Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Pflugers Arch ; 476(2): 229-242, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38036776

RESUMEN

Loss-of-function variants of SCN5A, encoding the sodium channel alpha subunit Nav1.5 are associated with high phenotypic variability and multiple cardiac presentations, while underlying mechanisms are incompletely understood. Here we investigated a family with individuals affected by Brugada Syndrome (BrS) of different severity and aimed to unravel the underlying genetic and electrophysiological basis.Next-generation sequencing was used to identify the genetic variants carried by family members. The index patient, who was severely affected by arrhythmogenic BrS, carried previously uncharacterized variants of Nav1.5 (SCN5A-G1661R) and glycerol-3-phosphate dehydrogenase-1-like protein (GPD1L-A306del) in a double heterozygous conformation. Family members exclusively carrying SCN5A-G1661R showed asymptomatic Brugada ECG patterns, while another patient solely carrying GPD1L-A306del lacked any clinical phenotype.To assess functional mechanisms, Nav1.5 channels were transiently expressed in HEK-293 cells in the presence and absence of GPD1L. Whole-cell patch-clamp recordings revealed loss of sodium currents after homozygous expression of SCN5A-G1661R, and reduction of current amplitude to ~ 50% in cells transfected with equal amounts of wildtype and mutant Nav1.5. Co-expression of wildtype Nav1.5 and GPD1L showed a trend towards increased sodium current amplitudes and a hyperpolarizing shift in steady-state activation and -inactivation compared to sole SCN5A expression. Application of the GPD1L-A306del variant shifted steady-state activation to more hyperpolarized and inactivation to more depolarized potentials.In conclusion, SCN5A-G1661R produces dysfunctional channels and associates with BrS. SCN5A mediated currents are modulated by co-expression of GDP1L and this interaction is altered by mutations in both proteins. Thus, additive genetic burden may aggravate disease severity, explaining higher arrhythmogenicity in double mutation carriers.


Asunto(s)
Síndrome de Brugada , Humanos , Síndrome de Brugada/genética , Síndrome de Brugada/metabolismo , Sodio/metabolismo , Células HEK293 , Mutación , Fenotipo , Canal de Sodio Activado por Voltaje NAV1.5/genética , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo
2.
Int J Mol Sci ; 23(13)2022 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-35806319

RESUMEN

Current protocols for the differentiation of human-induced pluripotent stem cells (hiPSC) into cardiomyocytes only generate a small amount of cardiac pacemaker cells. In previous work, we reported the generation of high amounts of cardiac pacemaker cells by co-culturing hiPSC with mouse visceral endoderm-like (END2) cells. However, potential medical applications of cardiac pacemaker cells generated according to this protocol, comprise an incalculable xenogeneic risk. We thus aimed to establish novel protocols maintaining the differentiation efficiency of the END2 cell-based protocol, yet eliminating the use of END2 cells. Three protocols were based on the activation and inhibition of the Wingless/Integrated (Wnt) signaling pathway, supplemented either with retinoic acid and the Wnt activator CHIR99021 (protocol B) or with the NODAL inhibitor SB431542 (protocol C) or with a combination of all three components (protocol D). An additional fourth protocol (protocol E) was used, which was originally developed by the manufacturer STEMCELL Technologies for the differentiation of hiPSC or hESC into atrial cardiomyocytes. All protocols (B, C, D, E) were compared to the END2 cell-based protocol A, serving as reference, in terms of their ability to differentiate hiPSC into cardiac pacemaker cells. Our analysis revealed that protocol E induced upregulation of 12 out of 15 cardiac pacemaker-specific genes. For comparison, reference protocol A upregulated 11, while protocols B, C and D upregulated 9, 10 and 8 cardiac pacemaker-specific genes, respectively. Cells differentiated according to protocol E displayed intense fluorescence signals of cardiac pacemaker-specific markers and showed excellent rate responsiveness to adrenergic and cholinergic stimulation. In conclusion, we characterized four novel and END2 cell-independent protocols for the differentiation of hiPSC into cardiac pacemaker cells, of which protocol E was the most efficient.


Asunto(s)
Células Madre Pluripotentes Inducidas , Animales , Diferenciación Celular , Línea Celular , Humanos , Ratones , Miocitos Cardíacos/metabolismo , Nodo Sinoatrial
3.
World J Stem Cells ; 12(10): 1133-1151, 2020 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-33178397

