Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Adv Exp Med Biol ; 1441: 1057-1090, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38884769

RESUMEN

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


Asunto(s)
Arritmias Cardíacas , Modelos Animales de Enfermedad , Animales , Humanos , Arritmias Cardíacas/genética , Arritmias Cardíacas/fisiopatología , Arritmias Cardíacas/metabolismo , Transducción de Señal/genética
2.
BMC Pediatr ; 23(1): 644, 2023 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-38114927

RESUMEN

BACKGROUND: Cantu syndrome is a rare and complex multisystem disorder characterized by hypertrichosis, facial dysmorphism, osteochondroplasia and cardiac abnormalities. With only 150 cases reported worldwide, Cantu syndrome is now gaining wider recognition due to molecular testing and a growing body of literature that further characterizes the syndrome and some of its most important features. Cardiovascular pathology previously described in the literature include cardiomegaly, pericardial effusion, vascular dilation and tortuosity, and other congenital heart defects. However, cardiovascular involvement is highly variable amongst individuals with Cantu syndrome. In some instances, it can be extensive and severe requiring surgical management and long term follow up. CASE PRESENTATION: Herein we report a case of a fourteen-year-old female who presented with worsening pericardial effusion of unknown etiology, and echocardiographic findings of concentric left ventricular hypertrophy, a mildly dilated aortic root and ascending aorta. Her medical history was notable for hemoptysis and an episode of pulmonary hemorrhage secondary to multiple aortopulmonary collaterals that were subsequently embolized in early childhood. She was initially managed with Ibuprofen and Colchicine but continued to worsen, and ultimately required a pericardial window for the management of refractory pericardial effusion. Imaging studies obtained on subsequent visits revealed multiple dilated and tortuous blood vessels in the head, neck, chest, and pelvis. A cardiomyopathy molecular studies panel was sent, and a pathogenic variant was identified in the ABCC9 gene, confirming the molecular diagnosis of autosomal dominant Cantu syndrome. CONCLUSIONS: Vascular anomalies and significant cardiac involvement are often present in Cantu syndrome, however there are currently no established screening recommendations or surveillance protocols in place. The triad of hypertrichosis, facial dysmorphism, and unexplained cardiovascular involvement in any patient should raise suspicion for Cantu syndrome and warrant further investigation. Initial cardiac evaluation and follow up should be indicated in any patient with a clinical and/or molecular diagnosis of Cantu syndrome. Furthermore, whole body imaging should be utilized to evaluate the extent of vascular involvement and dictate long term monitoring and care.


Asunto(s)
Anomalías Cardiovasculares , Hipertricosis , Osteocondrodisplasias , Derrame Pericárdico , Malformaciones Vasculares , Adolescente , Femenino , Humanos , Cardiomegalia/complicaciones , Cardiomegalia/genética , Cardiomegalia/patología , Hipertricosis/diagnóstico , Hipertricosis/genética , Hipertricosis/patología , Osteocondrodisplasias/complicaciones , Osteocondrodisplasias/diagnóstico , Osteocondrodisplasias/genética , Derrame Pericárdico/diagnóstico por imagen , Derrame Pericárdico/etiología
3.
Am J Physiol Cell Physiol ; 323(3): C920-C935, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35876283

RESUMEN

Kir6.1 and SUR2 are subunits of ATP-sensitive potassium (KATP) channels expressed in a wide range of tissues. Extensive study has implicated roles of these channel subunits in diverse physiological functions. Together they generate the predominant KATP conductance in vascular smooth muscle and are the target of vasodilatory drugs. Roles for Kir6.1/SUR2 dysfunction in disease have been suggested based on studies of animal models and human genetic discoveries. In recent years, it has become clear that gain-of-function (GoF) mutations in both genes result in Cantú syndrome (CS)-a complex, multisystem disorder. There is currently no targeted therapy for CS, but studies of mouse models of the disease reveal that pharmacological reversibility of cardiovascular and gastrointestinal pathologies can be achieved by administration of the KATP channel inhibitor, glibenclamide. Here we review the function, structure, and physiological and pathological roles of Kir6.1/SUR2B channels, with a focus on CS. Recent studies have led to much improved understanding of the underlying pathologies and the potential for treatment, but important questions remain: Can the study of genetically defined CS reveal new insights into Kir6.1/SUR2 function? Do these reveal new pathophysiological mechanisms that may be important in more common diseases? And is our pharmacological armory adequately stocked?


Asunto(s)
Hipertricosis , Osteocondrodisplasias , Adenosina Trifosfato , Animales , Cardiomegalia/genética , Humanos , Hipertricosis/genética , Canales KATP/genética , Ratones , Osteocondrodisplasias/genética , Receptores de Sulfonilureas/genética
4.
Am J Physiol Cell Physiol ; 323(4): C1018-C1035, 2022 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-35785984

RESUMEN

KATP channels function as negative regulators of active lymphatic pumping and lymph transport. This review summarizes and critiques the evidence for the expression of specific KATP channel subunits in lymphatic smooth muscle and endothelium, the roles that they play in normal lymphatic function, and their possible involvement in multiple diseases, including metabolic syndrome, lymphedema, and Cantú syndrome. For each of these topics, suggestions are made for directions for future research.


Asunto(s)
Hipertricosis , Osteocondrodisplasias , Adenosina Trifosfato , Cardiomegalia/metabolismo , Humanos , Hipertricosis/metabolismo , Canales KATP/genética , Canales KATP/metabolismo , Osteocondrodisplasias/metabolismo
5.
J Physiol ; 600(2): 299-312, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34820842

RESUMEN

ATP-sensitive potassium channels (KATP channels) are hetero-octameric nucleotide-gated ion channels that couple cellular metabolism to excitability in various tissues. In the heart, KATP channels are activated during ischaemia and potentially during adrenergic stimulation. In the vasculature, they are normally active at a low level, reducing vascular tone, but the ubiquitous nature of these channels leads to complex and poorly understood channelopathies as a result of gain- or loss-of-function mutations. Zebrafish (ZF) models of these channelopathies may provide insights to the link between molecular dysfunction and complex pathophysiology, but this requires understanding the tissue dependence of channel activity and subunit specificity. Thus far, direct analysis of ZF KATP expression and functional properties has only been performed in pancreatic ß-cells. Using a comprehensive combination of genetically modified fish, electrophysiology and gene expression analysis, we demonstrate that ZF cardiac myocytes (CM) and vascular smooth muscle (VSM) express functional KATP channels of similar subunit composition, structure and metabolic sensitivity to their mammalian counterparts. However, in contrast to mammalian cardiovascular KATP channels, ZF channels are insensitive to potassium channel opener drugs (pinacidil, minoxidil) in both chambers of the heart and in VSM. The results provide a first characterization of the molecular properties of fish KATP channels and validate the use of such genetically modified fish as models of human Cantú syndrome and ABCC9-related Intellectual Disability and Myopathy syndrome. KEY POINTS: Zebrafish cardiac myocytes (CM) and vascular smooth muscle (VSM) express functional KATP channels of similar subunit composition, structure and metabolic sensitivity to their mammalian counterparts. In contrast to mammalian cardiovascular KATP channels, zebrafish channels are insensitive to potassium channel opener drugs (pinacidil, minoxidil) in both chambers of the heart and in VSM. We provide a first characterization of the molecular properties of fish KATP channels and validate the use of such genetically modified fish as models of human Cantú syndrome and ABCC9-related Intellectual Disability and Myopathy syndrome.


