Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros











Intervalo de año de publicación
1.
Nat Commun ; 15(1): 6683, 2024 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-39107293

RESUMEN

Focal cortical dysplasia type I (FCD I) is the most common cause of pharmaco-resistant epilepsy with the poorest prognosis. To understand the epileptogenic mechanisms of FCD I, we obtained tissue resected from patients with FCD I epilepsy, and from tumor patients as control. Using whole-cell patch clamp in acute human brain slices, we investigated the cellular properties of fast-spiking interneurons (FSINs) and pyramidal neurons (PNs) within the ictal onset zone. In FCD I epilepsy, FSINs exhibited lower firing rates from slower repolarization and action potential broadening, while PNs had increased firing. Importantly, excitatory synaptic drive of FSINs increased progressively with the scale of cortical activation as a general property across species, but this relationship was inverted towards net inhibition in FCD I epilepsy. Further comparison with intracranial electroencephalography (iEEG) from the same patients revealed that the spatial extent of pathological high-frequency oscillations (pHFO) was associated with synaptic events at FSINs.


Asunto(s)
Potenciales de Acción , Epilepsia , Interneuronas , Células Piramidales , Humanos , Interneuronas/fisiología , Femenino , Masculino , Células Piramidales/fisiología , Potenciales de Acción/fisiología , Epilepsia/fisiopatología , Adulto , Malformaciones del Desarrollo Cortical/fisiopatología , Adolescente , Adulto Joven , Niño , Técnicas de Placa-Clamp , Sinapsis/fisiología , Preescolar , Epilepsia Refractaria/fisiopatología , Epilepsia Refractaria/cirugía , Electrocorticografía
2.
Ann Neurol ; 89(4): 740-752, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33415786

RESUMEN

OBJECTIVE: Discovery of a novel antibody would enable diagnosis and early treatment of autoimmune encephalitis. The aim was to discover a novel antibody targeting a synaptic receptor and characterize the pathogenic mechanism. METHOD: We screened for unknown antibodies in serum and cerebrospinal fluid samples from autoimmune encephalitis patients. Samples with reactivity to rat brain sections and no reactivity to conventional antibody tests underwent further processing for antibody discovery, using immunoprecipitation to primary neuronal cells, mass-spectrometry analysis, an antigen-binding assay on an antigen-overexpressing cell line, and an electrophysiological assay with cultured hippocampal neurons. RESULTS: Two patients had a novel antibody against CaV α2δ (voltage-gated calcium channel alpha-2/delta subunit). The patient samples stained neuropils of the hippocampus, basal ganglia, and cortex in rat brain sections and bound to a CaV α2δ-overexpressing cell line. Knockdown of CaV α2δ expression in cultured neurons turned off the immunoreactivity of the antibody from the patients to the neurons. The patients were associated with preceding meningitis or neuroendocrine carcinoma and responded to immunotherapy. In cultured neurons, the antibody reduced neurotransmitter release from presynaptic nerve terminals by interfering with tight coupling of calcium channels and exocytosis. INTERPRETATION: Here, we discovered a novel autoimmune encephalitis associated with anti-CaV α2δ antibody. Further analysis of the antibody in autoimmune encephalitis might promote early diagnosis and treatment. ANN NEUROL 2021;89:740-752.


Asunto(s)
Canales de Calcio/inmunología , Encefalitis/inmunología , Enfermedad de Hashimoto/inmunología , Adolescente , Anciano , Animales , Anticuerpos/líquido cefalorraquídeo , Células Cultivadas , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/psicología , Encefalitis/diagnóstico , Exocitosis , Femenino , Técnicas de Silenciamiento del Gen , Enfermedad de Hashimoto/diagnóstico , Hipocampo/inmunología , Humanos , Inmunoprecipitación , Masculino , Neuronas/inmunología , Neurópilo/inmunología , Terminales Presinápticos/inmunología , Ratas
3.
Mol Brain ; 11(1): 67, 2018 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-30413218

RESUMEN

Timing and temporal precision of action potential generation are thought to be important for encoding of information in the brain. The ability of single neurons to transform their input into output action potential is primarily determined by intrinsic excitability. Particularly, plastic changes in intrinsic excitability represent the cellular substrate for spatial memory formation in CA1 pyramidal neurons (CA1-PNs). Here, we report that synaptically activated mGluR5-signaling can modulate the intrinsic excitability of CA1-PNs. Specifically, high-frequency stimulation at CA3-CA1 synapses increased firing rate and advanced spike onset with an improvement of temporal precision. These changes are mediated by mGluR5 activation that induces cADPR/RyR-dependent Ca2+ release in the dendrites of CA1-PNs, which in turn causes an increase in persistent Na+ currents (INa,P) in the dendrites. When group I mGluRs in CA1-PNs are globally activated pharmacologically, afterdepolarization (ADP) generation as well as increased firing rate are observed. These effects are abolished by inhibiting mGluR5/cADPR/RyR-dependent Ca2+ release. However, the increase in firing rate, but not the generation of ADP is affected by inhibiting INa,P. The differences between local and global activation of mGluR5-signaling in CA1-PNs indicates that mGluR5-dependent modulation of intrinsic excitability is highly compartmentalized and a variety of ion channels are recruited upon their differential subcellular localizations. As mGluR5 activation is induced by physiologically plausible brief high-frequency stimulation at CA3-CA1 synapses, our results suggest that mGluR5-induced enhancement of dendritic INa,P in CA1-PNs may provide important implications for our understanding about place field formation in the hippocampus.