RESUMEN

BACKGROUND: Mesenchymal stem cells (MSC) modified by gene transfer to express cardiac pacemaker channels such as HCN2 or HCN4 were shown to elicit pacemaker function after intracardiac transplantation in experimental animal models. Human MSC derived from adipose tissue (haMSC) differentiate into cells with pacemaker properties in vitro, but little is known about their behavior after intracardiac transplantation. AIM: To investigate whether haMSC elicit biological pacemaker function in vivo after transplantation into pig hearts. METHODS: haMSC under native conditions (nhaMSC) or after pre-conditioning by medium differentiation (dhaMSC) (n = 6 pigs each, 5 × 106 cells/animal) were injected into the porcine left ventricular free wall. Animals receiving PBS injection served as controls (n = 6). Four weeks later, total atrioventricular (AV)-block was induced by radiofrequency catheter ablation, and electronic pacemaker devices were implanted for backup stimulation and heart rate monitoring. Ventricular rate and rhythm of pigs were evaluated during a follow-up of 15 d post ablation by 12-lead-ECG with heart rate assessment, 24-h continuous rate monitoring recorded by electronic pacemaker, assessment of escape recovery time, and pharmacological challenge to address catecholaminergic rate response. Finally, hearts were analyzed by histological and immunohistochemical investigations. RESULTS: In vivo transplantation of dhaMSC into the left ventricular free wall of pigs elicited spontaneous and regular rhythms that were pace-mapped to ventricular injection sites (mean heart rate 72.2 ± 3.6 bpm; n = 6) after experimental total AV block. Ventricular rhythms were stably detected over a 15-d period and were sensitive to catecholaminergic stimulation (mean maximum heart rate 131.0 ± 6.2 bpm; n = 6; P < 0.001). Pigs, which received nhaMSC or PBS presented significantly lower ventricular rates (mean heart rates 47.2 ± 2.5 bpm and 37.4 ± 3.2 bpm, respectively; n = 6 each; P < 0.001) and exhibited little sensitivity towards catecholaminergic stimulation (mean maximum heart rates 76.4 ± 3.1 bpm and 60.5 ± 3.1 bpm, respectively; n = 6 each; P < 0.05). Histological and immunohistochemical evaluation of hearts treated with dhaMSC revealed local clusters of transplanted cells at the injection sites that lacked macrophage or lymphocyte infiltrations or tumor formation. Intense fluorescence signals at these sites indicated membrane expression of HCN4 and other pacemaker-specific proteins involved in cardiac automaticity and impulse propagation. CONCLUSION: dhaMSC transplanted into pig left ventricles sustainably induced rate-responsive ventricular pacemaker activity after in vivo engraftment for four weeks. The data suggest that pre-conditioned MSC may further differentiate along a pacemaker-related lineage after myocardial integration and may establish superior pacemaker properties in vivo.

4.
Cell Transplant ; 29: 963689720914236, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32207339

RESUMEN

Engraftment and functional integration of stem cells or stem cell-derived cells within cardiac tissue is an important prerequisite for cell replacement therapy aiming at the treatment of heart disease. Recently, a novel intravenous approach for application of mesenchymal stromal cells (MSCs) to cardiac sites has been established using radiofrequency catheter ablation (RFCA)-guided targeting, bypassing the need for open chest surgery or direct myocardial cell injection. However, little is known about the quantitative efficacy and longevity of this strategy. We performed selective power-controlled RFCA with eight ablation pulses (30 W, 60 s each) to induce heat-mediated lesions at the right atrial appendices (RAAs) of pigs. Different concentrations of human bone marrow-derived MSCs (105 to 1.6 × 106 cells/kg bodyweight) labeled with superparamagnetic iron oxide (SPIO) particles were infused intravenously in nine pigs one d after RFCA treatment and hearts were explanted 8 d later to quantify the number of engrafted cells. Prussian blue staining revealed high numbers of SPIO-labeled cells in areas surrounding the RFCA-induced lesions. Cell numbers were evaluated by quantitative real-time polymerase chain reaction using specific primers for human MSCs (hMSCs), which indicated that up to 106 hMSCs, corresponding to ∼3.9% of the systemically applied human cells, engrafted within the RAAs of RFCA-treated pigs. Of note, infused hMSCs were observed in nontargeted organs, as well, but appeared at very low concentrations. To assess long-term deposition of MSCs, RAAs of three pigs were analyzed after 6 months, which revealed few persisting hMSCs at targeted sites. RFCA-mediated targeting of MSCs provides a novel minimal invasive strategy for cardiac stem cell engraftment. Qualitative and quantitative results of our large animal experiments indicate an efficient guidance of MSCs to selected cardiac regions, although only few cells remained at targeted sites 6 mo after cell transplantation.


Asunto(s)
Ablación por Catéter , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Miocitos Cardíacos/citología , Animales , Ablación por Catéter/métodos , Rastreo Celular/métodos , Imagen por Resonancia Magnética/métodos , Trasplante de Células Madre Mesenquimatosas/métodos , Coloración y Etiquetado , Porcinos
5.
Biochem Biophys Res Commun ; 519(1): 141-147, 2019 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-31481236

RESUMEN

Atrial fibrillation (AF) is the most frequent sustained arrhythmia and can lead to structural cardiac changes, known as tachycardia-induced cardiomyopathy (TIC). HCN4 is implicated in spontaneous excitation of the sinoatrial node, while channel dysfunction has been associated with sinus bradycardia, AF and structural heart disease. We here asked whether HCN4 mutations may contribute to the development of TIC, as well. Mutation scanning of HCN4 in 60 independent patients with AF and suspected TIC followed by panel sequencing in carriers of HCN4 variants identified the HCN4 variant P883R [minor allele frequency (MAF): 0,88%], together with the KCNE1 variant S38G (MAF: 65%) in three unrelated patients. Family histories revealed additional cases of AF, sudden cardiac death and cardiomyopathy. Patch-clamp recordings of HCN4-P883R channels expressed in HEK293 cells showed remarkable alterations of channel properties shifting the half-maximal activation voltage to more depolarized potentials, while channel deactivation was faster compared to wild-type (WT). Co-transfection of WT and mutant subunits, resembling the heterozygous cellular situation of our patients, revealed significantly higher current densities compared to WT. In conclusion HCN4-P883R may increase ectopic trigger and maintenance of AF by shifting the activation voltage of If to more positive potentials and producing higher current density. Together with the common KCNE1 variant S38G, previously proposed as a genetic modifier of AF, HCN4-P883R may provide a substrate for the development of AF and TIC.


Asunto(s)
Fibrilación Atrial/genética , Genes Modificadores , Predisposición Genética a la Enfermedad , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Proteínas Musculares/genética , Mutación/genética , Canales de Potasio/genética , Secuencia de Aminoácidos , Femenino , Pruebas Genéticas , Células HEK293 , Humanos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/química , Activación del Canal Iónico , Masculino , Proteínas Musculares/química , Linaje , Canales de Potasio/química
6.
Nat Commun ; 10(1): 3295, 2019 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-31337768

RESUMEN

HCN channels underlie the depolarizing funny current (If) that contributes importantly to cardiac pacemaking. If is upregulated in failing and infarcted hearts, but its implication in disease mechanisms remained unresolved. We generated transgenic mice (HCN4tg/wt) to assess functional consequences of HCN4 overexpression-mediated If increase in cardiomyocytes to levels observed in human heart failure. HCN4tg/wt animals exhibit a dilated cardiomyopathy phenotype with increased cellular arrhythmogenicity but unchanged heart rate and conduction parameters. If augmentation induces a diastolic Na+ influx shifting the Na+/Ca2+ exchanger equilibrium towards 'reverse mode' leading to increased [Ca2+]i. Changed Ca2+ homeostasis results in significantly higher systolic [Ca2+]i transients and stimulates apoptosis. Pharmacological inhibition of If prevents the rise of [Ca2+]i and protects from ventricular remodeling. Here we report that augmented myocardial If alters intracellular Ca2+ homeostasis leading to structural cardiac changes and increased arrhythmogenicity. Inhibition of myocardial If per se may constitute a therapeutic mechanism to prevent cardiomyopathy.


Asunto(s)
Calcio/metabolismo , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/fisiología , Proteínas Musculares/fisiología , Canales de Potasio/fisiología , Animales , Apoptosis , Electrofisiología Cardíaca , Perfilación de la Expresión Génica , Corazón/fisiología , Homeostasis , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Ratones Transgénicos , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/metabolismo , Canales de Potasio/genética , Canales de Potasio/metabolismo , Troponina I/genética , Troponina I/metabolismo , Troponina I/fisiología
7.
Life Sci ; 232: 116620, 2019 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-31291594

RESUMEN

AIMS: Cell-based biological pacemakers aim to overcome limitations and side effects of electronic pacemaker devices. We here developed and tested different approaches to achieve nodal-type differentiation using human adipose- and bone marrow-derived mesenchymal stem cells (haMSC, hbMSC). MAIN METHODS: haMSC and hbMSC were differentiated using customized protocols. Quantitative RT-PCR was applied for transcriptional pacemaker-gene profiling. Protein membrane expression was analyzed by immunocytochemistry. Pacemaker current (If) was studied in haMSC with and without lentiviral HCN4-transduction using patch clamp recordings. Functional characteristics were evaluated by co-culturing with neonatal rat ventricular myocytes (NRVM). KEY FINDINGS: Culture media-based differentiation for two weeks generated cells with abundant transcription of ion channel genes (Cav1.2, NCX1), transcription factors (TBX3, TBX18, SHOX2) and connexins (Cx31.9 and Cx45) characteristic for cardiac pacemaker tissue, but lack adequate HCN transcription. haMSC-derived cells revealed transcript levels, which were closer related to sinoatrial nodal cells than hbMSC-derived cells. To substitute for the lack of If, we performed lentiviral HCN4-transduction of haMSC resulting in stable If. Co-culturing with NRVM demonstrated that differentiated haMSC expressing HCN4 showed earlier onset of spontaneous contractions and higher beating regularity, synchrony and rate compared to co-cultures with non-HCN4-transduced haMSC or HCN4-transduced, non-differentiated haMSC. Confocal imaging indicated increased membrane expression of cardiac gap junctional proteins in differentiated haMSC. SIGNIFICANCE: By differentiation haMSC, rather than hbMSC attain properties favorable for cardiac pacemaking. In combination with lentiviral HCN4-transduction, a cellular phenotype was generated that sustainably controls and stabilizes rate in co-culture with NRVM.


Asunto(s)
Relojes Biológicos/fisiología , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Proteínas Musculares/metabolismo , Canales de Potasio/metabolismo , Tejido Adiposo/fisiología , Animales , Células de la Médula Ósea/fisiología , Diferenciación Celular/fisiología , Técnicas de Cocultivo , Humanos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/fisiología , Células Madre Mesenquimatosas/metabolismo , Células Musculares/metabolismo , Proteínas Musculares/fisiología , Miocitos Cardíacos/metabolismo , Técnicas de Placa-Clamp , Canales de Potasio/fisiología , Ratas , Nodo Sinoatrial
8.
Biochem Biophys Res Commun ; 512(4): 845-851, 2019 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-30929919

RESUMEN

Pathogenic long QT mutations often comprise high phenotypic variability and particularly variants in ANK2 (long QT syndrome 4) frequently lack QT prolongation. We sought to elucidate the genetic and functional background underlying the clinical diversity in a 3-generation family with different cardiac arrhythmias. Next-generation sequencing-based screening of patients with QT prolongation identified the index patient of the family carrying an ANK2-E1813K variant and a previously uncharacterized KCNH2-H562R mutation in a double heterozygous conformation. The patient presented with a severe clinical phenotype including a markedly prolonged QTc interval (544 ms), recurrent syncope due to Torsade de Pointes tachycardias, survived cardiopulmonary resuscitation, progressive cardiac conduction defect, and atrial fibrillation. Evaluation of other family members identified a sister and a niece solely carrying the ANK2-E1813K variant, who showed age-related conduction disease. An asymptomatic second sister solely carried the KCNH2-H562R mutation. Voltage-clamp recordings in Xenopus oocytes revealed that KCNH2-H562R subunits were non-functional but did not exert dominant-negative effects on wild-type subunits. Expression of KCNH2-H562R in HEK293 cells showed a trafficking deficiency. Co-expression of the C-terminal regulatory domain of ANK2 in Xenopus oocytes revealed that ANK2-E1813K diminished currents mediated by the combination of wild-type and H562R KCNH2 subunits. Our data suggest that ANK2 functionally interacts with KCNH2 leading to a stronger current suppression and marked aggravation of long QT syndrome in the patient carrying variants in both proteins.


Asunto(s)
Ancirinas/genética , Canal de Potasio ERG1/genética , Síndrome de QT Prolongado/genética , Mutación , Adulto , Anciano , Animales , Ancirinas/metabolismo , Canal de Potasio ERG1/metabolismo , Femenino , Células HEK293 , Humanos , Síndrome de QT Prolongado/etiología , Masculino , Persona de Mediana Edad , Oocitos/metabolismo , Linaje , Xenopus laevis
9.
Stem Cell Res Ther ; 8(1): 229, 2017 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-29037217

RESUMEN

BACKGROUND: Human induced pluripotent stem cells (hiPSC) harbor the potential to differentiate into diverse cardiac cell types. Previous experimental efforts were primarily directed at the generation of hiPSC-derived cells with ventricular cardiomyocyte characteristics. Aiming at a straightforward approach for pacemaker cell modeling and replacement, we sought to selectively differentiate cells with nodal-type properties. METHODS: hiPSC were differentiated into spontaneously beating clusters by co-culturing with visceral endoderm-like cells in a serum-free medium. Subsequent culturing in a specified fetal bovine serum (FBS)-enriched cell medium produced a pacemaker-type phenotype that was studied in detail using quantitative real-time polymerase chain reaction (qRT-PCR), immunocytochemistry, and patch-clamp electrophysiology. Further investigations comprised pharmacological stimulations and co-culturing with neonatal cardiomyocytes. RESULTS: hiPSC co-cultured in a serum-free medium with the visceral endoderm-like cell line END-2 produced spontaneously beating clusters after 10-12 days of culture. The pacemaker-specific genes HCN4, TBX3, and TBX18 were abundantly expressed at this early developmental stage, while levels of sarcomeric gene products remained low. We observed that working-type cardiomyogenic differentiation can be suppressed by transfer of early clusters into a FBS-enriched cell medium immediately after beating onset. After 6 weeks under these conditions, sinoatrial node (SAN) hallmark genes remained at high levels, while working-type myocardial transcripts (NKX2.5, TBX5) were low. Clusters were characterized by regular activity and robust beating rates (70-90 beats/min) and were triggered by spontaneous Ca2+ transients recapitulating calcium clock properties of genuine pacemaker cells. They were responsive to adrenergic/cholinergic stimulation and able to pace neonatal rat ventricular myocytes in co-culture experiments. Action potential (AP) measurements of cells individualized from clusters exhibited nodal-type (63.4%) and atrial-type (36.6%) AP morphologies, while ventricular AP configurations were not observed. CONCLUSION: We provide a novel culture media-based, transgene-free approach for targeted generation of hiPSC-derived pacemaker-type cells that grow in clusters and offer the potential for disease modeling, drug testing, and individualized cell-based replacement therapy of the SAN.


Asunto(s)
Relojes Biológicos , Diferenciación Celular , Células Madre Pluripotentes Inducidas/citología , Miocitos Cardíacos/citología , Potenciales de Acción , Animales , Señalización del Calcio , Células Cultivadas , Proteína Homeótica Nkx-2.5/genética , Proteína Homeótica Nkx-2.5/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Contracción Miocárdica , Miocitos Cardíacos/clasificación , Ratas , Nodo Sinoatrial/citología , Nodo Sinoatrial/metabolismo , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo
10.
J Perianesth Nurs ; 32(3): 188-198, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28527546

RESUMEN

PURPOSE: Inadvertent perioperative hypothermia is a common problem for patients undergoing surgery. Heat redistribution from the body's core to the periphery after induction of anesthesia is the major contributor. DESIGN: A prospective randomized controlled trial was conducted to determine if reflective blankets are more effective than cotton blankets in reducing the core-peripheral temperature gradient and increasing peripheral compartment heat content during the preoperative phase among adult patients undergoing elective surgery of less than 1 hour. About 328 adult patients undergoing general anesthesia were randomly allocated into two groups. METHODS: Data were analyzed using independent t tests for continuous variables and chi-square tests for categorical variables. FINDINGS: There was a significantly smaller reduction in temporal artery/foot temperature gradient (1.13 vs 1.64°C, P < .001) and a significant increase in foot temperature (0.64 vs 0.11°C, P < .001) in the reflective blanket group. CONCLUSIONS: Reflective blankets are more effective than cotton blankets in warming patients' periphery and reducing core-peripheral temperature gradient preoperatively. AUSTRALIAN NEW ZEALAND CLINICAL TRIALS REGISTRY NUMBER: ACTRN12614000931673 (retrospective registration).


Asunto(s)
Hipotermia/terapia , Atención Perioperativa , Humanos , Nueva Gales del Sur , Estudios Prospectivos
12.
J Am Coll Cardiol ; 64(8): 757-67, 2014 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-25145518

RESUMEN

BACKGROUND: Inherited arrhythmias were originally considered isolated electrical defects. There is growing evidence that ion channel dysfunction also contributes to myocardial disorders, but genetic overlap has not been reported for sinus node dysfunction (SND) and noncompaction cardiomyopathy (NCCM). OBJECTIVES: The study sought to investigate a familial electromechanical disorder characterized by SND and NCCM, and to identify the underlying genetic basis. METHODS: The index family and a cohort of unrelated probands with sinus bradycardia were examined by electrocardiography, Holter recording, exercise stress test, echocardiography, and/or cardiac magnetic resonance imaging. Targeted next-generation and direct sequencing were used for candidate gene analysis and mutation scanning. Ion channels were expressed in HEK293 cells and studied using patch-clamp recordings. RESULTS: SND and biventricular NCCM were diagnosed in multiple members of a German family. Segregation analysis suggested autosomal-dominant inheritance of the combined phenotype. When looking for potentially disease-causing gene variants with cosegregation, a novel hyperpolarization-activated cyclic nucleotide channel 4 (HCN4)-G482R mutation and a common cysteine and glycine-rich protein 3 (CSRP3)-W4R variant were identified. HCN4-G482R is located in the highly conserved channel pore domain. Mutant subunits were nonfunctional and exerted dominant-negative effects on wild-type current. CSRP3-W4R has previously been linked to dilated and hypertrophic cardiomyopathy, but was also found in healthy subjects. Moreover, different truncation (695X) and missense (P883R) HCN4 mutations segregated with a similar combined phenotype in an additional, unrelated family and a single unrelated proband respectively, which both lacked CSRP3-W4R. CONCLUSIONS: The symptom complex of SND and NCCM is associated with heritable HCN4 defects. The NCCM phenotype may be aggravated by a common CSRP3 variant in one of the families.


Asunto(s)
Cardiopatías Congénitas/genética , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Proteínas Musculares/genética , Canales de Potasio/genética , Síndrome del Seno Enfermo/genética , Adolescente , Animales , Ecocardiografía , Técnicas Electrofisiológicas Cardíacas , Femenino , Alemania/epidemiología , Células HEK293 , Cardiopatías Congénitas/diagnóstico por imagen , Cardiopatías Congénitas/epidemiología , Humanos , Masculino , Potenciales de la Membrana , Persona de Mediana Edad , Linaje , Fenotipo , Prevalencia , Síndrome del Seno Enfermo/diagnóstico por imagen , Síndrome del Seno Enfermo/epidemiología , Síndrome , Adulto Joven
13.
Eur J Hum Genet ; 22(1): 119-25, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23695275

RESUMEN

So far, the role of mutations in the δ-sarcogylcan (Sgcd) gene in causing autosomal dominant dilated cardiomyopathy (DCM) remains inconclusive. A p.S151A missense mutation in exon 6 of the Sgcd gene was reported to cause severe isolated autosomal dominant DCM without affecting skeletal muscle. This is controversial to our previous findings in a large consanguineous family where this p.S151A mutation showed no relevance for cardiac disease. In this study, the potential of the p.S151A mutation to cause DCM was investigated by using two different approaches: (1) engineering and characterization of heterozygous knock-in (S151A-) mice carrying the p.S151A mutation and (2) evaluation of the potential of adeno-associated virus (AAV) 9-based cardiac-specific transfer of p.S151A-mutated Sgcd cDNA to rescue the cardiac phenotype in Sgcd-deficient (Sgcd-null) mice as it has been demonstrated for intact, wild-type Sgcd cDNA. Heterozygous S151A knock-in mice developed a rather mild phenotype of cardiomyopathy. Increased heart to body weight suggests cardiac enlargement in 1-year-old S151A knock-in mice. However, at this age cardiac function, assessed by echocardiography, is maintained and histopathology completely absent. Myocardial expression of p.S151A cDNA, similar to intact Sgcd cDNA, restores cardiac function, although not being able to prevent myocardial histopathology in Sgcd-null mice completely. Our results suggest that the p.S151A mutation causes a mild, subclinical phenotype of cardiomyopathy, which is prone to be overseen in patients carrying such sequence variants. Furthermore, this study shows the suitability of an AAV-mediated cardiac gene transfer approach to analyze whether a sequence variant is a disease-causing mutation.