Asunto(s)
Hipertricosis , Canales KATP , Animales , Humanos , Canales KATP/genética , Músculo Liso Vascular , Miocitos Cardíacos , Receptores de Sulfonilureas/genética , Pez Cebra
6.
Am J Med Genet A ; 188(6): 1661-1666, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35243770

RESUMEN

Cantù syndrome (CS) is a rare multisystemic disorder, characterized by congenital hypertrichosis, macrocephaly, facial dysmorphisms, cardiomegaly, vascular, and skeletal anomalies. From the cognitive point of view, most of the patients show a mild speech delay and a few of them present intellectual disability and learning difficulties. To date, most CS-reported cases are caused by heterozygous ABCC9 gene mutations. Only three patients with CS and heterozygous KCNJ8 gene variants have been reported. The authors here present the fourth case of CS with a variant in KCNJ8 in a 6-month-old baby. Diagnosis was reached through Trio-Whole Exome analysis that revealed a de novo missense variant in KCNJ8.


Asunto(s)
Hipertricosis , Osteocondrodisplasias , Cardiomegalia/genética , Humanos , Hipertricosis/diagnóstico , Hipertricosis/genética , Lactante , Mutación Missense/genética , Osteocondrodisplasias/genética
7.
Proc Natl Acad Sci U S A ; 113(24): 6773-8, 2016 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-27247394

RESUMEN

Cantu syndrome (CS) is caused by gain-of-function (GOF) mutations in genes encoding pore-forming (Kir6.1, KCNJ8) and accessory (SUR2, ABCC9) KATP channel subunits. We show that patients with CS, as well as mice with constitutive (cGOF) or tamoxifen-induced (icGOF) cardiac-specific Kir6.1 GOF subunit expression, have enlarged hearts, with increased ejection fraction and increased contractility. Whole-cell voltage-clamp recordings from cGOF or icGOF ventricular myocytes (VM) show increased basal L-type Ca(2+) current (LTCC), comparable to that seen in WT VM treated with isoproterenol. Mice with vascular-specific expression (vGOF) show left ventricular dilation as well as less-markedly increased LTCC. Increased LTCC in KATP GOF models is paralleled by changes in phosphorylation of the pore-forming α1 subunit of the cardiac voltage-gated calcium channel Cav1.2 at Ser1928, suggesting enhanced protein kinase activity as a potential link between increased KATP current and CS cardiac pathophysiology.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Cardiomegalia/metabolismo , Ventrículos Cardíacos/metabolismo , Hipertricosis/metabolismo , Canales KATP/metabolismo , Contracción Miocárdica , Miocitos Cardíacos/metabolismo , Osteocondrodisplasias/metabolismo , Receptores de Sulfonilureas/metabolismo , Animales , Canales de Calcio Tipo L/genética , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/genética , Cardiomegalia/genética , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Femenino , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Humanos , Hipertricosis/genética , Hipertricosis/patología , Hipertricosis/fisiopatología , Isoproterenol/farmacología , Canales KATP/genética , Masculino , Ratones , Ratones Transgénicos , Miocitos Cardíacos/patología , Osteocondrodisplasias/genética , Osteocondrodisplasias/patología , Osteocondrodisplasias/fisiopatología , Receptores de Sulfonilureas/genética
8.
J Pharmacol Sci ; 135(1): 37-43, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28928055

RESUMEN

Genetic analysis of KCNJ8 has pointed a mutation (S422L) as a susceptible link to J wave syndrome (JWS). In vitro expression study indicated that the ATP-sensitive K+ (KATP) channel with the S422L mutation has the gain-of-function with reduced sensitivity to ATP. However, the electrophysiological impact of KCNJ8 has not been elucidated in vivo. Transgenic mouse strains overexpressing KCNJ8 S422L variant (TGmt) or WT (TGWT) in cardiomyocytes have been created to investigate the influence of KCNJ8 in cardiomyocytes and the JWS-related feature of the S422L variant on the cardiac electrophysiology. These TG strains demonstrated distinct changes in the J-ST segment of ECG with marked QT prolongation, which might be ascribed to the action potential prolongation resulting from the reduction of voltage-dependent K+ currents in ventricular cells. The pinacidil-induced KATP current was decreased in these TG myocytes and no obvious difference between TG and non-TG (WT) myocytes in the ATP sensitivity of the KATP channel was observed although the open probability of the KATP channels was significantly lower in TG myocytes than WT. These transgenic mouse strains with distinct ECG changes suggested that the S422L mutation in KCNJ8 gene is not a direct cause of JWS.


