Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
1.
Cell Death Dis ; 4: e843, 2013 Oct 10.
Article in English | MEDLINE | ID: mdl-24113177

ABSTRACT

Induced pluripotent stem cells (iPSC) offer a unique opportunity for developmental studies, disease modeling and regenerative medicine approaches in humans. The aim of our study was to create an in vitro 'patient-specific cell-based system' that could facilitate the screening of new therapeutic molecules for the treatment of catecholaminergic polymorphic ventricular tachycardia (CPVT), an inherited form of fatal arrhythmia. Here, we report the development of a cardiac model of CPVT through the generation of iPSC from a CPVT patient carrying a heterozygous mutation in the cardiac ryanodine receptor gene (RyR2) and their subsequent differentiation into cardiomyocytes (CMs). Whole-cell patch-clamp and intracellular electrical recordings of spontaneously beating cells revealed the presence of delayed afterdepolarizations (DADs) in CPVT-CMs, both in resting conditions and after ß-adrenergic stimulation, resembling the cardiac phenotype of the patients. Furthermore, treatment with KN-93 (2-[N-(2-hydroxyethyl)]-N-(4methoxybenzenesulfonyl)]amino-N-(4-chlorocinnamyl)-N-methylbenzylamine), an antiarrhythmic drug that inhibits Ca(2+)/calmodulin-dependent serine-threonine protein kinase II (CaMKII), drastically reduced the presence of DADs in CVPT-CMs, rescuing the arrhythmic phenotype induced by catecholaminergic stress. In addition, intracellular calcium transient measurements on 3D beating clusters by fast resolution optical mapping showed that CPVT clusters developed multiple calcium transients, whereas in the wild-type clusters, only single initiations were detected. Such instability is aggravated in the presence of isoproterenol and is attenuated by KN-93. As seen in our RyR2 knock-in CPVT mice, the antiarrhythmic effect of KN-93 is confirmed in these human iPSC-derived cardiac cells, supporting the role of this in vitro system for drug screening and optimization of clinical treatment strategies.


Subject(s)
Arrhythmias, Cardiac/drug therapy , Benzylamines/pharmacology , Benzylamines/therapeutic use , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Tachycardia, Ventricular/drug therapy , Adolescent , Adult , Animals , Arrhythmias, Cardiac/complications , Arrhythmias, Cardiac/enzymology , Arrhythmias, Cardiac/pathology , Base Sequence , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Differentiation/drug effects , Child , Child, Preschool , Female , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Male , Mice , Molecular Sequence Data , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Pedigree , Phenotype , Receptors, Adrenergic, beta/metabolism , Ryanodine Receptor Calcium Release Channel/genetics , Tachycardia, Ventricular/complications , Tachycardia, Ventricular/enzymology , Tachycardia, Ventricular/pathology
2.
Handb Exp Pharmacol ; (171): 267-86, 2006.
Article in English | MEDLINE | ID: mdl-16610348

ABSTRACT

The integration between molecular biology and clinical practice requires the achievement of fundamental steps to link basic science to diagnosis and management of patients. In the last decade, the study of genetic bases of human diseases has achieved several milestones, and it is now possible to apply the knowledge that stems from the identification of the genetic substrate of diseases to clinical practice. The first step along the process of linking molecular biology to clinical medicine is the identification of the genetic bases of inherited diseases. After this important goal is achieved, it becomes possible to extend research to understand the functional impairments of mutant protein(s) and to link them to clinical manifestations (genotype-phenotype correlation). In genetically heterogeneous diseases, it may be possible to identify locus-specific risk stratification and management algorithms. Finally, the most ambitious step in the study of genetic disease is to discover a novel pharmacological therapy targeted at correcting the inborn defect (locus-specific therapy) or even to "cure" the DNA abnormality by replacing the defective gene with gene therapy. At present, this curative goal has been successful only for very few diseases. In the field of inherited arrhythmogenic diseases, several genes have been discovered, and genetics is now emerging as a source of information contributing not only to a better diagnosis but also to risk stratification and management of patients. The functional characterization of mutant proteins has opened new perspectives about the possibility of performing gene-specific or mutation-specific therapy. In this chapter, we will briefly summarize the genetic bases of inherited arrhythmogenic conditions and we will point out how the information derived from molecular genetics has influenced the "optimal use of traditional therapies" and has paved the way to the development of gene-specific therapy.


Subject(s)
Arrhythmias, Cardiac/drug therapy , Arrhythmias, Cardiac/genetics , Long QT Syndrome/drug therapy , Animals , Defibrillators, Implantable , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels/genetics , Humans , Long QT Syndrome/genetics , Membrane Potentials/drug effects , Mutation , NAV1.5 Voltage-Gated Sodium Channel , Ryanodine Receptor Calcium Release Channel/drug effects , Ryanodine Receptor Calcium Release Channel/genetics , Sodium Channels/genetics , Tachycardia/drug therapy , Tachycardia/genetics
3.
Hong Kong Med J ; 10(1): 32-7, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14967853

ABSTRACT

OBJECTIVE: To study the clinical profile and genetic basis of Brugada syndrome in Chinese patients. DESIGN: Prospective observational study. SETTING: Seven regional public hospitals, Hong Kong. MAIN OUTCOME MEASURES: The clinical and follow-up data of 50 patients (47 men, 3 women; mean age, 53 years) were collected, and genetic data of 36 probands and eight family members of three genotyped probands were analysed. RESULTS: Eight patients survived sudden cardiac death (group A), 12 had syncope of unknown origin but no sudden death (group B), and 30 were asymptomatic before recognition of Brugada syndrome (group C). Programmed electrical stimulation induced sustained ventricular arrhythmias in 88% (7/8), 82% (9/11), and 27% (3/11) of patients in group A, group B, and group C, respectively. New arrhythmic events occurred in 50% (4/8) of patients in group A and 17% (2/12) of patients in group B after a mean follow-up period of 30 (standard deviation, 13) months and 25 (7) months, respectively. All group C patients remained asymptomatic during a mean follow-up period of 25 (standard deviation, 11) months. Five of 36 probands and three of eight family members who underwent genetic testing were found to have a mutation in their SCN5A gene. CONCLUSIONS: Chinese patients with Brugada syndrome who are symptomatic have a high likelihood of arrhythmia recurrence, whereas asymptomatic patients enjoy a good short-term prognosis. The prevalence of SCN5A mutation among probands is 14%. Thus, Chinese patients with Brugada syndrome share with their western counterparts similar clinical and genetic heterogeneity.


Subject(s)
Bundle-Branch Block/epidemiology , Bundle-Branch Block/genetics , Death, Sudden, Cardiac/epidemiology , Adult , Arrhythmias, Cardiac/epidemiology , Arrhythmias, Cardiac/genetics , Electrocardiography , Female , Hong Kong/epidemiology , Humans , Male , Middle Aged , Mutation , NAV1.5 Voltage-Gated Sodium Channel , Prospective Studies , Sex Distribution , Sodium Channels/genetics , Syncope/epidemiology , Syndrome
6.
Eur Heart J ; 23(12): 975-83, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12069453

ABSTRACT

AIMS: Differences in the sensitivity of the genotype of the congenital long QT syndrome to sympathetic stimulation have been suggested. This study compared the influence of sympathetic stimulation on continuous corrected QT (QTc) intervals between LQT1, LQT2 and LQT3 forms of the congenital long QT syndrome. METHODS AND RESULTS: We recorded a 12-lead electrocardiogram continuously before and after bolus injection (0.1 microg x kg(-1)) of epinephrine followed by continuous infusion (0.1 microg x kg(-1) min(-1)) in 12 LQT1, 10 LQT2, 6 LQT3, and 13 control patients. The QT intervals and previous RR intervals of all beats were measured semi-automatically, and the QTc intervals of all beats were calculated by Bazett's method. The dynamic response of the RR interval to epinephrine was no different between the four groups. The QTc was prolonged remarkably (477+/-42 to 631+/- 59 ms; P<0.0005, % delta prolongation =+32%) as the RR was maximally decreased (at peak of epinephrine), and remained prolonged at steady state conditions of epinephrine (556+/-56 ms; P<0.0005 vs baseline, +17%) in LQT1 patients. Epinephrine also prolonged the QTc dramatically (502+/-23 to 620+/-39 ms; P<0.0005, +24%) at peak of epinephrine in LQT2 patients, but this shortened to baseline levels at steady state (531+/-25 ms; P=ns vs baseline, +6%). The QTc was much less prolonged at peak of epinephrine in LQT3 (478+/-44 to 532+/-41 ms; P<0.05, +11%) and controls (394+/-21 to 456+/-18 ms; P<0.0005, +16%) than in LQT1 and LQT2 patients, and shortened to the baseline levels (LQT3; 466+/-49 ms, -3%, controls; 397+/-16 ms, +1%; P=ns vs baseline) at steady state. CONCLUSION: Our data suggest that the dynamic response of ventricular repolarization to sympathetic stimulation differs between LQT1, LQT2 and LQT3 syndromes, and may explain why the trigger of cardiac events differs between the genotypes.


Subject(s)
Long QT Syndrome/congenital , Long QT Syndrome/genetics , Adolescent , Adrenergic beta-Agonists/therapeutic use , Adult , Aged , Child , Child Welfare , Child, Preschool , Electrocardiography , Epinephrine/therapeutic use , Female , Gene Expression Regulation/drug effects , Heart Conduction System/drug effects , Humans , Infusions, Intravenous , Long QT Syndrome/drug therapy , Male , Middle Aged , Sensitivity and Specificity , Sympathetic Nervous System/drug effects , Treatment Outcome
7.
Europace ; 4(1): 3-18, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11858152

ABSTRACT

The European Society of Cardiology has convened a Task Force on Sudden Cardiac Death in order to provide a comprehensive, educational document on this important topic. The main document has been published in the European Heart Journal in August 2001. The Task Force has now summarized the most important clinical issues on sudden cardiac death and provided tables with recommendations for risk stratification and for prophylaxis of sudden cardiac death. The present recommendations are specifically intended to encourage the development and revision of national guidelines on prevention of sudden cardiac death. The common challenge for cardiologists, physicians of other medical specialties and health professionals throughout Europe is to realize the potential for sudden cardiac death prevention and to contribute to public health efforts to reduce its burden.


Subject(s)
Advisory Committees/standards , Death, Sudden, Cardiac/prevention & control , Practice Guidelines as Topic/standards , Societies, Medical/standards , Europe , Humans
8.
J Am Coll Cardiol ; 37(3): 911-9, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11693770

ABSTRACT

OBJECTIVES: The study compared the influence of sympathetic stimulation on transmural and spatial dispersion of repolarization between LQT1 and LQT2 forms of congenital long QT sYndrome (LQTS). BACKGROUND: Cardiac events are more associated with sympathetic stimulation in LQT1 than in LQT2 or LQT3 syndrome. Experimental studies have suggested that the interval between Tpeak and Tend (Tp-e) in the electrocardiogram (ECG) reflects transmural dispersion of repolarization across the ventricular wall. METHODS: We recorded 87-lead body-surface ECGs before and after epinephrine infusion (0.1 microg/kg/min) in 13 LQT1, 6 LQT2, and 7 control patients. The Q-Tend (QT-e), Q-Tpeak (QT-p), and Tp-e were measured automatically from 87-lead ECGs, corrected by Bazett's method (QTc-e, QTc-p, Tcp-e), and averaged among all 87-leads and among 24-leads, which reflect the potential from the left ventricular free wall. As an index of spatial dispersion of repolarization, the dispersion of QTc-e (QTc-eD) and QTc-p (QTc-pD) were obtained among 87-leads and among 24-leads, and were defined as the interval between the maximum and the minimum of the QTc-e and the QTc-p, respectively. RESULTS: Epinephrine significantly increased the mean QTc-e but not the mean QTc-p, resulting in a significant increase in the mean Tcp-e in both LQT1 and LQT2, but not in control patients. The epinephrine-induced increases in the mean QTc-e and Tcp-e were larger in LQT1 than in LQT2, and were more pronounced when the averaged data were obtained from 24-leads than from 87-leads. Epinephrine increased the maximum QTc-e but not the minimum QTc-e, producing a significant increase in the QTc-eD in both LQT1 and LQT2 patients, but not in control patients. The increase in the QTc-eD was larger in LQT1 than in LQT2 patients. CONCLUSIONS: Our data suggest that sympathetic stimulation produces a greater increase in both transmural and spatial dispersion of repolarization in LQT1 than in LQT2 syndrome, and this may explain why LQT1 patients are more sensitive to sympathetic stimulation.


Subject(s)
Epinephrine/pharmacology , Heart Conduction System/physiopathology , Long QT Syndrome/physiopathology , Sympathetic Nervous System/physiopathology , Sympathomimetics/pharmacology , Adult , Body Surface Potential Mapping , Electrocardiography , Female , Heart Conduction System/drug effects , Humans , Long QT Syndrome/congenital , Male , Middle Aged
9.
Lancet ; 358(9290): 1342-3, 2001 Oct 20.
Article in English | MEDLINE | ID: mdl-11684219

ABSTRACT

Although sudden infant death syndrome (SIDS) has been associated with long QT syndrome-a genetic disorder that causes arrhythmia-a causal link has not been shown. We screened genomic DNA from a child who died of SIDS and identified a de-novo mutation in KVLQT1, the gene most frequently associated with long QT syndrome. This mutation (C350T) had already been identified in an unrelated family that was affected by long QT syndrome. These results confirm the hypothesis that some deaths from SIDS are caused by long QT syndrome and support implementation of neonatal electrocardiographic screening.


Subject(s)
Long QT Syndrome/complications , Long QT Syndrome/genetics , Sudden Infant Death/etiology , Adolescent , Female , Humans , Infant , Italy , Male , Polymorphism, Genetic , Sudden Infant Death/diagnosis
12.
J Biol Chem ; 276(33): 30623-30, 2001 Aug 17.
Article in English | MEDLINE | ID: mdl-11410597

ABSTRACT

Defects of the SCN5A gene encoding the cardiac sodium channel alpha-subunit are associated with both the long QT-3 (LQT-3) subtype of long-QT syndrome and Brugada syndrome (BrS). One previously described SCN5A mutation (1795insD) in the C terminus results in a clinical phenotype combining QT prolongation and ST segment elevation, indicating a close interrelationship between the two disorders. Here we provide additional evidence that these two disorders are closely related. We report the analysis of two novel mutations on the same codon, Y1795C (LQT-3) and Y1795H (BrS), expressed in HEK 293 cells and characterized using whole-cell patch clamp procedures. We find marked and opposing effects on channel gating consistent with activity associated with the cellular basis of each clinical disorder. Y1795H speeds and Y1795C slows the onset of inactivation. The Y1795H, but not the Y1795C, mutation causes a marked negative shift in the voltage dependence of inactivation, and neither mutation affects the kinetics of the recovery from inactivation. Interestingly, both mutations increase the expression of sustained Na+ channel activity compared with wild type (WT) channels, although this effect is most pronounced for the Y1795C mutation, and both mutations promote entrance into an intermediate or a slowly developing inactivated state. These data confirm the key role of the C-terminal tail of the cardiac Na+ channel in the control of channel gating, illustrate how subtle changes in channel biophysics can have significant and distinct effects in human disease, and, additionally, provide further evidence of the close interrelationship between BrS and LQT-3 at the molecular level.


Subject(s)
Heart Block/genetics , Long QT Syndrome/genetics , Mutation , Sodium Channels/genetics , Sodium Channels/physiology , Humans , NAV1.5 Voltage-Gated Sodium Channel , Phenotype
13.
J Cardiovasc Electrophysiol ; 12(4): 455-61, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11332568

ABSTRACT

INTRODUCTION: Previous studies showed that diagnosing congenital long QT syndrome (LQTS) is difficult due to variable penetrance and genetic heterogeneity, especially when subjects from multiple families with diverse mutations are combined. We hypothesized that a combination of clinical and ECG techniques could identify gene carriers within a single family with congenital LQTS. METHODS AND RESULTS: One hundred one genotyped members of a family with LQTS, including 26 carriers of a HERG mutation, underwent history and ECG analysis. Forty-eight family members also underwent exercise testing with QT and T wave alternans (TWA) analysis and 24-hour Holter monitoring with QT and heart rate variability analysis. A logistic regression model, which included age, gender, QTc, and QTc by age, provided the best prediction of gene carrier status, although there was substantial overlap (78%) of QTc among subjects with and without the mutation. QTc was not helpful as a discriminator in children < or = 13 years. TWA (observed infrequently) did not add significantly to the model's ability to predict abnormal genotype. CONCLUSION: Even in this homogeneous LQTS population, the phenotype was so variable that clinical and detailed ECG analyses did not permit an accurate diagnosis of gene carrier status, especially in children. Sustained microvolt TWA was a specific (100%) but insensitive (18%) marker for LQTS. Its ability to predict risk of arrhythmia in this population remains to be determined. Genetic testing serves an essential role in screening for carriers of LQTS.


Subject(s)
Cation Transport Proteins , DNA-Binding Proteins , Electrocardiography/methods , Long QT Syndrome/congenital , Long QT Syndrome/genetics , Potassium Channels, Voltage-Gated , Trans-Activators , Adolescent , Adult , Aged , Child , ERG1 Potassium Channel , Electrocardiography, Ambulatory , Ether-A-Go-Go Potassium Channels , Exercise Test , Genetic Carrier Screening/methods , Genotype , Heart Rate , Humans , Middle Aged , Mutation , Phenotype , Potassium Channels/genetics , Prognosis , Sensitivity and Specificity , Transcriptional Regulator ERG
14.
Cardiovasc Res ; 50(2): 218-23, 2001 May.
Article in English | MEDLINE | ID: mdl-11334825

ABSTRACT

In approximately 6--10% of survivors of cardiac arrest no cardiac abnormality can be identified despite extensive clinical evaluation. Autopsy data confirm that in a similar percentage of victims of sudden death no structural heart disease can be identified at post mortem evaluation. Occurrence of cardiac arrest in the absence of a substrate is defined 'idiopathic ventricular fibrillation' thus admitting that the cause for the arrhythmic event has remained unknown. We present data supporting the hypothesis that incompletely penetrant genetic defects may underlie at least some of these unexplained deaths.


Subject(s)
Death, Sudden, Cardiac/etiology , Ventricular Fibrillation/genetics , Death, Sudden, Cardiac/pathology , Electrocardiography , Humans , Long QT Syndrome/complications , Pedigree
15.
Ital Heart J Suppl ; 2(3): 253-7, 2001 Mar.
Article in Italian | MEDLINE | ID: mdl-11307783

ABSTRACT

In clinical cardiology, resort has recently been made to molecular genetics in order to explain some mechanisms that underlie sudden cardiac death in young people with structurally normal hearts. It has become evident that genetic mutations regarding cardiac ion channels may disrupt the delicate balance of currents in the action potential, thus inducing malignant ventricular tachyarrhythmias. The cardiac sodium channel gene, SCN5A, is involved in two of such arrhythmogenic diseases, the Brugada syndrome and one form of the long QT syndrome (LQT3). It is believed that these syndromes result from opposite molecular effects: Brugada syndrome mutations cause a reduced sodium current, while LQT3 mutations are associated with a gain of function. The effects of class I antiarrhythmic drugs have been used to differentiate these diseases. Intravenous flecainide is used as a highly specific test to unmask the electrocardiographic phenotype of the Brugada syndrome. On the other hand, on the basis of experimental and clinical studies, the possibility that the same drugs act as a gene-specific therapy in this disorder by contrasting the effect of mutations in LQT3 has been explored. Recent evidence shows that phenotypic overlap may exist between the Brugada syndrome and LQT3. One large family with a SCN5A mutation and a "mixed" electrocardiographic pattern (prolonged QT interval and ST-segment elevation) has been reported. Moreover, our recent data showed that flecainide challenge may elicit ST-segment elevation in some LQT3 patients. The presence of "intermediate" phenotypes highlights a remarkable heterogeneity suggesting that clinical features may depend upon the single mutation. Only deepened understanding of the genotype-phenotype correlation will allow the definition of the individual patient's risk and the development of guidelines for clinical management.


Subject(s)
Death, Sudden, Cardiac , Long QT Syndrome/diagnosis , Syncope/diagnosis , Anti-Arrhythmia Agents/therapeutic use , Diagnosis, Differential , Humans , Long QT Syndrome/drug therapy , Syndrome
16.
Circ Res ; 88(7): 740-5, 2001 Apr 13.
Article in English | MEDLINE | ID: mdl-11304498

ABSTRACT

Variant 3 of the congenital long-QT syndrome (LQTS-3) is caused by mutations in the gene encoding the alpha subunit of the cardiac Na(+) channel. In the present study, we report a novel LQTS-3 mutation, E1295K (EK), and describe its functional consequences when expressed in HEK293 cells. The clinical phenotype of the proband indicated QT interval prolongation in the absence of T-wave morphological abnormalities and a steep QT/R-R relationship, consistent with an LQTS-3 lesion. However, biophysical analysis of mutant channels indicates that the EK mutation changes channel activity in a manner that is distinct from previously investigated LQTS-3 mutations. The EK mutation causes significant positive shifts in the half-maximal voltage (V(1/2)) of steady-state inactivation and activation (+5.2 and +3.4 mV, respectively). These gating changes shift the window of voltages over which Na(+) channels do not completely inactivate without altering the magnitude of these currents. The change in voltage dependence of window currents suggests that this alteration in the voltage dependence of Na(+) channel gating may cause marked changes in action potential duration because of the unique voltage-dependent rectifying properties of cardiac K(+) channels that underlie the plateau and terminal repolarization phases of the action potential. Na(+) channel window current is likely to have a greater effect on net membrane current at more positive potentials (EK channels) where total K(+) channel conductance is low than at more negative potentials (wild-type channels), where total K(+) channel conductance is high. These findings suggest a fundamentally distinct mechanism of arrhythmogenesis for congenital LQTS-3.


Subject(s)
Arrhythmias, Cardiac/diagnosis , Heart/physiopathology , Long QT Syndrome/diagnosis , Long QT Syndrome/genetics , Sodium Channels/genetics , Adolescent , Amino Acid Substitution , Arrhythmias, Cardiac/genetics , Cell Line , Conserved Sequence , DNA Mutational Analysis , Electrocardiography , Humans , Ion Channel Gating/drug effects , Ion Channel Gating/genetics , Kidney/cytology , Kidney/drug effects , Kidney/metabolism , Long QT Syndrome/physiopathology , Male , Mutation , NAV1.5 Voltage-Gated Sodium Channel , Patch-Clamp Techniques , Phenotype , Sodium/metabolism , Sodium Channels/metabolism , Tetrodotoxin/pharmacology , Transfection
18.
Circulation ; 103(2): 196-200, 2001 Jan 16.
Article in English | MEDLINE | ID: mdl-11208676

ABSTRACT

BACKGROUND: Catecholaminergic polymorphic ventricular tachycardia is a genetic arrhythmogenic disorder characterized by stress-induced, bidirectional ventricular tachycardia that may degenerate into cardiac arrest and cause sudden death. The electrocardiographic pattern of this ventricular tachycardia closely resembles the arrhythmias associated with calcium overload and the delayed afterdepolarizations observed during digitalis toxicity. We speculated that a genetically determined abnormality of intracellular calcium handling might be the substrate of the disease; therefore, we considered the human cardiac ryanodine receptor gene (hRyR2) a likely candidate for this genetically transmitted arrhythmic disorder. METHODS AND RESULTS: Twelve patients presenting with typical catecholaminergic polymorphic ventricular tachycardia in the absence of structural heart abnormalities were identified. DNA was extracted from peripheral blood lymphocytes, and single-strand conformation polymorphism analysis was performed on polymerase chain reaction-amplified exons of the hRyR2 gene. Four single nucleotide substitutions leading to missense mutations were identified in 4 probands affected by the disease. Genetic analysis of the asymptomatic parents revealed that 3 probands carried de novo mutations. In 1 case, the identical twin of the proband died suddenly after having suffered syncopal episodes. The fourth mutation was identified in the proband, in 4 clinically affected family members, and in none of 3 nonaffected family members in a kindred with 2 sudden deaths that occurred at 16 and 14 years, respectively, in the sisters of the proband. CONCLUSIONS: We demonstrated that, in agreement with our hypothesis, hRyR2 is a gene responsible for catecholaminergic polymorphic ventricular tachycardia.


Subject(s)
Mutation, Missense , Ryanodine Receptor Calcium Release Channel/genetics , Tachycardia, Ventricular/genetics , Adolescent , Adult , Base Sequence , Catecholamines , Child , Female , Humans , Male , Pedigree , Phenotype , Polymorphism, Single Nucleotide
19.
Circulation ; 103(1): 89-95, 2001 Jan 02.
Article in English | MEDLINE | ID: mdl-11136691

ABSTRACT

BACKGROUND: The congenital long-QT syndrome (LQTS) is caused by mutations on several genes, all of which encode cardiac ion channels. The progressive understanding of the electrophysiological consequences of these mutations opens unforeseen possibilities for genotype-phenotype correlation studies. Preliminary observations suggested that the conditions ("triggers") associated with cardiac events may in large part be gene specific. METHODS AND RESULTS: We identified 670 LQTS patients of known genotype (LQT1, n=371; LQT2, n=234; LQT3, n=65) who had symptoms (syncope, cardiac arrest, sudden death) and examined whether 3 specific triggers (exercise, emotion, and sleep/rest without arousal) differed according to genotype. LQT1 patients experienced the majority of their events (62%) during exercise, and only 3% occurred during rest/sleep. These percentages were almost reversed among LQT2 and LQT3 patients, who were less likely to have events during exercise (13%) and more likely to have events during rest/sleep (29% and 39%). Lethal and nonlethal events followed the same pattern. Corrected QT interval did not differ among LQT1, LQT2, and LQT3 patients (498, 497, and 506 ms, respectively). The percent of patients who were free of recurrence with ss-blocker therapy was higher and the death rate was lower among LQT1 patients (81% and 4%, respectively) than among LQT2 (59% and 4%, respectively) and LQT3 (50% and 17%, respectively) patients. CONCLUSIONS: Life-threatening arrhythmias in LQTS patients tend to occur under specific circumstances in a gene-specific manner. These data allow new insights into the mechanisms that relate the electrophysiological consequences of mutations on specific genes to clinical manifestations and offer the possibility of complementing traditional therapy with gene-specific approaches.


Subject(s)
Long QT Syndrome/genetics , Adrenergic beta-Antagonists/therapeutic use , Adult , Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/physiopathology , Death, Sudden, Cardiac/etiology , Electrocardiography , Emotions , Exercise , Female , Genotype , Humans , Ion Channels/genetics , Long QT Syndrome/classification , Long QT Syndrome/diagnosis , Long QT Syndrome/drug therapy , Long QT Syndrome/physiopathology , Male , Phenotype , Sex Factors , Sleep , Survival Rate , Syncope/etiology
SELECTION OF CITATIONS
SEARCH DETAIL
...