Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Pain ; 158(10): 2012-2024, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28767511

ABSTRACT

Several studies implicated cyclic adenosine monophosphate (cAMP) as an important second messenger for regulating nociceptor sensitization, but downstream targets of this signaling pathway which contribute to neuronal plasticity are not well understood. We used a Cre/loxP-based strategy to disable the function of either HCN2 or PKA selectively in a subset of peripheral nociceptive neurons and analyzed the nociceptive responses in both transgenic lines. A near-complete lack of sensitization was observed in both mutant strains when peripheral inflammation was induced by an intradermal injection of 8br-cAMP. The lack of HCN2 as well as the inhibition of PKA eliminated the cAMP-mediated increase of calcium transients in dorsal root ganglion neurons. Facilitation of Ih via cAMP, a hallmark of the Ih current, was abolished in neurons without PKA activity. Collectively, these results show a significant contribution of both genes to inflammatory pain and suggest that PKA-dependent activation of HCN2 underlies cAMP-triggered neuronal sensitization.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Potassium Channels/metabolism , Sensory Receptor Cells/metabolism , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Animals , Bradykinin/pharmacology , Calcium/metabolism , Cells, Cultured , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Ganglia, Spinal/cytology , Hyperalgesia/physiopathology , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Inflammation/chemically induced , Mice , Mice, Inbred C57BL , Mice, Transgenic , NAV1.8 Voltage-Gated Sodium Channel/genetics , NAV1.8 Voltage-Gated Sodium Channel/metabolism , Pain Threshold , Phosphorylation/drug effects , Phosphorylation/physiology , Potassium Channels/genetics , Proteins/genetics , Proteins/metabolism , Sensory Receptor Cells/drug effects , Signal Transduction
2.
Clin Exp Pharmacol Physiol ; 42(8): 874-80, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25933122

ABSTRACT

In critically ill patients regulation of heart-rate is often severely disturbed. Interaction of bacterial endotoxin (lipopolysaccharide, LPS) with hyperpolarization-activated cyclic nucleotide-gated cation-(HCN)-channels may interfere with heart-rate regulation. This study analyzes the effect of LPS, the HCN-channel blocker ivabradine or Ca(2+) -channel blockers (nifedipine, verapamil) on pacemaking in spontaneously beating neonatal rat cardiomyocytes (CM) in vitro. In vivo, the effect of LPS on the heart-rate of adult CD1-mice with and without autonomic blockade is analyzed telemetrically. LPS (100 ng/mL) and ivabradine (5 µg/mL) reduced the beating-rate of CM by 20.1% and 24.6%, respectively. Coincubation of CM with both, LPS and ivabradine, did not further reduce the beating-rate, indicating interaction of both compounds with HCN-channels, while coincubation with Ca(2+) -channel blockers and LPS caused additive beating-rate reduction. In CD1-mice (containing an active autonomic-nervous-system), injection of LPS (0.4 mg/kg) expectedly resulted in increased heart-rate. However, if the autonomic nervous system was blocked by propranolol and atropine, in line with the in vitro data, LPS induced a significant reduction of heart-rate, which was not additive to ivabradine. The in vivo and in vitro results indicate that LPS interacts with HCN-channels of cardiomyocytes. Thus, LPS indirectly sensitizes HCN-channels for sympathetic activation (tachycardic-effect), and in parallel directly inhibits channel activity (bradycardic-effect). Both effects may contribute to the detrimental effects of septic cardiomyopathy and septic autonomic dysfunction.


Subject(s)
Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Lipopolysaccharides/metabolism , Lipopolysaccharides/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/physiology , Animals , Benzazepines/pharmacology , Heart Rate/drug effects , Ivabradine , Male , Mice , Rats , Sympathetic Nervous System/physiopathology , Tachycardia/chemically induced , Tachycardia/metabolism , Tachycardia/physiopathology
3.
Stem Cell Reports ; 2(5): 592-605, 2014 May 06.
Article in English | MEDLINE | ID: mdl-24936448

ABSTRACT

Therapeutic approaches for "sick sinus syndrome" rely on electrical pacemakers, which lack hormone responsiveness and bear hazards such as infection and battery failure. These issues may be overcome via "biological pacemakers" derived from pluripotent stem cells (PSCs). Here, we show that forward programming of PSCs with the nodal cell inducer TBX3 plus an additional Myh6-promoter-based antibiotic selection leads to cardiomyocyte aggregates consisting of >80% physiologically and pharmacologically functional pacemaker cells. These induced sinoatrial bodies (iSABs) exhibited highly increased beating rates (300-400 bpm), coming close to those found in mouse hearts, and were able to robustly pace myocardium ex vivo. Our study introduces iSABs as highly pure, functional nodal tissue that is derived from PSCs and may be important for future cell therapies and drug testing in vitro.


Subject(s)
Cellular Reprogramming , Pluripotent Stem Cells/cytology , Sinoatrial Node/physiology , Animals , Biological Clocks , Calcium/metabolism , Cell Differentiation , Cell Line , Coculture Techniques , In Vitro Techniques , Mice , Models, Biological , Myocytes, Cardiac/metabolism , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Patch-Clamp Techniques , Pluripotent Stem Cells/metabolism , Sick Sinus Syndrome/metabolism , Sick Sinus Syndrome/pathology , Sick Sinus Syndrome/veterinary , Sinoatrial Node/cytology , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism
4.
Cardiovasc Res ; 99(4): 780-8, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23761399

ABSTRACT

AIMS: The complex molecular mechanisms underlying spontaneous cardiac pacemaking are not fully understood. Recent findings point to a co-ordinated interplay between intracellular Ca(2+) cycling and plasma membrane-localized cation transport determining the origin and periodicity of pacemaker potentials. The sodium-calcium exchanger (NCX1) is a key sarcolemmal protein for the maintenance of calcium homeostasis in the heart. Here, we investigated the contribution of NCX1 to cardiac pacemaking. METHODS AND RESULTS: We used an inducible and sinoatrial node-specific Cre transgene to create micelacking NCX1 selectively in cells of the cardiac pacemaking and conduction system (cpNCX1KO). RT-PCR and immunolabeling experiments confirmed the precise tissue-specific and temporally controlled deletion. Ablation of NCX1 resulted in a progressive slowing of heart rate accompanied by severe arrhythmias. Isolated sinoatrial tissue strips displayed a significantly decreased and irregular contraction rate underpinning a disturbed intrinsic pacemaker activity. Mutant animals displayed a gradual increase in the heart-to-body weight ratio and developed ventricular dilatation; however, their ventricular contractile performance was not significantly affected. Pacemaker cells from cpNCX1KO showed no NCX1 activity in response to caffeine-induced Ca(2+) release, determined by Ca(2+) imaging. Regular spontaneous Ca(2+) discharges were frequently seen in control, but only sporadically in knockout (KO) cells. The majority of NCX1KO cells displayed an irregular and a significantly reduced frequency of spontaneous Ca(2+) signals. Furthermore, Ca(2+) transients measured during electrical field stimulation were of smaller magnitude and decelerated kinetics in KO cells. CONCLUSIONS: Our results establish NCX1 as a critical target for the proper function of cardiac pacemaking.


Subject(s)
Heart Rate , Myocardium/metabolism , Sodium-Calcium Exchanger/physiology , Animals , Arrhythmias, Cardiac/etiology , Calcium/metabolism , Mice , Mice, Inbred C57BL , Sinoatrial Node/metabolism , Ventricular Remodeling
5.
Basic Res Cardiol ; 108(1): 312, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23184392

ABSTRACT

Common cardiovascular progenitor cells are characterized and induced by expression of the transcription factor MesP1. To characterize this population we used a 3.4-kb promoter fragment previously described by our group. This served to isolate MesP1-positive cells from differentiating ES stem cells via magnetic cell sorting based on a truncated CD4 surface marker. As this proximal promoter fragment omits a distal non-cardiovasculogenic enhancer region, we were able to achieve a synchronized fraction of highly enriched cardiovascular progenitors. These led to about 90% of cells representing the three cardiovascular lineages: cardiomyocytes, endothelial cells and smooth muscle cells as evident from protein and mRNA analyses. In addition, electrophysiological and pharmacological parameters of the cardiomyocytic fraction show that almost all correspond to the multipotent early/intermediate cardiomyocyte subtype at day 18 of differentiation. Further differentiation of these cells was not impaired as evident from strong and synchronous beating at later stages. Our work contributes to the understanding of the earliest cardiovasculogenic events and may become an important prerequisite for cell therapy, tissue engineering and pharmacological testing in the culture dish using pluripotent stem cell-derived as well as directly reprogrammed cardiovascular cell types. Likewise, these cells provide an ideal source for large-scale transcriptome and proteome analyses.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Cardiovascular System/cytology , Multipotent Stem Cells/cytology , Promoter Regions, Genetic , Animals , Cell Differentiation , Cell Separation , Endothelial Cells/cytology , Mice , Myocytes, Cardiac/cytology , Myocytes, Smooth Muscle/cytology
6.
Cardiovasc Res ; 95(3): 317-26, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22652004

ABSTRACT

AIMS: Cardiac hypertrophy is accompanied by reprogramming of gene expression, where the altered expression of ion channels decreases electrical stability and increases the risk of life-threatening arrhythmias. However, the underlying mechanisms are not fully understood. Here, we analysed the role of the depolarizing current I(f) which has been hypothesized to contribute to arrhythmogenesis in the hypertrophied ventricle. METHODS AND RESULTS: We used transverse aortic constriction in mice to induce ventricular hypertrophy. This resulted in an increased number of I(f) positive ventricular myocytes as well as a strongly enhanced and accelerated I(f) when compared with controls. Of the four HCN (hyperpolarization-activated cyclic nucleotide-gated channels) isoforms mediating I(f), HCN2 and HCN4 were the predominantly expressed subunits in healthy as well as hypertrophied hearts. Unexpectedly, only the HCN1 transcript was significantly upregulated in response to hypertrophy. However, the combined deletion of HCN2 and HCN4 disrupted ventricular I(f) completely. The lack of I(f) in hypertrophic double-knockouts resulted in a strong attenuation of pro-arrhythmogenic parameters characteristically observed in hypertrophic hearts. In particular, prolongation of the action potential was significantly decreased and lengthening of the QT interval was reduced. CONCLUSIONS: We suggest that the strongly increased HCN channel activity in hypertrophied myocytes prolongs the repolarization of the ventricular action potential and thereby may increase the arrhythmogenic potential. Our results provide for the first time a direct link between an upregulation of ventricular I(f) and a diminished repolarization reserve in cardiac hypertrophy.


Subject(s)
Arrhythmias, Cardiac/etiology , Cyclic Nucleotide-Gated Cation Channels/metabolism , Heart Ventricles/metabolism , Hypertrophy, Left Ventricular/complications , Myocytes, Cardiac/metabolism , Potassium Channels/metabolism , Ventricular Remodeling , Action Potentials , Animals , Aorta/surgery , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/pathology , Arrhythmias, Cardiac/physiopathology , Cyclic Nucleotide-Gated Cation Channels/deficiency , Cyclic Nucleotide-Gated Cation Channels/genetics , Disease Models, Animal , Electrocardiography , Gene Expression Regulation , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/pathology , Hypertrophy, Left Ventricular/physiopathology , Ion Channels/genetics , Ion Channels/metabolism , Ligation , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/pathology , Potassium Channels/deficiency , Potassium Channels/genetics , RNA, Messenger/metabolism , Time Factors
7.
J Clin Invest ; 122(3): 1119-30, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22354168

ABSTRACT

Cardiac pacemaker cells create rhythmic pulses that control heart rate; pacemaker dysfunction is a prevalent disorder in the elderly, but little is known about the underlying molecular causes. Popeye domain containing (Popdc) genes encode membrane proteins with high expression levels in cardiac myocytes and specifically in the cardiac pacemaking and conduction system. Here, we report the phenotypic analysis of mice deficient in Popdc1 or Popdc2. ECG analysis revealed severe sinus node dysfunction when freely roaming mutant animals were subjected to physical or mental stress. In both mutants, bradyarrhythmia developed in an age-dependent manner. Furthermore, we found that the conserved Popeye domain functioned as a high-affinity cAMP-binding site. Popdc proteins interacted with the potassium channel TREK-1, which led to increased cell surface expression and enhanced current density, both of which were negatively modulated by cAMP. These data indicate that Popdc proteins have an important regulatory function in heart rate dynamics that is mediated, at least in part, through cAMP binding. Mice with mutant Popdc1 and Popdc2 alleles are therefore useful models for the dissection of the mechanisms causing pacemaker dysfunction and could aid in the development of strategies for therapeutic intervention.


Subject(s)
Cell Adhesion Molecules/metabolism , Muscle Proteins/metabolism , Potassium Channels, Tandem Pore Domain/metabolism , Animals , Biological Clocks , Bradycardia/genetics , Electrocardiography/methods , Electrophysiology/methods , Heart Rate , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Phenotype , Protein Structure, Tertiary , Telemetry/methods , Time Factors
8.
Circ Res ; 109(9): 1015-23, 2011 Oct 14.
Article in English | MEDLINE | ID: mdl-21903939

ABSTRACT

RATIONALE: The hyperpolarization-activated current I(h) that is generated by hyperpolarization-activated cyclic nucleotide-gated channels (HCNs) plays a key role in the control of pacemaker activity in sinoatrial node cells of the heart. By contrast, it is unclear whether I(h) is also relevant for normal function of cardiac ventricles. OBJECTIVE: To study the role of the HCN3-mediated component of ventricular I(h) in normal ventricular function. METHODS AND RESULTS: To test the hypothesis that HCN3 regulates the ventricular action potential waveform, we have generated and analyzed a HCN3-deficient mouse line. At basal heart rate, mice deficient for HCN3 displayed a profound increase in the T-wave amplitude in telemetric electrocardiographic measurements. Action potential recordings on isolated ventricular myocytes indicate that this effect was caused by an acceleration of the late repolarization phase in epicardial myocytes. Furthermore, the resting membrane potential was shifted to more hyperpolarized potentials in HCN3-deficient mice. Cardiomyocytes of HCN3-deficient mice displayed approximately 30% reduction of total I(h). At physiological ionic conditions, the HCN3-mediated current had a reversal potential of approximately -35 mV and displayed ultraslow deactivation kinetics. CONCLUSIONS: We propose that HCN3 together with other members of the HCN channel family confer a depolarizing background current that regulates ventricular resting potential and counteracts the action of hyperpolarizing potassium currents in late repolarization. In conclusion, our data indicate that HCN3 plays an important role in shaping the cardiac action potential waveform.


Subject(s)
Action Potentials/physiology , Cyclic Nucleotide-Gated Cation Channels/physiology , Ventricular Function/physiology , Animals , Cyclic Nucleotide-Gated Cation Channels/deficiency , Cyclic Nucleotide-Gated Cation Channels/genetics , Electrocardiography , Heart Rate/physiology , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Myocytes, Cardiac/physiology , Potassium Channels , Sinoatrial Node/physiology
9.
Cardiovasc Res ; 84(2): 263-72, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19564151

ABSTRACT

AIMS: The proliferative potential of pluripotent stem cell-derived cardiomyocytes is limited, and reasonable yields for novel therapeutic options have yet to be achieved. In addition, various clinical applications will require the generation of specific cardiac cell types. Whereas early cardiovascular precursors appear to be important for novel approaches such as reseeding decellularized hearts, direct cell transplantation may require ventricular cells. Our recent work demonstrated that MesP1 represents a master regulator sufficient to induce cardiovasculogenesis in pluripotent cells. This led to our hypothesis that 'forward programming' towards specific subtypes may be feasible via overexpression of distinct early cardiovascular transcription factors. METHODS AND RESULTS: Here we demonstrate that forced expression of Nkx2.5 similar to MesP1 is sufficient to enhance cardiogenesis in murine embryonic stem cells (mES). In comparison to control transfected mES cells, a five-fold increased appearance of beating foci was observed as well as upregulated mRNA and protein expression levels. In contrast to MesP1, no increase of the endothelial lineage within the cardiovasculogenic mesoderm was observed. Likewise, Flk-1, the earliest known cardiovascular surface marker, was not induced via Nkx2.5 as opposed to MesP1. Detailed patch clamping analyses showed electrophysiological characteristics corresponding to all subtypes of cardiac ES cell differentiation in Nkx2.5 as well as MesP1 programmed embryoid bodies, but fractions of cardiomyocytes had distinct characteristics: MesP1 forced the appearance of early/intermediate type cardiomyocytes in comparison to control transfected ES cells whereas Nkx2.5 led to preferentially differentiated ventricular cells. CONCLUSION: Our findings show proof of principle for cardiovascular subtype-specific programming of pluripotent stem cells and confirm the molecular hierarchy for cardiovascular specification initiated via MesP1 with differentiation factors such as Nkx2.5 further downstream.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , Embryonic Stem Cells/metabolism , Homeodomain Proteins/metabolism , Myocardial Contraction , Myocytes, Cardiac/metabolism , Transcription Factors/metabolism , Action Potentials , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/genetics , Cell Line , Cell Lineage , Cell Proliferation , Gene Expression Regulation, Developmental , Genotype , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/genetics , Humans , Mice , Myocardial Contraction/genetics , Phenotype , RNA, Messenger/metabolism , Signal Transduction , Time Factors , Transcription Factors/genetics , Transfection
10.
Methods Find Exp Clin Pharmacol ; 30(8): 633-41, 2008 Oct.
Article in English | MEDLINE | ID: mdl-19088948

ABSTRACT

Ivabradine has been approved as a heart rate-lowering agent for use in the treatment of chronic stable angina pectoris in case of contraindication or intolerance to beta-blockers. This drug effectively lowers the heart rate by inhibiting the pacemaker current I(f) in the sinoatrial node. It appears to induce fewer adverse reactions than other drugs used for reducing the heart rate, such as calcium channel blockers or beta-blockers. Because of this favorable profile, ivabradine could become the first-choice drug when pure heart rate-lowering is the therapeutic goal. This review evaluates experimental and preclinical data to investigate the possibilities, as well as the limitations, of the clinical use of ivabradine. In experimental studies, it has been shown that ivabradine does have some unfavorable pharmacodynamic properties, such as the block of all four hyperpolarization-activated cyclic nucleotide-gated channels and block of other ion channels at high concentrations. Clinical studies, however, indicate that those properties do not result in clinical consequences as long as ivabradine is given at the recommended dose and contraindications are strictly observed.


Subject(s)
Benzazepines/pharmacology , Heart Rate/drug effects , Angina Pectoris/drug therapy , Angina Pectoris/physiopathology , Animals , Benzazepines/adverse effects , Benzazepines/therapeutic use , Chronic Disease , Cyclic Nucleotide-Gated Cation Channels/antagonists & inhibitors , Humans , Ion Channels/antagonists & inhibitors , Ivabradine
11.
J Mol Cell Cardiol ; 45(1): 62-9, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18538341

ABSTRACT

Temporally controlled gene deletion provides a powerful technique for examination of gene function in vivo. To permit use of this technology in the study of cardiac pacemaking, we attempted to generate a mouse line expressing an inducible Cre recombinase selectively in cardiac pacemaker cells. The tamoxifen-inducible CreER(T2) construct was 'knocked in' into the pacemaker channel HCN4 locus. In the absence of inducing agent, recombination was undetectable in HCN4-KiT mice. After injection of tamoxifen, highly selective and efficient recombination was observed in the sinoatrial and atrioventricular node. Expression of Cre and tamoxifen per se did not affect cardiac rhythm, basal heart rate and heart rate modulation. By crossing these animals with floxed HCN4 mice, complete deletion of this gene in the sinoatrial node could be achieved. HCN4-KiT mice represent the first tool for the temporally controlled inactivation of floxed target genes selectively in the conduction system of the murine heart.


Subject(s)
Estrogen Antagonists/pharmacology , Gene Deletion , Heart Conduction System/metabolism , Integrases/biosynthesis , Recombination, Genetic/drug effects , Tamoxifen/pharmacology , Animals , Cyclic Nucleotide-Gated Cation Channels/genetics , Cyclic Nucleotide-Gated Cation Channels/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Integrases/genetics , Mice , Mice, Transgenic , Quantitative Trait Loci/genetics , Recombination, Genetic/genetics
12.
Prog Biophys Mol Biol ; 98(2-3): 179-85, 2008.
Article in English | MEDLINE | ID: mdl-19351513

ABSTRACT

Pacemaker activity of the heart is generated by a small group of cells forming the sinoatrial node (SAN). Cells of the SAN are spontaneously active and generate action potentials with remarkable regularity and stability under all physiological conditions. The exact molecular mechanisms underlying pacemaker potentials in the SAN have not yet been fully elucidated. Several voltage-dependent ion channels as well as intracellular calcium cycling processes are thought to contribute to the pacemaker activity. Hyperpolarization-activated cation channels, which generate the I(f) current, have biophysical properties which seem ideally suited for the initiation of spontaneous electrical activity. This review describes recent work on several transgenic mice lacking different cardiac HCN channel subtypes. The role of I(f) for normal pacemaking and sinus node arrhythmia as revealed by these genetic models will be discussed. In addition, a new mouse line is described which enables gene targeting in a temporally-controlled manner selectively in SAN cells. Elucidating the function of HCN and other ion channels in well-controlled mouse models should ultimately lead to a better understanding of the mechanisms underlying human sinoatrial arrhythmias.


Subject(s)
Arrhythmia, Sinus/physiopathology , Ion Channels/physiology , Animals , Arrhythmia, Sinus/genetics , Cyclic Nucleotide-Gated Cation Channels/deficiency , Cyclic Nucleotide-Gated Cation Channels/genetics , Cyclic Nucleotide-Gated Cation Channels/physiology , Disease Models, Animal , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Ion Channels/deficiency , Ion Channels/genetics , Mice , Mice, Knockout , Mice, Transgenic
13.
EMBO J ; 26(21): 4423-32, 2007 Oct 31.
Article in English | MEDLINE | ID: mdl-17914461

ABSTRACT

Cardiac pacemaking involves a variety of ion channels, but their relative importance is controversial and remains to be determined. Hyperpolarization-activated, cyclic nucleotide-gated (HCN) channels, which underlie the I(f) current of sinoatrial cells, are thought to be key players in cardiac automaticity. In addition, the increase in heart rate following beta-adrenergic stimulation has been attributed to the cAMP-mediated enhancement of HCN channel activity. We have now studied mice in which the predominant sinoatrial HCN channel isoform HCN4 was deleted in a temporally controlled manner. Here, we show that deletion of HCN4 in adult mice eliminates most of sinoatrial I(f) and results in a cardiac arrhythmia characterized by recurrent sinus pauses. However, the mutants show no impairment in heart rate acceleration during sympathetic stimulation. Our results reveal that unexpectedly the channel does not play a role for the increase of the heart rate; however, HCN4 is necessary for maintaining a stable cardiac rhythm, especially during the transition from stimulated to basal cardiac states.


Subject(s)
Cyclic Nucleotide-Gated Cation Channels/metabolism , Gene Expression Regulation , Heart Rate , Animals , Cyclic AMP/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Ion Channel Gating , Ion Channels/metabolism , Isoproterenol/pharmacology , Mice , Mice, Knockout , Mice, Transgenic , Mutation , Protein Isoforms , Sinoatrial Node/metabolism , Sinoatrial Node/pathology
14.
Pflugers Arch ; 454(4): 517-22, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17549513

ABSTRACT

Hyperpolarization-activated cation currents termed I (f/h) are observed in many neurons and cardiac cells. Four genes (HCN1-4) encode the channels underlying these currents. New insights into the pathophysiological significance of HCN channels have been gained recently from analyses of mice engineered to be deficient in HCN genes. Lack of individual subunits results in markedly different phenotypes. Disruption of HCN1 impairs motor learning but enhances spatial learning and memory. Deletion of HCN2 results in absence epilepsy, ataxia, and sinus node dysfunction. Mice lacking HCN4 die during embryonic development and develop no sinoatrial node-like action potentials. In the present review, we summarize the physiology and pathophysiology of HCN channel family members based primarily on information from the transgenic mouse models and on data from human patients carrying defects in HCN4 channels.


Subject(s)
Ion Channels/physiopathology , Potassium Channels/physiology , Animals , Brain/physiopathology , Cardiovascular System/physiopathology , Cyclic Nucleotide-Gated Cation Channels , Gene Expression Regulation , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Ion Channels/genetics , Mice , Mice, Transgenic , Muscle Proteins/genetics , Neurons/physiology , Potassium Channels/genetics , Synaptic Transmission/physiology
15.
Mol Endocrinol ; 21(3): 753-64, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17158221

ABSTRACT

Hyperpolarization-activated cyclic nucleotide-modulated (HCN) channels mediate the pacemaker current (Ih or If) observed in electrically rhythmic cardiac and neuronal cells. Here we describe a hyperpolarization-activated time-dependent cationic current, beta-Ih, in pancreatic beta-cells. Transcripts for HCN1-4 were detected by RT-PCR and quantitative PCR in rat islets and MIN6 mouse insulinoma cells. beta-Ih in rat beta-cells and MIN6 cells displayed biophysical and pharmacological properties similar to those of HCN currents in cardiac and neuronal cells. Stimulation of cAMP production with forskolin/3-isobutyl-1-methylxanthine (50 microM) or dibutyryl-cAMP (1 mM) caused a significant rightward shift in the midpoint activation potential of beta-Ih, whereas expression of either specific small interfering (si)RNA against HCN2 (siHCN2b) or a dominant-negative HCN channel (HCN1-AAA) caused a near-complete inhibition of time-dependent beta-Ih. However, expression of siHCN2b in MIN6 cells had no affect on glucose-stimulated insulin secretion under normal or cAMP-stimulated conditions. Blocking beta-Ih in intact rat islets also did not affect membrane potential behavior at basal glucose concentrations. Taken together, our experiments provide the first evidence for functional expression of HCN channels in the pancreatic beta-cell.


Subject(s)
Insulin-Secreting Cells/metabolism , Potassium Channel Blockers/metabolism , Potassium Channels/metabolism , Animals , Benzazepines/pharmacology , Cells, Cultured , Cyclic AMP/physiology , Cyclic Nucleotide-Gated Cation Channels , Electrophysiology , Exocytosis/drug effects , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/physiology , Insulinoma/pathology , Membrane Potentials/drug effects , Mice , Piperidines/pharmacology , Pyrimidines/pharmacology , RNA, Small Interfering/pharmacology , Rats
16.
Mol Pharmacol ; 69(4): 1328-37, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16387796

ABSTRACT

Sinus node inhibitors reduce the heart rate presumably by blocking the pacemaker current If in the cardiac conduction system. This pacemaker current is carried by four hyperpolarization-activated, cyclic nucleotide-gated cation (HCN) channels. We tested the potential subtype-specificity of the sinus node inhibitors cilobradine, ivabradine, and zatebradine using cloned HCN channels. All three substances blocked the slow inward current through human HCN1, HCN2, HCN3, and HCN4 channels. There was no subtype-specificity for the steady-state block, with mean IC50 values of 0.99, 2.25, and 1.96 microM for cilobradine, ivabradine, and zatebradine, respectively. Native If, recorded from mouse sinoatrial node cells, was slightly more efficiently blocked by cilobradine (IC50 value of 0.62 microM) than were the HCN currents. The block of I(f) in sinoatrial node cells resulted in slower and dysrhythmic spontaneous action potentials. The in vivo action of these blockers was analyzed using telemetric ECG recordings in mice. Each compound reduced the heart rate dose-dependently from 600 to 200 bpm with ED50 values of 1.2, 4.7, and 1.8 mg/kg for cilobradine, ivabradine, and zatebradine, respectively. beta-Adrenergic stimulation or forced physical activity only partly reversed this bradycardia. In addition to bradycardia, all three drugs induced increasing arrhythmia at concentrations greater than 5 mg/kg for cilobradine, greater than 10 mg/kg for zatebradine, or greater than 15 mg/kg for ivabradine. This dysrhythmic heart rate is characterized by periodic fluctuations of the duration between the T and P wave, resembling a form of sick sinus syndrome in humans. Hence, all available sinus node inhibitors possess an as-yet-unrecognized proarrhythmic potential.


Subject(s)
Arrhythmias, Cardiac/chemically induced , Bradycardia/chemically induced , Sinoatrial Node/drug effects , Animals , Arrhythmias, Cardiac/physiopathology , Benzazepines/pharmacology , Bradycardia/physiopathology , Cardiotonic Agents/pharmacology , Cloning, Molecular , Electrocardiography , Humans , Ivabradine , Mice , Mice, Inbred C57BL , Piperidines/pharmacology , Up-Regulation
17.
J Biol Chem ; 280(41): 34635-43, 2005 Oct 14.
Article in English | MEDLINE | ID: mdl-16043489

ABSTRACT

Hyperpolarization-activated, cyclic nucleotide-gated cation (HCN) channels underlie the inward pacemaker current, termed I(f)/I(h), in a variety of tissues. Many details are known for the HCN subtypes 1, 2, and 4. We now successfully cloned the cDNA for HCN3 from human brain and compared the electrophysiological properties of hHCN3 to the other three HCN subtypes. Overexpression of human HCN3 channels in HEK293 cells resulted in a functional channel protein. Similar to hHCN2 channels, hHCN3 channels are activated with a rather slow time constant of 1244 +/- 526 ms at -100 mV, which is a greater time constant than that of HCN1 but a smaller one than that of HCN4 channels. The membrane potential for half-maximal activation V((1/2)) was -77 +/- 5.4 mV, and the reversal potential E(rev) was -20.5 +/- 4 mV, resulting in a permeability ratio P(Na)/P(K) of 0.3. Like all other HCNs, hHCN3 was inhibited rapidly and reversibly by extracellular cesium and slowly and irreversibly by extracellular applied ZD7288. Surprisingly, the human HCN3 channel was not modulated by intracellular cAMP, a hallmark of the other known HCN channels. Sequence comparison revealed >80% homology of the transmembrane segments, the pore region, and the cyclic nucleotide binding domain of hHCN3 with the other HCN channels. The missing response to cAMP distinguishes human HCN3 from both the well cAMP responding HCN subtypes 2 and 4 and the weak responding subtype 1.


Subject(s)
Gene Expression Regulation , Potassium Channels/biosynthesis , Potassium Channels/physiology , Blotting, Northern , Blotting, Western , Brain/metabolism , Cardiotonic Agents/pharmacology , Cations , Cell Line , Cell Membrane/metabolism , Cesium/chemistry , Cesium/pharmacology , Cloning, Molecular , Cyclic AMP/metabolism , Cyclic Nucleotide-Gated Cation Channels , DNA, Complementary/metabolism , Electrophysiology , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Ion Channels/chemistry , Kinetics , Membrane Potentials , Models, Biological , Plasmids/metabolism , Pyrimidines/pharmacology , RNA, Messenger/metabolism , Recombinant Fusion Proteins/chemistry , Sequence Analysis, DNA , Time Factors , Transfection
18.
EMBO J ; 23(21): 4222-31, 2004 Oct 27.
Article in English | MEDLINE | ID: mdl-15483626

ABSTRACT

Signalling by cGMP-dependent protein kinase type I (cGKI) relaxes various smooth muscles modulating thereby vascular tone and gastrointestinal motility. cGKI-dependent relaxation is possibly mediated by phosphorylation of the inositol 1,4,5-trisphosphate receptor I (IP(3)RI)-associated protein (IRAG), which decreases hormone-induced IP(3)-dependent Ca(2+) release. We show now that the targeted deletion of exon 12 of IRAG coding for the N-terminus of the coiled-coil domain disrupted in vivo the IRAG-IP(3)RI interaction and resulted in hypomorphic IRAG(Delta12/Delta12) mice. These mice had a dilated gastrointestinal tract and a disturbed gastrointestinal motility. Carbachol- and phenylephrine-contracted smooth muscle strips from colon and aorta, respectively, of IRAG(Delta12/Delta12) mice were not relaxed by cGMP, while cAMP-mediated relaxation was unperturbed. Norepinephrine-induced increases in [Ca(2+)](i) were not decreased by cGMP in aortic smooth muscle cells from IRAG(Delta12/Delta12) mice. In contrast, cGMP-induced relaxation of potassium-induced smooth muscle contraction was not abolished in IRAG(Delta12/Delta12) mice. We conclude that cGMP-dependent relaxation of hormone receptor-triggered smooth muscle contraction essentially depends on the interaction of cGKI-IRAG with IP(3)RI.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/metabolism , Cyclic GMP/analogs & derivatives , Muscle Contraction/physiology , Muscle Relaxation/physiology , Muscle, Smooth/physiology , Phosphoproteins , Animals , Aorta/anatomy & histology , Aorta/drug effects , COS Cells , Calcium/metabolism , Carbachol/pharmacology , Chlorocebus aethiops , Cholinergic Agonists/pharmacology , Colon/anatomy & histology , Colon/drug effects , Cyclic GMP/pharmacology , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Exons , Gadolinium/metabolism , Gastrointestinal Motility/drug effects , Gastrointestinal Tract/anatomy & histology , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/pathology , Gastrointestinal Tract/physiology , Gene Targeting , In Vitro Techniques , Membrane Proteins , Mice , Muscle Contraction/drug effects , Muscle Relaxation/drug effects , Muscle, Smooth/drug effects , Phosphoproteins/genetics , Phosphoproteins/metabolism
19.
Trends Cardiovasc Med ; 14(1): 23-8, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14720471

ABSTRACT

Cardiac pacemaker activity is regulated by at least five different classes of ion channels and by the opposing influence of sympathetic and parasympathetic stimulation. Inactivation of several genes, including a subunit coding for the potassium channel activated by the muscarinic receptor, I(KACh); the calcium channel, I(Ca,); and the hyperpolarization-activated channel, I(f), results in sinus node arrhythmia. Inactivation of the gene for the hyperpolarization-activated, cyclic nucleotide-gated channel isoform HCN2 or HCN4 and the use of pacemaker channel blockers show that (a) HCN2 prevents the diastolic membrane potential from becoming too negative, (b) HCN4 is the major channel mediating sympathetic stimulation of the pacemaker activity, and (3) complete blockage of the I(f) current is compatible with slow sinus node rhythm.


Subject(s)
Action Potentials , Heart Rate/genetics , Ion Channels/metabolism , Sinoatrial Node/physiology , Animals , Arrhythmia, Sinus/genetics , Arrhythmia, Sinus/physiopathology , Cyclic Nucleotide-Gated Cation Channels , Electrophysiology , Heart Rate/physiology , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Ion Channels/drug effects , Ion Channels/genetics , Mice , Mice, Knockout , Muscle Proteins/genetics , Potassium Channels , Rabbits , Risk Factors , Sensitivity and Specificity
20.
Proc Natl Acad Sci U S A ; 100(25): 15235-40, 2003 Dec 09.
Article in English | MEDLINE | ID: mdl-14657344

ABSTRACT

Hyperpolarization-activated, cyclic nucleotide-gated cation currents, termed If or Ih, are generated by four members of the hyperpolarization-activated, cyclic nucleotide-gated cation (HCN) channel family. These currents have been proposed to contribute to several functions including pacemaker activity in heart and brain, control of resting potential, and neuronal plasticity. Transcripts of the HCN4 isoform have been found in cardiomyocytes and neurons, but the physiological role of this channel is unknown. Here we show that HCN4 is essential for the proper function of the developing cardiac conduction system. In wild-type embryos, HCN4 is highly expressed in the cardiac region where the early sinoatrial node develops. Mice lacking HCN4 channels globally, as well as mice with a selective deletion of HCN4 in cardiomyocytes, died between embryonic days 9.5 and 11.5. On average, If in cardiomyocytes from mutant embryos is reduced by 85%. Hearts from HCN4-deficient embryos contracted significantly slower compared with wild type and could not be stimulated by cAMP. In both wild-type and HCN4-/- mice, cardiac cells with "primitive" pacemaker action potentials could be found. However, cardiac cells with "mature" pacemaker potentials, observed in wild-type embryos starting at day 9.0, were not detected in HCN4-deficient embryos. Thus, HCN4 channels are essential for the proper generation of pacemaker potentials in the emerging sinoatrial node.


Subject(s)
Action Potentials , Ion Channels/physiology , Muscle Proteins/physiology , Pacemaker, Artificial , Animals , Blotting, Western , Cardiovascular Agents/pharmacology , Cations , Cyclic AMP/metabolism , Cyclic Nucleotide-Gated Cation Channels , Dose-Response Relationship, Drug , Electrophysiology , Gene Deletion , Heart Rate , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Immunohistochemistry , In Situ Hybridization , Ion Channels/metabolism , Lac Operon , Membrane Potentials , Mice , Mice, Transgenic , Muscle Proteins/metabolism , Myocardium/cytology , Potassium Channels , Protein Isoforms , Pyrimidines/pharmacology , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sinoatrial Node , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...