Asunto(s)
Potenciales de Acción , Dendritas/metabolismo , Receptor del Glutamato Metabotropico 5/metabolismo , Canales de Sodio/metabolismo , Potenciales de Acción/efectos de los fármacos , Animales , Región CA1 Hipocampal/efectos de los fármacos , Región CA1 Hipocampal/fisiología , Calcio/metabolismo , ADP-Ribosa Cíclica/metabolismo , Dendritas/efectos de los fármacos , Estimulación Eléctrica , Metoxihidroxifenilglicol/análogos & derivados , Metoxihidroxifenilglicol/farmacología , Ratas Sprague-Dawley , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
4.
J Neurosci ; 35(50): 16479-93, 2015 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-26674872

RESUMEN

Expression of neuregulin-2 (NRG2) is intense in a few regions of the adult brain where neurogenesis persists; however, little is understood about its role in developments of newborn neurons. To study the role of NRG2 in synaptogenesis at different developmental stages, newborn granule cells in rat hippocampal slice cultures were labeled with retrovirus encoding tetracycline-inducible microRNA targeting NRG2 and treated with doxycycline (Dox) at the fourth or seventh postinfection day (dpi). The developmental increase of GABAergic postsynaptic currents (GPSCs) was suppressed by the early Dox treatment (4 dpi), but not by late treatment (7 dpi). The late Dox treatment was used to study the effect of NRG2 depletion specific to excitatory synaptogenesis. The Dox effect on EPSCs emerged 4 d after the impairment in dendritic outgrowth became evident (10 dpi). Notably, Dox treatment abolished the developmental increases of AMPA-receptor mediated EPSCs and the AMPA/NMDA ratio, indicating impaired maturation of glutamatergic synapses. In contrast to GPSCs, Dox effects on EPSCs and dendritic growth were independent of ErbB4 and rescued by concurrent overexpression of NRG2 intracellular domain. These results suggest that forward signaling of NRG2 mediates GABAergic synaptogenesis and its reverse signaling contributes to dendritic outgrowth and maturation of glutamatergic synapses. SIGNIFICANCE STATEMENT: The hippocampal dentate gyrus is one of special brain regions where neurogenesis persists throughout adulthood. Synaptogenesis is a critical step for newborn neurons to be integrated into preexisting neural network. Because neuregulin-2 (NRG2), a growth factor, is intensely expressed in these regions, we investigated whether it plays a role in synaptogenesis and dendritic growth. We found that NRG2 has dual roles in the development of newborn neurons. For GABAergic synaptogenesis, the extracellular domain of NRG2 acts as a ligand for a receptor on GABAergic neurons. In contrast, its intracellular domain was essential for dendritic outgrowth and glutamatergic synapse maturation. These results imply that NRG2 may play a critical role in network integration of newborn neurons.


Asunto(s)
Glutamatos/fisiología , Hipocampo/citología , Hipocampo/fisiología , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/fisiología , Sinapsis/fisiología , Ácido gamma-Aminobutírico/fisiología , Animales , Animales Recién Nacidos , Dendritas/efectos de los fármacos , Doxiciclina/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Femenino , Técnicas de Silenciamiento del Gen , Hipocampo/crecimiento & desarrollo , Masculino , Ratas , Ratas Sprague-Dawley , Receptor ErbB-4/genética , Receptor ErbB-4/metabolismo , Receptores AMPA/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/efectos de los fármacos
5.
Pflugers Arch ; 467(12): 2461-72, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26100674

RESUMEN

Na(+)/Ca(2+) exchangers are key players for Ca(2+) clearance in pancreatic ß-cells, but their molecular determinants and roles in insulin secretion are not fully understood. In the present study, we newly discovered that the Li(+)-permeable Na(+)/Ca(2+) exchangers (NCLX), which were known as mitochondrial Na(+)/Ca(2+) exchangers, contributed to the Na(+)-dependent Ca(2+) movement across the plasma membrane in rat INS-1 insulinoma cells. Na(+)/Ca(2+) exchange activity by NCLX was comparable to that by the Na(+)/Ca(2+) exchanger, NCX. We also confirmed the presence of NCLX proteins on the plasma membrane using immunocytochemistry and cell surface biotinylation experiments. We further investigated the role of NCLX on exocytosis function by measuring the capacitance increase in response to repetitive depolarization. Small interfering (si)RNA-mediated downregulation of NCLX did not affect the initial exocytosis, but significantly suppressed sustained exocytosis and recovery of exocytosis. XIP (NCX inhibitory peptide) or Na(+) replacement for inhibiting Na(+)-dependent Ca(2+) clearance also selectively suppressed sustained exocytosis. Consistent with the idea that sustained exocytosis requires ATP-dependent vesicle recruitment, mitochondrial function, assessed by mitochondrial membrane potential (ΔΨ), was impaired by siNCLX or XIP. However, depolarization-induced exocytosis was hardly affected by changes in intracellular Na(+) concentration, suggesting a negligible contribution of mitochondrial Na(+)/Ca(2+) exchanger. Taken together, our data indicate that Na(+)/Ca(2+) exchanger-mediated Ca(2+) clearance mediated by NCLX and NCX is crucial for optimizing mitochondrial function, which in turn contributes to vesicle recruitment for sustained exocytosis in pancreatic ß-cells.


Asunto(s)
Calcio/metabolismo , Membrana Celular/metabolismo , Exocitosis , Células Secretoras de Insulina/metabolismo , Litio/metabolismo , Intercambiador de Sodio-Calcio/metabolismo , Potenciales de Acción , Animales , Línea Celular Tumoral , Células Cultivadas , Células Secretoras de Insulina/efectos de los fármacos , Potencial de la Membrana Mitocondrial , Ratones , Ratones Endogámicos C57BL , Péptidos/farmacología , Ratas , Intercambiador de Sodio-Calcio/genética
6.
J Nat Prod ; 78(3): 363-7, 2015 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-25689430

RESUMEN

Two new benzophenones, acredinones A (1) and B (2), were isolated from a marine-sponge-associated Acremonium sp. fungus. Their chemical structures were elucidated on the interpretation of spectroscopic data. The structure of 1 was confirmed by palladium-catalyzed hydrogenation, followed by spectroscopic data analysis. Acredinones A (1) and B (2) inhibited the outward K(+) currents of the insulin secreting cell line INS-1 with IC50 values of 0.59 and 1.0 µM, respectively.


Asunto(s)
Acremonium/química , Benzofenonas/aislamiento & purificación , Benzofenonas/farmacología , Poríferos/microbiología , Bloqueadores de los Canales de Potasio/aislamiento & purificación , Bloqueadores de los Canales de Potasio/farmacología , Animales , Benzofenonas/química , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Concentración 50 Inhibidora , Insulina/metabolismo , Secreción de Insulina , Estructura Molecular , Resonancia Magnética Nuclear Biomolecular , Bloqueadores de los Canales de Potasio/química
7.
Biochem Biophys Res Commun ; 440(4): 539-44, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24103758

RESUMEN

Leptin regulates pancreatic ß-cell excitability through AMP-activated protein kinase (AMPK)-mediated ATP-sensitive potassium (KATP) channel trafficking. However, the signaling components connecting AMPK to KATP channel trafficking are not identified. In this study, we discovered that AMPK inhibits phosphatase and tensin homologue (PTEN) via glycogen synthase kinase 3ß (GSK3ß) and this signaling pathway is crucial for KATP channel trafficking in leptin-treated pancreatic ß-cells. Pharmacologic or genetic inhibition of AMPK or GSK3ß, but not casein kinase 2 (CK2), impaired leptin-induced PTEN inactivation and thereby KATP channel trafficking. The PTEN mutant lacking both protein and lipid phosphatase activity is sufficient to induce KATP channel trafficking without leptin. These results present a novel signaling mechanism that underlies leptin regulation of KATP channel trafficking in pancreatic ß-cells. Our findings assist in gaining a broader perspective on the peripheral action of leptin on pancreatic ß-cell physiology and glucose homeostasis.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Células Secretoras de Insulina/metabolismo , Canales KATP/metabolismo , Leptina/fisiología , Fosfohidrolasa PTEN/antagonistas & inhibidores , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Proteínas Quinasas Activadas por AMP/genética , Animales , Glucosa/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Homeostasis , Células Secretoras de Insulina/efectos de los fármacos , Leptina/farmacología , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Biosíntesis de Proteínas , Ratas
8.
Proc Natl Acad Sci U S A ; 110(31): 12673-8, 2013 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-23858470

RESUMEN

Leptin is a pivotal regulator of energy and glucose homeostasis, and defects in leptin signaling result in obesity and diabetes. The ATP-sensitive potassium (K(ATP)) channels couple glucose metabolism to insulin secretion in pancreatic ß-cells. In this study, we provide evidence that leptin modulates pancreatic ß-cell functions by promoting K(ATP) channel translocation to the plasma membrane via AMP-activated protein kinase (AMPK) signaling. K(ATP) channels were localized mostly to intracellular compartments of pancreatic ß-cells in the fed state and translocated to the plasma membrane in the fasted state. This process was defective in leptin-deficient ob/ob mice, but restored by leptin treatment. We discovered that the molecular mechanism of leptin-induced AMPK activation involves canonical transient receptor potential 4 and calcium/calmodulin-dependent protein kinase kinase ß. AMPK activation was dependent on both leptin and glucose concentrations, so at optimal concentrations of leptin, AMPK was activated sufficiently to induce K(ATP) channel trafficking and hyperpolarization of pancreatic ß-cells in a physiological range of fasting glucose levels. There was a close correlation between phospho-AMPK levels and ß-cell membrane potentials, suggesting that AMPK-dependent K(ATP) channel trafficking is a key mechanism for regulating ß-cell membrane potentials. Our results present a signaling pathway whereby leptin regulates glucose homeostasis by modulating ß-cell excitability.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Células Secretoras de Insulina/metabolismo , Leptina/metabolismo , Potenciales de la Membrana/fisiología , Transducción de Señal/fisiología , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Glucosa/metabolismo , Homeostasis/fisiología , Células Secretoras de Insulina/citología , Leptina/genética , Ratones , Ratones Obesos , Transporte de Proteínas/fisiología , ATPasa Intercambiadora de Sodio-Potasio/genética , Canales Catiónicos TRPC/genética , Canales Catiónicos TRPC/metabolismo
9.
Mol Ther ; 20(9): 1750-66, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22713697

RESUMEN

Loss of cardiomyocytes impairs cardiac function after myocardial infarction (MI). Recent studies suggest that cardiac stem/progenitor cells could repair the damaged heart. However, cardiac progenitor cells are difficult to maintain in terms of purity and multipotency when propagated in two-dimensional culture systems. Here, we investigated a new strategy that enhances potency and enriches progenitor cells. We applied the repeated sphere formation strategy (cardiac explant → primary cardiosphere (CS) formation → sphere-derived cells (SDCs) in adherent culture condition → secondary CS formation by three-dimensional culture). Cells in secondary CS showed higher differentiation potentials than SDCs. When transplanted into the infarcted myocardium, secondary CSs engrafted robustly, improved left ventricular (LV) dysfunction, and reduced infarct sizes more than SDCs did. In addition to the cardiovascular differentiation of transplanted secondary CSs, robust vascular endothelial growth factor (VEGF) synthesis and secretion enhanced neovascularization in the infarcted myocardium. Microarray pathway analysis and blocking experiments using E-selectin knock-out hearts, specific chemicals, and small interfering RNAs (siRNAs) for each pathway revealed that E-selectin was indispensable to sphere initiation and ERK/Sp1/VEGF autoparacrine loop was responsible for sphere maturation. These results provide a simple strategy for enhancing cellular potency for cardiac repair. Furthermore, this strategy may be implemented to other types of stem/progenitor cell-based therapy.


Asunto(s)
Técnicas de Cultivo de Célula , Infarto del Miocardio/terapia , Trasplante de Células Madre , Células Madre/citología , Disfunción Ventricular Izquierda/terapia , Animales , Diferenciación Celular , Selectina E/genética , Selectina E/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Expresión Génica , Humanos , Inmunoglobulinas/genética , Inmunoglobulinas/metabolismo , Masculino , Ratones , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Neovascularización Fisiológica , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Interferente Pequeño/genética , Transducción de Señal , Células Madre/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Disfunción Ventricular Izquierda/metabolismo , Disfunción Ventricular Izquierda/patología
10.
J Neurosci ; 32(17): 5953-63, 2012 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-22539855

RESUMEN

Alzheimer's disease (AD) in the early stages is characterized by memory impairment, which may be attributable to synaptic dysfunction. Oxidative stress, mitochondrial dysfunction, and Ca²âº dysregulation are key factors in the pathogenesis of AD, but the causal relationship between these factors and synaptic dysfunction is not clearly understood. We found that in the hippocampus of an AD mouse model (Tg2576), mitochondrial Ca²âº handling in dentate granule cells was impaired as early as the second postnatal month, and this Ca²âº dysregulation caused an impairment of post-tetanic potentiation in mossy fiber-CA3 synapses. The alteration of cellular Ca²âº clearance in Tg2576 mice is region-specific within hippocampus because in another region, CA1 pyramidal neuron, no significant difference in Ca²âº clearance was detected between wild-type and Tg2576 mice at this early stage. Impairment of mitochondrial Ca²âº uptake was associated with increased mitochondrial reactive oxygen species and depolarization of mitochondrial membrane potential. Mitochondrial dysfunctions in dentate granule cells and impairment of post-tetanic potentiation in mossy fiber-CA3 synapses were fully restored when brain slices obtained from Tg2576 were pretreated with antioxidant, suggesting that mitochondrial oxidative stress initiates other dysfunctions. Reversibility of early dysfunctions by antioxidants at the preclinical stage of AD highlights the importance of early diagnosis and antioxidant therapy to delay or prevent the disease processes.


Asunto(s)
Enfermedad de Alzheimer/patología , Giro Dentado/patología , Mitocondrias/patología , Fibras Musgosas del Hipocampo/fisiopatología , Plasticidad Neuronal/fisiología , Neuronas/ultraestructura , Transmisión Sináptica/fisiología , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/farmacología , Precursor de Proteína beta-Amiloide/genética , Animales , Animales Modificados Genéticamente , Antioxidantes/farmacología , Biofisica , Calcio/metabolismo , Cromanos/farmacología , Giro Dentado/metabolismo , Modelos Animales de Enfermedad , Interacciones Farmacológicas , Estimulación Eléctrica , Inhibidores Enzimáticos/farmacología , Ensayo de Inmunoadsorción Enzimática , Humanos , Técnicas In Vitro , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Potencial de la Membrana Mitocondrial/genética , Ratones , Mutación/genética , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/genética , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Técnicas de Placa-Clamp , Fragmentos de Péptidos/farmacología , ATPasas Transportadoras de Calcio de la Membrana Plasmática/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Compuestos de Rutenio/farmacología , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Intercambiador de Sodio-Calcio/metabolismo , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/genética
11.
PLoS One ; 6(10): e26625, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22028929

RESUMEN

Group I metabotropic glutamate receptors (group I mGluRs; mGluR1 and mGluR5) exert diverse effects on neuronal and synaptic functions, many of which are regulated by intracellular Ca(2+). In this study, we characterized the cellular mechanisms underlying Ca(2+) mobilization induced by (RS)-3,5-dihydroxyphenylglycine (DHPG; a specific group I mGluR agonist) in the somata of acutely dissociated rat hippocampal neurons using microfluorometry. We found that DHPG activates mGluR5 to mobilize intracellular Ca(2+) from ryanodine-sensitive stores via cyclic adenosine diphosphate ribose (cADPR), while the PLC/IP(3) signaling pathway was not involved in Ca(2+) mobilization. The application of glutamate, which depolarized the membrane potential by 28.5±4.9 mV (n = 4), led to transient Ca(2+) mobilization by mGluR5 and Ca(2+) influx through L-type Ca(2+) channels. We found no evidence that mGluR5-mediated Ca(2+) release and Ca(2+) influx through L-type Ca(2+) channels interact to generate supralinear Ca(2+) transients. Our study provides novel insights into the mechanisms of intracellular Ca(2+) mobilization by mGluR5 in the somata of hippocampal neurons.


Asunto(s)
Calcio/metabolismo , ADP-Ribosa Cíclica/metabolismo , Hipocampo/citología , Neuronas/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Animales , Canales de Calcio Tipo L/metabolismo , Fura-2/análogos & derivados , Fura-2/metabolismo , Ácido Glutámico/farmacología , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Potenciales de la Membrana/efectos de los fármacos , Neuronas/citología , Neuronas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptor del Glutamato Metabotropico 5 , Receptores AMPA/metabolismo , Rianodina/farmacología , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Transducción de Señal/efectos de los fármacos
12.
J Biol Chem ; 285(14): 10939-50, 2010 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-20123983

RESUMEN

M-type (KCNQ) potassium channels play an important role in regulating the action potential firing in neurons. Here, we investigated the effect of cholesterol on M current in superior cervical ganglion (SCG) sympathetic neurons, using the patch clamp technique. M current was inhibited in a dose-dependent manner by cholesterol loading with a methyl-beta-cyclodextrin-cholesterol complex. This effect was prevented when membrane cholesterol level was restored by including empty methyl-beta-cyclodextrin in the pipette solution. Dialysis of cells with AMP-PNP instead of ATP prevented cholesterol action on M currents. Protein kinase C (PKC) inhibitor, calphostin C, abolished cholesterol-induced inhibition whereas the PKC activator, PDBu, mimicked the inhibition of M currents by cholesterol. The in vitro kinase assay showed that KCNQ2 subunits of M channel can be phosphorylated by PKC. A KCNQ2 mutant that is defective in phosphorylation by PKC failed to show current inhibition not only by PDBu but also by cholesterol. These results indicate that cholesterol-induced inhibition of M currents is mediated by PKC phosphorylation. The inhibition of M currents by PDBu and cholesterol was completely blocked by PIP(2) loading, indicating that the decrease in PIP(2)-channel interaction underlies M channel inhibition by PKC-mediated phosphorylation. We conclude that cholesterol specifically regulates M currents in SCG neurons via PKC activation.


Asunto(s)
Colesterol/farmacología , Canal de Potasio KCNQ2/antagonistas & inhibidores , Riñón/efectos de los fármacos , Neuronas/efectos de los fármacos , Proteína Quinasa C/metabolismo , Ganglio Cervical Superior/efectos de los fármacos , Potenciales de Acción , Adenosina Trifosfato/farmacología , Adenilil Imidodifosfato/farmacología , Animales , Células Cultivadas , Electrofisiología , Humanos , Activación del Canal Iónico , Canal de Potasio KCNQ2/genética , Canal de Potasio KCNQ2/metabolismo , Riñón/citología , Riñón/metabolismo , Neuronas/citología , Neuronas/metabolismo , Técnicas de Placa-Clamp , Fosforilación/efectos de los fármacos , Ratas , Ganglio Cervical Superior/citología , Ganglio Cervical Superior/metabolismo , beta-Ciclodextrinas/farmacología
13.
J Mol Cell Cardiol ; 48(4): 773-80, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19883656

RESUMEN

The Ca(2+)-dependent facilitation (CDF) of L-type Ca(2+) channels, a major mechanism for force-frequency relationship of cardiac contraction, is mediated by Ca(2+)/CaM-dependent kinase II (CaMKII). Recently, CaMKII was shown to be activated by methionine oxidation. We investigated whether oxidation-dependent CaMKII activation is involved in the regulation of L-type Ca(2+) currents (I(Ca,L)) by H(2)O(2) and whether Ca(2+) is required in this process. Using patch clamp, I(Ca)(,L) was measured in rat ventricular myocytes. H(2)O(2) induced an increase in I(Ca,L) amplitude and slowed inactivation of I(Ca)(,L). This oxidation-dependent facilitation (ODF) of I(Ca)(,L) was abolished by a CaMKII blocker KN-93, but not by its inactive analog KN-92, indicating that CaMKII is involved in ODF. ODF was not affected by replacement of external Ca(2+) with Ba(2+) or presence of EGTA in the internal solutions. However, ODF was abolished by adding BAPTA to the internal solution or by depleting sarcoplasmic reticulum (SR) Ca(2+) stores using caffeine and thapsigargin. Alkaline phosphatase, beta-iminoadenosine 5'-triphosphate (AMP-PNP), an autophosphorylation inhibitor autocamtide-2-related inhibitory peptide (AIP), or a catalytic domain blocker (CaM-KIINtide) did not affect ODF. In conclusion, oxidation-dependent facilitation of L-type Ca(2+) channels is mediated by oxidation-dependent CaMKII activation, in which local Ca(2+) increases induced by SR Ca(2+) release is required.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Ventrículos Cardíacos/citología , Peróxido de Hidrógeno/química , Células Musculares/citología , Adenilil Imidodifosfato/química , Fosfatasa Alcalina/metabolismo , Animales , Bencilaminas/farmacología , Cafeína/química , Dominio Catalítico , Masculino , Oxígeno/química , Ratas , Ratas Sprague-Dawley , Sulfonamidas/farmacología , Tapsigargina/química
14.
Diabetes ; 58(12): 2813-9, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19720793

RESUMEN

OBJECTIVE: AMP-activated protein kinase (AMPK) and the ATP-sensitive K(+) (K(ATP)) channel are metabolic sensors that become activated during metabolic stress. AMPK is an important regulator of metabolism, whereas the K(ATP) channel is a regulator of cellular excitability. Cross talk between these systems is poorly understood. RESEARCH DESIGN AND METHODS: Rat pancreatic beta-cells or INS-1 cells were pretreated for 2 h at various concentrations of glucose. Maximum K(ATP) conductance (G(max)) was monitored by whole-cell measurements after intracellular ATP washout using ATP-free internal solutions. K(ATP) channel activity (NPo) was monitored by inside-out patch recordings in the presence of diazoxide. Distributions of K(ATP) channel proteins (Kir6.2 and SUR1) were examined using immunofluorescence imaging and surface biotinylation studies. Insulin secretion from rat pancreatic islets was measured using an enzyme immunoassay. RESULTS: G(max) and NPo in cells pretreated with glucose-free or 3 mmol/l glucose solutions were significantly higher than in cells pretreated in 11.1 mmol/l glucose solutions. Immunofluorescence imaging and biotinylation studies revealed that glucose deprivation induced an increase in the surface level of Kir6.2 without affecting the total cellular amount. Increases in G(max) and the surface level of Kir6.2 were inhibited by compound C, an AMPK inhibitor, and siAMPK transfection. The effects of glucose deprivation on K(ATP) channels were mimicked by an AMPK activator. Glucose deprivation reduced insulin secretion, but this response was attenuated by compound C. CONCLUSIONS: K(ATP) channel trafficking is regulated by energy status via AMPK, and this mechanism may play a key role in inhibiting insulin secretion under low energy status.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Glucosa/deficiencia , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Canales KATP/metabolismo , Animales , Biotinilación , Western Blotting , Electrofisiología , Técnica del Anticuerpo Fluorescente , Glucosa/metabolismo , Técnicas para Inmunoenzimas , Secreción de Insulina , Células Secretoras de Insulina/enzimología , Ratas
15.
Cell Calcium ; 45(5): 465-73, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19362367

RESUMEN

Calcium is a key regulator for expression of genes relevant to survival and maturation of newborn neurons. Mammalian hippocampal dentate gyrus generates new granule cells (GCs) throughout adult life. We identified young and mature GCs in hippocampi of young adult mice according to their electrical properties, and investigated contributions of Na/Ca exchanger (NCX), sarco-endoplasmic reticulum Ca(2+)-ATPase (SERCA), plasma membrane Ca(2+)-ATPase (PMCA) and mitochondria to Ca(2+) clearance in somata of GCs. Somatic Ca(2+) clearance was increased by about 50% as GCs matured. NCX activity increased proportionally during maturation with its relative contribution kept about 40% both in young and mature GCs. On the other hand, the developmental increases in activities of mitochondria and SERCA resulted in higher contributions to Ca(2+) clearance in mature GCs than in young GCs. Especially mitochondrial function was most highly enhanced during maturation. PMCA activity, however, did not increase during maturation. Low Ca(2+) clearance in immature GCs might facilitate higher Ca(2+) accumulation during network activity, which in turn help survival of young GCs.


Asunto(s)
Calcio/metabolismo , Hipocampo/citología , Neuronas/metabolismo , Animales , Calbindinas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/citología , Técnicas de Placa-Clamp , ATPasas Transportadoras de Calcio de la Membrana Plasmática/metabolismo , Proteína G de Unión al Calcio S100/genética , Proteína G de Unión al Calcio S100/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Intercambiador de Sodio-Calcio/metabolismo
16.
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
17.
Stem Cells ; 26(7): 1901-12, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18403756

RESUMEN

Stem cell transplantation in acute myocardial infarction (AMI) has emerged as a promising therapeutic option. We evaluated the impact of AMI on mesenchymal stem cell (MSC) differentiation into cardiomyocyte lineage. Cord blood-derived human MSCs were exposed to in vitro conditions simulating in vivo environments of the beating heart with acute ischemia, as follows: (a) myocardial proteins or serum obtained from sham-operated rats, and (b) myocardial proteins or serum from AMI rats, with or without application of oscillating pressure. Expression of cardiac-specific markers on MSCs was greatly induced by the infarcted myocardial proteins, compared with the normal proteins. It was also induced by application of oscillating pressure to MSCs. Treatment of MSCs with infarcted myocardial proteins and oscillating pressure greatly augmented expression of cardiac-specific genes. Such expression was blocked by inhibitor of transforming growth factor beta(1) (TGF-beta(1)) or bone morphogenetic protein-2 (BMP-2). In vitro cellular and electrophysiologic experiments showed that these differentiated MSCs expressing cardiomyocyte-specific markers were able to make a coupling with cardiomyocytes but not to selfbeat. The pathophysiologic significance of in vitro results was confirmed using the rat AMI model. The protein amount of TGF-beta(1) and BMP-2 in myocardium of AMI was significantly higher than that in normal myocardium. When MSCs were transplanted to the heart and analyzed 8 weeks later, they expressed cardiomyocyte-specific markers, leading to improved cardiac function. These in vitro and in vivo results suggest that infarct-related biological and physical factors in AMI induce commitment of MSCs to cardiomyocyte-like cells through TGF-beta/BMP-2 pathways.


Asunto(s)
Células Madre Mesenquimatosas/citología , Infarto del Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Células Madre/citología , Animales , Proteína Morfogenética Ósea 2 , Proteínas Morfogenéticas Óseas/metabolismo , Calcio/metabolismo , Diferenciación Celular , Uniones Comunicantes/metabolismo , Humanos , Oscilometría , Fenotipo , Ratas , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Venas Umbilicales/metabolismo
18.
Front Biosci ; 12: 1642-50, 2007 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-17127410

RESUMEN

Two-pore domain K+ (K2P) channels play a critical role in cellular responses to various stimuli, such as stretch or changes in pH and are considered to be important in pathological responses such as apoptosis and tumorigenesis. We investigated effects of H2O2 on various K2P channels expressed in CHO cells. Application of H2O2 did not affect TASK-1, TASK-3, TRAAK currents, but specifically increased TREK-2 currents recorded using a nystatin perforated whole cell technique. The H2O2-induced activation of TREK-2 currents was also observed at single channel levels in cell-attached patches, and the effect was reversed by the reducing agent, dithiothreitol. In contrast, TREK-2 currents recorded using ruptured whole cell technique or single channel recording in inside-out patches were not affected by H2O2. Furthermore, direct application of 5,5'-dithiobis-(2-nitrobenzoic acid) inhibited TREK-2, suggesting that the H2O2-induced activation does not result from direct oxidation of TREK-2 proteins. Among the cell signaling agents, myosin light chain kinase (MLCK) inhibitors significantly inhibited the H2O2-induced activation of TREK-2 currents. These results suggest that TREK-2 channels have a potential to play a specific role in cellular responses to reactive oxygen species and that MLCK activation is involved in this process.