Asunto(s)
Cardiomiopatías/genética , Mutación Missense , Sarcoglicanos/genética , Animales , Cardiomiopatías/etiología , Cardiomiopatías/patología , Dependovirus , Técnicas de Sustitución del Gen , Heterocigoto , Humanos , Ratones , Miocardio/patología , Fenotipo
14.
Eur Heart J ; 34(35): 2768-75, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23178648

RESUMEN

AIMS: HCN4 channels are involved in generation, regulation, and stabilization of heart rhythm and channel dysfunction is associated with inherited sinus bradycardia. We asked whether dysfunctional HCN4 channels also contribute to the generation of cardiac tachyarrhythmias. METHODS AND RESULTS: In a candidate gene approach, we screened 422 patients with atrial and/or ventricular tachyarrhythmias and detected a novel HCN4 gene mutation that replaced the positively charged lysine 530 with an asparagine (HCN4-K530N) in a highly conserved region of the C-linker. The index patient developed tachycardia-bradycardia syndrome and persistent atrial fibrillation (AF) in an age-dependent fashion. Pedigree analysis identified eight affected family members with a similar course of disease. Whole-cell patch clamp electrophysiology of HEK293 cells showed that homomeric mutant channels almost are indistinguishable from wild-type channels. In contrast, heteromeric channels composed of mutant and wild-type subunits displayed a significant hyperpolarizing shift in the half-maximal activation voltage. This may be caused by a shift in the equilibrium between the tonically inhibited nucleotide-free state of the C-terminal domain of HCN4 believed to consist of a 'dimer of dimers' and the activated ligand-bound tetrameric form, leading to an increased inhibition of activity in heteromeric channels. CONCLUSION: Altered C-linker oligomerization in heteromeric channels is considered to promote familial tachycardia-bradycardia syndrome and persistent AF, indicating that f-channel dysfunction contributes to the development of atrial tachyarrhythmias.


Asunto(s)
Fibrilación Atrial/genética , Bradicardia/genética , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Mutación/genética , Taquicardia/genética , Adulto , Análisis de Varianza , Electrocardiografía , Técnicas Electrofisiológicas Cardíacas , Femenino , Células HEK293 , Humanos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/química , Masculino , Persona de Mediana Edad , Linaje
15.
Heart Rhythm ; 9(2): 265-72, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21907172

RESUMEN

BACKGROUND: Atrial fibrillation (AF) is the most common sustained cardiac arrhythmia. Gene therapy-dependent modulation of atrial electrophysiology may provide a more specific alternative to pharmacological and ablative treatment strategies. OBJECTIVE: We hypothesized that genetic inactivation of atrial repolarizing ether-a-go-go-related gene (ERG) K(+) currents using a dominant-negative mutant would provide rhythm control in AF. METHODS: Ten domestic swine underwent pacemaker implantation and were subjected to atrial burst pacing to induce persistent AF. Animals were then randomized to receive either AdCERG-G627S to suppress ERG/I(Kr) currents or green fluorescent protein (AdGFP) as control. Adenoviruses were applied using a novel hybrid technique combining atrial virus injection and epicardial electroporation to increase transgene expression. RESULTS: In pigs treated with AdCERG-G627S, the onset of persistent AF was prevented (n = 2) or significantly delayed compared with AdGFP controls (12 ± 2.1 vs. 6.2 ± 1.3 days; P < .001) during 14-day follow-up. Effective refractory periods were prolonged in the AdCERG-G627S group compared with AdGFP animals (221.5 ± 4.7 ms vs. 197.0 ± 4.7 ms; P < .006). Impairment of left ventricular ejection fraction (LVEF) during AF was prevented by AdCERG-G627S application (LVEF(CERG-G627S) = 62.1% ± 4.0% vs. LVEF(GFP) = 30.3% ± 9.1%; P < .001). CONCLUSION: Inhibition of ERG function using atrial AdCERG-G627S gene transfer suppresses or delays the onset of persistent AF by prolongation of atrial refractoriness in a porcine model. Targeted gene therapy represents an alternative to pharmacological or ablative treatment of AF.


Asunto(s)
Fibrilación Atrial/terapia , Canales de Potasio Éter-A-Go-Go/genética , Terapia Genética/métodos , Adenoviridae , Animales , Fibrilación Atrial/genética , Electrocardiografía , Canales de Potasio Éter-A-Go-Go/efectos de los fármacos , Técnicas de Transferencia de Gen , Proteínas Fluorescentes Verdes/farmacología , Atrios Cardíacos/fisiopatología , Mutación , Sus scrofa
16.
PLoS One ; 6(1): e16469, 2011 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-21305030

RESUMEN

The balanced action of both pre- and postsynaptic organizers regulates the formation of neuromuscular junctions (NMJ). The precise mechanisms that control the regional specialization of acetylcholine receptor (AChR) aggregation, guide ingrowing axons and contribute to correct synaptic patterning are unknown. Synaptic activity is of central importance and to understand synaptogenesis, it is necessary to distinguish between activity-dependent and activity-independent processes. By engineering a mutated fetal AChR subunit, we used homologous recombination to develop a mouse line that expresses AChR with massively reduced open probability during embryonic development. Through histological and immunochemical methods as well as electrophysiological techniques, we observed that endplate anatomy and distribution are severely aberrant and innervation patterns are completely disrupted. Nonetheless, in the absence of activity AChRs form postsynaptic specializations attracting motor axons and permitting generation of multiple nerve/muscle contacts on individual fibers. This process is not restricted to a specialized central zone of the diaphragm and proceeds throughout embryonic development. Phenotypes can be attributed to separate activity-dependent and -independent pathways. The correct patterning of synaptic connections, prevention of multiple contacts and control of nerve growth require AChR-mediated activity. In contrast, myotube survival and acetylcholine-mediated dispersal of AChRs are maintained even in the absence of AChR-mediated activity. Because mouse models in which acetylcholine is entirely absent do not display similar effects, we conclude that acetylcholine binding to the AChR initiates activity-dependent and activity-independent pathways whereby the AChR modulates formation of the NMJ.


Asunto(s)
Receptores Colinérgicos/fisiología , Sinapsis/ultraestructura , Animales , Axones , Ratones , Modelos Animales , Neuronas Motoras/química , Neuronas Motoras/metabolismo , Neuronas Motoras/ultraestructura , Fibras Musculares Esqueléticas , Proteínas Mutantes , Unión Neuromuscular , Fenotipo , Agregación de Receptores , Receptores Colinérgicos/análisis , Receptores Colinérgicos/genética , Sinapsis/metabolismo
17.
Circ Arrhythm Electrophysiol ; 3(5): 542-52, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20693575

RESUMEN

BACKGROUND: HCN channels activate the pacemaker current I(f), which is thought to contribute significantly to generation and regulation of heart rhythm. HCN4 represents the dominant isotype in the sinoatrial node and binding of cAMP was suggested to be necessary for autonomic heart rate regulation. METHODS AND RESULTS: In a candidate gene approach, a heterozygous insertion of 13 nucleotides in exon 6 of the HCN4 gene leading to a truncated cyclic nucleotide-binding domain was identified in a 45-year-old woman with sinus bradycardia. Biophysical properties determined by whole-cell patch-clamp recording of HEK293 cells demonstrated that mutant subunits (HCN4-695X) were insensitive to cAMP. Heteromeric channels composed of wild-type and mutant subunits failed to respond to cAMP-like homomeric mutant channels, indicating a dominant-negative suppression of cAMP-induced channel activation by mutant subunits. Pedigree analysis identified 7 additional living carriers showing similar clinical phenotypes, that is, sinus node dysfunction with mean resting heart rate of 45.9±4.6 bpm (n=8) compared with 66.5±9.1 bpm of unaffected relatives (n=6; P<0.01). Clinical evaluation revealed no ischemic or structural heart disease in any family member. Importantly, mutant carriers exhibited normal heart rate variance and full ability to accelerate heart rate under physical activity or pharmacological stimulation. Moreover, mutant carriers displayed distinctive sinus arrhythmias and premature beats linked to adrenergic stress. CONCLUSIONS: In humans, cAMP responsiveness of I(f) determines basal heart rate but is not critical for maximum heart rate, heart rate variability, or chronotropic competence. Furthermore, cAMP-activated I(f) may stabilize heart rhythm during chronotropic response.


Asunto(s)
Sistema Nervioso Autónomo/fisiopatología , Estimulación Cardíaca Artificial/métodos , AMP Cíclico/metabolismo , Canales Catiónicos Regulados por Nucleótidos Cíclicos/genética , ADN/genética , Mutación , Canales de Potasio/genética , Síndrome del Seno Enfermo/genética , Adolescente , Adulto , Anciano , Niño , Canales Catiónicos Regulados por Nucleótidos Cíclicos/metabolismo , Femenino , Predisposición Genética a la Enfermedad , Frecuencia Cardíaca/genética , Humanos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Masculino , Persona de Mediana Edad , Proteínas del Tejido Nervioso , Canales de Potasio/metabolismo , Síndrome del Seno Enfermo/metabolismo , Síndrome del Seno Enfermo/terapia , Nodo Sinoatrial/metabolismo , Nodo Sinoatrial/fisiopatología , Adulto Joven
18.
J Mol Cell Cardiol ; 48(1): 230-7, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19540844

RESUMEN

Hereditary long QT syndrome (LQTS) is a cardiovascular disorder characterized by prolongation of the QT interval on the surface ECG and a high risk for arrhythmia-related sudden death. Mutations in a cardiac voltage-gated potassium channel, KCNQ1, account for the most common form of LQTS, LQTS1. The objective of this study was the characterization of a novel KCNQ1 mutation linked to LQTS. Electrophysiological properties and clinical features were determined and compared to characteristics of a different mutation at the same position. Single-strand conformation polymorphism analysis followed by direct sequencing was performed to screen LQTS genes for mutations. A novel missense mutation in the KCNQ1 gene, KCNQ1 P320H, was identified in the index patient presenting with recurrent syncope and aborted sudden death triggered by physical stress and swimming. Electrophysiological analyses of KCNQ1 P320H and the previously reported KCNQ1 P320A mutation indicate that both channels are non-functional and suppress wild type I(Ks) in a dominant-negative fashion. Based on homology modeling of the KCNQ1 channel pore region, we speculate that the proline residue at position 320 limits flexibility of the outer pore and is required to maintain the functional architecture of the selectivity filter/pore helix arrangement. Our observations on the KCNQ1 P320H mutation are consistent with previous studies indicating that pore mutations in potassium channel alpha-subunits are associated with more severe electrophysiological and clinical phenotypes than mutations in other regions of these proteins. This study emphasizes the significance of mutation screening for diagnosis, risk-assessment, and mutation-site specific management in LQTS patients.


Asunto(s)
Canal de Potasio KCNQ1/metabolismo , Síndrome de Romano-Ward/genética , Adulto , Análisis Mutacional de ADN , Electrofisiología , Femenino , Predisposición Genética a la Enfermedad , Humanos , Canal de Potasio KCNQ1/genética , Masculino , Mutación , Linaje , Canales de Potasio con Entrada de Voltaje/genética
19.
Basic Res Cardiol ; 104(6): 621-9, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19421833

RESUMEN

Hyperpolarization-activated ion channels, encoded by four mammalian genes (HCN1-4), contribute in an important way to the cardiac pacemaker current I(f). Here, we describe the transcription profiles of the four HCN genes, the NRSF, KCNE2 and Kir2.1 genes from embryonic stage E9.5 dpc to postnatal day 120 in the mouse. Embryonic atrium and ventricle revealed abundant HCN4 transcription but other HCN transcripts were almost absent. Towards birth, HCN4 was downregulated in the atrium and almost vanished from the ventricle. After birth, however, HCN isotype transcription changed remarkably, showing increased levels of HCN1, HCN2 and HCN4 in the atrium and of HCN2 and HCN4 in the ventricle. HCN3 showed highest transcription at early embryonic stages and was hardly detectable thereafter. At postnatal day 10, HCN4 was highest in the sinoatrial node, being twofold higher than HCN1 and fivefold higher than HCN2. In the atrium, HCN4 was similar to HCN1 and sevenfold higher than HCN2. In the ventricle, in contrast, HCN2 was sixfold higher than HCN4, while HCN1 was absent. Subsequently all HCN isotype transcripts declined to lower adult levels, while ratios of HCN isotypes remained stable. In conclusion, substantial changes of HCN isotype transcription throughout cardiac development suggest that a regulated pattern of HCN isotypes is required to establish and ensure a stable heart rhythm. Furthermore, constantly low HCN transcription in adult myocardium may be required to prevent atrial and ventricular arrhythmogenesis.


Asunto(s)
Corazón/embriología , Corazón/crecimiento & desarrollo , Miocardio/metabolismo , Canales de Potasio/biosíntesis , Canales de Potasio/genética , Animales , Canales Catiónicos Regulados por Nucleótidos Cíclicos/biosíntesis , Canales Catiónicos Regulados por Nucleótidos Cíclicos/genética , Expresión Génica , Perfilación de la Expresión Génica , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Canales Iónicos/biosíntesis , Canales Iónicos/genética , Ratones , Ratones Endogámicos C57BL , Canales de Potasio de Rectificación Interna/biosíntesis , Canales de Potasio de Rectificación Interna/genética , Canales de Potasio con Entrada de Voltaje/biosíntesis , Canales de Potasio con Entrada de Voltaje/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcripción Genética
20.
Biochem Biophys Res Commun ; 359(2): 209-13, 2007 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-17540338

RESUMEN

In a first step towards dissecting molecular mechanisms that contribute to the development of cardiac diseases, we have generated transgenic mice that express a Cre-GFP fusion protein under the transcriptional control of a 4.3kb murine cardiac Troponin I gene (cTnI) promoter. Cre-GFP expression, similar in three transgenic lines, is described in one line. In mouse embryos, transgenic for the Cre-GFP and ROSA lacZ reporter allele, first Cre-mediated recombination appeared at 16.5 dpc selectively at the heart. Like the endogenous cTnI gene, transgenic Cre expression showed a slow rise through fetal development that increased neonatally. Bitransgenic hearts, stained at 30 days of age, showed intense signals in ventricular and atrial myocytes while no recombination occurred in other tissues. The delayed onset of Cre activity in cTnI-Cre mice could provide a useful genetic tool to evaluate the function of loxP targeted cardiac genes without interference of recombination during early heart development.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Corazón/embriología , Integrasas/biosíntesis , Troponina I/genética , Alelos , Animales , Genes Reporteros , Humanos , Ratones , Ratones Transgénicos , Células Musculares/citología , Regiones Promotoras Genéticas , Recombinación Genética , Factores de Tiempo , Distribución Tisular , Troponina I/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA