Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
1.
Curr Cardiol Rep ; 25(7): 621-630, 2023 07.
Article in English | MEDLINE | ID: mdl-37227579

ABSTRACT

PURPOSE OF REVIEW: The sinoatrial node (SAN), the natural pacemaker of the heart, is responsible for generating electrical impulses and initiating each heartbeat. Sinoatrial node dysfunction (SND) causes various arrhythmias such as sinus arrest, SAN block, and tachycardia/bradycardia syndrome. Unraveling the underlying mechanisms of SND is of paramount importance in the pursuit of developing effective therapeutic strategies for patients with SND. This review provides a concise summary of the most recent progress in the signaling regulation of SND. RECENT FINDINGS: Recent studies indicate that SND can be caused by abnormal intercellular and intracellular signaling, various forms of heart failure (HF), and diabetes. These discoveries provide novel insights into the underlying mechanisms SND, advancing our understanding of its pathogenesis. SND can cause severe cardiac arrhythmias associated with syncope and an increased risk of sudden death. In addition to ion channels, the SAN is susceptible to the influence of various signalings including Hippo, AMP-activated protein kinase (AMPK), mechanical force, and natriuretic peptide receptors. New cellular and molecular mechanisms related to SND are also deciphered in systemic diseases such as HF and diabetes. Progress in these studies contributes to the development of potential therapeutics for SND.


Subject(s)
Heart Failure , Sinoatrial Node , Humans , Sinoatrial Node/metabolism , Sinoatrial Node/pathology , Sick Sinus Syndrome/metabolism , Sick Sinus Syndrome/pathology , Arrhythmias, Cardiac , Heart Rate/physiology , Ion Channels
2.
Proc Natl Acad Sci U S A ; 119(36): e2206708119, 2022 09 06.
Article in English | MEDLINE | ID: mdl-36044551

ABSTRACT

The sinoatrial node (SAN), the leading pacemaker region, generates electrical impulses that propagate throughout the heart. SAN dysfunction with bradyarrhythmia is well documented in heart failure (HF). However, the underlying mechanisms are not completely understood. Mitochondria are critical to cellular processes that determine the life or death of the cell. The release of Ca2+ from the ryanodine receptors 2 (RyR2) on the sarcoplasmic reticulum (SR) at mitochondria-SR microdomains serves as the critical communication to match energy production to meet metabolic demands. Therefore, we tested the hypothesis that alterations in the mitochondria-SR connectomics contribute to SAN dysfunction in HF. We took advantage of a mouse model of chronic pressure overload-induced HF by transverse aortic constriction (TAC) and a SAN-specific CRISPR-Cas9-mediated knockdown of mitofusin-2 (Mfn2), the mitochondria-SR tethering GTPase protein. TAC mice exhibited impaired cardiac function with HF, cardiac fibrosis, and profound SAN dysfunction. Ultrastructural imaging using electron microscope (EM) tomography revealed abnormal mitochondrial structure with increased mitochondria-SR distance. The expression of Mfn2 was significantly down-regulated and showed reduced colocalization with RyR2 in HF SAN cells. Indeed, SAN-specific Mfn2 knockdown led to alterations in the mitochondria-SR microdomains and SAN dysfunction. Finally, disruptions in the mitochondria-SR microdomains resulted in abnormal mitochondrial Ca2+ handling, alterations in localized protein kinase A (PKA) activity, and impaired mitochondrial function in HF SAN cells. The current study provides insights into the role of mitochondria-SR microdomains in SAN automaticity and possible therapeutic targets for SAN dysfunction in HF patients.


Subject(s)
Connectome , Heart Failure , Mitochondria, Heart , Sarcoplasmic Reticulum , Sick Sinus Syndrome , Sinoatrial Node , Animals , Heart Failure/pathology , Heart Failure/physiopathology , Mice , Mitochondria, Heart/ultrastructure , Myocytes, Cardiac/metabolism , Ryanodine Receptor Calcium Release Channel/genetics , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum/pathology , Sick Sinus Syndrome/pathology , Sick Sinus Syndrome/physiopathology , Sinoatrial Node/physiopathology
3.
Biochim Biophys Acta Mol Basis Dis ; 1866(7): 165757, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32147422

ABSTRACT

Atrial fibrillation (AF) is the most common arrhythmias, and patients with AF are facing increased risk of heart failure and ischemic stroke. However, the AF pathogenesis, especially the long noncoding RNAs (lncRNA)-related mechanism, has not been fully understood. In this study, we collected RNA sequencing data of the epicardial adipose tissues (EAT) from 6 AF and 6 sinus rhythm (SR) to identify the differentially expressed protein-coding genes (PCGs) and lncRNAs. Functionally, the differentially expressed PCGs were significantly enriched in bone development disease, chronic kidney failure, and kidney disease. Particularly, we found that homeobox (HOX) genes, especially the antisense RNAs, HOTAIRM1, HOXA-AS2 and HOXB-AS2, were significantly downregulated in EAT of AF. The biological function predictions for the dysregulated lncRNAs revealed that TNF signaling pathway was the most frequent pathway that the lncRNAs might participate in. In addition, SNHG16 and RP11-471B22.2 might participate in TGF-beta signaling and ECM-receptor interaction by interacting with the proteins involved in the pathways, respectively. Collectively, we provided some potentially pathogenic lncRNAs in AF, which might be useful for the related researchers to study their functionality and develop new therapeutics.


Subject(s)
Atrial Fibrillation/genetics , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Sick Sinus Syndrome/congenital , Adipose Tissue/metabolism , Adipose Tissue/pathology , Atrial Fibrillation/pathology , Female , Gene Expression Regulation/genetics , Genes, Homeobox/genetics , Genome, Human/genetics , Humans , Male , Pericardium/metabolism , RNA, Long Noncoding/classification , Sequence Analysis, RNA , Sick Sinus Syndrome/genetics , Sick Sinus Syndrome/pathology
4.
J Mol Cell Cardiol ; 138: 291-303, 2020 01.
Article in English | MEDLINE | ID: mdl-31751569

ABSTRACT

OBJECTIVE: Sick sinus syndrome (SSS) is associated with loss of HCN4 (hyperpolarization-activated cyclic nucleotide-gated potassium channel 4) function in the cardiac conduction system. The underlying mechanism for SSS remains elusive. This study is to investigate how mitochondrial oxidative stress induces HCN4 downregulation associated with in sick sinus syndrome. METHODS AND RESULTS: Trx2lox/lox mice were crossed with α-myosin heavy chain (α-Mhc)-Cre and Hcn4-CreERT2 deleter mice to generate Trx2 deletion mice in the whole heart (Trx2cKO) and in the conduction system (Trx2ccsKO), respectively. Echocardiography was applied to measure hemodynamics and heart rhythm. Histological analyses, gene profiling and chromatin immunoprecipitation were performed to define the mechanism by which thioredoxin-2 (Trx2) regulates HCN4 expression and cardiac function. Trx2cKO mice displayed dilated cardiomyopathy, low heart rate, and atrial ventricular block (AVB) phenotypes. Immunofluorescence revealed that HCN4 expression was specifically reduced within the sinoatrial node in Trx2cKO mice. Interestingly, Trx2ccsKO mice displayed low heart rate and AVB without dilated cardiomyopathy. Both mRNA and protein levels of HCN4 were reduced in the sinoatrial node, suggesting transcriptional HCN4 regulation upon Trx2 deletion. ChIP indicated that the binding of MEF2 to the HCN4 enhancer was not altered by Trx2 deletion; however, histone 3 acetylation at the MEF2 binding site was decreased, and expression of histone deacetylase 4 (HDAC4) was elevated following Trx2 deletion. Moreover, HDAC4 binding to the HCN4 enhancer was mediated by MEF2. Mitochondrial ROS were increased by Trx2 deletion and importantly, mitochondria-specific ROS scavenger MitoTEMPO suppressed HDAC4 elevation, HCN4 reduction, and sinus bradycardia in Trx2ccsKO mice. CONCLUSION: In the conduction system, Trx2 is critical for maintaining HCN4-mediated normal heart rate. Loss of Trx2 reduces HCN4 expression via a mitochondrial ROS-HDAC4-MEF2C pathway and subsequently induces sick sinus syndrome in mice.


Subject(s)
Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Mitochondria, Heart/metabolism , Oxidative Stress , Sick Sinus Syndrome/genetics , Sick Sinus Syndrome/pathology , Thioredoxins/metabolism , Animals , Bradycardia/complications , Bradycardia/metabolism , Bradycardia/pathology , Cardiomyopathy, Dilated/complications , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , Enhancer Elements, Genetic/genetics , Histone Deacetylases/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , MEF2 Transcription Factors/metabolism , Mice, Knockout , Models, Biological , Oxidative Stress/genetics , Phenotype , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Sick Sinus Syndrome/complications , Sinoatrial Node/metabolism , Sinoatrial Node/pathology
5.
Circ Genom Precis Med ; 12(1): e002238, 2019 01.
Article in English | MEDLINE | ID: mdl-30645171

ABSTRACT

BACKGROUND: Inherited forms of sinus node dysfunction (SND) clinically include bradycardia, sinus arrest, and chronotropic incompetence and may serve as disease models to understand sinus node physiology and impulse generation. Recently, a gain-of-function mutation in the G-protein gene GNB2 led to enhanced activation of the GIRK (G-protein activated inwardly rectifying K+ channel). Thus, human cardiac GIRK channels are important for heart rate regulation and subsequently, genes encoding their subunits Kir3.1 and Kir3.4 ( KCNJ3 and KCNJ5) are potential candidates for inherited SND in human. METHODS: We performed a combined approach of targeted sequencing of KCNJ3 and KCNJ5 in 52 patients with idiopathic SND and subsequent whole exome sequencing of additional family members in a genetically affected patient. A putative novel disease-associated gene variant was functionally analyzed by voltage-clamp experiments using various heterologous cell expression systems (Xenopus oocytes, CHO cells, and rat atrial cardiomyocytes). RESULTS: In a 3-generation family with SND we identified a novel variant in KCNJ5 which leads to an amino acid substitution (p.Trp101Cys) in the first transmembrane domain of the Kir3.4 subunit of the cardiac GIRK channel. The identified variant cosegregated with the disease in the family and was absent in the Exome Variant Server and Exome Aggregation Consortium databases. Expression of mutant Kir3.4 (±native Kir3.1) in different heterologous cell expression systems resulted in increased GIRK currents ( IK,ACh) and a reduced inward rectification which was not compensated by intracellular spermidine. Moreover, in silico modeling of heterotetrameric mutant GIRK channels indicates a structurally altered binding site for spermine. CONCLUSIONS: For the first time, an inherited gain-of-function mutation in the human GIRK3.4 causes familial human SND. The increased activity of GIRK channels is likely to lead to a sustained hyperpolarization of pacemaker cells and thereby reduces heart rate. Modulation of human GIRK channels may pave a way for further treatment of cardiac pacemaking.


Subject(s)
G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Gain of Function Mutation , Genetic Predisposition to Disease , Ion Channel Gating , Sick Sinus Syndrome/genetics , Sick Sinus Syndrome/pathology , Adolescent , Adult , Aged , Child , Female , Humans , Male , Membrane Potentials , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Young Adult
6.
Biomed Pharmacother ; 111: 778-784, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30612002

ABSTRACT

The Sick Sinus Syndrome (SSS) is a serious life-threatening heart disease. It is important to establish a credible and stable sinus node damage model. In this study, we use two methods to construct an SSS damage model in rats. One is to inject sodium hydroxide to the SSS area through internal jugular vein. Another is to cause ischemia-reperfusion injury on the SSS area. 43 healthy SD rats were randomly divided into 4 groups, namely ischemia-reperfusion injury group (IRIG), inject sodium hydroxide group (ISHG), and propranolol group (PG) and the control group (CG). The achievement ratio of modeling was 67% in the IRIG and 83% in the ISHG. The HR significantly decreased after operation in the IRIG and ISHG compared with pre-operation (P<0.01). The HR was reduced by above 30% in these 2 groups after modeling, while the reduction was better maintained in IRIG. Additionally, the sinoatrial node recovery time (SNRT) and sinoatrial conduction time (SACT) were significantly prolonged compared with pre-modeling in 2 groups (P < 0.01). Morphology results showed blurry in structure and boundaries with pale cytoplasm. It is speculated that IRIG and ISHG modeling might influence the calcium concentration and damage the sinus node function by decrease the expression of HCN4 and SCN5A, which impaired the driving ability of sinus node and leading to apoptosis. Ischemia reperfusion injury and sodium hydroxide injury could construct stable SSS models which could represent clinic pathological damage. Thus, both methods could be used for further studies of the SSS mechanisms and drugs.


Subject(s)
Disease Models, Animal , Reperfusion Injury/chemically induced , Reperfusion Injury/metabolism , Sick Sinus Syndrome/chemically induced , Sick Sinus Syndrome/metabolism , Sodium Hydroxide/toxicity , Animals , Heart Rate/drug effects , Heart Rate/physiology , Male , Rats , Rats, Sprague-Dawley , Reperfusion Injury/pathology , Sick Sinus Syndrome/pathology
7.
Sci Rep ; 8(1): 14565, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30275471

ABSTRACT

Genome-wide association studies have reported a strong association of the single nucleotide polymorphism (SNP) rs6817105 (T > C) on chromosome 4q25 with atrial fibrillation (AF), but phenotype alterations conferred by this SNP have not been described. We genotyped SNP rs6817105 and examined the relationships among rs6817105 genotype, clinical characteristics, echocardiographic parameters, and electrophysiological parameters in 574 AF patients and 1,554 non-AF controls. Further, multiple microRNAs (miRNAs) are reported to be involved in atrial remodeling and AF pathogenesis, so we investigated relationships between rs6817105 genotype and serum concentrations of 2555 miRNAs. The rs6817105 minor allele frequency was significantly higher in AF patients than non-AF controls (66% vs. 47%, odds ratio 2.12, p = 4.9 × 10-26). Corrected sinus node recovery time (CSRT) was longer and left atrial volume index (LAVI) was larger in AF patients with the rs6817105 minor allele than patient non-carriers (CSRT: CC 557 ± 315 ms, CT 486 ± 273 ms, TT 447 ± 234 ms, p = 0.001; LAVI: CC 43.6 ± 12.1, CT 42.4 ± 13.6, TT 39.8 ± 11.6, p = 0.030). There were no significant differences between rs6817105 genotype and the serum concentrations of miRNAs. These findings strongly implicate rs6817105 minor allele in sinus node dysfunction and left atrial enlargement.


Subject(s)
Atrial Fibrillation/genetics , Chromosomes, Human, Pair 4 , Genetic Loci , Genotype , Heart Atria/pathology , Sick Sinus Syndrome/genetics , Aged , Atrial Fibrillation/pathology , Echocardiography , Electrocardiography , Female , Gene Frequency , Humans , Male , MicroRNAs/blood , Middle Aged , Polymorphism, Single Nucleotide , Sick Sinus Syndrome/pathology
8.
Cell Physiol Biochem ; 42(5): 2021-2029, 2017.
Article in English | MEDLINE | ID: mdl-28803248

ABSTRACT

BACKGROUND/AIMS: Congenital Sick Sinus Syndrome (SSS) is a disorder associated with sudden cardiac death due to severe bradycardia and prolonged pauses. Mutations in HCN4, the gene encoding inward Na+/K+ current (If), have been described as a cause of congenital SSS. The objective of this study is to develop an SSS model in embryonic zebrafish, and use zebrafish as a moderate-throughput assay to functionally characterize HCN4 variants. METHODS: To determine the function of hcn4 in zebrafish, embryos were either bathed in the If -specific blocker (ZD-7288), or endogenous hcn4 expression was knocked down using splice-blocking morpholinos. To assess whether the zebrafish model discriminates benign from pathogenic variants, we tested four HCN4 mutations known to cause human SSS and four variants of unknown significance (VUS). RESULTS: Pharmacological blockade and knockdown of hcn4 in zebrafish phenocopied human SSS, displaying bradycardia and cardiac pauses in intact embryos and explanted hearts. The zebrafish assay correctly identified all disease-causing variants. Of the VUS, the assay predicted 2 as benign and 2 as hypomorphic variants. CONCLUSIONS: We conclude that our embryonic zebrafish assay is a novel and effective tool to functionally characterize human HCN4 variants, which can be translated into important clinical prognostic information.


Subject(s)
Genetic Variation , Sick Sinus Syndrome/pathology , Animals , Animals, Genetically Modified , Bradycardia/etiology , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Genotype , Heart/drug effects , Heart/physiology , Heart Rate/drug effects , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/antagonists & inhibitors , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , In Situ Hybridization , Morpholinos/metabolism , Muscle Proteins/antagonists & inhibitors , Muscle Proteins/genetics , Muscle Proteins/metabolism , Mutation , Patch-Clamp Techniques , Phenotype , Potassium Channels/genetics , Potassium Channels/metabolism , Pyrimidines/pharmacology , Sick Sinus Syndrome/genetics , Zebrafish/metabolism
9.
Cardiovasc Res ; 111(3): 274-86, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27097650

ABSTRACT

AIMS: Current mechanisms driving cardiac pacemaker function have focused on ion channel and gap junction channel function, which are essential for action potential generation and propagation between pacemaker cells. However, pacemaker cells also harbour desmosomes that structurally anchor pacemaker cells to each other in tissue, but their role in pacemaker function remains unknown. METHODS AND RESULTS: To determine the role of desmosomes in pacemaker function, we generated a novel mouse model harbouring cardiac conduction-specific ablation (csKO) of the central desmosomal protein, desmoplakin (DSP) using the Hcn4-Cre-ERT2 mouse line. Hcn4-Cre targets cells of the adult mouse sinoatrial node (SAN) and can ablate DSP expression in the adult DSP csKO SAN resulting in specific loss of desmosomal proteins and structures. Dysregulation of DSP via loss-of-function (adult DSP csKO mice) and mutation (clinical case of a patient harbouring a pathogenic DSP variant) in mice and man, respectively, revealed that desmosomal dysregulation is associated with a primary phenotype of increased sinus pauses/dysfunction in the absence of cardiomyopathy. Underlying defects in beat-to-beat regulation were also observed in DSP csKO mice in vivo and intact atria ex vivo. DSP csKO SAN exhibited migrating lead pacemaker sites associated with connexin 45 loss. In vitro studies exploiting ventricular cardiomyocytes that harbour DSP loss and concurrent early connexin loss phenocopied the loss of beat-to-beat regulation observed in DSP csKO mice and atria, extending the importance of DSP-associated mechanisms in driving beat-to-beat regulation of working cardiomyocytes. CONCLUSION: We provide evidence of a mechanism that implicates an essential role for desmosomes in cardiac pacemaker function, which has broad implications in better understanding mechanisms underlying beat-to-beat regulation as well as sinus node disease and dysfunction.


Subject(s)
Biological Clocks , Desmosomes , Heart Rate , Sick Sinus Syndrome/physiopathology , Sinoatrial Node/physiopathology , Action Potentials , Age Factors , Animals , Atrial Function , Cells, Cultured , Connexins/metabolism , Desmoplakins/deficiency , Desmoplakins/genetics , Desmosomes/metabolism , Desmosomes/ultrastructure , Genetic Predisposition to Disease , Humans , Mice, Knockout , Mutation , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/ultrastructure , Phenotype , Sick Sinus Syndrome/genetics , Sick Sinus Syndrome/metabolism , Sick Sinus Syndrome/pathology , Sinoatrial Node/metabolism , Sinoatrial Node/ultrastructure , Time Factors
10.
J Clin Invest ; 125(8): 3256-68, 2015 Aug 03.
Article in English | MEDLINE | ID: mdl-26193633

ABSTRACT

The sinoatrial node (SAN) maintains a rhythmic heartbeat; therefore, a better understanding of factors that drive SAN development and function is crucial to generation of potential therapies, such as biological pacemakers, for sinus arrhythmias. Here, we determined that the LIM homeodomain transcription factor ISL1 plays a key role in survival, proliferation, and function of pacemaker cells throughout development. Analysis of several Isl1 mutant mouse lines, including animals harboring an SAN-specific Isl1 deletion, revealed that ISL1 within SAN is a requirement for early embryonic viability. RNA-sequencing (RNA-seq) analyses of FACS-purified cells from ISL1-deficient SANs revealed that a number of genes critical for SAN function, including those encoding transcription factors and ion channels, were downstream of ISL1. Chromatin immunoprecipitation assays performed with anti-ISL1 antibodies and chromatin extracts from FACS-purified SAN cells demonstrated that ISL1 directly binds genomic regions within several genes required for normal pacemaker function, including subunits of the L-type calcium channel, Ank2, and Tbx3. Other genes implicated in abnormal heart rhythm in humans were also direct ISL1 targets. Together, our results demonstrate that ISL1 regulates approximately one-third of SAN-specific genes, indicate that a combination of ISL1 and other SAN transcription factors could be utilized to generate pacemaker cells, and suggest ISL1 mutations may underlie sick sinus syndrome.


Subject(s)
Cell Proliferation/physiology , Gene Expression Regulation, Developmental/physiology , LIM-Homeodomain Proteins/metabolism , Myocardial Contraction/physiology , Sinoatrial Node/embryology , Transcription Factors/metabolism , Animals , Ankyrins/genetics , Ankyrins/metabolism , Cell Survival , Chromatin/genetics , Chromatin/metabolism , Gene Deletion , LIM-Homeodomain Proteins/genetics , Mice , Mice, Transgenic , Protein Binding , Sick Sinus Syndrome/embryology , Sick Sinus Syndrome/genetics , Sick Sinus Syndrome/pathology , Sinoatrial Node/cytology , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Transcription Factors/genetics
11.
Int J Mol Sci ; 16(2): 3071-94, 2015 Jan 29.
Article in English | MEDLINE | ID: mdl-25642760

ABSTRACT

Since 2003, several loss-of-function mutations in the HCN4 gene, which encodes the HCN4 protein, have been associated with sinus node dysfunction. In human sinoatrial node (SAN), HCN4 is the most abundant of the four isoforms of the HCN family. Tetramers of HCN subunits constitute the ion channels that conduct the hyperpolarization-activated "funny" current (If), which plays an important modulating role in SAN pacemaker activity. Voltage-clamp experiments on HCN4 channels expressed in COS-7, CHO and HEK-293 cells, as well as in Xenopus oocytes have revealed changes in the expression and kinetics of mutant channels, but the extent to which especially the kinetic changes would affect If flowing during a human SAN action potential often remains unresolved. In our contribution to the Topical Collection on Human Single Nucleotide Polymorphisms and Disease Diagnostics, we provide an updated review of the mutation-induced changes in the expression and kinetics of HCN4 channels and provide an overview of their effects on If during the time course of a human SAN action potential, as assessed in simulated action potential clamp experiments. Future research may solve apparent inconsistencies between data from clinical studies and data from in vitro and in silico experiments.


Subject(s)
Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Sinoatrial Node/metabolism , Action Potentials , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Mutation , Potassium Channels, Voltage-Gated/genetics , Potassium Channels, Voltage-Gated/metabolism , Sick Sinus Syndrome/metabolism , Sick Sinus Syndrome/pathology
12.
Stem Cell Reports ; 2(5): 592-605, 2014 May 06.
Article in English | MEDLINE | ID: mdl-24936448

ABSTRACT

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


Subject(s)
Cellular Reprogramming , Pluripotent Stem Cells/cytology , Sinoatrial Node/physiology , Animals , Biological Clocks , Calcium/metabolism , Cell Differentiation , Cell Line , Coculture Techniques , In Vitro Techniques , Mice , Models, Biological , Myocytes, Cardiac/metabolism , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Patch-Clamp Techniques , Pluripotent Stem Cells/metabolism , Sick Sinus Syndrome/metabolism , Sick Sinus Syndrome/pathology , Sick Sinus Syndrome/veterinary , Sinoatrial Node/cytology , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism
14.
Circ Arrhythm Electrophysiol ; 6(2): 392-401, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23420830

ABSTRACT

BACKGROUND: Loss-of-function mutations in Na(v)1.5 cause sodium channelopathies, including Brugada syndrome, dilated cardiomyopathy, and sick sinus syndrome; however, no effective therapy exists. MOG1 increases plasma membrane (PM) expression of Na(v)1.5 and sodium current (I(Na)) density, thus we hypothesize that MOG1 can serve as a therapeutic target for sodium channelopathies. METHODS AND RESULTS: Knockdown of MOG1 expression using small interfering RNAs reduced Na(v)1.5 PM expression, decreased I(Na) densities by 2-fold in HEK/Na(v)1.5 cells and nearly abolished I(Na) in mouse cardiomyocytes. MOG1 did not affect Na(v)1.5 PM turnover. MOG1 small interfering RNAs caused retention of Na(v)1.5 in endoplasmic reticulum, disrupted the distribution of Na(v)1.5 into caveolin-3-enriched microdomains, and led to redistribution of Na(v)1.5 to noncaveolin-rich domains. MOG1 fully rescued the reduced PM expression and I(Na) densities by Na(v)1.5 trafficking-defective mutation D1275N associated with sick sinus syndrome/dilated cardiomyopathy/atrial arrhythmias. For Brugada syndrome mutation G1743R, MOG1 restored the impaired PM expression of the mutant protein and restored I(Na) in a heterozygous state (mixture of wild type and mutant Na(v)1.5) to a full level of a homozygous wild-type state. CONCLUSIONS: Use of MOG1 to enhance Na(v)1.5 trafficking to PM may be a potential personalized therapeutic approach for some patients with Brugada syndrome, dilated cardiomyopathy, and sick sinus syndrome in the future.


Subject(s)
Brugada Syndrome/genetics , Mutation , Myocytes, Cardiac/metabolism , NAV1.5 Voltage-Gated Sodium Channel/genetics , RNA/genetics , Sick Sinus Syndrome/genetics , Animals , Brugada Syndrome/metabolism , Brugada Syndrome/pathology , Cells, Cultured , DNA Mutational Analysis , Disease Models, Animal , Mice , Mice, Knockout , Myocytes, Cardiac/pathology , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Sick Sinus Syndrome/metabolism , Sick Sinus Syndrome/pathology
15.
J Appl Physiol (1985) ; 113(11): 1802-8, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-22898552

ABSTRACT

Sick Sinus Syndrome is a common and refractory arrhythmia, needing further study in which setting up a credible sinus node damage model is important. To explore the feasibility and superiority of an original formaldehyde pinpoint pressing permeation (FPPP) method for building a chronic sinus node damage (CSND) model, 5 rabbits were chosen from 35 as a sham-operation group, and the remaining were randomly divided into two groups: the formaldehyde wet compressing (FWC) group, in which models were established by applying a cotton bud dipped in 20% formaldehyde onto the sinus node (SN) area, and the FPPP group, in which models were established by injecting formaldehyde into the SN area through a self-made pinpointing and injecting electrode. We found that in both groups, the HR at 2 h, 24 h, 1 wk, and 2 wk after modeling decreased compared with premodeling; sinoatrial conduction time, sinus node recovery time, and corrected sinus node recovery time were prolonged compared with premodeling. The indexes mentioned shortened by 2 wk after modeling compared with 2 h in the FWC group, whereas they were stable after modeling in the FPPP group. The modeling achievement ratio in the FPPP group was higher and the death rate was lower. Under light microscope, paraffin sections of the SN tissue and cells showed severe injury in both groups. The results indicate that the CSND models in rabbits can be successfully established by the FPPP method, with higher achievement ratio, lower death rate, better stabilization effect, and less damaging comparing with the traditional method.


Subject(s)
Formaldehyde , Sick Sinus Syndrome/chemically induced , Sinoatrial Node/physiopathology , Action Potentials , Administration, Topical , Animals , Chronic Disease , Disease Models, Animal , Electrophysiologic Techniques, Cardiac , Feasibility Studies , Female , Formaldehyde/administration & dosage , Heart Rate , Injections , Male , Rabbits , Reproducibility of Results , Sick Sinus Syndrome/diagnosis , Sick Sinus Syndrome/pathology , Sick Sinus Syndrome/physiopathology , Sinoatrial Node/pathology , Time Factors
16.
J Biol Chem ; 287(36): 30268-81, 2012 Aug 31.
Article in English | MEDLINE | ID: mdl-22778271

ABSTRACT

Ankyrins (ankyrin-R, -B, and -G) are adapter proteins linked with defects in metazoan physiology. Ankyrin-B (encoded by ANK2) loss-of-function mutations are directly associated with human cardiovascular phenotypes including sinus node disease, atrial fibrillation, ventricular tachycardia, and sudden cardiac death. Despite the link between ankyrin-B dysfunction and monogenic disease, there are no data linking ankyrin-B regulation with common forms of human heart failure. Here, we report that ankyrin-B levels are altered in both ischemic and non-ischemic human heart failure. Mechanistically, we demonstrate that cardiac ankyrin-B levels are tightly regulated downstream of reactive oxygen species, intracellular calcium, and the calcium-dependent protease calpain, all hallmarks of human myocardial injury and heart failure. Surprisingly, ß(II)-spectrin, previously thought to mediate ankyrin-dependent modulation in the nervous system and heart, is not coordinately regulated with ankyrin-B or its downstream partners. Finally, our data implicate ankyrin-B expression as required for vertebrate myocardial protection as hearts deficient in ankyrin-B show increased cardiac damage and impaired function relative to wild-type mouse hearts following ischemia reperfusion. In summary, our findings provide the data of ankyrin-B regulation in human heart failure, provide insight into candidate pathways for ankyrin-B regulation in acquired human cardiovascular disease, and surprisingly, implicate ankyrin-B as a molecular component for cardioprotection following ischemia.


Subject(s)
Ankyrins/biosynthesis , Gene Expression Regulation , Heart Failure/metabolism , Muscle Proteins/metabolism , Myocardium/metabolism , Animals , Ankyrins/genetics , Calcium/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cells, Cultured , Heart Failure/genetics , Heart Failure/pathology , Humans , Mice , Mice, Mutant Strains , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Muscle Proteins/genetics , Myocardial Ischemia/genetics , Myocardial Ischemia/metabolism , Myocardial Ischemia/pathology , Myocardium/pathology , Reactive Oxygen Species/metabolism , Sick Sinus Syndrome/genetics , Sick Sinus Syndrome/metabolism , Sick Sinus Syndrome/pathology
18.
J Comp Pathol ; 146(2-3): 175-82, 2012.
Article in English | MEDLINE | ID: mdl-21612788

ABSTRACT

The hearts of seven elderly dogs in which bradycardia-tachycardia syndrome (BTS) had been diagnosed electrocardiographically were examined post mortem. The clinical basis of the underlying heart disease was invariably mitral or mitral and tricuspid regurgitation. Microscopical examination of the sinoatrial (SA) node and the SA junctional region consistently revealed depletion of SA nodal cells, with a corresponding increase in fibrous or fibro-fatty tissue that interrupted contiguity between the SA node and the surrounding atrial myocardium. The left and right atrial walls showed an increased amount of fibrous tissue in the myocardium and disruption of the muscle bundle architecture (interstitial myocardial fibrosis) to varying degrees. Qualitatively, these changes in the SA node and the SA node region resembled those associated with ageing in elderly people with or without BTS. Thus, it is possible that the pathological process affecting the SA node in these dogs was fundamentally related to ageing and may have caused BTS, in combination with atrial myocardial lesions caused by mitral and tricuspid regurgitation.


Subject(s)
Bradycardia/veterinary , Dog Diseases/pathology , Heart Valve Diseases/veterinary , Heart Valves/pathology , Sick Sinus Syndrome/veterinary , Tachycardia/veterinary , Animals , Bradycardia/complications , Bradycardia/pathology , Dogs , Heart Valve Diseases/complications , Heart Valve Diseases/pathology , Sick Sinus Syndrome/complications , Sick Sinus Syndrome/pathology , Tachycardia/complications , Tachycardia/pathology
19.
J Am Soc Echocardiogr ; 24(5): 556-564.e1, 2011 May.
Article in English | MEDLINE | ID: mdl-21353472

ABSTRACT

BACKGROUND: Long-term right ventricular apical pacing (RVAP) can lead to adverse clinical outcomes. Although left ventricular (LV) dyssynchrony is the major causative factor, other potential mechanisms are not fully understood. We sought to clarify whether RVAP elicits apical wall motion abnormalities that contribute to LV contractile dysfunction. METHODS: We studied annual echocardiographic data over a 5-year period after pacemaker implantation (PMI) for 74 patients who underwent RVAP. The patients were divided into two groups according to the percentage of ventricular pacing: right ventricular (RV) pacing < 50% and RV pacing ≥ 50%. We assessed LV ejection fraction, LV end-diastolic volume, and left atrial dimension. To assess regional wall motion abnormalities, the wall motion score index was calculated. RESULTS: LV wall motion abnormality was observed in 64% of the subjects and was more pronounced in apical segments than in other segments. At 2 years after PMI, brain natriuretic peptide levels were significantly higher in the group with RV pacing ≥ 50% than in the group with RV pacing < 50%. The subjects with RV pacing ≥ 50% had higher LV end-diastolic dimension and lower ejection fraction at 3 years after PMI. CONCLUSION: Long-term RVAP elicits apical wall motion abnormalities that could in part contribute to LV contractile dysfunction.


Subject(s)
Heart Atria/pathology , Heart Ventricles/pathology , Ventricular Dysfunction, Left/pathology , Ventricular Dysfunction, Right/pathology , Aged , Female , Heart Atria/diagnostic imaging , Heart Ventricles/diagnostic imaging , Humans , Male , Natriuretic Peptide, Brain , Prevalence , Retrospective Studies , Sick Sinus Syndrome/blood , Sick Sinus Syndrome/diagnostic imaging , Sick Sinus Syndrome/pathology , Statistics, Nonparametric , Stroke Volume , Time Factors , Ultrasonography , Ventricular Dysfunction, Left/diagnostic imaging , Ventricular Dysfunction, Right/diagnostic imaging , Ventricular Function, Left
20.
J Vet Cardiol ; 13(1): 63-70, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21288788

ABSTRACT

Using 24-h ambulatory electrocardiography, the RR intervals of all beats were determined in a West Highland white terrier with sick sinus syndrome characterized by long sinus pauses, bradycardia, supraventricular tachycardia (SVT) and varying degrees of atrioventricular (AV) heart block. Distinctive patterns of bradycardia and 1:1, 2:1, 3:1, 4:1 and 5:1 AV block associated with SVT were evident in the tachogram (RR interval distribution over time) and Poincaré plots (short-term heart rate variability plots of RRn versus RRn+1). These patterns differed from those of abrupt alteration in cycle length during long sinus pauses or bursts of supraventricular tachycardia. Recognition of such patterns may direct attention to time points for which close attention to the cardiac rhythm should be evaluated in the full-disclosure of the 24-h ECG recording.


Subject(s)
Atrioventricular Block/veterinary , Dog Diseases/pathology , Electrocardiography/veterinary , Sick Sinus Syndrome/veterinary , Tachycardia, Supraventricular/veterinary , Animals , Anti-Arrhythmia Agents/therapeutic use , Atenolol/therapeutic use , Atrioventricular Block/pathology , Atrioventricular Block/therapy , Circadian Rhythm , Dog Diseases/therapy , Dogs , Male , Pacemaker, Artificial/veterinary , Sick Sinus Syndrome/pathology , Sick Sinus Syndrome/therapy , Tachycardia, Supraventricular/pathology , Tachycardia, Supraventricular/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...