Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Artículo en Inglés | MEDLINE | ID: mdl-37348644

RESUMEN

The importance of cholesterol in hair follicle biology is underscored by its links to the pathogenesis of alopecias and hair growth disorders. Reports have associated defects in ABCA5, a membrane transporter, with altered keratinocyte cholesterol distribution in individuals with a form of congenital hypertrichosis, yet the biological basis for this defect in hair growth remains unknown. This study aimed to determine the impact of altered ABCA5 activity on hair follicle keratinocyte behaviour. Primary keratinocytes isolated from the outer root sheath of plucked human hair follicles were utilised as a relevant cell model. Following exogenous cholesterol loading, an increase in ABCA5 co-localisation to intracellular organelles was seen. Knockdown of ABCA5 revealed a dysregulation in cholesterol homeostasis, with LXR agonism leading to partial restoration of the homeostatic response. Filipin staining and live BODIPY cholesterol immunofluorescence microscopy revealed a reduction in endo-lysosomal cholesterol following ABCA5 knockdown. Analysis of oxysterols showed a significant increase in the fold change of 25-hydroxycholesterol and 7-ß-hydroxycholesterol following cholesterol loading in ORS keratinocytes, after ABCA5 knockdown. These data suggest a role for ABCA5 in the intracellular compartmentalisation of free cholesterol in primary hair follicle keratinocytes. The loss of normal homeostatic response, following the delivery of excess cholesterol after ABCA5 knockdown, suggests an impact on LXR-mediated transcriptional activity. The loss of ABCA5 in the hair follicle could lead to impaired endo-lysosomal cholesterol transport, impacting pathways known to influence hair growth. This avenue warrants further investigation.


Asunto(s)
Folículo Piloso , Hipertricosis , Humanos , Folículo Piloso/metabolismo , Queratinocitos/metabolismo , Hipertricosis/metabolismo , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Homeostasis , Colesterol/metabolismo
2.
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
3.
Proc Natl Acad Sci U S A ; 118(44)2021 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-34711681

RESUMEN

Vascular tone is dependent on smooth muscle KATP channels comprising pore-forming Kir6.1 and regulatory SUR2B subunits, in which mutations cause Cantú syndrome. Unique among KATP isoforms, they lack spontaneous activity and require Mg-nucleotides for activation. Structural mechanisms underlying these properties are unknown. Here, we determined cryogenic electron microscopy structures of vascular KATP channels bound to inhibitory ATP and glibenclamide, which differ informatively from similarly determined pancreatic KATP channel isoform (Kir6.2/SUR1). Unlike SUR1, SUR2B subunits adopt distinct rotational "propeller" and "quatrefoil" geometries surrounding their Kir6.1 core. The glutamate/aspartate-rich linker connecting the two halves of the SUR-ABC core is observed in a quatrefoil-like conformation. Molecular dynamics simulations reveal MgADP-dependent dynamic tripartite interactions between this linker, SUR2B, and Kir6.1. The structures captured implicate a progression of intermediate states between MgADP-free inactivated, and MgADP-bound activated conformations wherein the glutamate/aspartate-rich linker participates as mobile autoinhibitory domain, suggesting a conformational pathway toward KATP channel activation.


Asunto(s)
Adenosina Difosfato/metabolismo , Canales KATP/ultraestructura , Receptores de Sulfonilureas/ultraestructura , Adenosina Trifosfato/metabolismo , Cardiomegalia/metabolismo , Humanos , Hipertricosis/metabolismo , Canales KATP/genética , Canales KATP/metabolismo , Músculo Liso/metabolismo , Osteocondrodisplasias/metabolismo , Páncreas/metabolismo , Canales de Potasio/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Relación Estructura-Actividad , Receptores de Sulfonilureas/genética , Receptores de Sulfonilureas/metabolismo
4.
Cells ; 10(7)2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-34359961

RESUMEN

(1) Background: Cantu syndrome (CS) arises from gain-of-function (GOF) mutations in the ABCC9 and KCNJ8 genes, which encode ATP-sensitive K+ (KATP) channel subunits SUR2 and Kir6.1, respectively. Most CS patients have mutations in SUR2, the major component of skeletal muscle KATP, but the consequences of SUR2 GOF in skeletal muscle are unknown. (2) Methods: We performed in vivo and ex vivo characterization of skeletal muscle in heterozygous SUR2[A478V] (SUR2wt/AV) and homozygous SUR2[A478V] (SUR2AV/AV) CS mice. (3) Results: In SUR2wt/AV and SUR2AV/AV mice, forelimb strength and diaphragm amplitude movement were reduced; muscle echodensity was enhanced. KATP channel currents recorded in Flexor digitorum brevis fibers showed reduced MgATP-sensitivity in SUR2wt/AV, dramatically so in SUR2AV/AV mice; IC50 for MgATP inhibition of KATP currents were 1.9 ± 0.5 × 10-5 M in SUR2wt/AV and 8.6 ± 0.4 × 10-6 M in WT mice and was not measurable in SUR2AV/AV. A slight rightward shift of sensitivity to inhibition by glibenclamide was detected in SUR2AV/AV mice. Histopathological and qPCR analysis revealed atrophy of soleus and tibialis anterior muscles and up-regulation of atrogin-1 and MuRF1 mRNA in CS mice. (4) Conclusions: SUR2[A478V] "knock-in" mutation in mice impairs KATP channel modulation by MgATP, markedly so in SUR2AV/AV, with atrophy and non-inflammatory edema in different skeletal muscle phenotypes.


Asunto(s)
Cardiomegalia/genética , Cardiomegalia/metabolismo , Hipertricosis/genética , Hipertricosis/metabolismo , Complejo Mediador/metabolismo , Músculo Esquelético/metabolismo , Mutación/genética , Osteocondrodisplasias/genética , Osteocondrodisplasias/metabolismo , Animales , Atrofia/patología , Modelos Animales de Enfermedad , Mutación con Ganancia de Función/genética , Humanos , Ratones , Fenotipo
5.
JCI Insight ; 6(5)2021 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-33529173

RESUMEN

Cantu syndrome (CS) is caused by gain-of-function (GOF) mutations in pore-forming (Kir6.1, KCNJ8) and accessory (SUR2, ABCC9) ATP-sensitive potassium (KATP) channel subunits, the most common mutations being SUR2[R1154Q] and SUR2[R1154W], carried by approximately 30% of patients. We used CRISPR/Cas9 genome engineering to introduce the equivalent of the human SUR2[R1154Q] mutation into the mouse ABCC9 gene. Along with minimal CS disease features, R1154Q cardiomyocytes and vascular smooth muscle showed much lower KATP current density and pinacidil activation than WT cells. Almost complete loss of SUR2-dependent protein and KATP in homozygous R1154Q ventricles revealed underlying diazoxide-sensitive SUR1-dependent KATP channel activity. Surprisingly, sequencing of SUR2 cDNA revealed 2 distinct transcripts, one encoding full-length SUR2 protein; and the other with an in-frame deletion of 93 bases (corresponding to 31 amino acids encoded by exon 28) that was present in approximately 40% and approximately 90% of transcripts from hetero- and homozygous R1154Q tissues, respectively. Recombinant expression of SUR2A protein lacking exon 28 resulted in nonfunctional channels. CS tissue from SUR2[R1154Q] mice and human induced pluripotent stem cell-derived (hiPSC-derived) cardiomyocytes showed only full-length SUR2 transcripts, although further studies will be required in order to fully test whether SUR2[R1154Q] or other CS mutations might result in aberrant splicing and variable expressivity of disease features in human CS.


Asunto(s)
Cardiomegalia , Hipertricosis , Osteocondrodisplasias , Receptores de Sulfonilureas/genética , Animales , Cardiomegalia/genética , Cardiomegalia/metabolismo , Células Cultivadas , Femenino , Humanos , Hipertricosis/genética , Hipertricosis/metabolismo , Células Madre Pluripotentes Inducidas , Masculino , Ratones , Miocitos Cardíacos , Osteocondrodisplasias/genética , Osteocondrodisplasias/metabolismo
6.
J Clin Invest ; 130(3): 1116-1121, 2020 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-31821173

RESUMEN

Cantu syndrome (CS) is a complex disorder caused by gain-of-function (GoF) mutations in ABCC9 and KCNJ8, which encode the SUR2 and Kir6.1 subunits, respectively, of vascular smooth muscle (VSM) KATP channels. CS includes dilated vasculature, marked cardiac hypertrophy, and other cardiovascular abnormalities. There is currently no targeted therapy, and it is unknown whether cardiovascular features can be reversed once manifest. Using combined transgenic and pharmacological approaches in a knockin mouse model of CS, we have shown that reversal of vascular and cardiac phenotypes can be achieved by genetic downregulation of KATP channel activity specifically in VSM, and by chronic administration of the clinically used KATP channel inhibitor, glibenclamide. These findings demonstrate that VSM KATP channel GoF underlies CS cardiac enlargement and that CS-associated abnormalities are reversible, and provide evidence of in vivo efficacy of glibenclamide as a therapeutic agent in CS.


Asunto(s)
Cardiomegalia , Gliburida/farmacología , Hipertricosis , Canales KATP , Osteocondrodisplasias , Receptores de Sulfonilureas , Animales , Cardiomegalia/tratamiento farmacológico , Cardiomegalia/genética , Cardiomegalia/metabolismo , Cardiomegalia/patología , Modelos Animales de Enfermedad , Técnicas de Sustitución del Gen , Humanos , Hipertricosis/tratamiento farmacológico , Hipertricosis/genética , Hipertricosis/metabolismo , Hipertricosis/patología , Canales KATP/genética , Canales KATP/metabolismo , Ratones , Ratones Transgénicos , Osteocondrodisplasias/tratamiento farmacológico , Osteocondrodisplasias/genética , Osteocondrodisplasias/metabolismo , Osteocondrodisplasias/patología , Receptores de Sulfonilureas/genética , Receptores de Sulfonilureas/metabolismo
7.
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
8.
Horm Res Paediatr ; 91(5): 346-355, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30625464

RESUMEN

BACKGROUND: The term "H syndrome" was coined to denote the major clinical findings, which include hyperpigmentation, hypertrichosis, hearing loss, hepatosplenomegaly, hyperglycaemia, hypogonadism, hallux flexion contractures, and short height. OBJECTIVE: To report the clinical, endocrinological, histochemical, and genetic findings of three siblings. METHODS: Skin and liver biopsies were taken to investigate the histochemical characteristics of hyperpigmented hypertrichotic skin lesions and massive hepatomegaly. The levels of basal serum thyroid hormones, oestradiol, total testosterone, follicle-stimulating hormone, luteinising hormone, and stimulated growth hormone (GH) were measured to investigate the endocrine aspects of the syndrome. Mutation analysis was carried out in all six exons and exon-intron boundaries of SLC29A3 by direct sequencing. RESULTS: Physical examination of the patients revealed common charac-teristic findings of H syndrome. Additional clinical findings were sectorial iris atrophy in the younger sister. Laboratory evaluation revealed microcytic anaemia, markedly increased erythrocyte sedimentation rate and C-reactive protein levels, and humoral immune deficiency in the younger siblings, who presented with recurrent fever and sinopulmonary infection. Two different GH stimulation tests revealed GH deficiency in the younger sister with short stature. Liver and skin biopsies revealed polyclonal lymphohistiocytic and plasma cell infiltration. Sequencing of SLC29A3 in the three siblings revealed a novel homozygous mutation in exon 6, which caused the transition of arginine to tryptophan. CONCLUSION: This study not only extended the clinical and mutation spectrum of SLC29A3 in H syndrome, but also showed that short children should be assessed according to the guidelines for short stature in children.


Asunto(s)
Contractura , Pérdida Auditiva Sensorineural , Histiocitosis , Hiperpigmentación , Hipertricosis , Mutación , Proteínas de Transporte de Nucleósidos , Hermanos , Adolescente , Adulto , Niño , Contractura/diagnóstico , Contractura/genética , Contractura/metabolismo , Contractura/patología , Análisis Mutacional de ADN , Exones , Familia , Femenino , Pérdida Auditiva Sensorineural/diagnóstico , Pérdida Auditiva Sensorineural/genética , Pérdida Auditiva Sensorineural/metabolismo , Pérdida Auditiva Sensorineural/patología , Histiocitosis/diagnóstico , Histiocitosis/genética , Histiocitosis/metabolismo , Histiocitosis/patología , Humanos , Hiperpigmentación/diagnóstico , Hiperpigmentación/genética , Hiperpigmentación/metabolismo , Hiperpigmentación/patología , Hipertricosis/diagnóstico , Hipertricosis/genética , Hipertricosis/metabolismo , Hipertricosis/patología , Masculino , Proteínas de Transporte de Nucleósidos/genética , Proteínas de Transporte de Nucleósidos/metabolismo , Síndrome , Turquía
9.
Neuromolecular Med ; 20(3): 409-417, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30014449

RESUMEN

A growing number of histone modifiers are involved in human neurodevelopmental disorders, suggesting that proper regulation of chromatin state is essential for the development of the central nervous system. Among them, heterozygous de novo variants in KMT2A, a gene coding for histone methyltransferase, have been associated with Wiedemann-Steiner syndrome (WSS), a rare developmental disorder mainly characterized by intellectual disability (ID) and hypertrichosis. As KMT2A is known to regulate the expression of multiple target genes through methylation of lysine 4 of histone 3 (H3K4me), we sought to investigate the transcriptomic consequences of KMT2A variants involved in WSS. Using fibroblasts from four WSS patients harboring loss-of-function KMT2A variants, we performed RNA sequencing and identified a number of genes for which transcription was altered in KMT2A-mutated cells compared to the control ones. Strikingly, analysis of the pathways and biological functions significantly deregulated between patients with WSS and healthy individuals revealed a number of processes predicted to be altered that are relevant for hypertrichosis and intellectual disability, the cardinal signs of this disease.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/genética , Histonas/metabolismo , Hipertricosis/genética , Hipertricosis/metabolismo , Discapacidad Intelectual/genética , Discapacidad Intelectual/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/genética , Enfermedades Raras/genética , Enfermedades Raras/metabolismo , Adolescente , Adulto , Niño , Femenino , Fibroblastos , Regulación de la Expresión Génica , Humanos , Mutación con Pérdida de Función , Lisina/metabolismo , Masculino , Metilación , Persona de Mediana Edad , Óxido Nítrico Sintasa de Tipo III/metabolismo , Análisis de Secuencia de ARN , Transducción de Señal , Síndrome , Transcripción Genética , Adulto Joven
11.
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
12.
J Biol Chem ; 292(42): 17387-17398, 2017 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-28842488

RESUMEN

Cantu syndrome (CS) is a condition characterized by a range of anatomical defects, including cardiomegaly, hyperflexibility of the joints, hypertrichosis, and craniofacial dysmorphology. CS is associated with multiple missense mutations in the genes encoding the regulatory sulfonylurea receptor 2 (SUR2) subunits of the ATP-sensitive K+ (KATP) channel as well as two mutations (V65M and C176S) in the Kir6.1 (KCNJ8) subunit. Previous analysis of leucine and alanine substitutions at the Val-65-equivalent site (Val-64) in Kir6.2 indicated no major effects on channel function. In this study, we characterized the effects of both valine-to-methionine and valine-to-leucine substitutions at this position in both Kir6.1 and Kir6.2 using ion flux and patch clamp techniques. We report that methionine substitution, but not leucine substitution, results in increased open state stability and hence significantly reduced ATP sensitivity and a marked increase of channel activity in the intact cell irrespective of the identity of the coassembled SUR subunit. Sulfonylurea inhibitors, such as glibenclamide, are potential therapies for CS. However, as a consequence of the increased open state stability, both Kir6.1(V65M) and Kir6.2(V64M) mutations essentially abolish high-affinity sensitivity to the KATP blocker glibenclamide in both intact cells and excised patches. This raises the possibility that, at least for some CS mutations, sulfonylurea therapy may not prove to be successful and highlights the need for detailed pharmacogenomic analyses of CS mutations.


Asunto(s)
Cardiomegalia/metabolismo , Hipertricosis/metabolismo , Canales KATP/metabolismo , Mutación Missense , Osteocondrodisplasias/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Sustitución de Aminoácidos , Animales , Células COS , Cardiomegalia/genética , Chlorocebus aethiops , Gliburida/farmacología , Humanos , Hipertricosis/genética , Canales KATP/química , Canales KATP/genética , Ratones , Osteocondrodisplasias/genética , Técnicas de Placa-Clamp , Canales de Potasio de Rectificación Interna/química , Canales de Potasio de Rectificación Interna/genética , Estabilidad Proteica/efectos de los fármacos , Ratas
13.
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
14.
Future Med Chem ; 8(7): 789-802, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27161588

RESUMEN

ATP-sensitive potassium (KATP) channels play fundamental roles in the regulation of endocrine, neural and cardiovascular function. Small-molecule inhibitors (e.g., sulfonylurea drugs) or activators (e.g., diazoxide) acting on SUR1 or SUR2 have been used clinically for decades to manage the inappropriate secretion of insulin in patients with Type 2 diabetes, hyperinsulinism and intractable hypertension. More recently, the discovery of rare disease-causing mutations in KATP channel-encoding genes has highlighted the need for new therapeutics for the treatment of certain forms of neonatal diabetes mellitus, congenital hyperinsulinism and Cantu syndrome. Here, we provide a high-level overview of the pathophysiology of these diseases and discuss the development of a flexible high-throughput screening platform to enable the development of new classes of KATP channel modulators.


Asunto(s)
Canalopatías/tratamiento farmacológico , Canales KATP/metabolismo , Nucleótidos de Adenina/farmacología , Nucleótidos de Adenina/uso terapéutico , Animales , Cardiomegalia/tratamiento farmacológico , Cardiomegalia/metabolismo , Canalopatías/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Hiperinsulinismo/congénito , Hiperinsulinismo/tratamiento farmacológico , Hipertricosis/tratamiento farmacológico , Hipertricosis/metabolismo , Hipoglucemiantes/farmacología , Hipoglucemiantes/uso terapéutico , Activación del Canal Iónico , Canales KATP/antagonistas & inhibidores , Osteocondrodisplasias/tratamiento farmacológico , Osteocondrodisplasias/metabolismo , Transporte de Proteínas
15.
No To Hattatsu ; 48(5): 325-31, 2016 09.
Artículo en Japonés | MEDLINE | ID: mdl-30010274

RESUMEN

Multiple mutations in Kir6.x and SURx genes have implicated ATP-sensitive potassium (KATP) channels and, as a result, have led to diverse diseases, ranging from diabetes and hyperinsulinism to cardiac arrhythmias and cardiovascular disease. These diseases are referred to as KATP channelopathies. Recently, Cantú syndrome (CS), which was found to be caused by mutations in the ABCC9 or KCNJ8 gene, was newly added to the list of KATP channelopathies. CS is a rare multi-organ disease characterized by congenital hypertrichosis, characteristic face, persistent ductus arteriosus, cardiomegaly, intrauterine overgrowth, and skeletal abnormalities. Congenital hypertrichosis and coarse face have been confirmed in all CS patients. On the other hand, cardiovascular and skeletal abnormalities vary widely in severity, even in some familial cases and in isolated cases sharing the same mutation. Information about genotype-phenotype correlations in CS are described here.


Asunto(s)
Cardiomegalia/metabolismo , Hipertricosis/metabolismo , Canales KATP/metabolismo , Osteocondrodisplasias/metabolismo , Cardiomegalia/genética , Diseño de Fármacos , Humanos , Hipertricosis/genética , Canales KATP/genética , Mutación , Osteocondrodisplasias/genética
16.
J Gen Physiol ; 146(6): 527-40, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26621776

RESUMEN

Cantú syndrome (CS) is a rare disease characterized by congenital hypertrichosis, distinct facial features, osteochondrodysplasia, and cardiac defects. Recent genetic analysis has revealed that the majority of CS patients carry a missense mutation in ABCC9, which codes for the sulfonylurea receptor SUR2. SUR2 subunits couple with Kir6.x, inwardly rectifying potassium pore-forming subunits, to form adenosine triphosphate (ATP)-sensitive potassium (K(ATP)) channels, which link cell metabolism to membrane excitability in a variety of tissues including vascular smooth muscle, skeletal muscle, and the heart. The functional consequences of multiple uncharacterized CS mutations remain unclear. Here, we have focused on determining the functional consequences of three documented human CS-associated ABCC9 mutations: human P432L, A478V, and C1043Y. The mutations were engineered in the equivalent position in rat SUR2A (P429L, A475V, and C1039Y), and each was coexpressed with mouse Kir6.2. Using macroscopic rubidium ((86)Rb(+)) efflux assays, we show that K(ATP) channels formed with P429L, A475V, or C1039Y mutants enhance K(ATP) activity compared with wild-type (WT) channels. We used inside-out patch-clamp electrophysiology to measure channel sensitivity to ATP inhibition and to MgADP activation. For P429L and A475V mutants, sensitivity to ATP inhibition was comparable to WT channels, but activation by MgADP was significantly greater. C1039Y-dependent channels were significantly less sensitive to inhibition by ATP or by glibenclamide, but MgADP activation was comparable to WT. The results indicate that these three CS mutations all lead to overactive K(ATP) channels, but at least two mechanisms underlie the observed gain of function: decreased ATP inhibition and enhanced MgADP activation.


Asunto(s)
Cardiomegalia/genética , Hipertricosis/genética , Mutación Missense , Osteocondrodisplasias/genética , Receptores de Sulfonilureas/metabolismo , Potenciales de Acción , Adenosina Trifosfato/metabolismo , Secuencia de Aminoácidos , Animales , Células COS , Cardiomegalia/metabolismo , Chlorocebus aethiops , Humanos , Hipertricosis/metabolismo , Ratones , Datos de Secuencia Molecular , Osteocondrodisplasias/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Receptores de Sulfonilureas/química , Receptores de Sulfonilureas/genética
17.
Med Hypotheses ; 85(6): 969-71, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26392140

RESUMEN

It is hypothesized that a topical application of a sulfonylurea drug, which can inhibit the ATP-sensitive potassium-gated channels (Kir6.X/SUR) present in human hair bulb tissues, will inhibit hair growth in a targeted manner. Diazoxide is used to treat severe hypoglycemia due to hyperinsulinism of infancy. However, this often results in hypertrichosis that can be severe enough to prevent its use. Diazoxide blocks insulin release from the pancreas by opening the SUR1/Kir6.2 channels in ß-cells. Diazoxide can also act on two potassium-gated channels in the skin that affect hair growth, namely SUR1/Kir6.2 and SUR2B/Kir6.1, thus causing hypertrichosis. It is proposed that a topical sulfonylurea will inhibit the excessive hair growth due to diazoxide, but will not impact the beneficial effects of diazoxide on beta cells. This approach can also be applied to rare cases of Cantú syndrome, caused by mutations in ABCC9 (coding for SUR2) or in KCNJ8 (coding for Kir6.1) that is characterized by congenital hypertrichosis. More importantly, this approach may also be effective in treating other forms of hypertrichosis or hirsutism, that are quite common, yet very distressing to patients worldwide.


Asunto(s)
Diazóxido/administración & dosificación , Hipertricosis/tratamiento farmacológico , Compuestos de Sulfonilurea/administración & dosificación , Administración Tópica , Cardiomegalia/tratamiento farmacológico , Cabello , Humanos , Hipertricosis/metabolismo , Mutación , Osteocondrodisplasias/tratamiento farmacológico , Canales de Potasio/efectos de los fármacos , Receptores de Sulfonilureas/genética , Receptores de Sulfonilureas/metabolismo
18.
Ageing Res Rev ; 24(Pt B): 111-25, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26226329

RESUMEN

The ABCC9 gene and its polypeptide product, SUR2, are increasingly implicated in human neurologic disease, including prevalent diseases of the aged brain. SUR2 proteins are a component of the ATP-sensitive potassium ("KATP") channel, a metabolic sensor for stress and/or hypoxia that has been shown to change in aging. The KATP channel also helps regulate the neurovascular unit. Most brain cell types express SUR2, including neurons, astrocytes, oligodendrocytes, microglia, vascular smooth muscle, pericytes, and endothelial cells. Thus it is not surprising that ABCC9 gene variants are associated with risk for human brain diseases. For example, Cantu syndrome is a result of ABCC9 mutations; we discuss neurologic manifestations of this genetic syndrome. More common brain disorders linked to ABCC9 gene variants include hippocampal sclerosis of aging (HS-Aging), sleep disorders, and depression. HS-Aging is a prevalent neurological disease with pathologic features of both neurodegenerative (aberrant TDP-43) and cerebrovascular (arteriolosclerosis) disease. As to potential therapeutic intervention, the human pharmacopeia features both SUR2 agonists and antagonists, so ABCC9/SUR2 may provide a "druggable target", relevant perhaps to both HS-Aging and Alzheimer's disease. We conclude that more work is required to better understand the roles of ABCC9/SUR2 in the human brain during health and disease conditions.


Asunto(s)
Envejecimiento/fisiología , Encefalopatías , Cardiomegalia , Hipocampo , Hipertricosis , Osteocondrodisplasias , Receptores de Sulfonilureas , Animales , Encefalopatías/tratamiento farmacológico , Encefalopatías/metabolismo , Encefalopatías/patología , Cardiomegalia/genética , Cardiomegalia/metabolismo , Cardiomegalia/patología , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Hipertricosis/genética , Hipertricosis/metabolismo , Hipertricosis/patología , Mutación , Osteocondrodisplasias/genética , Osteocondrodisplasias/metabolismo , Osteocondrodisplasias/patología , Receptores de Sulfonilureas/agonistas , Receptores de Sulfonilureas/antagonistas & inhibidores , Receptores de Sulfonilureas/genética , Terapias en Investigación
20.
PLoS Genet ; 8(11): e1003002, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23133399

RESUMEN

Hereditary hypertrichoses are a group of hair overgrowth syndromes that are extremely rare in humans. We have previously demonstrated that a position effect on TRPS1 is associated with hypertrichosis in humans and mice. To gain insight into the functional role of Trps1, we analyzed the late morphogenesis vibrissae phenotype of Trps1(Δgt) mutant mice, which is characterized by follicle degeneration after peg downgrowth has been initiated. We found that Trps1 directly represses expression of the hair follicle stem cell regulator Sox9 to control proliferation of the follicle epithelium. Furthermore, we identified a copy number variation upstream of SOX9 in a family with hypertrichosis that significantly decreases expression of the gene in the hair follicle, providing new insights into the long-range regulation of SOX9. Our findings uncover a novel transcriptional hierarchy that regulates epithelial proliferation in the developing hair follicle and contributes to the pathology of hypertrichosis.


Asunto(s)
Proteínas de Unión al ADN , Folículo Piloso , Hipertricosis , Factor de Transcripción SOX9 , Factores de Transcripción , Animales , Proliferación Celular , Variaciones en el Número de Copia de ADN/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Regulación del Desarrollo de la Expresión Génica , Folículo Piloso/crecimiento & desarrollo , Folículo Piloso/metabolismo , Humanos , Hipertricosis/metabolismo , Hipertricosis/patología , Ratones , Morfogénesis , Mutación , Proteínas Represoras , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA