Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
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
2.
Am J Med Genet A ; 185(8): 2434-2444, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34056838

RESUMEN

Cantú syndrome (CS) is caused by pathogenic variants in ABCC9 and KCNJ8 encoding the regulatory and pore-forming subunits of ATP-sensitive potassium (KATP ) channels. CS is characterized by congenital hypertrichosis, distinctive facial features, peripheral edema, and cardiac and neurodevelopmental abnormalities. Behavioral and cognitive issues have been self-reported by some CS individuals, but results of formal standardized investigations have not been published. To assess the cognitive profile, social functioning, and psychiatric symptoms in a large group of CS subjects systematically in a cross-sectional manner, we invited 35 individuals (1-69 years) with confirmed ABCC9 variants and their relatives to complete various commonly applied standardized age-related questionnaires, including the Kaufman brief intelligence test 2, the social responsiveness scale-2, and the Achenbach system of empirically based assessment. The majority of CS individuals demonstrated average verbal and nonverbal intelligence compared to the general population. Fifteen percent of cases showed social functioning strongly associated with a clinical diagnosis of autism spectrum disorder. Both externalizing and internalizing problems were also present in this cohort. In particular, anxiety, anxiety or attention deficit hyperactivity disorder, and autism spectrum behaviors were predominantly observed in the younger subjects in the cohort (≥25%), but this percentage decreased markedly in adults.


Asunto(s)
Conducta , Cardiomegalia/diagnóstico , Cognición , Hipertricosis/diagnóstico , Osteocondrodisplasias/diagnóstico , Fenotipo , Adolescente , Adulto , Anciano , Alelos , Cardiomegalia/genética , Niño , Preescolar , Emociones , Femenino , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Hipertricosis/genética , Lactante , Masculino , Persona de Mediana Edad , Mutación , Osteocondrodisplasias/genética , Receptores de Sulfonilureas , Adulto Joven
3.
Am J Med Genet A ; 182(5): 1041-1052, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32100467

RESUMEN

Cantú syndrome (CS) was first described in 1982, and is caused by pathogenic variants in ABCC9 and KCNJ8 encoding regulatory and pore forming subunits of ATP-sensitive potassium (KATP ) channels, respectively. It is characterized by congenital hypertrichosis, osteochondrodysplasia, extensive cardiovascular abnormalities and distinctive facial anomalies including a broad nasal bridge, long philtrum, epicanthal folds, and prominent lips. Many genetic syndromes, such as CS, involve facial anomalies that serve as a significant clue in the initial identification of the respective disorder before clinical or molecular diagnosis are undertaken. However, an overwhelming number of CS patients receive misdiagnoses based on an evaluation of coarse facial features. By analyzing three-dimensional images of CS faces, we quantified facial dysmorphology in a cohort of both male and female CS patients with confirmed ABCC9 variants. Morphometric analysis of different regions of the face revealed gender-specific significant differences in face shape. Moreover, we show that 3D facial photographs can distinguish between CS and other genetic disorders with specific facial dysmorphologies that have been mistaken for CS-associated anomalies in the past, hence assisting in an earlier clinical and molecular diagnosis. This optimizes genetic counseling and reduces stress for patients and parents by avoiding unnecessary misdiagnosis.


Asunto(s)
Cardiomegalia/genética , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Hipertricosis/congénito , Canales KATP/genética , Osteocondrodisplasias/genética , Receptores de Sulfonilureas/genética , Adolescente , Adulto , Cardiomegalia/diagnóstico por imagen , Cardiomegalia/fisiopatología , Niño , Preescolar , Cara , Femenino , Enfermedades Genéticas Ligadas al Cromosoma X/diagnóstico por imagen , Enfermedades Genéticas Ligadas al Cromosoma X/fisiopatología , Predisposición Genética a la Enfermedad , Humanos , Hipertricosis/diagnóstico por imagen , Hipertricosis/genética , Hipertricosis/fisiopatología , Imagenología Tridimensional , Masculino , Mutación Missense/genética , Osteocondrodisplasias/diagnóstico por imagen , Osteocondrodisplasias/fisiopatología , Análisis de Componente Principal , Adulto Joven
4.
J Biol Chem ; 293(6): 2041-2052, 2018 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-29275331

RESUMEN

The complex disorder Cantu syndrome (CS) arises from gain-of-function mutations in either KCNJ8 or ABCC9, the genes encoding the Kir6.1 and SUR2 subunits of ATP-sensitive potassium (KATP) channels, respectively. Recent reports indicate that such mutations can increase channel activity by multiple molecular mechanisms. In this study, we determined the mechanism by which KATP function is altered by several substitutions in distinct structural domains of SUR2: D207E in the intracellular L0-linker and Y985S, G989E, M1060I, and R1154Q/R1154W in TMD2. We engineered substitutions at their equivalent positions in rat SUR2A (D207E, Y981S, G985E, M1056I, and R1150Q/R1150W) and investigated functional consequences using macroscopic rubidium (86Rb+) efflux assays and patch-clamp electrophysiology. Our results indicate that D207E increases KATP channel activity by increasing intrinsic stability of the open state, whereas the cluster of Y981S/G985E/M1056I substitutions, as well as R1150Q/R1150W, augmented Mg-nucleotide activation. We also tested the responses of these channel variants to inhibition by the sulfonylurea drug glibenclamide, a potential pharmacotherapy for CS. None of the D207E, Y981S, G985E, or M1056I substitutions had a significant effect on glibenclamide sensitivity. However, Gln and Trp substitution at Arg-1150 significantly decreased glibenclamide potency. In summary, these results provide additional confirmation that mutations in CS-associated SUR2 mutations result in KATP gain-of-function. They help link CS genotypes to phenotypes and shed light on the underlying molecular mechanisms, including consequences for inhibitory drug sensitivity, insights that may inform the development of therapeutic approaches to manage CS.


Asunto(s)
Cardiomegalia/genética , Mutación con Ganancia de Función , Hipertricosis/genética , Mutación Missense , Osteocondrodisplasias/genética , Receptores de Sulfonilureas/química , Receptores de Sulfonilureas/genética , Animales , Cardiomegalia/metabolismo , Gliburida/química , Gliburida/metabolismo , Humanos , Hipertricosis/metabolismo , Canales KATP/química , Canales KATP/genética , Canales KATP/metabolismo , Osteocondrodisplasias/metabolismo , Dominios Proteicos , Ratas , Receptores de Sulfonilureas/metabolismo
5.
Am J Med Genet C Semin Med Genet ; 181(4): 658-681, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31828977

RESUMEN

Cantú syndrome (CS), first described in 1982, is caused by pathogenic variants in ABCC9 and KCNJ8, which encode the regulatory and pore forming subunits of ATP-sensitive potassium (KATP ) channels, respectively. Multiple case reports of affected individuals have described the various clinical features of CS, but systematic studies are lacking. To define the effects of genetic variants on CS phenotypes and clinical outcomes, we have developed a standardized REDCap-based registry for CS. We report phenotypic features and associated genotypes on 74 CS subjects, with confirmed ABCC9 variants in 72 of the individuals. Hypertrichosis and a characteristic facial appearance are present in all individuals. Polyhydramnios during fetal life, hyperflexibility, edema, patent ductus arteriosus (PDA), cardiomegaly, dilated aortic root, vascular tortuosity of cerebral arteries, and migraine headaches are common features, although even with this large group of subjects, there is incomplete penetrance of CS-associated features, without clear correlation to genotype.


Asunto(s)
Cardiomegalia/epidemiología , Hipertricosis/epidemiología , Osteocondrodisplasias/epidemiología , Sistema de Registros , Adolescente , Adulto , Cardiomegalia/genética , Niño , Facies , Femenino , Humanos , Hipertricosis/genética , Masculino , Osteocondrodisplasias/genética , Fenotipo , Adulto Joven
6.
J Vis Exp ; (180)2022 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-35225267

RESUMEN

Zebrafish have long been used as a model vertebrate organism in cardiovascular research. The technical difficulties of isolating individual cells from the zebrafish cardiovascular tissues have been limiting in studying their electrophysiological properties. Previous methods have been described for dissection of zebrafish hearts and isolation of ventricular cardiac myocytes. However, the isolation of zebrafish atrial and vascular myocytes for electrophysiological characterization was not detailed. This work describes new and modified enzymatic protocols that routinely provide isolated juvenile and adult zebrafish ventricular and atrial cardiomyocytes, as well as vascular smooth muscle (VSM) cells from the bulbous arteriosus, suitable for patch-clamp experiments. There has been no literary evidence of electrophysiological studies on zebrafish cardiovascular tissues isolated at embryonic and larval stages of development. Partial dissociation techniques that allow patch-clamp experiments on individual cells from larval and embryonic hearts are demonstrated.


Asunto(s)
Músculo Liso Vascular , Pez Cebra , Animales , Ventrículos Cardíacos , Larva , Miocitos Cardíacos/fisiología
7.
BMJ Case Rep ; 14(7)2021 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-34253519

RESUMEN

This case report of a young adult with Cantú syndrome (CS) illustrates a remarkable journey of learning how to cope with symptom management and emotional impact associated with a rare skin condition. We describe a 20-year-old woman with a CS-related mutation in ABCC9 resulting in clinical manifestations, including congenital hypertrichosis, facial dysmorphism and cardiomegaly. As of yet, no treatment is available for CS.Little is known about the impact of CS and similar (skin) conditions on the life of affected individuals, and about their needs and preferences in this regard. Hence, we describe the psychosocial implications our case had to deal with immediately after her diagnosis. In addition, we outline her significant progress in managing disease-associated features and emotional stress prompted by considerable personal development and an increase in confidence. This example shows that a normal lifestyle is achievable for (newly diagnosed) individuals despite suffering from CS or a related skin disorder.


Asunto(s)
Hipertricosis , Osteocondrodisplasias , Cardiomegalia , Femenino , Humanos , Hipertricosis/diagnóstico , Hipertricosis/genética , Receptores de Sulfonilureas , Adulto Joven
8.
Trends Pharmacol Sci ; 42(4): 255-267, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33563480

RESUMEN

Currently, there are about 7000 identified rare diseases, together affecting 10% of the population. However, fewer than 6% of all rare diseases have an approved treatment option, highlighting their tremendous unmet needs in drug development. The process of repurposing drugs for new indications, compared with the development of novel orphan drugs, is a time-saving and cost-efficient method resulting in higher success rates, which can therefore drastically reduce the risk of drug development for rare diseases. Although drug repurposing is not novel, new strategies have been developed in recent years to do it in a systematic and rational way. Here, we review applied methodologies, recent accomplished progress, and the challenges associated in drug repurposing for rare diseases.


Asunto(s)
Reposicionamiento de Medicamentos , Enfermedades Raras , Humanos , Producción de Medicamentos sin Interés Comercial , Enfermedades Raras/tratamiento farmacológico
9.
Nat Commun ; 10(1): 4457, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31575858

RESUMEN

Mutations in genes encoding KATP channel subunits have been reported for pancreatic disorders and Cantú syndrome. Here, we report a syndrome in six patients from two families with a consistent phenotype of mild intellectual disability, similar facies, myopathy, and cerebral white matter hyperintensities, with cardiac systolic dysfunction present in the two oldest patients. Patients are homozygous for a splice-site mutation in ABCC9 (c.1320 + 1 G > A), which encodes the sulfonylurea receptor 2 (SUR2) subunit of KATP channels. This mutation results in an in-frame deletion of exon 8, which results in non-functional KATP channels in recombinant assays. SUR2 loss-of-function causes fatigability and cardiac dysfunction in mice, and reduced activity, cardiac dysfunction and ventricular enlargement in zebrafish. We term this channelopathy resulting from loss-of-function of SUR2-containing KATP channels ABCC9-related Intellectual disability Myopathy Syndrome (AIMS). The phenotype differs from Cantú syndrome, which is caused by gain-of-function ABCC9 mutations, reflecting the opposing consequences of KATP loss- versus gain-of-function.


Asunto(s)
Adenosina Trifosfato/metabolismo , Canalopatías/metabolismo , Predisposición Genética a la Enfermedad/genética , Discapacidad Intelectual/metabolismo , Enfermedades Musculares/metabolismo , Mutación , Receptores de Sulfonilureas/genética , Receptores de Sulfonilureas/metabolismo , Adolescente , Adulto , Secuencia de Aminoácidos , Animales , Cardiomegalia/genética , Cardiomegalia/metabolismo , Línea Celular , Niño , Modelos Animales de Enfermedad , Facies , Femenino , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Corazón , Cardiopatías/genética , Cardiopatías/metabolismo , Homocigoto , Humanos , Hipertricosis/genética , Hipertricosis/metabolismo , Discapacidad Intelectual/parasitología , Masculino , Complejo Mediador/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Enfermedades Musculares/genética , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/metabolismo , Trastornos del Neurodesarrollo/fisiopatología , Osteocondrodisplasias/genética , Osteocondrodisplasias/metabolismo , Linaje , Fenotipo , Rubidio , Secuenciación Completa del Genoma , Adulto Joven , Pez Cebra
10.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA