Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Pflugers Arch ; 476(2): 229-242, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38036776

ABSTRACT

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.


Subject(s)
Brugada Syndrome , Humans , Brugada Syndrome/genetics , Brugada Syndrome/metabolism , Sodium/metabolism , HEK293 Cells , Mutation , Phenotype , NAV1.5 Voltage-Gated Sodium Channel/genetics , NAV1.5 Voltage-Gated Sodium Channel/metabolism
2.
Neurobiol Dis ; 187: 106306, 2023 Oct 15.
Article in English | MEDLINE | ID: mdl-37734623

ABSTRACT

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.
Life Sci ; 281: 119769, 2021 Sep 15.
Article in English | MEDLINE | ID: mdl-34186046

ABSTRACT

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.


Subject(s)
Heart Failure/physiopathology , Histone Deacetylase 2/metabolism , Ventricular Remodeling , Action Potentials , Animals , Animals, Newborn , Disease Models, Animal , Gene Knockdown Techniques , Histone Deacetylase 2/genetics , Mice , Potassium Channels, Voltage-Gated/genetics , RNA, Small Interfering/genetics , Reproducibility of Results , Swine
4.
Basic Res Cardiol ; 116(1): 13, 2021 02 25.
Article in English | MEDLINE | ID: mdl-33630168

ABSTRACT

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.


Subject(s)
Action Potentials , Atrial Fibrillation/enzymology , Epigenesis, Genetic , Heart Atria/enzymology , Heart Rate , Histone Deacetylase 2/metabolism , Myocytes, Cardiac/enzymology , Potassium Channels/metabolism , Repressor Proteins/metabolism , Adult , Aged , Animals , Atrial Fibrillation/genetics , Atrial Fibrillation/physiopathology , Atrial Remodeling , Case-Control Studies , Cell Line , Disease Models, Animal , Female , Heart Atria/physiopathology , Histone Deacetylase 2/genetics , Humans , Male , Middle Aged , Potassium Channels/genetics , Repressor Proteins/genetics , Sus scrofa , Time Factors
5.
Biochem Biophys Res Commun ; 519(1): 141-147, 2019 10 29.
Article in English | MEDLINE | ID: mdl-31481236

ABSTRACT

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.


Subject(s)
Atrial Fibrillation/genetics , Genes, Modifier , Genetic Predisposition to Disease , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Muscle Proteins/genetics , Mutation/genetics , Potassium Channels/genetics , Amino Acid Sequence , Female , Genetic Testing , HEK293 Cells , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/chemistry , Ion Channel Gating , Male , Muscle Proteins/chemistry , Pedigree , Potassium Channels/chemistry
6.
Nat Commun ; 10(1): 3295, 2019 07 23.
Article in English | MEDLINE | ID: mdl-31337768

ABSTRACT

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.


Subject(s)
Calcium/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/physiology , Muscle Proteins/physiology , Potassium Channels/physiology , Animals , Apoptosis , Cardiac Electrophysiology , Gene Expression Profiling , Heart/physiology , Homeostasis , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Mice, Transgenic , Muscle Proteins/genetics , Muscle Proteins/metabolism , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Potassium Channels/genetics , Potassium Channels/metabolism , Troponin I/genetics , Troponin I/metabolism , Troponin I/physiology
7.
Life Sci ; 232: 116620, 2019 Sep 01.
Article in English | MEDLINE | ID: mdl-31291594

ABSTRACT

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.


Subject(s)
Biological Clocks/physiology , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Muscle Proteins/metabolism , Potassium Channels/metabolism , Adipose Tissue/physiology , Animals , Bone Marrow Cells/physiology , Cell Differentiation/physiology , Coculture Techniques , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/physiology , Mesenchymal Stem Cells/metabolism , Muscle Cells/metabolism , Muscle Proteins/physiology , Myocytes, Cardiac/metabolism , Patch-Clamp Techniques , Potassium Channels/physiology , Rats , Sinoatrial Node
8.
Neuroscience ; 393: 258-272, 2018 11 21.
Article in English | MEDLINE | ID: mdl-30075244

ABSTRACT

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.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Optic Neuritis/immunology , Retina/immunology , Retinal Ganglion Cells/immunology , Animals , Axons/immunology , Disease Models, Animal , Female , Myelin-Oligodendrocyte Glycoprotein/immunology , Optic Nerve/immunology , Rats
9.
Basic Res Cardiol ; 112(1): 8, 2017 01.
Article in English | MEDLINE | ID: mdl-28005193

ABSTRACT

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.


Subject(s)
Atrial Fibrillation/metabolism , Heart Failure/metabolism , Potassium Channels, Tandem Pore Domain/metabolism , Adenoviridae , Adult , Aged , Animals , Atrial Fibrillation/physiopathology , Disease Models, Animal , Down-Regulation , Female , Genetic Therapy/methods , Genetic Vectors , Heart Failure/physiopathology , Humans , Male , Middle Aged , Potassium Channels, Tandem Pore Domain/genetics , Random Allocation , Swine
10.
Histochem Cell Biol ; 145(6): 637-46, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26780567

ABSTRACT

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.


Subject(s)
Glycine/metabolism , Neurons/cytology , Neurons/metabolism , Spinal Cord/cytology , Symporters/deficiency , Symporters/metabolism , Synapses/metabolism , Cells, Cultured , Humans
11.
Thromb Haemost ; 113(5): 1095-108, 2015 May.
Article in English | MEDLINE | ID: mdl-25608503

ABSTRACT

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.


Subject(s)
CD40 Ligand/metabolism , Endothelial Cells/metabolism , von Willebrand Factor/metabolism , Animals , Arteries/metabolism , Atherosclerosis/metabolism , Blood Platelets/metabolism , Calcium/chemistry , Carotid Artery, Common/pathology , Cell Adhesion , Electrophysiology , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Inflammation , Ischemia/pathology , Mice , Microscopy, Fluorescence , Monocytes/cytology , Monocytes/metabolism , P-Selectin/metabolism , Perfusion , Recombinant Proteins/metabolism , Shear Strength , Signal Transduction , Stroke , Type C Phospholipases/metabolism , Weibel-Palade Bodies/metabolism
12.
J Am Coll Cardiol ; 64(8): 757-67, 2014 Aug 26.
Article in English | MEDLINE | ID: mdl-25145518

ABSTRACT

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.


Subject(s)
Heart Defects, Congenital/genetics , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Muscle Proteins/genetics , Potassium Channels/genetics , Sick Sinus Syndrome/genetics , Adolescent , Animals , Echocardiography , Electrophysiologic Techniques, Cardiac , Female , Germany/epidemiology , HEK293 Cells , Heart Defects, Congenital/diagnostic imaging , Heart Defects, Congenital/epidemiology , Humans , Male , Membrane Potentials , Middle Aged , Pedigree , Phenotype , Prevalence , Sick Sinus Syndrome/diagnostic imaging , Sick Sinus Syndrome/epidemiology , Syndrome , Young Adult
13.
EMBO J ; 33(19): 2231-46, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25097251

ABSTRACT

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.


Subject(s)
Gene Expression Regulation , Hippocampus/metabolism , Homeostasis/physiology , MicroRNAs/genetics , Neurons/metabolism , RNA-Binding Proteins/metabolism , Synapses/physiology , Animals , Blotting, Western , Cells, Cultured , Electrophysiology , Fluorescent Antibody Technique , Hippocampus/embryology , Immunoprecipitation , Neuronal Plasticity , Neurons/cytology , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Receptors, AMPA/genetics , Receptors, AMPA/metabolism , Reverse Transcriptase Polymerase Chain Reaction
14.
Eur J Pharmacol ; 702(1-3): 44-55, 2013 Feb 28.
Article in English | MEDLINE | ID: mdl-23376566

ABSTRACT

Alzheimer's disease is accompanied by increased brain levels of soluble amyloid-ß (Aß) oligomers. It has been suggested that oligomers directly impair synaptic function, thereby causing cognitive deficits in Alzheimer's disease patients. Recently, it has been shown that synthetic Aß oligomers directly modulate P/Q-type calcium channels, possibly leading to excitotoxic cascades and subsequent synaptic decline. Using whole-cell recordings we studied the modulation of recombinant presynaptic calcium channels in HEK293 cells after application of a stable Aß oligomer preparation (Aß1-42 globulomer). Aß globulomer shifted the half-activation voltage of P/Q-type and N-type calcium channels to more hyperpolarized values (by 11.5 and 7.5 mV). Application of non-aggregated Aß peptides had no effect. We then analyzed the potential of calcium channel blockers to prevent Aß globulomer-induced synaptic decline in hippocampal slice cultures. Specific block of P/Q-type or N-type calcium channels with peptide toxins completely reversed Aß globulomer-induced deficits in glutamatergic neurotransmission. Two state-dependent low molecular weight P/Q-type and N-type calcium channel blockers also protected neurons from Aß-induced alterations. On the contrary, inhibition of L-type calcium channels failed to reverse the deficit. Our data show that Aß globulomer directly modulates recombinant P/Q-type and N-type calcium channels in HEK293 cells. Block of presynaptic calcium channels with both state-dependent and state-independent modulators can reverse Aß-induced functional deficits in synaptic transmission. These findings indicate that presynaptic calcium channel blockers may be a therapeutic strategy for the treatment of Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels, N-Type/physiology , Calcium Channels/physiology , Peptide Fragments/pharmacology , Synapses/drug effects , Animals , Calcium/physiology , HEK293 Cells , Hippocampus/drug effects , Hippocampus/physiology , Humans , Rats , Rats, Wistar , Synapses/physiology , omega-Agatoxin IVA/pharmacology , omega-Conotoxins/pharmacology
15.
Comb Chem High Throughput Screen ; 16(3): 233-43, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23228050

ABSTRACT

Development of calcium channel blockers is attractive, but has in the past been hampered by lack of high throughput electrophysiological technology. This limitation has been overcome by the implementation of automated patch clamp systems that allow identification of state-dependent compounds, which preferentially target pathologically overactive channels. We recently presented a fluorescence-based high-throughput screen for P/Q-type calcium channels followed by automated electrophysiology. Here, we provide a detailed description of the development of the secondary screen, and show the full analysis of the inactivation kinetics of the recombinant P/Q channel that served as a basis for the automated patch clamp protocol. Increasing the length of pre-depolarization shifted the inactivation to more hyperpolarized potentials. No steady-state inactivation was reached up to pre-depolarization durations of 3 min, while stability of the recordings progressively declined. As a compromise, a 3s pre-depolarization protocol was proposed for functional screening. In order to validate the electrophysiological screening, we compared kinetics and pharmacology of recombinant P/Q-type channels between automated and manual patch clamp measurements. Channel activation was similar under both conditions. By contrast, inactivation occurred at more hyperpolarized potentials in the automated system. Therefore, P/Q-type calcium channel inactivation is sensitive to the applied technological platform and needs to be adjusted when performing automated patch clamp recordings. Our results indicate that a thorough analysis of the inactivation kinetics is mandatory, when establishing an electrophysiological screening protocol for calcium channel blockers. As some data obtained by automated recordings may not be identical to manual patch clamp analysis, we recommend a proper initial validation of the screening assay and--if necessary--a posthoc adjustment of automated patch clamp values. The protocol presented here supports hit-to-lead and lead optimization efforts during the development of novel P/Q-type calcium channel blockers, and may be valuable for the generation of assays in other ion channel programs.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Channels, P-Type/metabolism , Calcium Channels, Q-Type/metabolism , Drug Evaluation, Preclinical/methods , Cell Line , Humans , Patch-Clamp Techniques/methods , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism
16.
Eur Heart J ; 34(35): 2768-75, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23178648

ABSTRACT

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.


Subject(s)
Atrial Fibrillation/genetics , Bradycardia/genetics , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Mutation/genetics , Tachycardia/genetics , Adult , Analysis of Variance , Electrocardiography , Electrophysiologic Techniques, Cardiac , Female , HEK293 Cells , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/chemistry , Male , Middle Aged , Pedigree
17.
Neurobiol Aging ; 33(7): 1356-63, 2012 Jul.
Article in English | MEDLINE | ID: mdl-21257232

ABSTRACT

The mechanisms which mediate cortical map plasticity and functional recovery following stroke remain a matter of debate. Readjustment of the excitatory-inhibitory balance may support cortical map plasticity in perilesional areas. Here we studied cortical net inhibition in the vicinity of photothrombotically-induced cortical lesions in young adult (3 months) and aged (24 months) male rats. Field potentials were recorded in cortical layer II/III following application of paired-pulse stimulation at layer VI/white matter in coronal brain slices. Additionally, we analyzed the regional distribution of 5 major gamma-aminobutyric acid A (GABA(A)) receptor subunits (α1, α2, α3, α5, and γ2) by immunohistochemistry. Paired-pulse inhibition in the perilesional parietal cortex was decreased in young rats but was increased in aged rats. As a consequence of the diminished intrinsic net inhibition in aged control animals, the excitatory-inhibitory balance was readjusted to an age-independent similar level in young and aged lesioned rats in a homeostatic-like fashion. These physiological changes in neuronal activity were accompanied by age-specific laminar alterations of the gamma-aminobutyric acid A (GABA(A)) receptor subunit composition, most prominently of the subunit α5. The present study suggests that the mechanisms underlying functional reorganization in aged animals may be distinctly different from those in young animals.


Subject(s)
Aging/physiology , Excitatory Postsynaptic Potentials/physiology , Neural Inhibition/physiology , Stroke/physiopathology , Aging/metabolism , Animals , Brain/metabolism , Brain/physiology , Male , Rats , Rats, Wistar , Receptors, GABA-A/physiology , Stroke/metabolism
18.
J Neurosci ; 31(20): 7300-11, 2011 May 18.
Article in English | MEDLINE | ID: mdl-21593314

ABSTRACT

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.


Subject(s)
Chloride-Bicarbonate Antiporters/physiology , Glutamic Acid/metabolism , Sodium-Bicarbonate Symporters/physiology , Sodium/metabolism , Synapses/metabolism , Animals , Animals, Newborn , Cells, Cultured , Female , Hydrogen-Ion Concentration , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Patch-Clamp Techniques , Sodium-Bicarbonate Symporters/deficiency , Sodium-Bicarbonate Symporters/genetics
19.
Eur J Neurosci ; 32(8): 1300-9, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20846322

ABSTRACT

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.


Subject(s)
Dentate Gyrus/physiology , Neural Inhibition/physiology , Neurons/physiology , gamma-Aminobutyric Acid/physiology , Action Potentials/physiology , Animals , Electrophysiology , Mice , Receptors, GABA-A/physiology
20.
Circ Arrhythm Electrophysiol ; 3(5): 542-52, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20693575

ABSTRACT

BACKGROUND: HCN channels activate the pacemaker current I(f), which is thought to contribute significantly to generation and regulation of heart rhythm. HCN4 represents the dominant isotype in the sinoatrial node and binding of cAMP was suggested to be necessary for autonomic heart rate regulation. METHODS AND RESULTS: In a candidate gene approach, a heterozygous insertion of 13 nucleotides in exon 6 of the HCN4 gene leading to a truncated cyclic nucleotide-binding domain was identified in a 45-year-old woman with sinus bradycardia. Biophysical properties determined by whole-cell patch-clamp recording of HEK293 cells demonstrated that mutant subunits (HCN4-695X) were insensitive to cAMP. Heteromeric channels composed of wild-type and mutant subunits failed to respond to cAMP-like homomeric mutant channels, indicating a dominant-negative suppression of cAMP-induced channel activation by mutant subunits. Pedigree analysis identified 7 additional living carriers showing similar clinical phenotypes, that is, sinus node dysfunction with mean resting heart rate of 45.9±4.6 bpm (n=8) compared with 66.5±9.1 bpm of unaffected relatives (n=6; P<0.01). Clinical evaluation revealed no ischemic or structural heart disease in any family member. Importantly, mutant carriers exhibited normal heart rate variance and full ability to accelerate heart rate under physical activity or pharmacological stimulation. Moreover, mutant carriers displayed distinctive sinus arrhythmias and premature beats linked to adrenergic stress. CONCLUSIONS: In humans, cAMP responsiveness of I(f) determines basal heart rate but is not critical for maximum heart rate, heart rate variability, or chronotropic competence. Furthermore, cAMP-activated I(f) may stabilize heart rhythm during chronotropic response.


Subject(s)
Autonomic Nervous System/physiopathology , Cardiac Pacing, Artificial/methods , Cyclic AMP/metabolism , Cyclic Nucleotide-Gated Cation Channels/genetics , DNA/genetics , Mutation , Potassium Channels/genetics , Sick Sinus Syndrome/genetics , Adolescent , Adult , Aged , Child , Cyclic Nucleotide-Gated Cation Channels/metabolism , Female , Genetic Predisposition to Disease , Heart Rate/genetics , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Male , Middle Aged , Nerve Tissue Proteins , Potassium Channels/metabolism , Sick Sinus Syndrome/metabolism , Sick Sinus Syndrome/therapy , Sinoatrial Node/metabolism , Sinoatrial Node/physiopathology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...