triciribine in this (inducible caPI3Kα expression) model did not prevent the upregulation of myocardial ion channel subunits. These combined observations demonstrate that chronic exercise training and enhanced PI3Kα expression/activity result in transcriptional upregulation of myocardial ion channel subunits independent of cellular hypertrophy and Akt signaling.


Asunto(s)
Miocardio/metabolismo , Miocitos Cardíacos/citología , Fosfatidilinositol 3-Quinasa/metabolismo , Condicionamiento Físico Animal , Canales de Potasio/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Animales , Cardiomegalia , Corazón/fisiología , Ventrículos Cardíacos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/citología , Miocitos Cardíacos/metabolismo , Técnicas de Placa-Clamp , Canales de Potasio/biosíntesis , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/deficiencia , Proteínas Proto-Oncogénicas c-akt/genética , Ribonucleósidos/farmacología , Regulación hacia Arriba
13.
Mol Pharmacol ; 81(1): 21-30, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21984254

RESUMEN

Large conductance, Ca(2+)-activated K channel proteins are involved in a wide range of physiological activities, so there is considerable interest in the pharmacology of large conductance calcium-activated K (BK) channels. One potent activator of BK channels is mallotoxin (MTX), which produces a very large hyperpolarizing shift of the voltage gating of heterologously expressed BK channels and causes a dramatic increase in the activity of BK channels in human smooth muscle cells. However, we found that MTX shifted the steady-state activation of BK channels in native parotid acinar cells by only 6 mV. This was not because the parotid BK isoform (parSlo) is inherently insensitive to MTX as MTX shifted the activation of heterologously expressed parSlo channels by 70 mV. Even though MTX had a minimal effect on steady-state activation of parotid BK channels, it produced an approximate 2-fold speeding of the channel-gating kinetics. The BK channels in parotid acinar cells have a much more hyperpolarized voltage activation range than BK channels in most other cell types. We found that this is probably attributable to an accessory protein, LRRC26, which is expressed in parotid glands: expressed parSlo + LRRC26 channels were resistant to the actions of MTX. Another class of BK activators is the benzimidazalones that includes 1,3-dihydro-1-(2-hydroxy-5-(trifluoromethyl)phenyl)-5-(trifluoromethyl)-2H-benzimidazol-2-one (NS-1619). Although the LRRC26 accessory protein strongly inhibited the ability of MTX to activate BK channels, we found that it had only a small effect on the action of NS-1619 on BK channels. Thus, the LRRC26 BK channel accessory protein selectively alters the pharmacology of BK channels.


Asunto(s)
Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Proteínas de Neoplasias/fisiología , Células Acinares/efectos de los fármacos , Células Acinares/metabolismo , Animales , Bencimidazoles/metabolismo , Bencimidazoles/farmacología , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Humanos , Canales de Potasio de Gran Conductancia Activados por el Calcio/agonistas , Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Ratones , Glándula Parótida/efectos de los fármacos , Glándula Parótida/metabolismo , Técnicas de Placa-Clamp , Picrotoxina/análogos & derivados , Picrotoxina/toxicidad , Canales de Potasio/agonistas , Canales de Potasio/biosíntesis
14.
Mol Endocrinol ; 26(1): 153-64, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22135067

RESUMEN

Pituitary cells fire action potentials independently of external stimuli, and such spontaneous electrical activity is modulated by a large variety of hypothalamic and intrapituitary agonists. Here, we focused on the potential role of hyperpolarization-activated and cyclic nucleotide-gated (HCN) channels in electrical activity of cultured rat anterior pituitary cells. Quantitative RT-PCR analysis showed higher level of expression of mRNA transcripts for HCN2 and HCN3 subunits and lower expression of HCN1 and HCN4 subunits in these cells. Western immunoblot analysis of lysates from normal and GH(3) immortalized pituitary cells showed bands with appropriate molecular weights for HCN2, HCN3, and HCN4. Electrophysiological experiments showed the presence of a slowly developing hyperpolarization-activated inward current, which was blocked by Cs(+) and ZD7288, in gonadotrophs, thyrotrophs, somatotrophs, and a fraction of lactotrophs, as well as in other unidentified pituitary cell types. Stimulation of adenylyl cyclase and addition of 8-Br-cAMP enhanced this current and depolarized the cell membrane, whereas 8-Br-cGMP did not alter the current and hyperpolarized the cell membrane. Both inhibition of basal adenylyl cyclase activity and stimulation of phospholipase C signaling pathway inhibited this current. Inhibition of HCN channels affected the frequency of firing but did not abolish spontaneous electrical activity. These experiments indicate that cAMP and cGMP have opposite effects on the excitability of endocrine pituitary cells, that basal cAMP production in cultured cells is sufficient to integrate the majority of HCN channels in electrical activity, and that depletion of phosphatidylinositol 4,5-bisphosphate caused by activation of phospholipase C silences them.


Asunto(s)
Canales Catiónicos Regulados por Nucleótidos Cíclicos/metabolismo , Células Endocrinas/metabolismo , Adenohipófisis/metabolismo , Canales de Potasio/metabolismo , 8-Bromo Monofosfato de Adenosina Cíclica/farmacología , Potenciales de Acción/efectos de los fármacos , Adenilil Ciclasas/biosíntesis , Animales , Membrana Celular/metabolismo , Células Cultivadas , Cesio/farmacología , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacología , Canales Catiónicos Regulados por Nucleótidos Cíclicos/biosíntesis , Femenino , Gonadotrofos/metabolismo , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Lactotrofos/metabolismo , Potenciales de la Membrana/efectos de los fármacos , Fosfatidilinositol 4,5-Difosfato/deficiencia , Adenohipófisis/citología , Canales de Potasio/biosíntesis , Pirimidinas/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Somatotrofos/metabolismo , Tirotrofos/metabolismo , Fosfolipasas de Tipo C/biosíntesis , Fosfolipasas de Tipo C/metabolismo
15.
J Bioenerg Biomembr ; 43(3): 311-22, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21523406

RESUMEN

Whole-cell patch-clamp analysis revealed a resting membrane potential of -60 mV in primary osteoblasts and in the MG-63 osteoblast-like cells. Depolarization-induced action potentials were characterized by duration of 60 ms, a minimal peak-to-peak distance of 180 ms, a threshold value of -20 mV and a repolarization between the spikes to -45 mV. Expressed channels were characterized by application of voltage pulses between -150 mV and 90 mV in 10 mV steps, from a holding potential of -40 mV. Voltages below -60 mV induced an inward current. Depolarizing voltages above -30 mV evoked two currents: (a) a fast activated and inactivated inward current at voltages between -30 and 30 mV, and (b) a delayed-activated outward current that was induced by voltages above -30 mV. Electrophysiological and pharmacological parameters indicated that hyperpolarization activated strongly rectifying K(+) (K(ir)) channels, whereas depolarization activated tetrodotoxin sensitive voltage gated Na(+) (Na(v)) channels as well as delayed, slowly activated, non-inactivating, and tetraethylammonium sensitive voltage gated K(+) (K(v)) channels. In addition, RT-PCR showed expression of Na(v)1.3, Na(v)1.4, Na(v)1.5, Na(v)1.6, Na(v)1.7, and K(ir)2.1, K(ir)2.3, and K(ir)2.4 as well as K(v)2.1. We conclude that osteoblasts express channels that allow firing of action potentials.


Asunto(s)
Osteoblastos/fisiología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Neoplasias Óseas , Línea Celular , Línea Celular Tumoral , Humanos , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteosarcoma , Técnicas de Placa-Clamp , Canales de Potasio/biosíntesis , Canales de Potasio/fisiología , Canales de Sodio/biosíntesis , Canales de Sodio/fisiología
16.
Epilepsia ; 51 Suppl 3: 52-5, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20618401

RESUMEN

Cortical dysplasia is often associated with intractable seizures. Studies in animal models have described changes in inhibitory and excitatory synaptic function that contribute to hyperexcitability. The role of changes in intrinsic excitability and abnormal dendritic properties has received less attention. Changes in hyperpolarization-activated nonselective cation (HCN) channels have been implicated in several models of epilepsy. Herein we review evidence for alterations in HCN channels and dendritic morphology in the rat freeze-lesion model of cortical dysplasia. Immunocytochemical HCN1 staining, typically seen in the apical dendrites of layer V pyramidal cells in normal cortex, was greatly reduced in the region adjacent to the freeze-induced microgyrus. Although staining was preserved in layer I, fewer dendrites were stained in upper cortical layers. Deeper cortical layers were virtually devoid of immunoreactivity. Examination of biocytin-labeled pyramidal cells revealed markedly altered dendritic trees in the lesioned animals. In addition, resting membrane properties were altered and a subpopulation of neurons with abnormal dendritic arbors was present. These changes are likely to interact with the previously reported synaptic changes in this model of cortical dysplasia. HCN channel alterations are a potentially important cellular mechanism underlying hyperexcitability in cortical dysplasia.


Asunto(s)
Canales Catiónicos Regulados por Nucleótidos Cíclicos/fisiología , Epilepsia/etiología , Malformaciones del Desarrollo Cortical/patología , Canales de Potasio/fisiología , Células Piramidales/patología , Animales , Animales Recién Nacidos , Canales Catiónicos Regulados por Nucleótidos Cíclicos/biosíntesis , Modelos Animales de Enfermedad , Epilepsia/patología , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Malformaciones del Desarrollo Cortical/complicaciones , Canales de Potasio/biosíntesis , Células Piramidales/fisiopatología , Ratas , Ratas Sprague-Dawley
17.
J Appl Toxicol ; 30(6): 497-512, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20583319

RESUMEN

Ion channels play essential roles in human physiology and toxicology. Cardiac contraction, neural transmission, temperature sensing, insulin release, regulation of apoptosis, cellular pH and oxidative stress, as well as detection of active compounds from chilli, are some of the processes in which ion channels have an important role. Regulation of ion channels by several chemicals including those found in air, water and soil represents an interesting potential link between environmental pollution and human diseases; for instance, de novo expression of ion channels in response to exposure to carcinogens is being considered as a potential tool for cancer diagnosis and therapy. Non-specific binding of several drugs to ion channels is responsible for a huge number of undesirable side-effects, and testing guidelines for several drugs now require ion channel screening for pharmaceutical safety. Animal toxins targeting human ion channels have serious effects on the population and have also provided a remarkable tool to study the molecular structure and function of ion channels. In this review, we will summarize the participation of ion channels in biological processes extensively used in toxicological studies, including cardiac function, apoptosis and cell proliferation. Major findings on the adverse effects of drugs on ion channels as well as the regulation of these proteins by different chemicals, including some pesticides, are also reviewed. Association of ion channels and toxicology in several biological processes strongly suggests these proteins to be excellent candidates to follow the toxic effects of xenobiotics, and as potential early indicators of life-threatening situations including chronic degenerative diseases.


Asunto(s)
Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Contaminantes Ambientales/toxicidad , Canales Iónicos , Toxicología/métodos , Toxinas Biológicas/toxicidad , Animales , Apoptosis , Canales de Calcio/biosíntesis , Canales de Calcio/metabolismo , Canales de Calcio/fisiología , Canales de Cloruro/biosíntesis , Canales de Cloruro/metabolismo , Canales de Cloruro/fisiología , Humanos , Canales Iónicos/biosíntesis , Canales Iónicos/metabolismo , Canales Iónicos/fisiología , Neoplasias/inducido químicamente , Neoplasias/metabolismo , Neoplasias/patología , Canales de Potasio/biosíntesis , Canales de Potasio/metabolismo , Canales de Potasio/fisiología , Unión Proteica , Canales de Sodio/biosíntesis , Canales de Sodio/metabolismo , Canales de Sodio/fisiología
18.
Mol Pharmacol ; 77(3): 469-82, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20028740

RESUMEN

bTREK-1 K(+) channels set the resting membrane potential of bovine adrenal zona fasciculata (AZF) cells and function pivotally in the physiology of cortisol secretion. Adrenocorticotropic hormone controls the function and expression of bTREK-1 channels through signaling mechanisms that may involve cAMP and downstream effectors including protein kinase A (PKA) and exchange protein 2 directly activated by cAMP (Epac2). Using patch-clamp and Northern blot analysis, we explored the regulation of bTREK-1 mRNA and K(+) current expression by cAMP analogs and several of their putative metabolites in bovine AZF cells. At concentrations sufficient to activate both PKA and Epac2, 8-bromoadenosine-cAMP enhanced the expression of both bTREK-1 mRNA and K(+) current. N(6)-Benzoyladenosine-cAMP, which activates PKA but not Epac, also enhanced the expression of bTREK-1 mRNA and K(+) current measured at times from 24 to 96 h. An Epac-selective cAMP analog, 8-(4-chlorophenylthio)-2'-O-methyl-cAMP (8CPT-2'-OMe-cAMP), potently stimulated bTREK-1 mRNA and K(+) current expression, whereas the nonhydrolyzable Epac activator 8-(4-chlorophenylthio)-2'-O-methyl-cAMP, Sp-isomer was ineffective. Metabolites of 8CPT-2'-OMe-cAMP, including 8-(4-chlorophenylthio)-2'-O-methyladenosine-5'-O-monophosphate and 8CPT-2'-OMe-adenosine, promoted the expression of bTREK-1 transcripts and ion current with a temporal pattern, potency, and effectiveness resembling that of the parent compound. Likewise, at low concentrations, 8-(4-chlorophenylthio)-cAMP (8CPT-cAMP; 30 microM) but not its nonhydrolyzable analog 8-(4-chlorophenylthio)-cAMP, Sp-isomer, enhanced the expression of bTREK-1 mRNA and current. 8CPT-cAMP metabolites, including 8CPT-adenosine and 8CPT-adenine, also increased bTREK-1 expression. These results indicate that cAMP increases the expression of bTREK-1 mRNA and K(+) current through a cAMP-dependent but Epac2-independent mechanism. They further demonstrate that one or more metabolites of 8-(4-chlorophenylthio)-cAMP analogs potently stimulate bTREK-1 expression by activation of a novel cAMP-independent mechanism. These findings raise significant questions regarding the specificity of 8-(4-chlorophenylthio)-cAMP analogs as cAMP mimetics.


Asunto(s)
Corteza Suprarrenal/metabolismo , AMP Cíclico/análogos & derivados , AMP Cíclico/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Canales de Potasio de Dominio Poro en Tándem/biosíntesis , Canales de Potasio/biosíntesis , ARN Mensajero/biosíntesis , Corteza Suprarrenal/citología , Corteza Suprarrenal/efectos de los fármacos , Animales , Bovinos , AMP Cíclico/farmacología , Canales de Potasio/agonistas , Canales de Potasio de Dominio Poro en Tándem/agonistas
19.
Pflugers Arch ; 458(6): 1061-8, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19629520

RESUMEN

Thyroid hormone (TH) markedly modulates cardiovascular function and heart rate. The pacemaker current I(f) and encoding hyperpolarization-activated cation (HCN) genes have been identified as TH targets. To analyze the specific contribution and functional significance of thyroid receptor isoforms responsible for HCN gene transactivation, we generated transgenic neonatal rat cardiomyocytes with adenovirus-mediated overexpression of the thyroid receptors alpha1 (TR alpha 1) and beta1 (TR beta 1), and analyzed native I(f) current and expression levels of the underlying molecular components HCN2 and HCN4. Initial results revealed that spontaneous beating activity was higher in TR alpha 1- and lower in TR beta 1-expressing cardiomyocytes. This was associated with accelerated depolarization velocity and abbreviated action potential duration in cells overexpressing TR alpha 1, while TR beta 1 suppressed phase 4 depolarization and prolonged action potentials. Consistently, TR alpha 1-infected myocytes exhibited larger I(f) current densities along with increased HCN2 and HCN4 mRNA and protein levels. In contrast, HCN2 gene expression was not significantly affected by TR beta 1. TR beta 1 exclusively suppressed HCN4 transcription. T3 application led to significant effects only in controls and TR alpha 1-infected cardiomyocytes; whereas, no ligand-dependent actions were observed in TR beta 1-expressing neonatal cardiomyocytes. Our results demonstrate that TR alpha 1 and TR beta 1 divergently regulate cardiac pacing activity. TH-induced positive chronotropic effects are likely to be mediated by TR alpha 1 through enhanced expression of I(f) pacemaker current and its underlying genes.


Asunto(s)
Canales Catiónicos Regulados por Nucleótidos Cíclicos/genética , Corazón/fisiología , Canales Iónicos/genética , Proteínas Musculares/genética , Canales de Potasio/genética , Receptores alfa de Hormona Tiroidea/fisiología , Receptores beta de Hormona Tiroidea/fisiología , Potenciales de Acción/efectos de los fármacos , Animales , Canales Catiónicos Regulados por Nucleótidos Cíclicos/biosíntesis , Fenómenos Electrofisiológicos , Frecuencia Cardíaca/efectos de los fármacos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Canales Iónicos/biosíntesis , Proteínas Musculares/biosíntesis , Miocitos Cardíacos/metabolismo , Canales de Potasio/biosíntesis , Ratas
20.
Biochem Biophys Res Commun ; 379(4): 1048-53, 2009 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-19141293

RESUMEN

Aryl hydrocarbon receptor nuclear translocator (ARNT) has been known to participate in cellular responses to xenobiotic and hypoxic stresses, as a common partner of aryl hydrocarbon receptor and hypoxia inducible factor-1/2alpha. Recently, it was reported that ARNT is essential for adequate insulin secretion in response to glucose input and that its expression is downregulated in the pancreatic islets of diabetic patients. In the present study, the authors addressed the mechanism by which ARNT regulates insulin secretion in the INS-1 insulinoma cell line. In ARNT knock-down cells, basal insulin release was elevated, but insulin secretion was not further stimulated by a high-glucose challenge. Electrophysiological analyses revealed that glucose-dependent membrane depolarization was impaired in these cells. Furthermore, K(ATP) channel activity and expression were reduced. Of two K(ATP) channel subunits, Kir6.2 was found to be positively regulated by ARNT at the mRNA and protein levels. Based on these results, the authors suggest that ARNT expresses K(ATP) channel and by so doing regulates glucose-dependent insulin secretion.


Asunto(s)
Translocador Nuclear del Receptor de Aril Hidrocarburo/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Canales de Potasio de Rectificación Interna/biosíntesis , Canales de Potasio/biosíntesis , Animales , Translocador Nuclear del Receptor de Aril Hidrocarburo/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Línea Celular Tumoral , Técnicas de Silenciamiento del Gen , Glucosa/farmacología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/efectos de los fármacos , Insulinoma , Canales de Potasio/genética , Canales de Potasio de Rectificación Interna/genética , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA