Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.346
Filter
1.
PLoS One ; 19(7): e0305248, 2024.
Article in English | MEDLINE | ID: mdl-38968219

ABSTRACT

Long QT Syndrome type 8 (LQT8) is a cardiac arrhythmic disorder associated with Timothy Syndrome, stemming from mutations in the CACNA1C gene, particularly the G406R mutation. While prior studies hint at CACNA1C mutations' role in ventricular arrhythmia genesis, the mechanisms, especially in G406R presence, are not fully understood. This computational study explores how the G406R mutation, causing increased transmural dispersion of repolarization, induces and sustains reentrant ventricular arrhythmias. Using three-dimensional numerical simulations on an idealized left-ventricular model, integrating the Bidomain equations with the ten Tusscher-Panfilov ionic model, we observe that G406R mutation with 11% and 50% heterozygosis significantly increases transmural dispersion of repolarization. During S1-S4 stimulation protocols, these gradients facilitate conduction blocks, triggering reentrant ventricular tachycardia. Sustained reentry pathways occur only with G406R mutation at 50% heterozygosis, while neglecting transmural heterogeneities of action potential duration prevents stable reentry, regardless of G406R mutation presence.


Subject(s)
Action Potentials , Calcium Channels, L-Type , Computer Simulation , Long QT Syndrome , Syndactyly , Humans , Long QT Syndrome/genetics , Long QT Syndrome/physiopathology , Calcium Channels, L-Type/genetics , Syndactyly/genetics , Syndactyly/physiopathology , Mutation , Autistic Disorder/genetics , Autistic Disorder/physiopathology , Heart Ventricles/physiopathology , Models, Cardiovascular , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/physiopathology , Tachycardia, Ventricular/genetics , Tachycardia, Ventricular/physiopathology
2.
Oncol Nurs Forum ; 51(4): 332-348, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38950091

ABSTRACT

OBJECTIVES: To evaluate for associations between the occurrence of palpitations reported by women prior to breast cancer surgery and single nucleotide polymorphisms (SNPs) for neurotransmitter genes. SAMPLE & SETTING: A total of 398 women, who were scheduled for unilateral breast cancer surgery, provided detailed information on demographic and clinical characteristics and the occurrence of palpitations prior to breast cancer surgery. METHODS & VARIABLES: The occurrence of palpitations was assessed using a single item (i.e., "heart races/pounds" in the past week ["yes"/"no"]). Blood samples were collected for genomic analyses. Multiple logistic regression analyses were used to identify associations between the occurrence of palpitations and variations in neurotransmitter genes. RESULTS: Nine SNPs and two haplotypes among 11 candidate genes were associated with the occurrence of palpitations. These genes encode for a number of neurotransmitters and/or their receptors, including serotonin, norepinephrine, dopamine, gamma-amino butyric acid, Substance P, and neurokinin. IMPLICATIONS FOR NURSING: These findings suggest that alterations in a variety of neurotransmitters contribute to the development of this symptom.


Subject(s)
Breast Neoplasms , Neurotransmitter Agents , Polymorphism, Single Nucleotide , Humans , Female , Breast Neoplasms/genetics , Middle Aged , Adult , Aged , Arrhythmias, Cardiac/genetics
3.
Mol Med ; 30(1): 97, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38937697

ABSTRACT

BACKGROUND: Ubiquitin-specific protease 38 (USP38), belonging to the USP family, is recognized for its role in controlling protein degradation and diverse biological processes. Ventricular arrhythmias (VAs) following heart failure (HF) are closely linked to ventricular electrical remodeling, yet the specific mechanisms underlying VAs in HF remain inadequately explored. In this study, we examined the impact of USP38 on VAs in pressure overload-induced HF. METHODS: Cardiac-specific USP38 knockout mice, cardiac-specific USP38 transgenic mice and their matched control littermates developed HF induced by aortic banding (AB) surgery. After subjecting the mice to AB surgery for a duration of four weeks, comprehensive investigations were conducted, including pathological analysis and electrophysiological assessments, along with molecular analyses. RESULTS: We observed increased USP38 expression in the left ventricle of mice with HF. Electrocardiogram showed that the USP38 knockout shortened the QRS interval and QTc, while USP38 overexpression prolonged these parameters. USP38 knockout decreased the susceptibility of VAs by shortening action potential duration (APD) and prolonging effective refractory period (ERP). In addition, USP38 knockout increased ion channel and Cx43 expression in ventricle. On the contrary, the increased susceptibility of VAs and the decreased expression of ventricular ion channels and Cx43 were observed with USP38 overexpression. In both in vivo and in vitro experiments, USP38 knockout inhibited TBK1/AKT/CAMKII signaling, whereas USP38 overexpression activated this pathway. CONCLUSION: Our data indicates that USP38 increases susceptibility to VAs after HF through TBK1/AKT/CAMKII signaling pathway, Consequently, USP38 may emerge as a promising therapeutic target for managing VAs following HF.


Subject(s)
Heart Failure , Mice, Knockout , Ubiquitin-Specific Proteases , Ventricular Remodeling , Animals , Mice , Ventricular Remodeling/genetics , Heart Failure/metabolism , Heart Failure/etiology , Heart Failure/genetics , Heart Failure/physiopathology , Ubiquitin-Specific Proteases/metabolism , Ubiquitin-Specific Proteases/genetics , Disease Models, Animal , Male , Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/genetics , Heart Ventricles/metabolism , Heart Ventricles/physiopathology , Mice, Transgenic , Signal Transduction , Electrocardiography
4.
Eur J Psychotraumatol ; 15(1): 2366055, 2024.
Article in English | MEDLINE | ID: mdl-38912597

ABSTRACT

Background: Emerging evidence has linked childhood maltreatment with cardiovascular disease risk; however, the association between childhood maltreatment and cardiac arrhythmias remains unclear. Moreover, any genetic predispositions to atrial fibrillation (AF), a common cardiac arrhythmia associated with an elevated risk of stroke, heart failure, and mortality, that modify such associations have been undocumented.Purpose: To examine the associations between childhood maltreatment and incident arrhythmias, and whether a genetic predisposition to arrhythmias modifies these associations.Methods: This prospective analysis included 151,741 participants from the UK Biobank (mean age 55.8 years, 43.4% male). Childhood maltreatment, including five types, was measured using the Childhood Trauma Screener (CTS). Incident arrhythmias (AF, ventricular arrhythmias [VA], and bradyarrhythmia [BA]) were documented through linked hospital admission and death registry. Weighted AF genetic risk score was calculated. Cox proportional hazard models were conducted to test for associations between childhood maltreatment and incident arrhythmias.Results: During a median follow-up of 12.21 years (interquartile range, 11.49-12.90 years), 6,588 AF, 2,093 BA, and 742 VA events occurred. Compared with the absence of childhood maltreatment, having 3-5 types of childhood maltreatment was associated with an increased risk of incident AF (HR, 1.23; 95%CI 1.09-1.37), VA (HR, 1.39; 95%CI 1.03-1.89), and BA (HR, 1.32; 95%CI 1.09-1.61) after adjusting demographic, socioeconomic and lifestyle factors. The associations between cumulative type of childhood maltreatment and the risk of AF (Poverall < .001; Pnonlinear = .674) and BA (Poverall = .007; Pnonlinear = .377) demonstrated a linear pattern. There was a gradient association between childhood maltreatment and AF risks across the intermediate and high genetic risk groups (both Ptrend < .05) but not within the low genetic risk group (Ptrend = .378), irrespective of non-significant interaction effect (Pinteraction = .204).Conclusion: Childhood maltreatment was associated with higher risks of incident arrhythmias, especially AF and BA. Genetic risk of AF did not modify these associations.


Previous studies indicate that childhood maltreatment is associated with cardiovascular disease risk.Childhood maltreatment was associated with an increased risk of incident arrhythmias, particularly atrial fibrillation and bradyarrhythmia. Genetic predisposition to atrial fibrillation did not significantly modify these associations.Childhood maltreatment could be a new psychological risk factor for cardiac arrhythmias in later life. Inquiries into childhood maltreatment and subsequent referral to psychological services may be helpful.


Subject(s)
Arrhythmias, Cardiac , Humans , Male , Female , Prospective Studies , Arrhythmias, Cardiac/epidemiology , Arrhythmias, Cardiac/genetics , Middle Aged , United Kingdom/epidemiology , Risk Factors , Genetic Predisposition to Disease , Adult , Cohort Studies , Adult Survivors of Child Abuse/statistics & numerical data , Child Abuse/statistics & numerical data
5.
Int J Mol Sci ; 25(11)2024 May 27.
Article in English | MEDLINE | ID: mdl-38892025

ABSTRACT

Pathogenic variants in LMNA have been associated with a wide spectrum of muscular conditions: the laminopathies. LMNA-related congenital muscular dystrophy is a laminopathy characterised by the early onset of symptoms and often leads to a fatal outcome at young ages. Children face a heightened risk of malignant arrhythmias. No established paediatric protocols for managing this condition are available. We review published cases and provide insights into disease progression in two twin sisters with LMNA-related muscular dystrophy. Our objective is to propose a cardiac surveillance and management plan tailored specifically for paediatric patients. We present a family of five members, including two twin sisters with LMNA-related muscular dystrophy. A comprehensive neuromuscular and cardiac work-up was performed in all family members. Genetic analysis using massive sequencing technology was performed in both twins. Clinical assessment showed that only the twins showed diagnoses of LMNA-related muscular dystrophy. Follow-up showed an early onset of symptoms and life-threatening arrhythmias, with differing disease progressions despite both twins passing away. Genetic analysis identified a de novo rare missense deleterious variant in the LMNA gene. Other additional rare variants were identified in genes associated with myasthenic syndrome. Early-onset neuromuscular symptoms could be related to a prognosis of worse life-threatening arrhythmias in LMNA related muscular dystrophy. Being a carrier of other rare variants may be a modifying factor in the progression of the phenotype, although further studies are needed. There is a pressing need for specific cardiac recommendations tailored to the paediatric population to mitigate the risk of malignant arrhythmias.


Subject(s)
Lamin Type A , Muscular Dystrophies , Twins, Monozygotic , Humans , Lamin Type A/genetics , Twins, Monozygotic/genetics , Female , Muscular Dystrophies/genetics , Muscular Dystrophies/therapy , Male , Child , Pedigree , Child, Preschool , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/etiology
6.
Europace ; 26(6)2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38875491

ABSTRACT

AIMS: Patients with mutations in SCN5A encoding NaV1.5 often display variable severity of electrical and structural alterations, but the underlying mechanisms are not fully elucidated. We here investigate the combined modulatory effect of genetic background and age on disease severity in the Scn5a1798insD/+ mouse model. METHODS AND RESULTS: In vivo electrocardiogram and echocardiograms, ex vivo electrical and optical mapping, and histological analyses were performed in adult (2-7 months) and aged (8-28 months) wild-type (WT) and Scn5a1798insD/+ (mutant, MUT) mice from the FVB/N and 129P2 inbred strains. Atrio-ventricular (AV) conduction, ventricular conduction, and ventricular repolarization are modulated by strain, genotype, and age. An aging effect was present in MUT mice, with aged MUT mice of both strains showing prolonged QRS interval and right ventricular (RV) conduction slowing. 129P2-MUT mice were severely affected, with adult and aged 129P2-MUT mice displaying AV and ventricular conduction slowing, prolonged repolarization, and spontaneous arrhythmias. In addition, the 129P2 strain appeared particularly susceptible to age-dependent electrical, functional, and structural alterations including RV conduction slowing, reduced left ventricular (LV) ejection fraction, RV dilatation, and myocardial fibrosis as compared to FVB/N mice. Overall, aged 129P2-MUT mice displayed the most severe conduction defects, RV dilatation, and myocardial fibrosis, in addition to the highest frequency of spontaneous arrhythmia and inducible arrhythmias. CONCLUSION: Genetic background and age both modulate disease severity in Scn5a1798insD/+ mice and hence may explain, at least in part, the variable disease expressivity observed in patients with SCN5A mutations. Age- and genetic background-dependent development of cardiac structural alterations furthermore impacts arrhythmia risk. Our findings therefore emphasize the importance of continued assessment of cardiac structure and function in patients carrying SCN5A mutations.


Subject(s)
Arrhythmias, Cardiac , Disease Models, Animal , Fibrosis , Genetic Predisposition to Disease , Mutation , NAV1.5 Voltage-Gated Sodium Channel , Animals , NAV1.5 Voltage-Gated Sodium Channel/genetics , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/physiopathology , Age Factors , Severity of Illness Index , Heart Conduction System/physiopathology , Action Potentials , Electrocardiography , Phenotype , Genetic Background , Mice, 129 Strain , Male , Heart Rate/genetics , Myocardium/pathology , Aging/genetics
7.
Adv Exp Med Biol ; 1441: 1033-1055, 2024.
Article in English | MEDLINE | ID: mdl-38884768

ABSTRACT

Inherited forms of cardiac arrhythmias mostly are rare diseases (prevalence <1:2000) and considered to be either "primary electrical heart disorders" due to the absence of structural heart abnormalities or "cardiac ion channel disorders" due to the myocellular structures involved. Precise knowledge of the electrocardiographic features of these diseases and their genetic classification will enable early disease recognition and prevention of cardiac events including sudden cardiac death.The genetic background of these diseases is complex and heterogeneous. In addition to the predominant "private character" of a mutation in each family, locus heterogeneity involving many ion channel genes for the same familial arrhythmia syndrome is typical. Founder pathogenic variants or mutational hot spots are uncommon. Moreover, phenotypes may vary and overlap even within the same family and mutation carriers. For the majority of arrhythmias, the clinical phenotype of an ion channel mutation is restricted to cardiac tissue, and therefore, the disease is nonsyndromic.Recent and innovative methods of parallel DNA analysis (so-called next-generation sequencing, NGS) will enhance further mutation and other variant detection as well as arrhythmia gene identification.


Subject(s)
Arrhythmias, Cardiac , Genetic Predisposition to Disease , Mutation , Humans , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/physiopathology , Genetic Predisposition to Disease/genetics , Ion Channels/genetics , Phenotype , Electrocardiography
8.
JCI Insight ; 9(11)2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38855866

ABSTRACT

TANGO2-deficiency disorder (TDD) is an autosomal-recessive genetic disease caused by biallelic loss-of-function variants in the TANGO2 gene. TDD-associated cardiac arrhythmias are recalcitrant to standard antiarrhythmic medications and constitute the leading cause of death. Disease modeling for TDD has been primarily carried out using human dermal fibroblast and, more recently, in Drosophila by multiple research groups. No human cardiomyocyte system has been reported, which greatly hinders the investigation and understanding of TDD-associated arrhythmias. Here, we established potentially novel patient-derived induced pluripotent stem cell differentiated cardiomyocyte (iPSC-CM) models that recapitulate key electrophysiological abnormalities in TDD. These electrophysiological abnormalities were rescued in iPSC-CMs with either adenoviral expression of WT-TANGO2 or correction of the pathogenic variant using CRISPR editing. Our natural history study in patients with TDD suggests that the intake of multivitamin/B complex greatly diminished the risk of cardiac crises in patients with TDD. In agreement with the clinical findings, we demonstrated that high-dose folate (vitamin B9) virtually abolishes arrhythmias in TDD iPSC-CMs and that folate's effect was blocked by the dihydrofolate reductase inhibitor methotrexate, supporting the need for intracellular folate to mediate antiarrhythmic effects. In summary, data from TDD iPSC-CM models together with clinical observations support the use of B vitamins to mitigate cardiac crises in patients with TDD, providing potentially life-saving treatment strategies during life-threatening events.


Subject(s)
Arrhythmias, Cardiac , Folic Acid , Induced Pluripotent Stem Cells , Myocytes, Cardiac , Humans , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Folic Acid/metabolism , Folic Acid/therapeutic use , Arrhythmias, Cardiac/drug therapy , Arrhythmias, Cardiac/genetics , Male , Female , Child
9.
Adv Exp Med Biol ; 1441: 1057-1090, 2024.
Article in English | MEDLINE | ID: mdl-38884769

ABSTRACT

Arrhythmias account for over 300,000 annual deaths in the United States, and approximately half of all deaths are associated with heart disease. Mechanisms underlying arrhythmia risk are complex; however, work in humans and animal models over the past 25 years has identified a host of molecular pathways linked with both arrhythmia substrates and triggers. This chapter will focus on select arrhythmia pathways solved by linking human clinical and genetic data with animal models.


Subject(s)
Arrhythmias, Cardiac , Disease Models, Animal , Animals , Humans , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/physiopathology , Arrhythmias, Cardiac/metabolism , Signal Transduction/genetics
10.
Int J Mol Sci ; 25(11)2024 Jun 02.
Article in English | MEDLINE | ID: mdl-38892333

ABSTRACT

The sodium channel NaV1.8, encoded by the SCN10A gene, has recently emerged as a potential regulator of cardiac electrophysiology. We have previously shown that NaV1.8 contributes to arrhythmogenesis by inducing a persistent Na+ current (late Na+ current, INaL) in human atrial and ventricular cardiomyocytes (CM). We now aim to further investigate the contribution of NaV1.8 to human ventricular arrhythmogenesis at the CM-specific level using pharmacological inhibition as well as a genetic knockout (KO) of SCN10A in induced pluripotent stem cell CM (iPSC-CM). In functional voltage-clamp experiments, we demonstrate that INaL was significantly reduced in ventricular SCN10A-KO iPSC-CM and in control CM after a specific pharmacological inhibition of NaV1.8. In contrast, we did not find any effects on ventricular APD90. The frequency of spontaneous sarcoplasmic reticulum Ca2+ sparks and waves were reduced in SCN10A-KO iPSC-CM and control cells following the pharmacological inhibition of NaV1.8. We further analyzed potential triggers of arrhythmias and found reduced delayed afterdepolarizations (DAD) in SCN10A-KO iPSC-CM and after the specific inhibition of NaV1.8 in control cells. In conclusion, we show that NaV1.8-induced INaL primarily impacts arrhythmogenesis at a subcellular level, with minimal effects on systolic cellular Ca2+ release. The inhibition or knockout of NaV1.8 diminishes proarrhythmic triggers in ventricular CM. In conjunction with our previously published results, this work confirms NaV1.8 as a proarrhythmic target that may be useful in an anti-arrhythmic therapeutic strategy.


Subject(s)
Arrhythmias, Cardiac , Heart Ventricles , Induced Pluripotent Stem Cells , Myocytes, Cardiac , NAV1.8 Voltage-Gated Sodium Channel , NAV1.8 Voltage-Gated Sodium Channel/metabolism , NAV1.8 Voltage-Gated Sodium Channel/genetics , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Heart Ventricles/metabolism , Heart Ventricles/cytology , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/etiology , Action Potentials/drug effects
11.
Genome Med ; 16(1): 73, 2024 05 30.
Article in English | MEDLINE | ID: mdl-38816749

ABSTRACT

BACKGROUND: KCNE1 encodes a 129-residue cardiac potassium channel (IKs) subunit. KCNE1 variants are associated with long QT syndrome and atrial fibrillation. However, most variants have insufficient evidence of clinical consequences and thus limited clinical utility. METHODS: In this study, we leveraged the power of variant effect mapping, which couples saturation mutagenesis with high-throughput sequencing, to ascertain the function of thousands of protein-coding KCNE1 variants. RESULTS: We comprehensively assayed KCNE1 variant cell surface expression (2554/2709 possible single-amino-acid variants) and function (2534 variants). Our study identified 470 loss- or partial loss-of-surface expression and 574 loss- or partial loss-of-function variants. Of the 574 loss- or partial loss-of-function variants, 152 (26.5%) had reduced cell surface expression, indicating that most functionally deleterious variants affect channel gating. Nonsense variants at residues 56-104 generally had WT-like trafficking scores but decreased functional scores, indicating that the latter half of the protein is dispensable for protein trafficking but essential for channel function. 22 of the 30 KCNE1 residues (73%) highly intolerant of variation (with > 70% loss-of-function variants) were in predicted close contact with binding partners KCNQ1 or calmodulin. Our functional assay data were consistent with gold standard electrophysiological data (ρ = - 0.64), population and patient cohorts (32/38 presumed benign or pathogenic variants with consistent scores), and computational predictors (ρ = - 0.62). Our data provide moderate-strength evidence for the American College of Medical Genetics/Association of Molecular Pathology functional criteria for benign and pathogenic variants. CONCLUSIONS: Comprehensive variant effect maps of KCNE1 can both provide insight into I Ks channel biology and help reclassify variants of uncertain significance.


Subject(s)
Calmodulin , Potassium Channels, Voltage-Gated , Potassium Channels, Voltage-Gated/genetics , Potassium Channels, Voltage-Gated/metabolism , Humans , Calmodulin/genetics , Calmodulin/metabolism , Arrhythmias, Cardiac/genetics , High-Throughput Nucleotide Sequencing , Genetic Variation , Protein Transport , HEK293 Cells
12.
J Am Heart Assoc ; 13(10): e030467, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38761081

ABSTRACT

BACKGROUND: Many cardiomyopathy-associated FLNC pathogenic variants are heterozygous truncations, and FLNC pathogenic variants are associated with arrhythmias. Arrhythmia triggers in filaminopathy are incompletely understood. METHODS AND RESULTS: We describe an individual with biallelic FLNC pathogenic variants, p.Arg650X and c.970-4A>G, with peripartum cardiomyopathy and ventricular arrhythmias. We also describe clinical findings in probands with FLNC variants including Val2715fs87X, Glu2458Serfs71X, Phe106Leu, and c.970-4A>G with hypertrophic and dilated cardiomyopathy, atrial fibrillation, and ventricular tachycardia. Induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) were generated. The FLNC truncation, Arg650X/c.970-4A>G, showed a marked reduction in filamin C protein consistent with biallelic loss of function mutations. To assess loss of filamin C, gene editing of a healthy control iPSC line was used to generate a homozygous FLNC disruption in the actin binding domain. Because filamin C has been linked to protein quality control, we assessed the necessity of filamin C in iPSC-CMs for response to the proteasome inhibitor bortezomib. After exposure to low-dose bortezomib, FLNC-null iPSC-CMs showed an increase in the chaperone proteins BAG3, HSP70 (heat shock protein 70), and HSPB8 (small heat shock protein B8) and in the autophagy marker LC3I/II. FLNC null iPSC-CMs had prolonged electric field potential, which was further prolonged in the presence of low-dose bortezomib. FLNC null engineered heart tissues had impaired function after low-dose bortezomib. CONCLUSIONS: FLNC pathogenic variants associate with a predisposition to arrhythmias, which can be modeled in iPSC-CMs. Reduction of filamin C prolonged field potential, a surrogate for action potential, and with bortezomib-induced proteasome inhibition, reduced filamin C led to greater arrhythmia potential and impaired function.


Subject(s)
Filamins , Proteostasis , Filamins/genetics , Filamins/metabolism , Humans , Female , Induced Pluripotent Stem Cells/metabolism , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/physiopathology , Arrhythmias, Cardiac/etiology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Cardiomyopathies/physiopathology , Male , Adult , Mutation , Bortezomib/pharmacology
14.
Biochem Biophys Res Commun ; 720: 150105, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-38754163

ABSTRACT

BACKGROUND: Dexmedetomidine (DEX), a highly selective α2-adrenoceptor agonist, can decrease the incidence of arrhythmias, such as catecholaminergic polymorphic ventricular tachycardia (CPVT). However, the underlying mechanisms by which DEX affects cardiac electrophysiological function remain unclear. METHODS: Ryanodine receptor (RyR2) heterozygous R2474S mice were used as a model for CPVT. WT and RyR2R2474S/+ mice were treated with isoproterenol (ISO) and DEX, and electrocardiograms were continuously monitored during both in vivo and ex vivo experiments. Dual-dye optical mapping was used to explore the anti-arrhythmic mechanism of DEX. RESULTS: DEX significantly reduced the occurrence and duration of ISO-induced of VT/VF in RyR2R2474S/+ mice in vivo and ex vivo. DEX remarkably prolonged action potential duration (APD80) and calcium transient duration (CaTD80) in both RyR2R2474S/+ and WT hearts, whereas it reduced APD heterogeneity and CaT alternans in RyR2R2474S/+ hearts. DEX inhibited ectopy and reentry formation, and stabilized voltage-calcium latency. CONCLUSION: DEX exhibited an antiarrhythmic effect through stabilizing membrane voltage and intracellular Ca2+. DEX can be used as a beneficial perioperative anesthetic for patients with CPVT or other tachy-arrhythmias.


Subject(s)
Arrhythmias, Cardiac , Calcium , Dexmedetomidine , Ryanodine Receptor Calcium Release Channel , Animals , Dexmedetomidine/pharmacology , Ryanodine Receptor Calcium Release Channel/metabolism , Ryanodine Receptor Calcium Release Channel/genetics , Calcium/metabolism , Mice , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/drug therapy , Arrhythmias, Cardiac/genetics , Membrane Potentials/drug effects , Isoproterenol/pharmacology , Tachycardia, Ventricular/metabolism , Tachycardia, Ventricular/genetics , Tachycardia, Ventricular/drug therapy , Anti-Arrhythmia Agents/pharmacology , Male , Action Potentials/drug effects , Mice, Inbred C57BL
16.
Card Electrophysiol Clin ; 16(2): 195-202, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38749641

ABSTRACT

The case series reviews differential diagnosis of a genetic arrhythmia syndrome when evaluating a patient with prolonged QTc. Making the correct diagnosis requires: detailed patient history, family history, and careful review of the electrocardiogram (ECG). Signs and symptoms and ECG characteristics can often help clinicians make the diagnosis before genetic testing results return. These skills can help clinicians make an accurate and timely diagnosis and prevent life-threatening events.


Subject(s)
Arrhythmias, Cardiac , Electrocardiography , Long QT Syndrome , Humans , Diagnosis, Differential , Arrhythmias, Cardiac/diagnosis , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/physiopathology , Long QT Syndrome/diagnosis , Long QT Syndrome/genetics , Long QT Syndrome/physiopathology , Child , Male , Female , Adolescent , Genetic Testing
17.
Card Electrophysiol Clin ; 16(2): 211-218, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38749643

ABSTRACT

The following case series presents three different pediatric patients with SCN5A-related disease. In addition, family members are presented to demonstrate the variable penetrance that is commonly seen. Identifying features of this disease is important, because even in the very young, SCN5A disorders can cause lethal arrhythmias and sudden death.


Subject(s)
Arrhythmias, Cardiac , Long QT Syndrome , NAV1.5 Voltage-Gated Sodium Channel , Humans , NAV1.5 Voltage-Gated Sodium Channel/genetics , Long QT Syndrome/genetics , Long QT Syndrome/physiopathology , Male , Female , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/physiopathology , Arrhythmias, Cardiac/diagnosis , Child , Electrocardiography , Child, Preschool , Adolescent , Infant
18.
J Clin Invest ; 134(10)2024 May 15.
Article in English | MEDLINE | ID: mdl-38747296

ABSTRACT

Arrhythmogenic cardiomyopathy (ACM) is an inherited cardiac condition characterized by cardiac remodeling and life-threatening ventricular arrhythmias. In this issue of the JCI, Chelko, Penna, and colleagues mechanistically addressed the intricate contribution of immune-mediated injury in ACM pathogenesis. Inhibition of nuclear factor κ-B (NF-κB) and infiltration of monocyte-derived macrophages expressing C-C motif chemokine receptor-2 (CCR2) alleviated the phenotypic ACM features (i.e., fibrofatty replacement, contractile dysfunction, and ventricular arrhythmias) in desmoglein 2-mutant (Dsg2mut/mut) mice. These findings pave the way for efficacious and targetable immune therapy for patients with ACM.


Subject(s)
Desmoglein 2 , Macrophages , Receptors, CCR2 , Animals , Macrophages/metabolism , Macrophages/immunology , Macrophages/pathology , Mice , Humans , Desmoglein 2/genetics , Desmoglein 2/metabolism , Desmoglein 2/immunology , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Receptors, CCR2/antagonists & inhibitors , NF-kappa B/metabolism , NF-kappa B/genetics , Arrhythmias, Cardiac/pathology , Arrhythmias, Cardiac/immunology , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/metabolism , Arrhythmogenic Right Ventricular Dysplasia/genetics , Arrhythmogenic Right Ventricular Dysplasia/pathology , Arrhythmogenic Right Ventricular Dysplasia/metabolism , Cardiomyopathies/genetics , Cardiomyopathies/pathology , Cardiomyopathies/immunology , Cardiomyopathies/metabolism
19.
Sci Rep ; 14(1): 11980, 2024 05 25.
Article in English | MEDLINE | ID: mdl-38796549

ABSTRACT

Pathogenic BAG5 variants recently linked to dilated cardiomyopathy (DCM) prompt further investigation into phenotypic, mutational, and pathomechanistic aspects. We explored the clinical and molecular characteristics of DCM associated with BAG5 variants, uncovering the consistently severe manifestations of the disease and its impact on the endoplasmic reticulum (ER) stress response. The analysis involved three siblings affected by DCM and arrhythmia, along with their four unaffected siblings, their unaffected father, and their mother who exhibited arrhythmia. The parents were consanguineous. Exome and Sanger sequencing identified a novel BAG5 variant, c.444_445delGA (p.Lys149AsnfsTer6), homozygous in affected siblings and heterozygous in parents and unaffected siblings. We generated heterozygous and homozygous Bag5 point mutant knock-in (KI) mice and evaluated cardiac pathophysiology under stress conditions, including tunicamycin (TN) administration. Bag5-/- mice displayed no abnormalities up to 12 months old and showed no anomalies during an exercise stress test. However, following TN injection, Bag5-/- mice exhibited significantly reduced left ventricular fractional shortening (LVFS) and ejection fraction (LVEF). Their cardiac tissues exhibited a notable increase in apoptotic cells, despite non-distinctive changes in CHOP and GRP78 levels. Interestingly, only Bag5 KI male mice demonstrated arrhythmia, which was more pronounced in Bag5-/- than in Bag5+/-males. Here, our study reveals a novel BAG5 mutation causing DCM by impairing the ER stress response, with observed sex-specific arrhythmia differences.


Subject(s)
Arrhythmias, Cardiac , Cardiomyopathy, Dilated , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress , Animals , Cardiomyopathy, Dilated/genetics , Endoplasmic Reticulum Stress/genetics , Humans , Arrhythmias, Cardiac/genetics , Male , Female , Mice , Pedigree , Mice, Knockout , Adult , Apoptosis/genetics , Mutation
20.
Card Electrophysiol Clin ; 16(2): 203-210, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38749642

ABSTRACT

Bidirectional ventricular tachycardia is a unique arrhythmia that can herald lethal arrhythmia syndromes. Using cases based on real patient stories, this article examines 3 different presentations to help clinicians learn the differential diagnosis associated with this condition. Each associated genetic disorder will be briefly discussed, and valuable tips for distinguishing them from each other will be provided.


Subject(s)
Tachycardia, Ventricular , Child , Humans , Male , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/diagnosis , Arrhythmias, Cardiac/physiopathology , Diagnosis, Differential , Electrocardiography , Long QT Syndrome/genetics , Long QT Syndrome/diagnosis , Long QT Syndrome/physiopathology , Tachycardia, Ventricular/diagnosis , Tachycardia, Ventricular/genetics , Tachycardia, Ventricular/physiopathology , Adolescent
SELECTION OF CITATIONS
SEARCH DETAIL
...