Asunto(s)
Fenómenos Electrofisiológicos/genética , Expresión Génica/genética , Canales KATP/genética , Mutación , Miocitos Cardíacos/fisiología , Adenosina Trifosfato/metabolismo , Animales , Arritmias Cardíacas/genética , Electrocardiografía , Predisposición Genética a la Enfermedad/genética , Canales KATP/metabolismo , Ratones Transgénicos , Miocitos Cardíacos/metabolismo , Técnicas de Placa-Clamp , Síndrome
9.
Hum Mutat ; 35(7): 809-13, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24700710

RESUMEN

ATP-sensitive potassium (KATP ) channels, composed of inward-rectifying potassium channel subunits (Kir6.1 and Kir6.2, encoded by KCNJ8 and KCNJ11, respectively) and regulatory sulfonylurea receptor (SUR1 and SUR2, encoded by ABCC8 and ABCC9, respectively), couple metabolism to excitability in multiple tissues. Mutations in ABCC9 cause Cantú syndrome (CS), a distinct multiorgan disease, potentially via enhanced KATP channel activity. We screened KCNJ8 in an ABCC9 mutation-negative patient who also exhibited clinical hallmarks of CS (hypertrichosis, macrosomia, macrocephaly, coarse facial appearance, cardiomegaly, and skeletal abnormalities). We identified a de novo missense mutation encoding Kir6.1[p.Cys176Ser] in the patient. Kir6.1[p.Cys176Ser] channels exhibited markedly higher activity than wild-type channels, as a result of reduced ATP sensitivity, whether coexpressed with SUR1 or SUR2A subunits. Our results identify a novel causal gene in CS, but also demonstrate that the cardinal features of the disease result from gain of KATP channel function, not from a Kir6-independent SUR2 function.


Asunto(s)
Cardiomegalia/genética , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Hipertricosis/genética , Canales KATP/genética , Mutación , Osteocondrodisplasias/genética , Adolescente , Cardiomegalia/diagnóstico , Análisis Mutacional de ADN , Facies , Enfermedades Genéticas Ligadas al Cromosoma X/diagnóstico , Humanos , Hipertricosis/diagnóstico , Canales KATP/química , Canales KATP/metabolismo , Masculino , Potenciales de la Membrana , Modelos Moleculares , Mutación Missense , Osteocondrodisplasias/diagnóstico , Fenotipo , Conformación Proteica
10.
Function (Oxf) ; 5(5)2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-38984978

RESUMEN

Cantú syndrome (CS), a multisystem disease with a complex cardiovascular phenotype, is caused by gain-of-function (GoF) variants in the Kir6.1/SUR2 subunits of ATP-sensitive potassium (KATP) channels and is characterized by low systemic vascular resistance, as well as tortuous, dilated, vessels, and decreased pulse-wave velocity. Thus, CS vascular dysfunction is multifactorial, with both hypomyotonic and hyperelastic components. To dissect whether such complexities arise cell autonomously within vascular smooth muscle cells (VSMCs) or as secondary responses to the pathophysiological milieu, we assessed electrical properties and gene expression in human induced pluripotent stem cell-derived VSMCs (hiPSC-VSMCs), differentiated from control and CS patient-derived hiPSCs, and in native mouse control and CS VSMCs. Whole-cell voltage clamp of isolated aortic and mesenteric arterial VSMCs isolated from wild-type (WT) and Kir6.1[V65M] (CS) mice revealed no clear differences in voltage-gated K+ (Kv) or Ca2+ currents. Kv and Ca2+ currents were also not different between validated hiPSC-VSMCs differentiated from control and CS patient-derived hiPSCs. While pinacidil-sensitive KATP currents in control hiPSC-VSMCs were similar to those in WT mouse VSMCs, they were considerably larger in CS hiPSC-VSMCs. Under current-clamp conditions, CS hiPSC-VSMCs were also hyperpolarized, consistent with increased basal K conductance and providing an explanation for decreased tone and decreased vascular resistance in CS. Increased compliance was observed in isolated CS mouse aortae and was associated with increased elastin mRNA expression. This was consistent with higher levels of elastin mRNA in CS hiPSC-VSMCs and suggesting that the hyperelastic component of CS vasculopathy is a cell-autonomous consequence of vascular KATP GoF. The results show that hiPSC-VSMCs reiterate expression of the same major ion currents as primary VSMCs, validating the use of these cells to study vascular disease. Results in hiPSC-VSMCs derived from CS patient cells suggest that both the hypomyotonic and hyperelastic components of CS vasculopathy are cell-autonomous phenomena driven by KATP overactivity within VSMCs .


Asunto(s)
Hipertricosis , Células Madre Pluripotentes Inducidas , Canales KATP , Músculo Liso Vascular , Miocitos del Músculo Liso , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Músculo Liso Vascular/metabolismo , Hipertricosis/genética , Hipertricosis/metabolismo , Hipertricosis/fisiopatología , Hipertricosis/patología , Animales , Ratones , Miocitos del Músculo Liso/metabolismo , Canales KATP/genética , Canales KATP/metabolismo , Osteocondrodisplasias/genética , Osteocondrodisplasias/metabolismo , Osteocondrodisplasias/patología , Osteocondrodisplasias/fisiopatología , Mutación , Diferenciación Celular/genética , Técnicas de Placa-Clamp , Cardiomegalia , Receptores de Sulfonilureas
11.
Dev Cell ; 57(11): 1383-1399.e7, 2022 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-35588738

RESUMEN

Loss- or gain-of-function mutations in ATP-sensitive potassium channel (K-ATP)-encoding genes, KCNJ8 and ABCC9, cause human central nervous system disorders with unknown pathogenesis. Here, using mice, zebrafish, and cell culture models, we investigated cellular and molecular causes of brain dysfunctions derived from altered K-ATP channel function. We show that genetic/chemical inhibition or activation of KCNJ8/ABCC9-containing K-ATP channel function leads to brain-selective suppression or promotion of arterial/arteriolar vascular smooth muscle cell (VSMC) differentiation, respectively. We further show that brain VSMCs develop from KCNJ8/ABCC9-containing K-ATP channel-expressing mural cell progenitor and that K-ATP channel cell autonomously regulates VSMC differentiation through modulation of intracellular Ca2+ oscillation via voltage-dependent calcium channels. Consistent with defective VSMC development, Kcnj8 knockout mice showed deficiency in vasoconstrictive capacity and neuronal-evoked vasodilation leading to local hyperemia. Our results demonstrate a role for KCNJ8/ABCC9-containing K-ATP channels in the differentiation of brain VSMC, which in turn is necessary for fine-tuning of cerebral blood flow.


Asunto(s)
Canales KATP/metabolismo , Músculo Liso Vascular , Acoplamiento Neurovascular , Receptores de Sulfonilureas/metabolismo , Adenosina Trifosfato , Animales , Encéfalo/metabolismo , Canales KATP/genética , Ratones , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Receptores de Sulfonilureas/química , Receptores de Sulfonilureas/genética , Pez Cebra/metabolismo
12.
Function (Oxf) ; 1(1): zqaa004, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32865539

RESUMEN

Dramatic cardiomegaly arising from gain-of-function (GoF) mutations in the ATP-sensitive potassium (KATP) channels genes, ABCC9 and KCNJ8, is a characteristic feature of Cantú syndrome (CS). How potassium channel over-activity results in cardiac hypertrophy, as well as the long-term consequences of cardiovascular remodeling in CS, is unknown. Using genome-edited mouse models of CS, we therefore sought to dissect the pathophysiological mechanisms linking KATP channel GoF to cardiac remodeling. We demonstrate that chronic reduction of systemic vascular resistance in CS is accompanied by elevated renin-angiotensin signaling, which drives cardiac enlargement and blood volume expansion. Cardiac enlargement in CS results in elevation of basal cardiac output, which is preserved in aging. However, the cardiac remodeling includes altered gene expression patterns that are associated with pathological hypertrophy and are accompanied by decreased exercise tolerance, suggestive of reduced cardiac reserve. Our results identify a high-output cardiac hypertrophy phenotype in CS which is etiologically and mechanistically distinct from other myocardial hypertrophies, and which exhibits key features of high-output heart failure (HOHF). We propose that CS is a genetically-defined HOHF disorder and that decreased vascular smooth muscle excitability is a novel mechanism for HOHF pathogenesis.


Asunto(s)
Mutación con Ganancia de Función , Canales KATP , Ratones , Animales , Canales KATP/genética , Mutación con Ganancia de Función/genética , Remodelación Ventricular , Receptores de Sulfonilureas/genética , Cardiomegalia/genética , Adenosina Trifosfato
13.
Dis Model Mech ; 11(10)2018 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-30355756

RESUMEN

The zebrafish (Danio rerio) has become a popular vertebrate model organism to study organ formation and function due to its optical clarity and rapid embryonic development. The use of genetically modified zebrafish has also allowed identification of new putative therapeutic drugs. So far, most studies have relied on broad overexpression of transgenes harboring patient-derived mutations or loss-of-function mutants, which incompletely model the human disease allele in terms of expression levels or cell-type specificity of the endogenous gene of interest. Most human genetically inherited conditions are caused by alleles carrying single nucleotide changes resulting in altered gene function. Introduction of such point mutations in the zebrafish genome would be a prerequisite to recapitulate human disease but remains challenging to this day. We present an effective approach to introduce small nucleotide changes in the zebrafish genome. We generated four different knock-in lines carrying distinct human cardiovascular-disorder-causing missense mutations in their zebrafish orthologous genes by combining CRISPR/Cas9 with a short template oligonucleotide. Three of these lines carry gain-of-function mutations in genes encoding the pore-forming (Kir6.1, KCNJ8) and regulatory (SUR2, ABCC9) subunits of an ATP-sensitive potassium channel (KATP) linked to Cantú syndrome (CS). Our heterozygous zebrafish knock-in lines display significantly enlarged ventricles with enhanced cardiac output and contractile function, and distinct cerebral vasodilation, demonstrating the causality of the introduced mutations for CS. These results demonstrate that introducing patient alleles in their zebrafish orthologs promises a broad application for modeling human genetic diseases, paving the way for new therapeutic strategies using this model organism.


Asunto(s)
Proteína 9 Asociada a CRISPR/metabolismo , Sistemas CRISPR-Cas/genética , Enfermedades Cardiovasculares/genética , Edición Génica , Nucleótidos/genética , Pez Cebra/genética , Animales , Secuencia de Bases , Modelos Animales de Enfermedad , Técnicas de Sustitución del Gen , Pruebas Genéticas , Heterocigoto , Humanos , Mutación/genética
14.
Heart Rhythm ; 12(11): 2316-24, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26142302

RESUMEN

BACKGROUND: Gain-of-function (GOF) mutations in the KATP channel subunits Kir6.1 and SUR2 cause Cantu syndrome (CS), a disease characterized by multiple cardiovascular abnormalities. OBJECTIVE: The purpose of this study was to better determine the electrophysiologic consequences of such GOF mutations in the heart. METHODS: We generated transgenic mice (Kir6.1-GOF) expressing ATP-insensitive Kir6.1[G343D] subunits under α-myosin heavy chain (α-MHC) promoter control, to target gene expression specifically in cardiomyocytes, and performed patch-clamp experiments on isolated ventricular myocytes and invasive electrophysiology on anesthetized mice. RESULTS: In Kir6.1-GOF ventricular myocytes, KATP channels showed decreased ATP sensitivity but no significant change in current density. Ambulatory ECG recordings on Kir6.1-GOF mice revealed AV nodal conduction abnormalities and junctional rhythm. Invasive electrophysiologic analyses revealed slowing of conduction and conduction failure through the AV node but no increase in susceptibility to atrial or ventricular ectopic activity. Surface ECGs recorded from CS patients also demonstrated first-degree AV block and fascicular block. CONCLUSION: The primary electrophysiologic consequence of cardiac KATP GOF is on the conduction system, particularly the AV node, resulting in conduction abnormalities in CS patients who carry KATP GOF mutations.


Asunto(s)
Bloqueo Atrioventricular/genética , Cardiomegalia/genética , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/genética , Regulación del Desarrollo de la Expresión Génica , Hipertricosis/genética , Canales KATP/genética , Osteocondrodisplasias/genética , Animales , Síndrome de Brugada/genética , Trastorno del Sistema de Conducción Cardíaco , Cardiomegalia/diagnóstico por imagen , Células Cultivadas , Preescolar , Modelos Animales de Enfermedad , Ecocardiografía Doppler , Electrocardiografía , Fenómenos Electrofisiológicos/genética , Humanos , Hipertricosis/diagnóstico por imagen , Masculino , Ratones , Ratones Transgénicos , Mutación , Miocitos Cardíacos/citología , Miocitos Cardíacos/fisiología , Osteocondrodisplasias/diagnóstico por imagen , Distribución Aleatoria , Enfermedades Raras , Muestreo
15.
J Am Heart Assoc ; 2(4): e000365, 2013 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-23974906

RESUMEN

BACKGROUND: KATP channels, assembled from pore-forming (Kir6.1 or Kir6.2) and regulatory (SUR1 or SUR2) subunits, link metabolism to excitability. Loss of Kir6.2 results in hypoglycemia and hyperinsulinemia, whereas loss of Kir6.1 causes Prinzmetal angina-like symptoms in mice. Conversely, overactivity of Kir6.2 induces neonatal diabetes in mice and humans, but consequences of Kir6.1 overactivity are unknown. METHODS AND RESULTS: We generated transgenic mice expressing wild-type (WT), ATP-insensitive Kir6.1 [Gly343Asp] (GD), and ATP-insensitive Kir6.1 [Gly343Asp,Gln53Arg] (GD-QR) subunits, under Cre-recombinase control. Expression was induced in smooth muscle cells by crossing with smooth muscle myosin heavy chain promoter-driven tamoxifen-inducible Cre-recombinase (SMMHC-Cre-ER) mice. Three weeks after tamoxifen induction, we assessed blood pressure in anesthetized and conscious animals, as well as contractility of mesenteric artery smooth muscle and KATP currents in isolated mesenteric artery myocytes. Both systolic and diastolic blood pressures were significantly reduced in GD and GD-QR mice but normal in mice expressing the WT transgene and elevated in Kir6.1 knockout mice as well as in mice expressing dominant-negative Kir6.1 [AAA] in smooth muscle. Contractile response of isolated GD-QR mesenteric arteries was blunted relative to WT controls, but nitroprusside relaxation was unaffected. Basal KATP conductance and pinacidil-activated conductance were elevated in GD but not in WT myocytes. CONCLUSIONS: KATP overactivity in vascular muscle can lead directly to reduced vascular contractility and lower blood pressure. We predict that gain of vascular KATP function in humans would lead to a chronic vasodilatory phenotype, as indeed has recently been demonstrated in Cantu syndrome.


Asunto(s)
Presión Sanguínea , Hipotensión/metabolismo , Canales KATP/metabolismo , Músculo Liso Vascular/metabolismo , Animales , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/genética , Relación Dosis-Respuesta a Droga , Predisposición Genética a la Enfermedad , Hipotensión/genética , Hipotensión/fisiopatología , Canales KATP/genética , Potenciales de la Membrana , Arterias Mesentéricas/metabolismo , Arterias Mesentéricas/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Noqueados , Ratones Transgénicos , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/fisiopatología , Mutación , Fenotipo , Potasio/metabolismo , Vasoconstricción , Vasodilatación , Vasodilatadores/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA