Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 6.465
Filtrar
1.
J Gen Physiol ; 156(9)2024 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-39110119

RESUMO

JGP study (Si et al. https://doi.org/10.1085/jgp.202413578) reveals that, although they are present at low levels and only generate small currents in the sinoatrial node, Kv1.1 channels have a significant impact on cardiac pacemaking.


Assuntos
Canal de Potássio Kv1.1 , Nó Sinoatrial , Animais , Canal de Potássio Kv1.1/metabolismo , Canal de Potássio Kv1.1/genética , Nó Sinoatrial/metabolismo , Nó Sinoatrial/fisiologia , Potenciais de Ação/fisiologia , Humanos , Frequência Cardíaca/fisiologia
2.
Nat Commun ; 15(1): 6550, 2024 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-39095365

RESUMO

The cardiac conduction system (CCS) is a network of specialized cardiomyocytes that coordinates electrical impulse generation and propagation for synchronized heart contractions. Although the components of the CCS, including the sinoatrial node, atrioventricular node, His bundle, bundle branches, and Purkinje fibers, were anatomically discovered more than 100 years ago, their molecular constituents and regulatory mechanisms remain incompletely understood. Here, we demonstrate the transcriptomic landscape of the postnatal mouse CCS at a single-cell resolution with spatial information. Integration of single-cell and spatial transcriptomics uncover region-specific markers and zonation patterns of expression. Network inference shows heterogeneous gene regulatory networks across the CCS. Notably, region-specific gene regulation is recapitulated in vitro using neonatal mouse atrial and ventricular myocytes overexpressing CCS-specific transcription factors, Tbx3 and/or Irx3. This finding is supported by ATAC-seq of different CCS regions, Tbx3 ChIP-seq, and Irx motifs. Overall, this study provides comprehensive molecular profiles of the postnatal CCS and elucidates gene regulatory mechanisms contributing to its heterogeneity.


Assuntos
Sistema de Condução Cardíaco , Proteínas de Homeodomínio , Miócitos Cardíacos , Proteínas com Domínio T , Animais , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Camundongos , Miócitos Cardíacos/metabolismo , Sistema de Condução Cardíaco/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Redes Reguladoras de Genes , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Regulação da Expressão Gênica , Animais Recém-Nascidos , Análise de Célula Única , Transcriptoma , Ramos Subendocárdicos/metabolismo , Ramos Subendocárdicos/fisiologia , Nó Atrioventricular/metabolismo , Nó Sinoatrial/metabolismo , Fascículo Atrioventricular/metabolismo
3.
Geroscience ; 46(5): 5085-5101, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-38967697

RESUMO

Aging is a major risk factor for sinoatrial node (SAN) dysfunction, which can impair heart rate (HR) control and heart rate variability (HRV). HR and HRV are determined by intrinsic SAN function and its regulation by the autonomic nervous system (ANS). The purpose of this study was to use multi-scale multi-fractal detrended fluctuation analysis (MSMFDFA; a complexity-based approach to analyze multi-fractal dynamics) to longitudinally assess changes in multi-fractal HRV properties and SAN function in ECG time series recorded repeatedly across the full adult lifespan in mice. ECGs were recorded in anesthetized mice in baseline conditions and after autonomic nervous system blockade every three months beginning at 6 months of age until the end of life. MSMFDFA was used to assess HRV and SAN function every three months between 6 and 27 months of age. Intrinsic HR (i.e. HR during ANS blockade) remained relatively stable until 15 months of age, and then progressively declined until study endpoint at 27 months of age. MSMFDFA revealed sudden and rapid changes in multi-fractal properties of the ECG RR interval time series in aging mice. In particular, multi-fractal spectrum width (MFSW, a measure of multi-fractality) was relatively stable between 6 months and 15 months of age and then progressively increased at 27 months of age. These changes in MFSW were evident in baseline conditions and during ANS blockade. Thus, intrinsic SAN function declines progressively during aging and is manifested by age-associated changes in multi-fractal HRV across the lifespan in mice, which can be accurately quantified by MSMFDFA.


Assuntos
Envelhecimento , Sistema Nervoso Autônomo , Eletrocardiografia , Frequência Cardíaca , Nó Sinoatrial , Animais , Frequência Cardíaca/fisiologia , Nó Sinoatrial/fisiopatologia , Nó Sinoatrial/fisiologia , Envelhecimento/fisiologia , Sistema Nervoso Autônomo/fisiopatologia , Sistema Nervoso Autônomo/fisiologia , Camundongos , Masculino , Camundongos Endogâmicos C57BL , Estudos Longitudinais
4.
J Gen Physiol ; 156(9)2024 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-39037413

RESUMO

The heartbeat originates from spontaneous action potentials in specialized pacemaker cells within the sinoatrial node (SAN) of the right atrium. Voltage-gated potassium channels in SAN myocytes mediate outward K+ currents that regulate cardiac pacemaking by controlling action potential repolarization, influencing the time between heartbeats. Gene expression studies have identified transcripts for many types of voltage-gated potassium channels in the SAN, but most remain of unknown functional significance. One such gene is Kcna1, which encodes epilepsy-associated voltage-gated Kv1.1 K+ channel α-subunits that are important for regulating action potential firing in neurons and cardiomyocytes. Here, we investigated the functional contribution of Kv1.1 to cardiac pacemaking at the whole heart, SAN, and SAN myocyte levels by performing Langendorff-perfused isolated heart preparations, multielectrode array recordings, patch clamp electrophysiology, and immunocytochemistry using Kcna1 knockout (KO) and wild-type (WT) mice. Our results showed that either genetic or pharmacological ablation of Kv1.1 significantly decreased the SAN firing rate, primarily by impairing SAN myocyte action potential repolarization. Voltage-clamp electrophysiology and immunocytochemistry revealed that Kv1.1 exerts its effects despite contributing only a small outward K+ current component, which we term IKv1.1, and despite apparently being present in low abundance at the protein level in SAN myocytes. These findings establish Kv1.1 as the first identified member of the Kv1 channel family to play a role in sinoatrial function, thereby rendering it a potential candidate and therapeutic targeting of sinus node dysfunction. Furthermore, our results demonstrate that small currents generated via low-abundance channels can still have significant impacts on cardiac pacemaking.


Assuntos
Potenciais de Ação , Canal de Potássio Kv1.1 , Miócitos Cardíacos , Nó Sinoatrial , Animais , Canal de Potássio Kv1.1/metabolismo , Canal de Potássio Kv1.1/genética , Camundongos , Nó Sinoatrial/metabolismo , Nó Sinoatrial/fisiologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/fisiologia , Camundongos Knockout , Masculino , Camundongos Endogâmicos C57BL
6.
BMC Anesthesiol ; 24(1): 263, 2024 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-39085782

RESUMO

BACKGROUND: Dexmedetomidine and propofol are common sedatives in intensive care units and for interventional procedures. Both may compromise sinus node function and atrioventricular conduction. The objective of this prospective, randomized study is to compare the effect of dexmedetomidine with propofol on sinus node function and atrioventricular conduction. METHODS: In a tertiary care center in Switzerland we included from September 2019 to October 2020 160 patients (65 ± 11 years old; 32% female) undergoing first ablation for atrial fibrillation by cryoballoon ablation or by radiofrequency ablation. Patients were randomly assigned to deep sedation with dexmedetomidine (DEX group) versus propofol (PRO group). A standard electrophysiological study was performed after pulmonary vein isolation with the patients still deeply sedated and hemodynamically stable. RESULTS: Eighty patients each were randomized to the DEX and PRO group. DEX group patients had higher baseline sinus cycle length (1022 vs. 1138 ms; p = 0.003) and longer sinus node recovery time (SNRT400; 1597 vs. 1412 ms; p = 0.042). However, both corrected SNRT and normalized SNRT did not differ. DEX group patients had longer PR interval (207 vs. 186 ms; p = 0.002) and AH interval (111 vs. 95 ms, p = 0.008), longer Wenckebach cycle length of the atrioventricular node (512 vs. 456 ms; p = 0.005), and longer atrioventricular node effective refractory period (390 vs. 344 ms; p = 0.009). QRS width and HV interval were not different. An arrhythmia, mainly atrial fibrillation, was induced in 33 patients during the electrophysiological study, without differences among groups (20% vs. 15%, p = 0.533). CONCLUSIONS: Dexmedetomidine has a more pronounced slowing effect on sinus rate and suprahissian AV conduction than propofol, but not on infrahissian AV conduction and ventricular repolarization. These differences need to be taken into account when using these sedatives. TRIAL REGISTRATION: ClinicalTrials.gov number NCT03844841, 19/02/2019.


Assuntos
Fibrilação Atrial , Sedação Profunda , Dexmedetomidina , Hipnóticos e Sedativos , Propofol , Humanos , Dexmedetomidina/farmacologia , Dexmedetomidina/administração & dosagem , Propofol/administração & dosagem , Propofol/farmacologia , Feminino , Masculino , Estudos Prospectivos , Hipnóticos e Sedativos/administração & dosagem , Hipnóticos e Sedativos/farmacologia , Idoso , Fibrilação Atrial/cirurgia , Fibrilação Atrial/fisiopatologia , Fibrilação Atrial/tratamento farmacológico , Pessoa de Meia-Idade , Sedação Profunda/métodos , Nó Sinoatrial/efeitos dos fármacos
8.
Int J Mol Sci ; 25(12)2024 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-38928314

RESUMO

A large diversity of epigenetic factors, such as microRNAs and histones modifications, are known to be capable of regulating gene expression without altering DNA sequence itself. In particular, miR-1 is considered the first essential microRNA in cardiac development. In this study, miR-1 potential role in early cardiac chamber differentiation was analyzed through specific signaling pathways. For this, we performed in chick embryos functional experiments by means of miR-1 microinjections into the posterior cardiac precursors-of both primitive endocardial tubes-committed to sinoatrial region fates. Subsequently, embryos were subjected to whole mount in situ hybridization, immunohistochemistry and RT-qPCR analysis. As a relevant novelty, our results revealed that miR-1 increased Amhc1, Tbx5 and Gata4, while this microRNA diminished Mef2c and Cripto expressions during early differentiation of the cardiac sinoatrial region. Furthermore, we observed in this developmental context that miR-1 upregulated CrabpII and Rarß and downregulated CrabpI, which are three crucial factors in the retinoic acid signaling pathway. Interestingly, we also noticed that miR-1 directly interacted with Hdac4 and Calm1/Calmodulin, as well as with Erk2/Mapk1, which are three key factors actively involved in Mef2c regulation. Our study shows, for the first time, a key role of miR-1 as an epigenetic regulator in the early differentiation of the cardiac sinoatrial region through orchestrating opposite actions between retinoic acid and Mef2c, fundamental to properly assign cardiac cells to their respective heart chambers. A better understanding of those molecular mechanisms modulated by miR-1 will definitely help in fields applied to therapy and cardiac regeneration and repair.


Assuntos
Diferenciação Celular , Epigênese Genética , Regulação da Expressão Gênica no Desenvolvimento , MicroRNAs , Animais , MicroRNAs/genética , MicroRNAs/metabolismo , Diferenciação Celular/genética , Embrião de Galinha , Fatores de Transcrição MEF2/metabolismo , Fatores de Transcrição MEF2/genética , Nó Sinoatrial/metabolismo , Nó Sinoatrial/citologia , Transdução de Sinais , Coração/embriologia , Coração/fisiologia
9.
In Vitro Cell Dev Biol Anim ; 60(7): 815-823, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38898365

RESUMO

Sinoatrial node (SAN) is the pacemaker of the heart in charge of initiating spontaneous electronical activity and controlling heart rate. Myocytes from SAN can generate spontaneous rhythmic action potentials, which propagate through the myocardium, thereby triggering cardiac myocyte contraction. Acutely, the method for isolating sinoatrial node myocytes (SAMs) is critical in studying the protein expression and function of myocytes in SAN. Currently, the SAMs were isolated by transferring SAN tissue directly into the digestion solution, but it is difficult to judge the degree of digestion, and the system was unstable. Here, we present a modified protocol for the isolation of SAMs in mice, based on the collagenase II and protease perfusion of the heart using a Langendorff apparatus and subsequent dissociation of SAMs. The appearance and droplet flow rate of the heart could be significantly changed during enzymatic digestion via perfusion, which allowed us to easily judge the degree of digestion and avoid incomplete or excessive digestion. The SAMs with stable yield and viability achieved from our optimized approach would facilitate the follow-up experiments.


Assuntos
Separação Celular , Miócitos Cardíacos , Nó Sinoatrial , Animais , Nó Sinoatrial/citologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Separação Celular/métodos , Camundongos , Camundongos Endogâmicos C57BL , Masculino , Perfusão
10.
Sci Rep ; 14(1): 14041, 2024 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-38890395

RESUMO

The sinus node (SN) serves as the primary pacemaker of the heart and is the first component of the cardiac conduction system. Due to its anatomical properties and sample scarcity, the cellular composition of the human SN has been historically challenging to study. Here, we employed a novel deep learning deconvolution method, namely Bulk2space, to characterise the cellular heterogeneity of the human SN using existing single-cell datasets of non-human species. As a proof of principle, we used Bulk2Space to profile the cells of the bulk human right atrium using publicly available mouse scRNA-Seq data as a reference. 18 human cell populations were identified, with cardiac myocytes being the most abundant. Each identified cell population correlated to its published experimental counterpart. Subsequently, we applied the deconvolution to the bulk transcriptome of the human SN and identified 11 cell populations, including a population of pacemaker cardiomyocytes expressing pacemaking ion channels (HCN1, HCN4, CACNA1D) and transcription factors (SHOX2 and TBX3). The connective tissue of the SN was characterised by adipocyte and fibroblast populations, as well as key immune cells. Our work unravelled the unique single cell composition of the human SN by leveraging the power of a novel machine learning method.


Assuntos
Miócitos Cardíacos , Análise de Célula Única , Nó Sinoatrial , Humanos , Nó Sinoatrial/citologia , Nó Sinoatrial/metabolismo , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/citologia , Análise de Célula Única/métodos , Camundongos , Animais , Inteligência Artificial , Transcriptoma , Átrios do Coração/metabolismo , Átrios do Coração/citologia , Aprendizado Profundo
11.
Adv Exp Med Biol ; 1441: 185-200, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38884712

RESUMO

The electrical impulses that coordinate the sequential, rhythmic contractions of the atria and ventricles are initiated and tightly regulated by the specialized tissues of the cardiac conduction system. In the mature heart, these impulses are generated by the pacemaker cardiomyocytes of the sinoatrial node, propagated through the atria to the atrioventricular node where they are delayed and then rapidly propagated to the atrioventricular bundle, right and left bundle branches, and finally, the peripheral ventricular conduction system. Each of these specialized components arise by complex patterning events during embryonic development. This chapter addresses the origins and transcriptional networks and signaling pathways that drive the development and maintain the function of the cardiac conduction system.


Assuntos
Sistema de Condução Cardíaco , Animais , Humanos , Nó Atrioventricular/fisiologia , Nó Atrioventricular/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Sistema de Condução Cardíaco/fisiologia , Miócitos Cardíacos/fisiologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/citologia , Transdução de Sinais , Nó Sinoatrial/fisiologia , Nó Sinoatrial/embriologia
12.
Expert Opin Ther Targets ; 28(5): 385-400, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38700451

RESUMO

INTRODUCTION: The cardiac conduction system (CCS) is crucial for maintaining adequate cardiac frequency at rest and modulation during exercise. Furthermore, the atrioventricular node and His-Purkinje system are essential for maintaining atrioventricular and interventricular synchrony and consequently maintaining an adequate cardiac output. AREAS COVERED: In this review article, we examine the anatomy, physiology, and pathophysiology of the CCS. We then discuss in detail the most common genetic mutations and the molecular mechanisms of cardiac conduction disease (CCD) and provide our perspectives on future research and therapeutic opportunities in this field. EXPERT OPINION: Significant advancement has been made in understanding the molecular mechanisms of CCD, including the recognition of the heterogeneous signaling at the subcellular levels of sinoatrial node, the involvement of inflammatory and autoimmune mechanisms, and the potential impact of epigenetic regulations on CCD. However, the current treatment of CCD manifested as bradycardia still relies primarily on cardiovascular implantable electronic devices (CIEDs). On the other hand, an If specific inhibitor was developed to treat inappropriate sinus tachycardia and sinus tachycardia in heart failure patients with reduced ejection fraction. More work is needed to translate current knowledge into pharmacologic or genetic interventions for the management of CCDs.


Assuntos
Doença do Sistema de Condução Cardíaco , Sistema de Condução Cardíaco , Terapia de Alvo Molecular , Humanos , Animais , Sistema de Condução Cardíaco/fisiopatologia , Doença do Sistema de Condução Cardíaco/fisiopatologia , Doença do Sistema de Condução Cardíaco/terapia , Doença do Sistema de Condução Cardíaco/tratamento farmacológico , Mutação , Desenvolvimento de Medicamentos , Insuficiência Cardíaca/fisiopatologia , Insuficiência Cardíaca/terapia , Insuficiência Cardíaca/tratamento farmacológico , Epigênese Genética , Nó Sinoatrial/fisiopatologia
13.
J Cardiovasc Electrophysiol ; 35(7): 1393-1400, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38741382

RESUMO

INTRODUCTION: Inappropriate sinus tachycardia (IST) is a common condition with frequently not tolerated beta-blockers or ivabradine and a high rate of complication in ablation strategy; we describe an alternative anatomical approach of sinus node (SN) modulation. METHODS: This retrospective study describes a case series of 6 patients from two centers diagnosed with symptomatic IST undergoing SN ablation. RESULTS: The mean age was 40.6 ± 13.9 years; five of the six patients were female, 100% of patients reported heart palpitations, and 66% reported dizziness, the average heart rate (HR) on a 24-h Holter was 93.2 ± 7.9 bpm. HR during the first stage of a stress test using a standard Bruce protocol was 150 ± 70 bpm, The average HR on 24-h Holter postablation was 75 ± 5.6 bpm, the sinus rate HR during stage 1 of a Bruce protocol exercise stress test was 120 ± 10 bpm. CONCLUSION: This is the first case series reporting the acute and long-term results of a novel anatomical approach for SN modulation to treat IST targeting the arcuate ridge (AR) under intracardiac echography (ICE) guidance. The novel anatomic ICE-guided catheter ablation approach aimed to identify the earliest activation at the AR with an extension of RF lesions toward its septal region seems effective and safe to modulate the SN in symptomatic patients with IST refractory to medical treatment.


Assuntos
Ablação por Cateter , Frequência Cardíaca , Taquicardia Sinusal , Humanos , Feminino , Taquicardia Sinusal/cirurgia , Taquicardia Sinusal/fisiopatologia , Estudos Retrospectivos , Masculino , Pessoa de Meia-Idade , Adulto , Resultado do Tratamento , Potenciais de Ação , Valor Preditivo dos Testes , Antiarrítmicos/uso terapêutico , Fatores de Tempo , Ultrassonografia de Intervenção , Eletrocardiografia Ambulatorial , Resistência a Medicamentos , Nó Sinoatrial/cirurgia , Nó Sinoatrial/fisiopatologia , Ecocardiografia
14.
Circ Res ; 134(10): 1348-1378, 2024 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-38723033

RESUMO

Loss or dysregulation of the normally precise control of heart rate via the autonomic nervous system plays a critical role during the development and progression of cardiovascular disease-including ischemic heart disease, heart failure, and arrhythmias. While the clinical significance of regulating changes in heart rate, known as the chronotropic effect, is undeniable, the mechanisms controlling these changes remain not fully understood. Heart rate acceleration and deceleration are mediated by increasing or decreasing the spontaneous firing rate of pacemaker cells in the sinoatrial node. During the transition from rest to activity, sympathetic neurons stimulate these cells by activating ß-adrenergic receptors and increasing intracellular cyclic adenosine monophosphate. The same signal transduction pathway is targeted by positive chronotropic drugs such as norepinephrine and dobutamine, which are used in the treatment of cardiogenic shock and severe heart failure. The cyclic adenosine monophosphate-sensitive hyperpolarization-activated current (If) in pacemaker cells is passed by hyperpolarization-activated cyclic nucleotide-gated cation channels and is critical for generating the autonomous heartbeat. In addition, this current has been suggested to play a central role in the chronotropic effect. Recent studies demonstrate that cyclic adenosine monophosphate-dependent regulation of HCN4 (hyperpolarization-activated cyclic nucleotide-gated cation channel isoform 4) acts to stabilize the heart rate, particularly during rapid rate transitions induced by the autonomic nervous system. The mechanism is based on creating a balance between firing and recently discovered nonfiring pacemaker cells in the sinoatrial node. In this way, hyperpolarization-activated cyclic nucleotide-gated cation channels may protect the heart from sinoatrial node dysfunction, secondary arrhythmia of the atria, and potentially fatal tachyarrhythmia of the ventricles. Here, we review the latest findings on sinoatrial node automaticity and discuss the physiological and pathophysiological role of HCN pacemaker channels in the chronotropic response and beyond.


Assuntos
Frequência Cardíaca , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização , Nó Sinoatrial , Humanos , Animais , Nó Sinoatrial/metabolismo , Nó Sinoatrial/fisiopatologia , Nó Sinoatrial/fisiologia , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/metabolismo , Relógios Biológicos
15.
BMC Cardiovasc Disord ; 24(1): 217, 2024 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-38643100

RESUMO

BACKGROUND: During normal sinus rhythm, atrial depolarization is conducted from right atrium to left atrium through Bachmann's bundle, and a normal P wave axis which is measured on the frontal plane is between 0º and + 75º. The change of P wave polarity is helpful for the analysis of origin point. CASE PRESENTATION: We report a patient with negative P wave in lead I. The characteristics of QRS complex in leads V1 to V6 are helpful to preliminarily differential diagnosis. The 12-lead electrocardiogram (ECG) with correct limb leads (right arm-left arm) placement shows sinus rhythm with complete right bundle branch block (RBBB). CONCLUSIONS: The change of P wave polarity as well as characteristics of QRS complex can help identify limb-lead reversals.


Assuntos
Bloqueio de Ramo , Eletrocardiografia , Humanos , Bloqueio de Ramo/diagnóstico , Nó Sinoatrial , Átrios do Coração , Nó Atrioventricular
16.
Cardiovasc Res ; 120(8): 927-942, 2024 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-38661182

RESUMO

AIMS: In patients with heart failure (HF), concomitant sinus node dysfunction (SND) is an important predictor of mortality, yet its molecular underpinnings are poorly understood. Using proteomics, this study aimed to dissect the protein and phosphorylation remodelling within the sinus node in an animal model of HF with concurrent SND. METHODS AND RESULTS: We acquired deep sinus node proteomes and phosphoproteomes in mice with heart failure and SND and report extensive remodelling. Intersecting the measured (phospho)proteome changes with human genomics pharmacovigilance data, highlighted downregulated proteins involved in electrical activity such as the pacemaker ion channel, Hcn4. We confirmed the importance of ion channel downregulation for sinus node physiology using computer modelling. Guided by the proteomics data, we hypothesized that an inflammatory response may drive the electrophysiological remodeling underlying SND in heart failure. In support of this, experimentally induced inflammation downregulated Hcn4 and slowed pacemaking in the isolated sinus node. From the proteomics data we identified proinflammatory cytokine-like protein galectin-3 as a potential target to mitigate the effect. Indeed, in vivo suppression of galectin-3 in the animal model of heart failure prevented SND. CONCLUSION: Collectively, we outline the protein and phosphorylation remodeling of SND in heart failure, we highlight a role for inflammation in electrophysiological remodelling of the sinus node, and we present galectin-3 signalling as a target to ameliorate SND in heart failure.


Assuntos
Modelos Animais de Doenças , Insuficiência Cardíaca , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização , Camundongos Endogâmicos C57BL , Proteômica , Síndrome do Nó Sinusal , Nó Sinoatrial , Animais , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/fisiopatologia , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/patologia , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/metabolismo , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/genética , Nó Sinoatrial/metabolismo , Nó Sinoatrial/fisiopatologia , Fosforilação , Síndrome do Nó Sinusal/metabolismo , Síndrome do Nó Sinusal/fisiopatologia , Síndrome do Nó Sinusal/genética , Masculino , Mediadores da Inflamação/metabolismo , Inflamação/metabolismo , Inflamação/fisiopatologia , Inflamação/patologia , Frequência Cardíaca , Canais de Potássio/metabolismo , Canais de Potássio/genética , Simulação por Computador , Modelos Cardiovasculares , Humanos , Transdução de Sinais , Potenciais de Ação
17.
J Vet Intern Med ; 38(3): 1305-1324, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38682817

RESUMO

BACKGROUND: Sinus node dysfunction because of abnormal impulse generation or sinoatrial conduction block causes bradycardia that can be difficult to differentiate from high parasympathetic/low sympathetic modulation (HP/LSM). HYPOTHESIS: Beat-to-beat relationships of sinus node dysfunction are quantifiably distinguishable by Poincaré plots, machine learning, and 3-dimensional density grid analysis. Moreover, computer modeling establishes sinoatrial conduction block as a mechanism. ANIMALS: Three groups of dogs were studied with a diagnosis of: (1) balanced autonomic modulation (n = 26), (2) HP/LSM (n = 26), and (3) sinus node dysfunction (n = 21). METHODS: Heart rate parameters and Poincaré plot data were determined [median (25%-75%)]. Recordings were randomly assigned to training or testing. Supervised machine learning of the training data was evaluated with the testing data. The computer model included impulse rate, exit block probability, and HP/LSM. RESULTS: Confusion matrices illustrated the effectiveness in diagnosing by both machine learning and Poincaré density grid. Sinus pauses >2 s differentiated (P < .0001) HP/LSM (2340; 583-3947 s) from sinus node dysfunction (8503; 7078-10 050 s), but average heart rate did not. The shortest linear intervals were longer with sinus node dysfunction (315; 278-323 ms) vs HP/LSM (260; 251-292 ms; P = .008), but the longest linear intervals were shorter with sinus node dysfunction (620; 565-698 ms) vs HP/LSM (843; 799-888 ms; P < .0001). CONCLUSIONS: Number and duration of pauses, not heart rate, differentiated sinus node dysfunction from HP/LSM. Machine learning and Poincaré density grid can accurately identify sinus node dysfunction. Computer modeling supports sinoatrial conduction block as a mechanism of sinus node dysfunction.


Assuntos
Doenças do Cão , Frequência Cardíaca , Aprendizado de Máquina , Animais , Cães , Doenças do Cão/diagnóstico , Doenças do Cão/fisiopatologia , Frequência Cardíaca/fisiologia , Bloqueio Sinoatrial/veterinária , Bloqueio Sinoatrial/diagnóstico , Bloqueio Sinoatrial/fisiopatologia , Masculino , Feminino , Nó Sinoatrial/fisiopatologia , Síndrome do Nó Sinusal/veterinária , Síndrome do Nó Sinusal/diagnóstico , Síndrome do Nó Sinusal/fisiopatologia , Eletrocardiografia/veterinária
18.
Exp Neurol ; 376: 114773, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38599368

RESUMO

BACKGROUND: Arrhythmia is the most common cardiac complication after ischemic stroke. Connexin 40 is the staple component of gap junctions, which influences the propagation of cardiac electrical signals in the sinoatrial node. However, the role of connexin 40 in post-stroke arrhythmia remains unclear. METHODS: In this study, a permanent middle cerebral artery occlusion model was used to simulate the occurrence of an ischemic stroke. Subsequently, an electrocardiogram was utilized to record and assess variations in electrocardiogram measures. In addition, optical tissue clearing and whole-mount immunofluorescence staining were used to confirm the anatomical localization of the sinoatrial node, and the sinoatrial node tissue was collected for RNA sequencing to screen for potential pathological mechanisms. Lastly, the rAAV9-Gja5 virus was injected with ultrasound guidance into the heart to increase Cx40 expression in the sinoatrial node. RESULTS: We demonstrated that the mice suffering from a permanent middle cerebral artery occlusion displayed significant arrhythmia, including atrial fibrillation, premature ventricular contractions, atrioventricular block, and abnormal electrocardiogram parameters. Of note, we observed a decrease in connexin 40 expression within the sinoatrial node after the ischemic stroke via RNA sequencing and western blot. Furthermore, rAAV9-Gja5 treatment ameliorated the occurrence of arrhythmia following stroke. CONCLUSIONS: In conclusion, decreased connexin 40 expression in the sinoatrial node contributed to the ischemic stroke-induced cardiac arrhythmia. Therefore, enhancing connexin 40 expression holds promise as a potential therapeutic approach for ischemic stroke-induced arrhythmia.


Assuntos
Arritmias Cardíacas , Proteína alfa-5 de Junções Comunicantes , AVC Isquêmico , Nó Sinoatrial , Animais , Camundongos , Arritmias Cardíacas/etiologia , Arritmias Cardíacas/genética , Conexinas/genética , Conexinas/metabolismo , Proteína alfa-5 de Junções Comunicantes/genética , Proteína alfa-5 de Junções Comunicantes/metabolismo , AVC Isquêmico/complicações , AVC Isquêmico/genética , AVC Isquêmico/metabolismo , AVC Isquêmico/patologia , Camundongos Endogâmicos C57BL , Nó Sinoatrial/metabolismo , Nó Sinoatrial/patologia
19.
Int J Mol Sci ; 25(6)2024 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-38542361

RESUMO

Human-induced pluripotent stem cell (hiPSC)-derived cardiomyocytes raise the possibility of generating pluripotent stem cells from a wide range of human diseases. In the cardiology field, hiPSCs have been used to address the mechanistic bases of primary arrhythmias and in investigations of drug safety. These studies have been focused primarily on atrial and ventricular pathologies. Consequently, many hiPSC-based cardiac differentiation protocols have been developed to differentiate between atrial- or ventricular-like cardiomyocytes. Few protocols have successfully proposed ways to obtain hiPSC-derived cardiac pacemaker cells, despite the very limited availability of human tissues from the sinoatrial node. Providing an in vitro source of pacemaker-like cells would be of paramount importance in terms of furthering our understanding of the mechanisms underlying sinoatrial node pathophysiology and testing innovative clinical strategies against sinoatrial node dysfunction (i.e., biological pacemakers and genetic- and pharmacological- based therapy). Here, we summarize and detail the currently available protocols used to obtain patient-derived pacemaker-like cells.


Assuntos
Células-Tronco Pluripotentes Induzidas , Células-Tronco Pluripotentes , Humanos , Miócitos Cardíacos , Diferenciação Celular/fisiologia , Nó Sinoatrial
20.
Commun Biol ; 7(1): 385, 2024 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-38553636

RESUMO

Shox2 plays a vital role in the morphogenesis and physiological function of the sinoatrial node (SAN), the primary cardiac pacemaker, manifested by the formation of a hypoplastic SAN and failed differentiation of pacemaker cells in Shox2 mutants. Shox2 and Nkx2-5 are co-expressed in the developing SAN and regulate the fate of the pacemaker cells through a Shox2-Nkx2-5 antagonistic mechanism. Here we show that simultaneous inactivation of Nkx2-5 in the SAN of Shox2 mutants (dKO) rescued the pacemaking cell fate but not the hypoplastic defects, indicating uncoupling of SAN cell fate determination and morphogenesis. Single-cell RNA-seq revealed that the presumptive SAN cells of Shox2-/- mutants failed to activate pacemaking program but remained in a progenitor state preceding working myocardium, while both wildtype and dKO SAN cells displayed normal pacemaking cell fate with similar cellular state. Shox2 thus acts as a safeguard but not a determinant to ensure the pacemaking cell fate through the Shox2-Nkx2-5 antagonistic mechanism, which is segregated from its morphogenetic regulatory function in SAN development.


Assuntos
Proteínas de Homeodomínio , Nó Sinoatrial , Proteínas de Homeodomínio/metabolismo , Nó Sinoatrial/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Miócitos Cardíacos/metabolismo , Morfogênese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA