Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Dis Markers ; 2018: 6924608, 2018.
Article in English | MEDLINE | ID: mdl-30581499

ABSTRACT

BACKGROUND: Left atrial enlargement is a mortality and heart failure risk factor in primary mitral regurgitation (MR) patients. Pig models of MR have shown differential expression of genes linked to the renin-angiotensin system. Therefore, the aim of this study was to investigate the key genes of the renin-angiotensin that are expressed differentially in the left atrial myocardium in MR patients. METHODS: Quantitative RT-PCR was used to compare gene expression in the renin-angiotensin system in the left atrium in MR patients, aortic valve disease patients, and normal subjects. RESULTS: Plasma angiotensin II concentrations did not significantly differ between MR patients and aortic valve disease patients (P = 0.582). Compared to normal controls, however, MR patients had significantly downregulated expressions of angiotensin-converting enzyme, angiotensin I converting enzyme 2, type 1 angiotensin II receptor, glutamyl aminopeptidase, angiotensinogen, cathepsin A (CTSA), thimet oligopeptidase 1, neurolysin, alanyl aminopeptidase, cathepsin G, leucyl/cystinyl aminopeptidase (LNPEP), neprilysin, and carboxypeptidase A3 in the left atrium. The MR patients also had significantly upregulated expressions of MAS1 oncogene (MAS1) and mineralocorticoid receptor compared to normal controls. Additionally, in comparison with aortic valve disease patients, MR patients had significantly downregulated CTSA and LNPEP expression and significantly upregulated MAS1 expression in the left atrium. CONCLUSIONS: Expressions of genes in the renin-angiotensin system, especially CTSA, LNPEP, and MAS1, in the left atrium in MR patients significantly differed from expressions of these genes in aortic valve disease patients and normal controls. Notably, differences in expression were independent of circulating angiotensin II levels. The results of this study provide a rationale for pharmacological therapies or posttranslational regulation therapies targeting genes expressed differentially in the renin-angiotensin system to remedy structural remodeling associated with atrial enlargement and heart failure progression in patients with MR.


Subject(s)
Atrial Function/genetics , Heart Atria , Mitral Valve Insufficiency/genetics , Renin-Angiotensin System/genetics , Aged , Angiotensin II/analysis , Angiotensin II/blood , Case-Control Studies , Cathepsin A/genetics , Cystinyl Aminopeptidase/genetics , Female , Heart Failure/genetics , Heart Valve Diseases/genetics , Humans , Male , Middle Aged , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , Receptors, G-Protein-Coupled/genetics
2.
Int J Mol Sci ; 18(9)2017 Aug 30.
Article in English | MEDLINE | ID: mdl-28867785

ABSTRACT

Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) hold great promise for studying inherited cardiac arrhythmias and developing drug therapies to treat such arrhythmias. Unfortunately, until now, action potential (AP) measurements in hiPSC-CMs have been hampered by the virtual absence of the inward rectifier potassium current (IK1) in hiPSC-CMs, resulting in spontaneous activity and altered function of various depolarising and repolarising membrane currents. We assessed whether AP measurements in "ventricular-like" and "atrial-like" hiPSC-CMs could be improved through a simple, highly reproducible dynamic clamp approach to provide these cells with a substantial IK1 (computed in real time according to the actual membrane potential and injected through the patch-clamp pipette). APs were measured at 1 Hz using perforated patch-clamp methodology, both in control cells and in cells treated with all-trans retinoic acid (RA) during the differentiation process to increase the number of cells with atrial-like APs. RA-treated hiPSC-CMs displayed shorter APs than control hiPSC-CMs and this phenotype became more prominent upon addition of synthetic IK1 through dynamic clamp. Furthermore, the variability of several AP parameters decreased upon IK1 injection. Computer simulations with models of ventricular-like and atrial-like hiPSC-CMs demonstrated the importance of selecting an appropriate synthetic IK1. In conclusion, the dynamic clamp-based approach of IK1 injection has broad applicability for detailed AP measurements in hiPSC-CMs.


Subject(s)
Action Potentials/physiology , Arrhythmias, Cardiac/physiopathology , Induced Pluripotent Stem Cells/physiology , Myocytes, Cardiac/physiology , Action Potentials/genetics , Arrhythmias, Cardiac/drug therapy , Arrhythmias, Cardiac/genetics , Atrial Function/genetics , Cell Differentiation/drug effects , Cell Differentiation/physiology , Heart Ventricles/physiopathology , Humans , Induced Pluripotent Stem Cells/drug effects , Patch-Clamp Techniques , Potassium/metabolism , Potassium Channels, Inwardly Rectifying , Tretinoin/administration & dosage
3.
Ann Med ; 49(6): 496-503, 2017 09.
Article in English | MEDLINE | ID: mdl-28294644

ABSTRACT

INTRODUCTION: The cardiac sodium channel SCN5A regulates atrioventricular and ventricular depolarization as well as cardiac conduction. Patients with cardiac electrical abnormalities have an increased risk of sudden cardiac death (SCD) and cardio-embolic stroke. Optimal management of cardiac disease includes the understanding of association between the causative mutations and the clinical phenotype. A 12-lead electrocardiogram (ECG) is an easy and inexpensive tool for finding risk patients. MATERIALS AND METHODS: A blood sample for DNA extraction was obtained in a Finnish family with 43 members; systematic 12-lead ECG analysis was performed in 13 of the family members carrying an SCN5A D1275N mutation. Conduction defects and supraventricular arrhythmias, including atrial fibrillation/flutter, atrioventricular nodal re-entry tachycardia (AVNRT) and junctional rhythm were searched for. RESULTS: Five (38%) mutation carriers had fascicular or bundle branch block, 10 had atrial arrhythmias; no ventricular arrhythmias were found. Notching of the R- and S waves - including initial QRS fragmentation - and prolonged S-wave upstroke were present in all the affected family members. Notably, four (31%) affected family members had a stroke before the age of 31 and two experienced premature death. CONCLUSIONS: A 12-lead ECG can be used to predict arrhythmias in SCN5A D1275N mutation carriers. Key messages The 12-lead ECG may reveal cardiac abnormalities even before clinical symptoms occur. Specific ECG findings - initial QRS fragmentation, prolonged S-wave upstroke as well as supraventricular arrhythmias - were frequently encountered in all SCN5A D1257N mutation carriers. ECG follow-up is recommended for all SCN5A D1275N mutation carriers.


Subject(s)
Arrhythmias, Cardiac/genetics , Electrocardiography/methods , Mutation , NAV1.5 Voltage-Gated Sodium Channel/blood , Pedigree , Adolescent , Adult , Arrhythmias, Cardiac/diagnosis , Atrial Function/genetics , Child , Female , Finland , Genotype , Heart Atria/diagnostic imaging , Heart Conduction System/physiopathology , Humans , Male , Middle Aged , Phenotype , Young Adult
4.
Sci Transl Med ; 8(354): 354ra115, 2016 08 31.
Article in English | MEDLINE | ID: mdl-27582060

ABSTRACT

Cardiac rhythm is extremely robust, generating 2 billion contraction cycles during the average human life span. Transcriptional control of cardiac rhythm is poorly understood. We found that removal of the transcription factor gene Tbx5 from the adult mouse caused primary spontaneous and sustained atrial fibrillation (AF). Atrial cardiomyocytes from the Tbx5-mutant mice exhibited action potential abnormalities, including spontaneous depolarizations, which were rescued by chelating free calcium. We identified a multitiered transcriptional network that linked seven previously defined AF risk loci: TBX5 directly activated PITX2, and TBX5 and PITX2 antagonistically regulated membrane effector genes Scn5a, Gja1, Ryr2, Dsp, and Atp2a2 In addition, reduced Tbx5 dose by adult-specific haploinsufficiency caused decreased target gene expression, myocardial automaticity, and AF inducibility, which were all rescued by Pitx2 haploinsufficiency in mice. These results defined a transcriptional architecture for atrial rhythm control organized as an incoherent feed-forward loop, driven by TBX5 and modulated by PITX2. TBX5/PITX2 interplay provides tight control of atrial rhythm effector gene expression, and perturbation of the co-regulated network caused AF susceptibility. This work provides a model for the molecular mechanisms underpinning the genetic implication of multiple AF genome-wide association studies loci and will contribute to future efforts to stratify patients for AF risk by genotype.


Subject(s)
Gene Regulatory Networks , Heart Rate/genetics , Homeodomain Proteins/genetics , T-Box Domain Proteins/genetics , Transcription Factors/genetics , Animals , Atrial Fibrillation/genetics , Atrial Fibrillation/physiopathology , Atrial Function/genetics , Atrial Function/physiology , Calcium Signaling , Disease Models, Animal , Genetic Predisposition to Disease , Genome-Wide Association Study , Haploinsufficiency , Heart Rate/physiology , Homeodomain Proteins/physiology , Humans , Mice , Mice, Knockout , Myocardial Contraction/genetics , Myocardial Contraction/physiology , T-Box Domain Proteins/deficiency , T-Box Domain Proteins/physiology , Transcription Factors/deficiency , Transcription Factors/physiology , Translational Research, Biomedical , Homeobox Protein PITX2
5.
J Cardiovasc Electrophysiol ; 27(12): 1420-1428, 2016 12.
Article in English | MEDLINE | ID: mdl-27572111

ABSTRACT

INTRODUCTION: Arrhythmogenic right ventricular dysplasia/cardiomyopathy (ARVD/C) is associated with desmosomal mutations. Although desmosomal disruption affects both ventricles and atria, little is known about atrial involvement in ARVD/C. OBJECTIVE: To describe the extent and clinical significance of structural atrial involvement and atrial arrhythmias (AA) in ARVD/C stratified by genotype. METHODS: We included 71 patients who met ARVD/C Task Force Criteria and underwent cardiac magnetic resonance (CMR) imaging and molecular genetic analysis. Indexed atrial end-diastolic volume and area-length-ejection-fraction (ALEF) were evaluated on CMR and compared to controls with idiopathic right ventricular outflow tract tachycardia (n = 40). The primary outcome was occurrence of AA (atrial fibrillation or atrial flutter) during follow-up, recorded by 12-lead ECG, Holter monitoring or implantable cardioverter defibrillator (ICD) interrogation. RESULTS: Patients harbored a desmosomal plakophilin-2 (PKP2) (n = 37) or nondesmosomal phospholamban (PLN) (n = 14) mutation. In 20 subjects, no pathogenic mutation was identified. Compared to controls, right atrial (RA) volumes were reduced in PKP2 (P = 0.002) and comparable in PLN (P = 0.441) mutation carriers. In patients with no mutation identified, RA (P = 0.011) and left atrial (P = 0.034) volumes were increased. Bi-atrial ALEF showed no significant difference between the groups. AA were experienced by 27% of patients and occurred equally among PKP2 (30%) and no mutation identified patients (30%), but less among PLN mutation carriers (14%). CONCLUSION: Genotype influences atrial volume and occurrence of AA in ARVD/C. While the incidence of AA is similar in PKP2 mutation carriers and patients with no mutation identified, PKP2 mutation carriers have significantly smaller atria. This suggests a different arrhythmogenic mechanism.


Subject(s)
Arrhythmogenic Right Ventricular Dysplasia/genetics , Atrial Fibrillation/genetics , Atrial Flutter/genetics , Atrial Function/genetics , Calcium-Binding Proteins/genetics , Heart Atria/physiopathology , Mutation , Plakophilins/genetics , Adult , Arrhythmogenic Right Ventricular Dysplasia/diagnosis , Arrhythmogenic Right Ventricular Dysplasia/physiopathology , Atrial Fibrillation/diagnosis , Atrial Fibrillation/physiopathology , Atrial Flutter/diagnosis , Atrial Flutter/physiopathology , Case-Control Studies , Cross-Sectional Studies , DNA Mutational Analysis , Electrocardiography, Ambulatory , Female , Genetic Predisposition to Disease , Heart Atria/diagnostic imaging , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Phenotype , Registries , Risk Factors
6.
Eur Heart J ; 36(11): 686-97, 2015 Mar 14.
Article in English | MEDLINE | ID: mdl-24216388

ABSTRACT

AIMS: Loss-of-function mutations in Calsequestrin 2 (CASQ2) are associated with catecholaminergic polymorphic ventricular tachycardia (CPVT). CPVT patients also exhibit bradycardia and atrial arrhythmias for which the underlying mechanism remains unknown. We aimed to study the sinoatrial node (SAN) dysfunction due to loss of CASQ2. METHODS AND RESULTS: In vivo electrocardiogram (ECG) monitoring, in vitro high-resolution optical mapping, confocal imaging of intracellular Ca(2+) cycling, and 3D atrial immunohistology were performed in wild-type (WT) and Casq2 null (Casq2(-/-)) mice. Casq2(-/-) mice exhibited bradycardia, SAN conduction abnormalities, and beat-to-beat heart rate variability due to enhanced atrial ectopic activity both at baseline and with autonomic stimulation. Loss of CASQ2 increased fibrosis within the pacemaker complex, depressed primary SAN activity, and conduction, but enhanced atrial ectopic activity and atrial fibrillation (AF) associated with macro- and micro-reentry during autonomic stimulation. In SAN myocytes, CASQ2 deficiency induced perturbations in intracellular Ca(2+) cycling, including abnormal Ca(2+) release, periods of significantly elevated diastolic Ca(2+) levels leading to pauses and unstable pacemaker rate. Importantly, Ca(2+) cycling dysfunction occurred not only at the SAN cellular level but was also globally manifested as an increased delay between action potential (AP) and Ca(2+) transient upstrokes throughout the atrial pacemaker complex. CONCLUSIONS: Loss of CASQ2 causes abnormal sarcoplasmic reticulum Ca(2+) release and selective interstitial fibrosis in the atrial pacemaker complex, which disrupt SAN pacemaking but enhance latent pacemaker activity, create conduction abnormalities and increase susceptibility to AF. These functional and extensive structural alterations could contribute to SAN dysfunction as well as AF in CPVT patients.


Subject(s)
Atrial Fibrillation/genetics , Bradycardia/genetics , Calsequestrin/genetics , Gene Deletion , Sarcoplasmic Reticulum/metabolism , Sinoatrial Node/physiology , Action Potentials/physiology , Animals , Atrial Function/genetics , Calcium/metabolism , Calsequestrin/deficiency , Cardiomegaly/genetics , Fibrosis/genetics , Gene Knockout Techniques , Mice, Transgenic , Sinoatrial Node/pathology
7.
Cardiovasc Res ; 91(2): 223-31, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21427120

ABSTRACT

The development of the heart is a complex process during which different cell types progressively contribute to shape a four-chambered pumping organ. Over the last decades, our understanding of the specification and transcriptional regulation of cardiac development has been greatly augmented as has our understanding of the functional bases of cardiac electrophysiology during embryogenesis. The nascent heart gradually acquires distinct cellular and functional characteristics, such as the formation of contractile structures, the development of conductive capabilities, and soon thereafter the co-ordinated conduction of the electrical impulse, in order to fulfil its functional properties. Over the last decade, we have learnt about the consequences of impairing cardiac morphogenesis, which in many cases leads to congenital heart defects; however, we are not yet aware of the consequences of impairing electrical function during cardiogenesis. The most prevalent cardiac arrhythmia is atrial fibrillation (AF), although its genetic aetiology remains rather elusive. Recent genome-wide association studies have identified several genetic variants highly associated with AF. Among them are genetic variants located on chromosome 4q25 adjacent to PITX2, a transcription factor known to play a critical role in left-right asymmetry and cardiogenesis. Here, we review new insights into the cellular and molecular links between PITX2 and AF.


Subject(s)
Atrial Fibrillation/genetics , Atrial Function/genetics , Gene Expression Regulation, Developmental , Heart Conduction System/embryology , Heart Rate/genetics , Homeodomain Proteins/genetics , Transcription Factors/genetics , Action Potentials , Animals , Atrial Fibrillation/embryology , Atrial Fibrillation/metabolism , Atrial Fibrillation/physiopathology , Genetic Predisposition to Disease , Heart Conduction System/metabolism , Heart Conduction System/physiopathology , Homeodomain Proteins/metabolism , Humans , Kinetics , Morphogenesis , Phenotype , Transcription Factors/metabolism , Homeobox Protein PITX2
8.
Trends Cardiovasc Med ; 19(4): 123-30, 2009 May.
Article in English | MEDLINE | ID: mdl-19818948

ABSTRACT

Atrial fibrillation (AF) is the most common cardiac arrhythmia encountered in clinical practice. The abnormal rhythm is associated not only with a variety of symptoms, such as palpitations, dizziness, or shortness of breath, but also with increased risk of stroke, heart failure, and mortality. A genetic predisposition is suggested by the fact that the relative risk for the development of AF is estimated at 85% in individuals with at least one parent with a history of AF. Current therapeutic strategies include control of rate or rhythm with medication and catheter ablation procedures. Especially in the pathophysiology of paroxysmal AF, ectopic electrical activity originating in the myocardial sleeves surrounding the pulmonary veins is considered causal. In these cases, ablation is applied to isolate the pulmonary venous myocardium from the remainder of the left atrial myocardium. Other recent evidence has shown that genetic and developmental defects can be involved in the development of AF. In this review, it is our aim to discuss the possible underlying causes of AF from a combined genetic and cardiac developmental view.


Subject(s)
Atrial Fibrillation/etiology , Animals , Anti-Arrhythmia Agents/therapeutic use , Atrial Fibrillation/embryology , Atrial Fibrillation/genetics , Atrial Fibrillation/therapy , Atrial Function/genetics , Catheter Ablation , Gene Expression Regulation, Developmental , Genetic Predisposition to Disease , Genotype , Humans , Pedigree , Phenotype , Polymorphism, Genetic , Risk Assessment , Risk Factors
10.
Circ J ; 73(9): 1691-7, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19609041

ABSTRACT

BACKGROUND: Mutations of cardiac troponin C (cTnC) can cause dilated cardiomyopathy in humans. METHODS AND RESULTS: Plasmids were constructed such that the reverse tetracycline-controlled transactivator (rtTA) was driven by the cardiac myosin light chain 2 promoter. This heart-specific rtTA bound another bidirectional promoter to express the green fluorescence protein reporter gene and the antisense RNA of cTnC in the presence of doxycycline. A transgenic line of zebrafish (CA17) with cTnC dysfunction was also generated. The heart rates of the embryos in the CA17 line were significantly slower than those of embryos in the control T03 transgenic line at 6 and 12 days post fertilization (dpf). In the CA17 line, cardiac chambers in the F2 embryos were significantly greater and the ventricular ejection fraction was lower than those in the T03 at both 6 and 12 dpf. The mortality rate of F2 adult fish of the CA17 line was also significantly higher (P<0.001). CONCLUSIONS: Using conditional expression of antisense RNA of zebrafish cTnC, a new animal model with phenotypes simulating dilated cardiomyopathy has been created.


Subject(s)
Cardiomyopathy, Dilated/genetics , Gene Knockdown Techniques , RNA, Antisense/biosynthesis , Troponin C/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Amino Acid Sequence , Animals , Animals, Genetically Modified , Atrial Function/genetics , Base Sequence , Blotting, Western , Cardiac Myosins/genetics , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , Cardiomyopathy, Dilated/physiopathology , Disease Models, Animal , Genotype , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Heart Rate/genetics , Molecular Sequence Data , Myocardial Contraction/genetics , Myocardium/metabolism , Myocardium/pathology , Myosin Light Chains/genetics , Phenotype , Promoter Regions, Genetic , Stroke Volume/genetics , Transcription, Genetic , Troponin C/metabolism , Ventricular Function/genetics , Zebrafish Proteins/metabolism
11.
Clin Exp Pharmacol Physiol ; 36(4): 386-93, 2009 Apr.
Article in English | MEDLINE | ID: mdl-18986326

ABSTRACT

1. ClC-3 has been proposed as a molecular candidate responsible for volume-sensitive outwardly rectifying anion channels (VSOAC) in cardiac and smooth muscle cells. To further test this hypothesis, we produced a novel line of transgenic mice with cardiac-specific overexpression of the human short ClC-3 isoform (hsClC-3). 2. Northern and western blot analyses demonstrated that mRNA and protein levels of the short isoform (sClC-3) in the heart were significantly increased in hsClC-3-overexpressing (OE) mice compared with wild-type (WT) mice. Heart weight : bodyweight ratios for OE mice were significantly smaller compared with age-matched WT mice. 3. Electrocardiogram recordings indicated no difference at rest, whereas echocardiographic recordings revealed consistent reductions in left ventricular diastolic diameter, left ventricular posterior wall thickness at end of diastole and interventricular septum thickness in diastole in OE mice. 4. The VSOAC current densities in atrial cardiomyocytes were significantly increased by ClC-3 overexpression compared with WT cells. No differences in VSOAC current properties in OE and WT atrial myocytes were observed in terms of outward rectification, anion permeability (I(-) > Cl(-) > Asp(-)) and inhibition by 4,4'-diisothiocyanatostilbene-2,2'-disulphonic acid and glibenclamide. The VSOAC in atrial myocytes from both groups were totally abolished by phorbol-12,13-dibutyrate (a protein kinase C activator) and by intracellular dialysis of an N-terminal anti-ClC-3 antibody. 5. Cardiac cell volume measurements revealed a significant acceleration of the rate of regulatory volume decrease (RVD) in OE myocytes compared with WT. 6. In conclusion, enhanced VSOAC currents and acceleration of the time-course of RVD in atrial myocytes of OE mice is strong evidence supporting an essential role of sClC-3 in native VSOAC function in mouse atrial myocytes.


Subject(s)
Chloride Channels/genetics , Myocardium/metabolism , Animals , Atrial Function/genetics , Chloride Channels/metabolism , Electrophysiology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Organ Specificity/genetics , Patch-Clamp Techniques , Phenotype , Protein Isoforms/genetics , Protein Isoforms/metabolism , Up-Regulation/genetics
12.
Exp Biol Med (Maywood) ; 233(5): 558-74, 2008 May.
Article in English | MEDLINE | ID: mdl-18375832

ABSTRACT

Accumulating evidence links calcium-overload and oxidative stress to atrial remodeling during atrial fibrillation (AF). Furthermore, atrial remodeling appears to increase atrial thrombogeneity, characterized by increased expression of adhesion molecules. The aim of this study was to assess mitochondrial dysfunction and oxidative stress-activated signal transduction (nuclear factor-kappaB [NF-kappa B], lectin-like oxidized low-density lipoprotein receptor [LOX-1], intercellular adhesion molecule-1 [ICAM-1], and hemeoxgenase-1 [HO-1]) in atrial tissue during AF. Ex vivo atrial tissue from patients with and without AF and, additionally, rapid pacing of human atrial tissue slices were used to study mitochondrial structure by electron microscopy and mitochondrial respiration. Furthermore, quantitative reverse transcription polymerase chain reaction (RT-PCR), immunoblot analyses, gel-shift assays, and enzyme-linked immunosorbent assay (ELISA) were applied to measure nuclear amounts of NF-kappa B target gene expression. Using ex vivo atrial tissue samples from patients with AF we demonstrated oxidative stress and impaired mitochondrial structure and respiration, which was accompanied by nuclear accumulation of NF-kappa B and elevated expression levels of the adhesion molecule ICAM-1 and the oxidative stress-induced markers HO-1 and LOX-1. All these changes were reproduced by rapid pacing for 24 hours of human atrial tissue slices. Furthermore, the blockade of calcium inward current with verapamil effectively prevented both the mitochondrial changes and the activation of NF-kappa B signaling and target gene expression. The latter appeared also diminished by the antioxidants apocynin and resveratrol (an inhibitor of NF-kappa B), or the angiotensin II receptor type 1 antagonist, olmesartan. This study demonstrates that calcium inward current via L-type calcium channels contributes to oxidative stress and increased expression of oxidative stress markers and adhesion molecules during cardiac tachyarrhythmia.


Subject(s)
Atrial Function , Mitochondrial Diseases/metabolism , Signal Transduction , Tachycardia/metabolism , Aged , Atrial Function/genetics , Cell Respiration , Female , Fibrosis/metabolism , Gene Expression Regulation/drug effects , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Humans , Intercellular Adhesion Molecule-1/genetics , Male , Microscopy, Electron , Mitochondrial Diseases/genetics , Mitochondrial Diseases/pathology , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Oxidation-Reduction , Oxidative Stress , Protein Carbonylation , Scavenger Receptors, Class E/genetics , Tachycardia/genetics , Tachycardia/pathology
13.
Genes Dev ; 21(9): 1098-112, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17473172

ABSTRACT

The sinoatrial node initiates the heartbeat and controls the rate and rhythm of contraction, thus serving as the pacemaker of the heart. Despite the crucial role of the sinoatrial node in heart function, the mechanisms that underlie its specification and formation are not known. Tbx3, a transcriptional repressor required for development of vertebrates, is expressed in the developing conduction system. Here we show that Tbx3 expression delineates the sinoatrial node region, which runs a gene expression program that is distinct from that of the bordering atrial cells. We found lineage segregation of Tbx3-negative atrial and Tbx3-positive sinoatrial node precursor cells as soon as cardiac cells turn on the atrial gene expression program. Tbx3 deficiency resulted in expansion of expression of the atrial gene program into the sinoatrial node domain, and partial loss of sinoatrial node-specific gene expression. Ectopic expression of Tbx3 in mice revealed that Tbx3 represses the atrial phenotype and imposes the pacemaker phenotype on the atria. The mice displayed arrhythmias and developed functional ectopic pacemakers. These data identify a Tbx3-dependent pathway for the specification and formation of the sinoatrial node, and show that Tbx3 regulates the pacemaker gene expression program and phenotype.


Subject(s)
Atrial Function/genetics , Sinoatrial Node/embryology , Sinoatrial Node/physiology , T-Box Domain Proteins/genetics , Animals , Base Sequence , Cell Differentiation , DNA Primers/genetics , Gene Expression Regulation, Developmental , Heart Atria/cytology , Heart Atria/embryology , Mice , Mice, Knockout , Mice, Transgenic , Myoblasts, Cardiac/cytology , Myoblasts, Cardiac/physiology , Sinoatrial Node/cytology , T-Box Domain Proteins/deficiency , T-Box Domain Proteins/physiology
14.
Heart Rhythm ; 2(10): 1128-34, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16188595

ABSTRACT

BACKGROUND: Congenital atrial standstill has been linked to SCN5A. Incomplete penetrance observed in atrial standstill has been attributed in part to the digenic inheritance of polymorphisms in the atrial-specific gap junction connexin 40 (Cx40) in conjunction with an SCN5A mutation. OBJECTIVES: The purpose of this study was to determine the clinical and biophysical characteristics of a novel SCN5A mutation identified in a family with atrial standstill. METHODS: Family members of an apparently sporadic case of atrial standstill underwent genetic screening of SCN5A and atrial-specific genes including Cx40. Biophysical properties of the wild-type (WT) and mutant SCN5A channels in a heterologous expression system were studied using the whole-cell patch clamp technique. RESULTS: The novel SCN5A mutation L212P was identified in the proband (age 11 years) and his father. The father was in normal sinus rhythm. The proband had no P waves on surface ECG, and his right atrium could not be captured by pacing. The recombinant L212P Na channel showed a large hyperpolarizing shift in both the voltage dependence of activation (WT: -48.1 +/- 0.9 mV; L212P: -63.5 +/- 1.5 mV; P < .001) and inactivation (WT: -86.6 +/- 0.9 mV; L212P: -95.6 +/- 0.8 mV; P < .001) and delayed recovery from inactivation. Further screenings for genetic variations that might mitigate L212P dysfunction revealed that the proband, but not his father, carries Cx40 polymorphisms inherited from his asymptomatic mother. CONCLUSION: These results suggest that genetic defects in SCN5A most likely underlie atrial standstill. Coinheritance of Cx40 polymorphisms is a possible genetic factor that modifies the clinical manifestation of this inherited arrhythmia.


Subject(s)
Bradycardia/genetics , Connexins/genetics , Muscle Proteins/genetics , Mutation, Missense/genetics , Polymorphism, Genetic/genetics , Sodium Channels/genetics , Alleles , Atrial Fibrillation/genetics , Atrial Fibrillation/physiopathology , Atrial Fibrillation/prevention & control , Atrial Function/genetics , Bradycardia/physiopathology , Bradycardia/prevention & control , Child, Preschool , DNA Mutational Analysis , Electrocardiography , Family Health , Female , Genetic Predisposition to Disease/genetics , Genetic Testing , Heart Atria/pathology , Heart Atria/physiopathology , Heart Rate/genetics , Humans , Male , NAV1.5 Voltage-Gated Sodium Channel , Pedigree , Phenotype , Gap Junction alpha-5 Protein
15.
Proteomics ; 4(1): 261-70, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14730688

ABSTRACT

We examine the reliability and accuracy of gene array technology in analyzing differences in gene expression between human non-diseased left atrium and left ventricle. We have used cDNA gene arrays and validated those data by carefully designed quantitative real-time polymerase chain reaction (PCR). We have identified pitfalls using cDNA gene array technology based on comparisons with other gene array studies and with changes reported for the levels of expression of the genes corresponding to these cDNAs. The high error rate reported here underscores the cautionary comments reported by others in this field.


Subject(s)
Atrial Function/genetics , Gene Expression/physiology , Ventricular Function/genetics , Atrial Function/physiology , DNA, Complementary , Gene Expression Profiling , Humans , Oligonucleotide Array Sequence Analysis , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Terminology as Topic , Ventricular Function/physiology
16.
Jpn Heart J ; 44(1): 101-10, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12622442

ABSTRACT

UNLABELLED: Thyroid hormone has been shown to modulate the gene expression of cardiac potassium channels, however, it is not known if gene expression is different between the atrium and the ventricle. The long-term effects of thyroid hormone on nuclear thyroid hormone receptors are also not known. Triiodothyronine (T3) at 25 microg/100 g of body weight or propylthiouracil (PTU) at 4 mg/100 g of body weight was given to adult rats via a gastric tube for 14 days. The levels of mRNA of Kv1.2. Kv1.4, Kv1.5, Kv2.1, Kv4.2, erg, LQT1, and minK were assayed by RNase protection assay. The mRNA of nuclear T3-receptor-al and T3-receptor-beta1 were also assayed for 15 days. After T3 (or PTU), plasma free T3 and free T4 increased (or decreased) significantly. The mRNA levels of Kv1.2 and Kv1.4 were reduced after T3 in the atrium and the ventricle. while PTU increased the levels in both chambers. Kv1.5 was significantly up-regulated by T3 in the atrium and the ventricle (P < 0.02 for both) and PTU decreased its expression in the ventricle (P < 0.02). Kv2.1 and Kv4.2 were not affected by T3 or PTU. mRNA of erg was not affected by T3 in the atrium but decreased in the ventricle (P < 0.01). After PTU, erg mRNA was decreased in the atrium (P < 0.02) but increased in the ventricle (P < 0.01). LQT1 was decreased by T3 in both chambers (P < 0.01) and not affected by PTU. minK was not detectable in the control state and was up-regulated only in the atrium: a peak on the 4th day followed by a decline to the undetectable level on the 10-15th days. During T3 treatment, nuclear T3-receptor-alpha1 and beta1 mRNA were decreased in the initial 3 days but returned to control levels thereafter. CONCLUSIONS: Between the atrium and ventricle of the adult rat heart, the responses of gene expression of voltage-gated potassium channels to T3 or PTU were quantitatively or qualitatively different and the differential responses may explain cardiac manifestations of hyperthyroidism, which is a frequent complication of supraventricular arrhythmia.


Subject(s)
Atrial Function/genetics , Gene Expression Regulation/drug effects , Potassium Channels, Voltage-Gated/drug effects , Triiodothyronine/pharmacology , Ventricular Function/genetics , Animals , Atrial Function/drug effects , Female , Heart Rate , Rats , Rats, Inbred WKY , Ventricular Function/drug effects
17.
Circ Res ; 92(1): 14-22, 2003 Jan 10.
Article in English | MEDLINE | ID: mdl-12522116

ABSTRACT

Atrial standstill (AS) is a rare arrhythmia that occasionally appears to be genetically determined. This study investigates the genetic background of this arrhythmogenic disorder in a large family. Forty-four family members were clinically evaluated. One deceased and three living relatives were unambiguously affected by AS. All other relatives appeared unaffected. Candidate gene screening revealed a novel mutation in the cardiac sodium channel gene SCN5A (D1275N) in all three affected living relatives and in five unaffected relatives, and the deceased relative was an obligate carrier. In addition, two closely linked polymorphisms were detected within regulatory regions of the gene for the atrial-specific gap junction protein connexin40 (Cx40) at nucleotides -44 (G-->A) and +71 (A-->G). Eight relatives were homozygous for both polymorphisms, which occurred in only approximately 7% of control subjects, and three of these relatives were affected by AS. The three living AS patients exclusively coinherited both the rare Cx40 genotype and the SCN5A-D1275N mutation. SCN5A-D1275N channels showed a small depolarizing shift in activation compared with wild-type channels. Rare Cx40 genotype reporter gene analysis showed a reduction in reporter gene expression compared with the more common Cx40 genotype. In this study, familial AS was associated with the concurrence of a cardiac sodium channel mutation and rare polymorphisms in the atrial-specific Cx40 gene. We propose that, although the functional effect of each genetic change is relatively benign, the combined effect of genetic changes eventually progresses to total AS.


Subject(s)
Arrhythmias, Cardiac/genetics , Atrial Function/genetics , Connexins/genetics , Mutation , Sodium Channels/genetics , Adolescent , Adult , Aged , Amino Acid Substitution , Animals , Arrhythmias, Cardiac/diagnosis , Arrhythmias, Cardiac/physiopathology , DNA Mutational Analysis , Dizziness/etiology , Electrocardiography , Female , Genotype , Humans , Male , NAV1.5 Voltage-Gated Sodium Channel , Netherlands , Oocytes/metabolism , Patch-Clamp Techniques , Pedigree , Phenotype , Polymorphism, Genetic , Sodium Channels/metabolism , Syncope/etiology , Transfection , Xenopus laevis , Gap Junction alpha-5 Protein
18.
Circulation ; 101(16): 2007-14, 2000 Apr 25.
Article in English | MEDLINE | ID: mdl-10779469

ABSTRACT

BACKGROUND: Atrial fibrillation causes electrophysiological changes of the atrium, thereby facilitating its maintenance. Although the expression of ion channels is modulated in chronic atrial fibrillation, it is yet unknown whether paroxysmal atrial fibrillation can also lead to electrical remodeling by affecting gene expression. METHODS AND RESULTS: To examine the short-term effects of rapid pacing on the mRNA level of voltage-dependent K(+) channels, high-rate atrial pacing was performed in Sprague-Dawley rat hearts. Total RNA was prepared from the atrial appendages from 0 to 8 hours after the onset of pacing, and mRNA levels of Kv1.2, Kv1. 4, Kv1.5, Kv2.1, Kv4.2, Kv4.3, erg, KvLQT1, and minK were determined by RNase protection assay. Among these 9 genes, the mRNA level of the Kv1.5 channel immediately and transiently increased, with bimodal peaks at 0.5 and 2 hours after the onset of pacing. Conversely, the pacing gradually and progressively decreased the mRNA levels of the Kv4.2 and Kv4.3 channels. The increase of Kv1.5 and the decrease of Kv4.2 and Kv4.3 mRNA levels were both rate dependent. In correspondence with the changes in the mRNA level, Kv1. 5 channel protein transiently increased in the membrane fraction of the atrium during a 2- to 8-hour pacing period. Electrophysiological findings that the shortening of the action potential produced by 4-hour pacing was almost abolished by a low concentration of 4-aminopyridine implied that the increased Kv1.5 protein was functioning. CONCLUSIONS: Even short-term high-rate atrial excitation could differentially alter the mRNA levels of Kv1.5, Kv4.2, and Kv4.3 in a rate-dependent manner. In particular, increased Kv1.5 gene expression, having a transient nature, implied the possible biochemical electrical remodeling unique to paroxysmal tachycardia.


Subject(s)
Atrial Function/genetics , Myocardium/metabolism , Pacemaker, Artificial , Potassium Channels, Voltage-Gated , Potassium Channels/genetics , Tachycardia, Ectopic Atrial/physiopathology , Action Potentials/physiology , Acute Disease , Animals , Antisense Elements (Genetics) , Atrial Fibrillation/genetics , Atrial Fibrillation/physiopathology , Blotting, Western , Chronic Disease , DNA Primers , Delayed Rectifier Potassium Channels , ERG1 Potassium Channel , Electrophysiology , Ether-A-Go-Go Potassium Channels , Gene Expression/physiology , Heart Atria/chemistry , Heart Atria/metabolism , Heart Conduction System/physiology , Heart Rate/physiology , KCNQ Potassium Channels , KCNQ1 Potassium Channel , Kv1.2 Potassium Channel , Kv1.4 Potassium Channel , Kv1.5 Potassium Channel , Myocardium/chemistry , Potassium Channels/analysis , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Shab Potassium Channels , Shal Potassium Channels , Tachycardia, Ectopic Atrial/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...