Asunto(s)
Peróxido de Hidrógeno/farmacología , Quinasa de Cadena Ligera de Miosina/metabolismo , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Animales , Células CHO , Cricetinae , Cricetulus , Conductividad Eléctrica , Proteínas del Tejido Nervioso/metabolismo , Técnicas de Placa-Clamp , Transducción de Señal
19.
Arch Pharm Res ; 29(4): 310-7, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16681037

RESUMEN

We investigated the effects of trinitrobenzene sulfonic acid (TNBS), an amino-group reagent, on the human ether-a-go-go-related gene (HERG) K+ channels expressed in Xenopus oocytes. TNBS neutralizes the positively charged amino-groups of peptide N-terminal and lysine residues. External application of TNBS at 10 mM for 5 min irreversibly shifted the curves for currents at the end of the pulse and tail currents of HERG to a more negative potential and decreased the maximal amplitude of the I(tail) curve (I(tail,max)). TNBS had little effect on either the activated current-voltage relationship or the reversal potential of HERG current, indicating that TNBS did not change ion selectivity properties. TNBS shifted the time constant curves of both activation and deactivation of the HERG current to a more hyperpolarized potential; TNBS's effect was greater on channel opening than channel closing. External H+ is known to inhibit HERG current by shifting V(1/2) to the right and decreasing I(tail,max). TNBS enhanced the blockade of external H+ by exaggerating the effect of H+ on I(tail,max), not on V(1/2). Our data provide evidence for the presence of essential amino-groups that are associated with the normal functioning of the HERG channel and evidence that these groups modify the blocking effect of external H+ on the current.


Asunto(s)
Canales de Potasio Éter-A-Go-Go/genética , Ácido Trinitrobencenosulfónico/farmacología , Animales , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go/biosíntesis , Canales de Potasio Éter-A-Go-Go/química , Humanos , Concentración de Iones de Hidrógeno , Técnicas In Vitro , Activación del Canal Iónico/efectos de los fármacos , Lisina/química , Potenciales de la Membrana , Oocitos/metabolismo , Técnicas de Placa-Clamp , Transfección , Xenopus laevis
20.
Am J Physiol Endocrinol Metab ; 287(5): E1008-18, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15265758

RESUMEN

A low-taurine diet during fetal or early postnatal life causes abnormal pancreatic beta-cell development. Tissue and plasma taurine concentrations can also be low in diabetic patients. We examined the effect of taurine on impaired glucose responses in diabetic rat beta-cells adenovirally overexpressing uncoupling protein (UCP)2, which is upregulated in obesity-related type 2 diabetes. We found that taurine pretreatment restored the ATP-to-ADP (ATP/ADP) ratio and glucose-stimulated insulin secretion in UCP2-infected islets. ATP-sensitive K(+) channel sensitivity to dihydroxyacetone, another insulin secretagogue, was similar in both UCP2-infected and control beta-cells. In freshly isolated mitochondria from UCP2-overexpressing insulin-secreting (INS)-1 beta-cells, methyl pyruvate-mediated mitochondrial Ca(2+) increase was significantly ameliorated by taurine. A mitochondrial Ca(2+) uniporter blocker, ruthenium red, inhibited the action of taurine. This study suggests that taurine enhances the glucose sensitivity of UCP2-overexpressing beta-cells, probably by increasing mitochondrial Ca(2+) influx through the Ca(2+) uniporter, thereby enhancing mitochondrial metabolic function and increasing the ATP/ADP ratio.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Intolerancia a la Glucosa/metabolismo , Glucosa/metabolismo , Islotes Pancreáticos/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Taurina/fisiología , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Calcio/metabolismo , Células Cultivadas , Regulación de la Expresión Génica , Insulina/metabolismo , Secreción de Insulina , Canales Iónicos , Islotes Pancreáticos/citología , Masculino , Potenciales de la Membrana/fisiología , Proteínas de Transporte de Membrana/genética , Proteínas Mitocondriales/genética , Canales de Potasio/metabolismo , Ratas , Ratas Sprague-Dawley , Transfección , Proteína Desacopladora 2
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA