Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Pflugers Arch ; 476(2): 229-242, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38036776

RESUMEN

Loss-of-function variants of SCN5A, encoding the sodium channel alpha subunit Nav1.5 are associated with high phenotypic variability and multiple cardiac presentations, while underlying mechanisms are incompletely understood. Here we investigated a family with individuals affected by Brugada Syndrome (BrS) of different severity and aimed to unravel the underlying genetic and electrophysiological basis.Next-generation sequencing was used to identify the genetic variants carried by family members. The index patient, who was severely affected by arrhythmogenic BrS, carried previously uncharacterized variants of Nav1.5 (SCN5A-G1661R) and glycerol-3-phosphate dehydrogenase-1-like protein (GPD1L-A306del) in a double heterozygous conformation. Family members exclusively carrying SCN5A-G1661R showed asymptomatic Brugada ECG patterns, while another patient solely carrying GPD1L-A306del lacked any clinical phenotype.To assess functional mechanisms, Nav1.5 channels were transiently expressed in HEK-293 cells in the presence and absence of GPD1L. Whole-cell patch-clamp recordings revealed loss of sodium currents after homozygous expression of SCN5A-G1661R, and reduction of current amplitude to ~ 50% in cells transfected with equal amounts of wildtype and mutant Nav1.5. Co-expression of wildtype Nav1.5 and GPD1L showed a trend towards increased sodium current amplitudes and a hyperpolarizing shift in steady-state activation and -inactivation compared to sole SCN5A expression. Application of the GPD1L-A306del variant shifted steady-state activation to more hyperpolarized and inactivation to more depolarized potentials.In conclusion, SCN5A-G1661R produces dysfunctional channels and associates with BrS. SCN5A mediated currents are modulated by co-expression of GDP1L and this interaction is altered by mutations in both proteins. Thus, additive genetic burden may aggravate disease severity, explaining higher arrhythmogenicity in double mutation carriers.


Asunto(s)
Síndrome de Brugada , Humanos , Síndrome de Brugada/genética , Síndrome de Brugada/metabolismo , Sodio/metabolismo , Células HEK293 , Mutación , Fenotipo , Canal de Sodio Activado por Voltaje NAV1.5/genética , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo
2.
Neurobiol Dis ; 187: 106306, 2023 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-37734623

RESUMEN

Glial glutamate transporters actively participate in neurotransmission and have a fundamental role in determining the ambient glutamate concentration in the extracellular space. Their expression is dynamically regulated in many diseases, including experimental autoimmune encephalomyelitis (EAE), a model of multiple sclerosis. In EAE, a downregulation has been reported which may render neurons more susceptible to glutamate excitotoxicity. In this study, we have investigated the expression of GLAST (EAAT1) and GLT-1 (EAAT2) in the retina of Brown Norway rats following induction of myelin oligodendrocyte glycoprotein (MOG)-EAE, which results in retinal ganglion cell (RGC) degeneration and dysfunction. In addition, we tested whether AAV-mediated overexpression of GLAST in the retina can protect RGCs from degeneration. To address the impact of glutamate transporter modulation on RGCs, we performed whole-cell recordings and measured tonic NMDA receptor-mediated currents in the absence and presence of a glutamate-uptake blocker. We report that αOFF-RGCs show larger tonic glutamate-induced currents than αON-RGCs, in line with their greater vulnerability under neuroinflammatory conditions. We further show that increased AAV-mediated expression of GLAST in the retina does indeed protect RGCs from degeneration during the inflammatory disease. Collectively, our study highlights the neuroprotective role of glutamate transporters in the EAE retina and provides a characterization of tonic glutamate-currents of αRGCs. The larger effects of increased extracellular glutamate concentration on the αOFF-subtype may underlie its enhanced vulnerability to degeneration.

3.
Basic Res Cardiol ; 116(1): 13, 2021 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-33630168

RESUMEN

Atrial fibrillation (AF) is associated with electrical remodeling, leading to cellular electrophysiological dysfunction and arrhythmia perpetuation. Emerging evidence suggests a key role for epigenetic mechanisms in the regulation of ion channel expression. Histone deacetylases (HDACs) control gene expression through deacetylation of histone proteins. We hypothesized that class I HDACs in complex with neuron-restrictive silencer factor (NRSF) determine atrial K+ channel expression. AF was characterized by reduced atrial HDAC2 mRNA levels and upregulation of NRSF in humans and in a pig model, with regional differences between right and left atrium. In vitro studies revealed inverse regulation of Hdac2 and Nrsf in HL-1 atrial myocytes. A direct association of HDAC2 with active regulatory elements of cardiac K+ channels was revealed by chromatin immunoprecipitation. Specific knock-down of Hdac2 and Nrsf induced alterations of K+ channel expression. Hdac2 knock-down resulted in prolongation of action potential duration (APD) in neonatal rat cardiomyocytes, whereas inactivation of Nrsf induced APD shortening. Potential AF-related triggers were recapitulated by experimental tachypacing and mechanical stretch, respectively, and exerted differential effects on the expression of class I HDACs and K+ channels in cardiomyocytes. In conclusion, HDAC2 and NRSF contribute to AF-associated remodeling of APD and K+ channel expression in cardiomyocytes via direct interaction with regulatory chromatin regions. Specific modulation of these factors may provide a starting point for the development of more individualized treatment options for atrial fibrillation.


Asunto(s)
Potenciales de Acción , Fibrilación Atrial/enzimología , Epigénesis Genética , Atrios Cardíacos/enzimología , Frecuencia Cardíaca , Histona Desacetilasa 2/metabolismo , Miocitos Cardíacos/enzimología , Canales de Potasio/metabolismo , Proteínas Represoras/metabolismo , Adulto , Anciano , Animales , Fibrilación Atrial/genética , Fibrilación Atrial/fisiopatología , Remodelación Atrial , Estudios de Casos y Controles , Línea Celular , Modelos Animales de Enfermedad , Femenino , Atrios Cardíacos/fisiopatología , Histona Desacetilasa 2/genética , Humanos , Masculino , Persona de Mediana Edad , Canales de Potasio/genética , Proteínas Represoras/genética , Sus scrofa , Factores de Tiempo
4.
Biochem Biophys Res Commun ; 519(1): 141-147, 2019 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-31481236

RESUMEN

Atrial fibrillation (AF) is the most frequent sustained arrhythmia and can lead to structural cardiac changes, known as tachycardia-induced cardiomyopathy (TIC). HCN4 is implicated in spontaneous excitation of the sinoatrial node, while channel dysfunction has been associated with sinus bradycardia, AF and structural heart disease. We here asked whether HCN4 mutations may contribute to the development of TIC, as well. Mutation scanning of HCN4 in 60 independent patients with AF and suspected TIC followed by panel sequencing in carriers of HCN4 variants identified the HCN4 variant P883R [minor allele frequency (MAF): 0,88%], together with the KCNE1 variant S38G (MAF: 65%) in three unrelated patients. Family histories revealed additional cases of AF, sudden cardiac death and cardiomyopathy. Patch-clamp recordings of HCN4-P883R channels expressed in HEK293 cells showed remarkable alterations of channel properties shifting the half-maximal activation voltage to more depolarized potentials, while channel deactivation was faster compared to wild-type (WT). Co-transfection of WT and mutant subunits, resembling the heterozygous cellular situation of our patients, revealed significantly higher current densities compared to WT. In conclusion HCN4-P883R may increase ectopic trigger and maintenance of AF by shifting the activation voltage of If to more positive potentials and producing higher current density. Together with the common KCNE1 variant S38G, previously proposed as a genetic modifier of AF, HCN4-P883R may provide a substrate for the development of AF and TIC.


Asunto(s)
Fibrilación Atrial/genética , Genes Modificadores , Predisposición Genética a la Enfermedad , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Proteínas Musculares/genética , Mutación/genética , Canales de Potasio/genética , Secuencia de Aminoácidos , Femenino , Pruebas Genéticas , Células HEK293 , Humanos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/química , Activación del Canal Iónico , Masculino , Proteínas Musculares/química , Linaje , Canales de Potasio/química
5.
EMBO J ; 33(19): 2231-46, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25097251

RESUMEN

Neurons employ a set of homeostatic plasticity mechanisms to counterbalance altered levels of network activity. The molecular mechanisms underlying homeostatic plasticity in response to increased network excitability are still poorly understood. Here, we describe a sequential homeostatic synaptic depression mechanism in primary hippocampal neurons involving miRNA-dependent translational regulation. This mechanism consists of an initial phase of synapse elimination followed by a reinforcing phase of synaptic downscaling. The activity-regulated microRNA miR-134 is necessary for both synapse elimination and the structural rearrangements leading to synaptic downscaling. Results from miR-134 inhibition further uncover a differential requirement for GluA1/2 subunits for the functional expression of homeostatic synaptic depression. Downregulation of the miR-134 target Pumilio-2 in response to chronic activity, which selectively occurs in the synapto-dendritic compartment, is required for miR-134-mediated homeostatic synaptic depression. We further identified polo-like kinase 2 (Plk2) as a novel target of Pumilio-2 involved in the control of GluA2 surface expression. In summary, we have described a novel pathway of homeostatic plasticity that stabilizes neuronal circuits in response to increased network activity.


Asunto(s)
Regulación de la Expresión Génica , Hipocampo/metabolismo , Homeostasis/fisiología , MicroARNs/genética , Neuronas/metabolismo , Proteínas de Unión al ARN/metabolismo , Sinapsis/fisiología , Animales , Western Blotting , Células Cultivadas , Electrofisiología , Técnica del Anticuerpo Fluorescente , Hipocampo/embriología , Inmunoprecipitación , Plasticidad Neuronal , Neuronas/citología , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores AMPA/genética , Receptores AMPA/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
6.
Basic Res Cardiol ; 112(1): 8, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-28005193

RESUMEN

Atrial fibrillation (AF) is the most common cardiac arrhythmia. Concomitant heart failure (HF) poses a particular therapeutic challenge and is associated with prolonged atrial electrical refractoriness compared with non-failing hearts. We hypothesized that downregulation of atrial repolarizing TREK-1 (K2P2.1) K+ channels contributes to electrical remodeling during AF with HF, and that TREK-1 gene transfer would provide rhythm control via normalization of atrial effective refractory periods in this AF subset. In patients with chronic AF and HF, atrial TREK-1 mRNA levels were reduced by 82% (left atrium) and 81% (right atrium) compared with sinus rhythm (SR) subjects. Human findings were recapitulated in a porcine model of atrial tachypacing-induced AF and reduced left ventricular function. TREK-1 mRNA (-66%) and protein (-61%) was suppressed in AF animals at 14-day follow-up compared with SR controls. Downregulation of repolarizing TREK-1 channels was associated with prolongation of atrial effective refractory periods versus baseline conditions, consistent with prior observations in humans with HF. In a preclinical therapeutic approach, pigs were randomized to either atrial Ad-TREK-1 gene therapy or sham treatment. Gene transfer effectively increased TREK-1 protein levels and attenuated atrial effective refractory period prolongation in the porcine AF model. Ad-TREK-1 increased the SR prevalence to 62% during follow-up in AF animals, compared to 35% in the untreated AF group. In conclusion, TREK-1 downregulation and rhythm control by Ad-TREK-1 transfer suggest mechanistic and potential therapeutic significance of TREK-1 channels in a subgroup of AF patients with HF and prolonged atrial effective refractory periods. Functional correction of ionic remodeling through TREK-1 gene therapy represents a novel paradigm to optimize and specify AF management.


Asunto(s)
Fibrilación Atrial/metabolismo , Insuficiencia Cardíaca/metabolismo , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Adenoviridae , Adulto , Anciano , Animales , Fibrilación Atrial/fisiopatología , Modelos Animales de Enfermedad , Regulación hacia Abajo , Femenino , Terapia Genética/métodos , Vectores Genéticos , Insuficiencia Cardíaca/fisiopatología , Humanos , Masculino , Persona de Mediana Edad , Canales de Potasio de Dominio Poro en Tándem/genética , Distribución Aleatoria , Porcinos
7.
Histochem Cell Biol ; 145(6): 637-46, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26780567

RESUMEN

Synaptic inhibition in the spinal cord is mediated mainly by strychnine-sensitive glycine (GlyRs) and by γ-aminobutyric acid type A receptors (GABAAR). During neuronal maturation, neonatal GlyRs containing α2 subunits are replaced by adult-type GlyRs harboring α1 and α3 subunits. At the same time period of postnatal development, the transmembrane chloride gradient is changed due to increased expression of the potassium-chloride cotransporter (KCC2), thereby shifting the GABA- and glycine-mediated synaptic currents from mostly excitatory depolarization to inhibitory hyperpolarization. Here, we used RNA interference to suppress KCC2 expression during in vitro maturation of spinal cord neurons. Morphological analysis revealed reduced numbers and size of dendritic GlyR clusters containing α1 subunits but not of clusters harboring neonatal α2 subunits. The morphological changes were accompanied by decreased frequencies and amplitudes of glycinergic miniature inhibitory currents, whereas GABAergic synapses appeared functionally unaltered. Our data indicate that KCC2 exerts specific functions for the maturation of glycinergic synapses in cultured spinal cord neurons.


Asunto(s)
Glicina/metabolismo , Neuronas/citología , Neuronas/metabolismo , Médula Espinal/citología , Simportadores/deficiencia , Simportadores/metabolismo , Sinapsis/metabolismo , Células Cultivadas , Humanos
8.
Eur Heart J ; 34(35): 2768-75, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23178648

RESUMEN

AIMS: HCN4 channels are involved in generation, regulation, and stabilization of heart rhythm and channel dysfunction is associated with inherited sinus bradycardia. We asked whether dysfunctional HCN4 channels also contribute to the generation of cardiac tachyarrhythmias. METHODS AND RESULTS: In a candidate gene approach, we screened 422 patients with atrial and/or ventricular tachyarrhythmias and detected a novel HCN4 gene mutation that replaced the positively charged lysine 530 with an asparagine (HCN4-K530N) in a highly conserved region of the C-linker. The index patient developed tachycardia-bradycardia syndrome and persistent atrial fibrillation (AF) in an age-dependent fashion. Pedigree analysis identified eight affected family members with a similar course of disease. Whole-cell patch clamp electrophysiology of HEK293 cells showed that homomeric mutant channels almost are indistinguishable from wild-type channels. In contrast, heteromeric channels composed of mutant and wild-type subunits displayed a significant hyperpolarizing shift in the half-maximal activation voltage. This may be caused by a shift in the equilibrium between the tonically inhibited nucleotide-free state of the C-terminal domain of HCN4 believed to consist of a 'dimer of dimers' and the activated ligand-bound tetrameric form, leading to an increased inhibition of activity in heteromeric channels. CONCLUSION: Altered C-linker oligomerization in heteromeric channels is considered to promote familial tachycardia-bradycardia syndrome and persistent AF, indicating that f-channel dysfunction contributes to the development of atrial tachyarrhythmias.


Asunto(s)
Fibrilación Atrial/genética , Bradicardia/genética , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Mutación/genética , Taquicardia/genética , Adulto , Análisis de Varianza , Electrocardiografía , Técnicas Electrofisiológicas Cardíacas , Femenino , Células HEK293 , Humanos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/química , Masculino , Persona de Mediana Edad , Linaje
9.
J Neurosci ; 31(20): 7300-11, 2011 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-21593314

RESUMEN

On the one hand, neuronal activity can cause changes in pH; on the other hand, changes in pH can modulate neuronal activity. Consequently, the pH of the brain is regulated at various levels. Here we show that steady-state pH and acid extrusion were diminished in cultured hippocampal neurons of mice with a targeted disruption of the Na(+)-driven Cl(-)/HCO(3)(-) exchanger Slc4a8. Because Slc4a8 was found to predominantly localize to presynaptic nerve endings, we hypothesize that Slc4a8 is a key regulator of presynaptic pH. Supporting this hypothesis, spontaneous glutamate release in the CA1 pyramidal layer was reduced but could be rescued by increasing the intracellular pH. The reduced excitability in vitro correlated with an increased seizure threshold in vivo. Together with the altered kinetics of stimulated synaptic vesicle release, these data suggest that Slc4a8 modulates glutamate release in a pH-dependent manner.


Asunto(s)
Antiportadores de Cloruro-Bicarbonato/fisiología , Ácido Glutámico/metabolismo , Simportadores de Sodio-Bicarbonato/fisiología , Sodio/metabolismo , Sinapsis/metabolismo , Animales , Animales Recién Nacidos , Células Cultivadas , Femenino , Concentración de Iones de Hidrógeno , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Técnicas de Placa-Clamp , Simportadores de Sodio-Bicarbonato/deficiencia , Simportadores de Sodio-Bicarbonato/genética
10.
Life Sci ; 281: 119769, 2021 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-34186046

RESUMEN

AIMS: Heart failure (HF) is linked to electrical remodeling that promotes ventricular arrhythmias. Underlying molecular signaling is insufficiently understood, in particular concerning patients with early disease stages. Previous observations suggest a key role for epigenetic mechanisms in cardiac remodeling processes. We hypothesized that histone deacetylases (HDACs) 1 and 2 contribute to cellular electrophysiological dysregulation in ventricular cardiomyocytes during HF development. MATERIALS AND METHODS: HDAC and ion channel expression was quantified in a porcine model of early HF induced by short-term atrial tachypacing, resulting in atrial fibrillation with rapid ventricular rate response. Anti-Hdac1 and anti-Hdac2 siRNA treatment was employed in neonatal murine cardiomyocytes (NMCM) to study effects of HDACs on ion channel mRNA expression and action potential duration (APD). KEY FINDINGS: Early HF was characterized by mild reduction of left ventricular ejection fraction, prolonged QTc intervals, and increased ventricular effective refractory periods. Delayed repolarization was linked to significant downregulation of HDAC2 in left ventricular (LV) tissue. In addition, there was a tendency towards reduced transcript expression of KCNJ2/Kir2.1 K+ channels. In NMCM, knock-down of Hdac2 recapitulated AP prolongation. Finally, siRNA-mediated suppression of Hdac2 reduced Kcnh2/Kv11.1 K+ channel expression. SIGNIFICANCE: Suppression of HDAC2 is linked to ventricular electrical remodeling of APD and ion channel expression in early stages of heart failure. This previously unrecognized mechanism may serve as basis for future approaches to prevention and treatment of ventricular arrhythmias.


Asunto(s)
Insuficiencia Cardíaca/fisiopatología , Histona Desacetilasa 2/metabolismo , Remodelación Ventricular , Potenciales de Acción , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Técnicas de Silenciamiento del Gen , Histona Desacetilasa 2/genética , Ratones , Canales de Potasio con Entrada de Voltaje/genética , ARN Interferente Pequeño/genética , Reproducibilidad de los Resultados , Porcinos
11.
Eur J Neurosci ; 32(8): 1300-9, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20846322

RESUMEN

The dentate gyrus is the main hippocampal input structure receiving strong excitatory cortical afferents via the perforant path. Therefore, inhibition at this 'hippocampal gate' is important, particularly during postnatal development, when the hippocampal network is prone to seizures. The present study describes the development of tonic GABAergic inhibition in mouse dentate gyrus. A prominent tonic GABAergic component was already present at early postnatal stages (postnatal day 3), in contrast to the slowly developing phasic postsynaptic GABAergic currents. Tonic currents were mediated by GABA(A) receptors containing α(5)- and δ-subunits, which are sensitive to low ambient GABA concentrations. The extracellular GABA level was determined by synaptic GABA release and GABA uptake via the GABA transporter 1. The contribution of these main regulatory components was surprisingly stable during postnatal granule cell maturation. Throughout postnatal development, tonic GABAergic signals were inhibitory. They increased the action potential threshold of granule cells and reduced network excitability, starting as early as postnatal day 3. Thus, tonic inhibition is already functional at early developmental stages and plays a key role in regulating the excitation/inhibition balance of both the adult and the maturing dentate gyrus.


Asunto(s)
Giro Dentado/fisiología , Inhibición Neural/fisiología , Neuronas/fisiología , Ácido gamma-Aminobutírico/fisiología , Potenciales de Acción/fisiología , Animales , Electrofisiología , Ratones , Receptores de GABA-A/fisiología
12.
J Neurosci ; 28(4): 788-97, 2008 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-18216187

RESUMEN

Abnormal accumulation of soluble oligomers of amyloid beta (Abeta) is believed to cause malfunctioning of neurons in Alzheimer's disease. It has been shown that Abeta oligomers impair synaptic plasticity, thereby altering the ability of the neuron to store information. We examined the underlying cellular mechanism of Abeta oligomer-induced synaptic modifications by using a recently described stable oligomeric Abeta preparation called "Abeta(1-42) globulomer." Synthetically prepared Abeta(1-42) globulomer has been shown to localize to neurons and impairs long-term potentiation (Barghorn et al., 2005). Here, we demonstrate that Abeta(1-42) globulomer does not affect intrinsic neuronal properties, as assessed by measuring input resistance and discharge characteristics, excluding an unspecific alteration of membrane properties. We provide evidence that Abeta(1-42) globulomer, at concentrations as low as 8 nM, specifically suppresses spontaneous synaptic activity resulting from a reduction of vesicular release at terminals of both GABAergic and glutamatergic synapses. EPSCs and IPSCs were primarily unaffected. A detailed search for the precise molecular target of Abeta(1-42) globulomer revealed a specific inhibition of presynaptic P/Q calcium currents, whereas other voltage-activated calcium currents remained unaltered. Because intact P/Q calcium currents are needed for synaptic plasticity, the disruption of such currents by Abeta(1-42) globulomer may cause deficits in cellular mechanisms of information storage in brains of Alzheimer's disease patients. The inhibitory effect of Abeta(1-42) globulomer on synaptic vesicle release could be reversed by roscovitine, a specific enhancer of P/Q currents. Selective enhancement of the P/Q calcium current may provide a promising strategy in the treatment of Alzheimer's disease.


Asunto(s)
Péptidos beta-Amiloides/química , Bloqueadores de los Canales de Calcio/química , Canales de Calcio Tipo P/metabolismo , Canales de Calcio Tipo Q/metabolismo , Inhibición Neural/fisiología , Fragmentos de Péptidos/química , Transmisión Sináptica/fisiología , Péptidos beta-Amiloides/fisiología , Animales , Células Cultivadas , Ácido Glutámico/fisiología , Hipocampo/efectos de los fármacos , Hipocampo/fisiología , Fragmentos de Péptidos/fisiología , Ratas , Ratas Wistar , Transmisión Sináptica/efectos de los fármacos , Ácido gamma-Aminobutírico/fisiología
13.
Nat Commun ; 10(1): 3295, 2019 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-31337768

RESUMEN

HCN channels underlie the depolarizing funny current (If) that contributes importantly to cardiac pacemaking. If is upregulated in failing and infarcted hearts, but its implication in disease mechanisms remained unresolved. We generated transgenic mice (HCN4tg/wt) to assess functional consequences of HCN4 overexpression-mediated If increase in cardiomyocytes to levels observed in human heart failure. HCN4tg/wt animals exhibit a dilated cardiomyopathy phenotype with increased cellular arrhythmogenicity but unchanged heart rate and conduction parameters. If augmentation induces a diastolic Na+ influx shifting the Na+/Ca2+ exchanger equilibrium towards 'reverse mode' leading to increased [Ca2+]i. Changed Ca2+ homeostasis results in significantly higher systolic [Ca2+]i transients and stimulates apoptosis. Pharmacological inhibition of If prevents the rise of [Ca2+]i and protects from ventricular remodeling. Here we report that augmented myocardial If alters intracellular Ca2+ homeostasis leading to structural cardiac changes and increased arrhythmogenicity. Inhibition of myocardial If per se may constitute a therapeutic mechanism to prevent cardiomyopathy.


Asunto(s)
Calcio/metabolismo , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/fisiología , Proteínas Musculares/fisiología , Canales de Potasio/fisiología , Animales , Apoptosis , Electrofisiología Cardíaca , Perfilación de la Expresión Génica , Corazón/fisiología , Homeostasis , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Ratones Transgénicos , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/metabolismo , Canales de Potasio/genética , Canales de Potasio/metabolismo , Troponina I/genética , Troponina I/metabolismo , Troponina I/fisiología
14.
Life Sci ; 232: 116620, 2019 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-31291594

RESUMEN

AIMS: Cell-based biological pacemakers aim to overcome limitations and side effects of electronic pacemaker devices. We here developed and tested different approaches to achieve nodal-type differentiation using human adipose- and bone marrow-derived mesenchymal stem cells (haMSC, hbMSC). MAIN METHODS: haMSC and hbMSC were differentiated using customized protocols. Quantitative RT-PCR was applied for transcriptional pacemaker-gene profiling. Protein membrane expression was analyzed by immunocytochemistry. Pacemaker current (If) was studied in haMSC with and without lentiviral HCN4-transduction using patch clamp recordings. Functional characteristics were evaluated by co-culturing with neonatal rat ventricular myocytes (NRVM). KEY FINDINGS: Culture media-based differentiation for two weeks generated cells with abundant transcription of ion channel genes (Cav1.2, NCX1), transcription factors (TBX3, TBX18, SHOX2) and connexins (Cx31.9 and Cx45) characteristic for cardiac pacemaker tissue, but lack adequate HCN transcription. haMSC-derived cells revealed transcript levels, which were closer related to sinoatrial nodal cells than hbMSC-derived cells. To substitute for the lack of If, we performed lentiviral HCN4-transduction of haMSC resulting in stable If. Co-culturing with NRVM demonstrated that differentiated haMSC expressing HCN4 showed earlier onset of spontaneous contractions and higher beating regularity, synchrony and rate compared to co-cultures with non-HCN4-transduced haMSC or HCN4-transduced, non-differentiated haMSC. Confocal imaging indicated increased membrane expression of cardiac gap junctional proteins in differentiated haMSC. SIGNIFICANCE: By differentiation haMSC, rather than hbMSC attain properties favorable for cardiac pacemaking. In combination with lentiviral HCN4-transduction, a cellular phenotype was generated that sustainably controls and stabilizes rate in co-culture with NRVM.


Asunto(s)
Relojes Biológicos/fisiología , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Proteínas Musculares/metabolismo , Canales de Potasio/metabolismo , Tejido Adiposo/fisiología , Animales , Células de la Médula Ósea/fisiología , Diferenciación Celular/fisiología , Técnicas de Cocultivo , Humanos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/fisiología , Células Madre Mesenquimatosas/metabolismo , Células Musculares/metabolismo , Proteínas Musculares/fisiología , Miocitos Cardíacos/metabolismo , Técnicas de Placa-Clamp , Canales de Potasio/fisiología , Ratas , Nodo Sinoatrial
15.
Neurosci Lett ; 442(1): 44-9, 2008 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-18602451

RESUMEN

Synaptic transmission is triggered by presynaptic calcium influx through voltage-gated calcium channels. Axon terminals of central neurons express a diverse set of homologous calcium channels, giving rise to P/Q-, N-, and R-type calcium currents. The relative contribution of these components to presynaptic calcium signalling is heterogeneous and incompletely understood. Here we report that chronic block of N-type calcium channels in developing cultured rat hippocampal neurons leads to a compensatory up-regulation of P/Q-type calcium currents. This increase was measured directly by recording whole-cell calcium currents as well as in spontaneous inhibitory postsynaptic currents, indicating a global functional up-regulation of the P/Q-component. In contrast, immunocytochemical stainings as well as quantitative real-time PCR analysis did not reveal an increased expression of Ca(v) 2.1, the underlying calcium channel alpha-subunit. We conclude that developing hippocampal neurons can compensate for the loss of one calcium current component by up-regulation of alternative isoforms at the post-translational level.


Asunto(s)
Canales de Calcio Tipo N/metabolismo , Canales de Calcio Tipo P/metabolismo , Canales de Calcio Tipo Q/metabolismo , Neuronas/metabolismo , Transmisión Sináptica/fisiología , Animales , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo N/efectos de los fármacos , Células Cultivadas , Hipocampo/metabolismo , Inmunohistoquímica , Potenciales Postsinápticos Inhibidores/fisiología , Técnicas de Placa-Clamp , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , omega-Conotoxinas/farmacología
16.
Neuroscience ; 393: 258-272, 2018 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-30075244

RESUMEN

Retinal ganglion cells (RGCs), a diverse body of neurons which relay visual signals from the retina to the higher processing regions of the brain, are susceptible to neurodegenerative processes in several diseases affecting the retina. Previous evidence shows that RGCs are damaged at early stages of autoimmune optic neuritis (AON), prior to subsequent degeneration of the optic nerve. In order to study cell type-specific vulnerability of RGCs we performed immunohistochemical and patch-clamp electrophysiological analyses of RGCs following induction of AON using the experimental autoimmune encephalomyelitis model in Brown Norway rats. We report that αRGCs are more susceptible to degeneration than the global RGC population as a whole, with functional and structural changes beginning even prior to demyelination and inflammatory infiltration of the optic nerve (where the RGC axons reside). Functional classification of αRGCs into OFF-sustained, OFF-transient and ON-sustained subtypes revealed that αOFF RGCs (both sustained and transient subtypes) are more vulnerable than αON RGCs, as indicated by reductions in light-evoked post-synaptic currents and retraction of dendritic arbours. Classification of neuronal susceptibility is a first step in furthering our understanding of what underlies a neuron's vulnerability to degenerative processes, necessary for the future development of effective neuroprotective strategies.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Neuritis Óptica/inmunología , Retina/inmunología , Células Ganglionares de la Retina/inmunología , Animales , Axones/inmunología , Modelos Animales de Enfermedad , Femenino , Glicoproteína Mielina-Oligodendrócito/inmunología , Nervio Óptico/inmunología , Ratas
17.
Brain Res ; 1186: 56-64, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17996219

RESUMEN

The dentate gyrus is the main target for cortical inputs to the hippocampal formation and is particularly strongly controlled by synaptic inhibition. Many GABAergic interneurons migrate from the dentate molecular layer towards their final position in the hilus during the first two postnatal weeks. During this critical period of development we monitored the intrinsic and synaptic properties of developing interneurons in the molecular layer of mouse hippocampal slices. We focussed on multipolar cells in the middle portion of the molecular layer. With increasing age, input resistance decreased and action potential waveform changed to larger amplitude and shorter duration. Repetitive spiking was scarce at early stages, while trains of action potentials could be readily elicited after the first postnatal week. At all ages, we observed spontaneous postsynaptic currents which were almost exclusively GABA(A) receptor-mediated and increased in frequency with age. All developmental changes in intrinsic and synaptic properties occurred between p 6-8 and p 9-11, indicating a rapid functional maturation at the end of the first postnatal week. Parallel immunohistochemical experiments revealed that calretinin positive cells formed the major part of developing interneurons in the middle molecular layer. Together, the data shows a rapid functional maturation of intrinsic and synaptic properties of interneurons in the dentate molecular layer and an early integration into synaptic networks with clear prevalence of inhibitory synaptic inputs.


Asunto(s)
Giro Dentado/crecimiento & desarrollo , Interneuronas/metabolismo , Potenciales de la Membrana/fisiología , Vías Nerviosas/crecimiento & desarrollo , Sinapsis/metabolismo , Factores de Edad , Animales , Giro Dentado/citología , Giro Dentado/metabolismo , Inmunohistoquímica , Técnicas In Vitro , Interneuronas/citología , Ratones , Ratones Endogámicos C57BL , Vías Nerviosas/citología , Vías Nerviosas/metabolismo , Receptores de GABA-A/metabolismo , Ácido gamma-Aminobutírico/metabolismo
18.
Thromb Haemost ; 113(5): 1095-108, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25608503

RESUMEN

Co-stimulation via CD154 binding to CD40, pivotal for both innate and adaptive immunity, may also link haemostasis to vascular remodelling. Here we demonstrate that human platelet-bound or recombinant soluble CD154 (sCD154) elicit the release from and tethering of ultra-large (UL) von Willebrand factor (vWF) multimers to the surface of human cultured endothelial cells (ECs) exposed to shear stress. This CD40-mediated ULVWF multimer release from the Weibel-Palade bodies was triggered by consecutive activation of TRAF6, the tyrosine kinase c-Src and phospholipase Cγ1 followed by inositol-1,4,5 trisphosphate-mediated calcium mobilisation. Subsequent exposure to human washed platelets caused ULVWF multimer-platelet string formation on the EC surface in a shear stress-dependent manner. Platelets tethered to these ULVWF multimers exhibited P-selectin on their surface and captured labelled monocytes from the superfusate. When exposed to shear stress and sCD154, native ECs from wild-type but not CD40 or vWF-deficient mice revealed a comparable release of ULVWF multimers to which murine washed platelets rapidly adhered, turning P-selectin-positive and subsequently capturing monocytes from the perfusate. This novel CD154-provoked ULVWF multimer-platelet string formation at normal to fast flow may contribute to vascular remodelling processes requiring the perivascular or intravascular accumulation of pro-inflammatory macrophages such as arteriogenesis or atherosclerosis.


Asunto(s)
Ligando de CD40/metabolismo , Células Endoteliales/metabolismo , Factor de von Willebrand/metabolismo , Animales , Arterias/metabolismo , Aterosclerosis/metabolismo , Plaquetas/metabolismo , Calcio/química , Arteria Carótida Común/patología , Adhesión Celular , Electrofisiología , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inflamación , Isquemia/patología , Ratones , Microscopía Fluorescente , Monocitos/citología , Monocitos/metabolismo , Selectina-P/metabolismo , Perfusión , Proteínas Recombinantes/metabolismo , Resistencia al Corte , Transducción de Señal , Accidente Cerebrovascular , Fosfolipasas de Tipo C/metabolismo , Cuerpos de Weibel-Palade/metabolismo
19.
J Comp Neurol ; 478(2): 176-88, 2004 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-15349978

RESUMEN

Animal models of focal ischemic infarcts reveal an impaired GABAergic (gamma-aminobutyric acid) neurotransmission. GABA, the main inhibitory neurotransmitter, is primarily taken up by specific sodium-dependent transporters. As these transporters play a crucial role in maintaining levels of GABA concentration, they may be functionally involved in ischemic processes. We investigated whether the mRNA and protein expression of GAT-1, the dominant neuronal GABA transporter, is altered after cortical infarct induced by photothrombosis in Wistar rats. In situ hybridization was performed to analyze GAT-1 mRNA-positive cells in cortical brain regions and the hippocampus. The lesion dramatically raised the number of GABA transporter mRNA-expressing cells in all investigated cortical regions. Double-labeling studies with a general neuronal marker and a marker for astrocytes revealed that cells expressing GAT-1 mRNA after photothrombosis are neurons. The mRNA expression pattern of all hippocampal subfields remained unchanged. In contrast, cortical GAT-1 protein density was only slightly affected and surprisingly in the opposite way. In the primary and secondary somatosensory cortex, density values were significantly reduced. Immunoreactivity was not altered in all investigated hippocampal areas. We found a marked discordance between the increased number of cells expressing GAT-1 mRNA in the cortex and the reduced tissue GAT-1 protein content. Focal brain ischemia obviously triggers mechanisms that interfere with GAT-1 transcriptional regulation and protein synthesis or turnover.


Asunto(s)
Encéfalo/metabolismo , Infarto Cerebral/metabolismo , Proteínas de Transporte de Membrana/biosíntesis , Neuronas/metabolismo , Animales , Infarto Cerebral/fisiopatología , Modelos Animales de Enfermedad , Proteínas Transportadoras de GABA en la Membrana Plasmática , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Hibridación in Situ , Masculino , ARN Mensajero/análisis , Ratas , Ratas Wistar
20.
J Am Coll Cardiol ; 64(8): 757-67, 2014 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-25145518

RESUMEN

BACKGROUND: Inherited arrhythmias were originally considered isolated electrical defects. There is growing evidence that ion channel dysfunction also contributes to myocardial disorders, but genetic overlap has not been reported for sinus node dysfunction (SND) and noncompaction cardiomyopathy (NCCM). OBJECTIVES: The study sought to investigate a familial electromechanical disorder characterized by SND and NCCM, and to identify the underlying genetic basis. METHODS: The index family and a cohort of unrelated probands with sinus bradycardia were examined by electrocardiography, Holter recording, exercise stress test, echocardiography, and/or cardiac magnetic resonance imaging. Targeted next-generation and direct sequencing were used for candidate gene analysis and mutation scanning. Ion channels were expressed in HEK293 cells and studied using patch-clamp recordings. RESULTS: SND and biventricular NCCM were diagnosed in multiple members of a German family. Segregation analysis suggested autosomal-dominant inheritance of the combined phenotype. When looking for potentially disease-causing gene variants with cosegregation, a novel hyperpolarization-activated cyclic nucleotide channel 4 (HCN4)-G482R mutation and a common cysteine and glycine-rich protein 3 (CSRP3)-W4R variant were identified. HCN4-G482R is located in the highly conserved channel pore domain. Mutant subunits were nonfunctional and exerted dominant-negative effects on wild-type current. CSRP3-W4R has previously been linked to dilated and hypertrophic cardiomyopathy, but was also found in healthy subjects. Moreover, different truncation (695X) and missense (P883R) HCN4 mutations segregated with a similar combined phenotype in an additional, unrelated family and a single unrelated proband respectively, which both lacked CSRP3-W4R. CONCLUSIONS: The symptom complex of SND and NCCM is associated with heritable HCN4 defects. The NCCM phenotype may be aggravated by a common CSRP3 variant in one of the families.


Asunto(s)
Cardiopatías Congénitas/genética , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Proteínas Musculares/genética , Canales de Potasio/genética , Síndrome del Seno Enfermo/genética , Adolescente , Animales , Ecocardiografía , Técnicas Electrofisiológicas Cardíacas , Femenino , Alemania/epidemiología , Células HEK293 , Cardiopatías Congénitas/diagnóstico por imagen , Cardiopatías Congénitas/epidemiología , Humanos , Masculino , Potenciales de la Membrana , Persona de Mediana Edad , Linaje , Fenotipo , Prevalencia , Síndrome del Seno Enfermo/diagnóstico por imagen , Síndrome del Seno Enfermo/epidemiología , Síndrome , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA