Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Physiol ; 596(24): 6205-6217, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30179258

RESUMEN

KEY POINTS: ß-Cell KATP channels are partially open in the absence of metabolic substrates, whereas cardiac KATP channels are closed. Using cloned channels heterologously expressed in Xenopus oocytes we measured the effect of MgADP on the MgATP concentration-inhibition curve immediately after patch excision. MgADP caused a far more striking reduction in ATP inhibition of Kir6.2/SUR1 channels than Kir6.2/SUR2A channels; this effect declined rapidly after patch excision. Exchanging the final 42 amino acids of SUR was sufficient to switch the Mg-nucleotide regulation of Kir6.2/SUR1 and Kir6.2/SUR2A channels, and partially switch their sensitivity to metabolic inhibition. Deletion of the C-terminal 42 residues of SUR abolished MgADP activation of both Kir6.2/SUR1 and Kir6.2/SUR2A channels. We conclude that the different metabolic sensitivity of Kir6.2/SUR1 and Kir6.2/SUR2A channels is at least partially due to their different regulation by Mg-nucleotides, which is determined by the final 42 amino acids. ABSTRACT: ATP-sensitive potassium (KATP ) channels couple the metabolic state of a cell to its electrical activity and play important physiological roles in many tissues. In contrast to ß-cell (Kir6.2/SUR1) channels, which open when extracellular glucose levels fall, cardiac (Kir6.2/SUR2A) channels remain closed. This is due to differences in the SUR subunit rather than cell metabolism. As ATP inhibition and MgADP activation are similar for both types of channels, we investigated channel inhibition by MgATP in the presence of 100 µm MgADP immediately after patch excision [when the channel open probability (PO ) is near maximal]. The results were strikingly different: 100 µm MgADP substantially reduced MgATP inhibition of Kir6.2/SUR1, but had no effect on MgATP inhibition of Kir6.2/SUR2A. Exchanging the final 42 residues of SUR2A with that of SUR1 switched the channel phenotype (and vice versa), and deleting this region abolished Mg-nucleotide activation. This suggests the C-terminal 42 residues are important for the ability of MgADP to influence ATP inhibition at Kir6.2. This region was also necessary, but not sufficient, for activation of the KATP channel in intact cells by metabolic inhibition (azide). We conclude that the ability of MgADP to impair ATP inhibition at Kir6.2 accounts, in part, for the differential metabolic sensitivities of ß-cell and cardiac KATP channels.


Asunto(s)
Adenosina Difosfato/farmacología , Células Secretoras de Insulina/fisiología , Canales KATP/metabolismo , Canales de Potasio de Rectificación Interna/farmacología , Receptores de Sulfonilureas/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Regulación de la Expresión Génica/efectos de los fármacos , Canales KATP/genética , Oocitos , Técnicas de Placa-Clamp , Xenopus laevis
2.
Diabetologia ; 59(7): 1430-1436, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27118464

RESUMEN

AIMS/HYPOTHESIS: The pancreatic ATP-sensitive potassium (KATP) channel plays a pivotal role in linking beta cell metabolism to insulin secretion. Mutations in KATP channel genes can result in hypo- or hypersecretion of insulin, as in neonatal diabetes mellitus and congenital hyperinsulinism, respectively. To date, all patients affected by neonatal diabetes due to a mutation in the pore-forming subunit of the channel (Kir6.2, KCNJ11) are heterozygous for the mutation. Here, we report the first clinical case of neonatal diabetes caused by a homozygous KCNJ11 mutation. METHODS: A male patient was diagnosed with diabetes shortly after birth. At 5 months of age, genetic testing revealed he carried a homozygous KCNJ11 mutation, G324R, (Kir6.2-G324R) and he was successfully transferred to sulfonylurea therapy (0.2 mg kg(-1) day(-1)). Neither heterozygous parent was affected. Functional properties of wild-type, heterozygous and homozygous mutant KATP channels were examined after heterologous expression in Xenopus oocytes. RESULTS: Functional studies indicated that the Kir6.2-G324R mutation reduces the channel ATP sensitivity but that the difference in ATP inhibition between homozygous and heterozygous channels is remarkably small. Nevertheless, the homozygous patient developed neonatal diabetes, whereas the heterozygous parents were, and remain, unaffected. Kir6.2-G324R channels were fully shut by the sulfonylurea tolbutamide, which explains why the patient's diabetes was well controlled by sulfonylurea therapy. CONCLUSIONS/INTERPRETATION: The data demonstrate that tiny changes in KATP channel activity can alter beta cell electrical activity and insulin secretion sufficiently to cause diabetes. They also aid our understanding of how the Kir6.2-E23K variant predisposes to type 2 diabetes.


Asunto(s)
Adenosina Trifosfato/metabolismo , Diabetes Mellitus Tipo 2/genética , Canales de Potasio de Rectificación Interna/metabolismo , Adulto , Animales , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Electrofisiología , Femenino , Predisposición Genética a la Enfermedad/genética , Heterocigoto , Homocigoto , Humanos , Lactante , Insulina/metabolismo , Masculino , Mutación/genética , Canales de Potasio de Rectificación Interna/genética , Compuestos de Sulfonilurea/uso terapéutico , Xenopus
3.
Biochem Soc Trans ; 43(5): 901-7, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26517901

RESUMEN

Sulphonylureas stimulate insulin secretion from pancreatic ß-cells primarily by closing ATP-sensitive K(+) channels in the ß-cell plasma membrane. The mechanism of channel inhibition by these drugs is unusually complex. As direct inhibitors of channel activity, sulphonylureas act only as partial antagonists at therapeutic concentrations. However, they also exert an additional indirect inhibitory effect via modulation of nucleotide-dependent channel gating. In this review, we summarize current knowledge and recent advances in our understanding of the molecular mechanism of action of these drugs.


Asunto(s)
Adenosina Trifosfato/metabolismo , Activación del Canal Iónico/efectos de los fármacos , Canales KATP/metabolismo , Compuestos de Sulfonilurea/farmacología , Diabetes Mellitus/tratamiento farmacológico , Diabetes Mellitus/metabolismo , Humanos , Recién Nacido , Insulina/metabolismo , Secreción de Insulina , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Potenciales de la Membrana/efectos de los fármacos
4.
Nat Commun ; 15(1): 4173, 2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38755204

RESUMEN

Potassium channels of the Two-Pore Domain (K2P) subfamily, KCNK1-KCNK18, play crucial roles in controlling the electrical activity of many different cell types and represent attractive therapeutic targets. However, the identification of highly selective small molecule drugs against these channels has been challenging due to the high degree of structural and functional conservation that exists not only between K2P channels, but across the whole K+ channel superfamily. To address the issue of selectivity, here we generate camelid antibody fragments (nanobodies) against the TREK-2 (KCNK10) K2P K+ channel and identify selective binders including several that directly modulate channel activity. X-ray crystallography and CryoEM data of these nanobodies in complex with TREK-2 also reveal insights into their mechanisms of activation and inhibition via binding to the extracellular loops and Cap domain, as well as their suitability for immunodetection. These structures facilitate design of a biparatropic inhibitory nanobody with markedly improved sensitivity. Together, these results provide important insights into TREK channel gating and provide an alternative, more selective approach to modulation of K2P channel activity via their extracellular domains.


Asunto(s)
Canales de Potasio de Dominio Poro en Tándem , Anticuerpos de Dominio Único , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Anticuerpos de Dominio Único/metabolismo , Anticuerpos de Dominio Único/inmunología , Anticuerpos de Dominio Único/química , Humanos , Cristalografía por Rayos X , Animales , Microscopía por Crioelectrón , Células HEK293 , Modelos Moleculares
5.
J Clin Invest ; 119(1): 80-90, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19065048

RESUMEN

Neonatal diabetes is a rare monogenic form of diabetes that usually presents within the first six months of life. It is commonly caused by gain-of-function mutations in the genes encoding the Kir6.2 and SUR1 subunits of the plasmalemmal ATP-sensitive K+ (KATP) channel. To better understand this disease, we generated a mouse expressing a Kir6.2 mutation (V59M) that causes neonatal diabetes in humans and we used Cre-lox technology to express the mutation specifically in pancreatic beta cells. These beta-V59M mice developed severe diabetes soon after birth, and by 5 weeks of age, blood glucose levels were markedly increased and insulin was undetectable. Islets isolated from beta-V59M mice secreted substantially less insulin and showed a smaller increase in intracellular calcium in response to glucose. This was due to a reduced sensitivity of KATP channels in pancreatic beta cells to inhibition by ATP or glucose. In contrast, the sulfonylurea tolbutamide, a specific blocker of KATP channels, closed KATP channels, elevated intracellular calcium levels, and stimulated insulin release in beta-V59M beta cells, indicating that events downstream of KATP channel closure remained intact. Expression of the V59M Kir6.2 mutation in pancreatic beta cells alone is thus sufficient to recapitulate the neonatal diabetes observed in humans. beta-V59M islets also displayed a reduced percentage of beta cells, abnormal morphology, lower insulin content, and decreased expression of Kir6.2, SUR1, and insulin mRNA. All these changes are expected to contribute to the diabetes of beta-V59M mice. Their cause requires further investigation.


Asunto(s)
Diabetes Mellitus/genética , Modelos Animales de Enfermedad , Células Secretoras de Insulina/metabolismo , Mutación , Canales de Potasio de Rectificación Interna/genética , Subunidades de Proteína/genética , Adenosina Trifosfato/metabolismo , Animales , Calcio/metabolismo , Diabetes Mellitus/metabolismo , Femenino , Humanos , Hipoglucemiantes/farmacología , Recién Nacido , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/efectos de los fármacos , Masculino , Ratones , Ratones Transgénicos , Técnicas de Placa-Clamp , Canales de Potasio de Rectificación Interna/metabolismo , Subunidades de Proteína/metabolismo , Tolbutamida/farmacología
6.
Nat Genet ; 54(10): 1534-1543, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36195757

RESUMEN

Sleep apnea is a common disorder that represents a global public health burden. KCNK3 encodes TASK-1, a K+ channel implicated in the control of breathing, but its link with sleep apnea remains poorly understood. Here we describe a new developmental disorder with associated sleep apnea (developmental delay with sleep apnea, or DDSA) caused by rare de novo gain-of-function mutations in KCNK3. The mutations cluster around the 'X-gate', a gating motif that controls channel opening, and produce overactive channels that no longer respond to inhibition by G-protein-coupled receptor pathways. However, despite their defective X-gating, these mutant channels can still be inhibited by a range of known TASK channel inhibitors. These results not only highlight an important new role for TASK-1 K+ channels and their link with sleep apnea but also identify possible therapeutic strategies.


Asunto(s)
Mutación con Ganancia de Función , Síndromes de la Apnea del Sueño , Niño , Discapacidades del Desarrollo , Humanos , Mutación/genética , Proteínas del Tejido Nervioso , Canales de Potasio de Dominio Poro en Tándem , Síndromes de la Apnea del Sueño/genética
7.
PLoS One ; 16(10): e0258275, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34618865

RESUMEN

In addition to the classical voltage-dependent behavior mediated by the voltage-sensing-domains (VSD) of ion channels, a growing number of voltage-dependent gating behaviors are being described in channels that lack canonical VSDs. A common thread in their mechanism of action is the contribution of the permeating ion to this voltage sensing process. The polymodal K2P K+ channel, TREK2 responds to membrane voltage through a gating process mediated by the interaction of K+ with its selectivity filter. Recently, we found that this action can be modulated by small molecule agonists (e.g. BL1249) which appear to have an electrostatic influence on K+ binding within the inner cavity and produce an increase in the single-channel conductance of TREK-2 channels. Here, we directly probed this K+-dependent gating process by recording both macroscopic and single-channel currents of TREK-2 in the presence of high concentrations of internal K+. Surprisingly we found TREK-2 is inhibited by high internal K+ concentrations and that this is mediated by the concomitant increase in ionic-strength. However, we were still able to determine that the increase in single channel conductance in the presence of BL1249 was blunted in high ionic-strength, whilst its activatory effect (on channel open probability) persisted. These effects are consistent with an electrostatic mechanism of action of negatively charged activators such as BL1249 on permeation, but also suggest that their influence on channel gating is complex.


Asunto(s)
Permeabilidad de la Membrana Celular , Activación del Canal Iónico , Concentración Osmolar , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Aniones , Células HEK293 , Humanos , Presión Osmótica , Conformación Proteica
8.
J Food Drug Anal ; 29(4): 653-675, 2021 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-35649140

RESUMEN

Oxytocin (Oxt) is known to regulate social communication, stress and body weight. The activation of Oxt receptors (OTR) has clinical potential to abate stress disorders and metabolic syndrome. Kamikihito (KKT) is a traditional Japanese medicine used to treat psychological stress-related disorders. We investigated the effects of KKT, its ingredients and chemical components on Oxt neurons and OTR. C-Fos expression was examined after oral and peripheral administration of KKT in rats. Electrophysiological change of Oxt neurons and Oxt release upon application of KKT were measured in rat brain slice. The direct effect of KKT, its ingredients and its chemical components were examined by cytosolic Ca2+([Ca2+]i) measurement in Oxt neurons and OTR-expressing HEK293 cells. Both intraperitoneal and oral administration of KKT in rats induced c-Fos expression in neurons of the paraventricular nucleus (PVN) including Oxt neurons. Application of KKT induced activation of Oxt neurons and Oxt release. KKT increased [Ca2+]i in OTR-expressing HEK293 cells, and failed to activate with OTR antagonist. KKT-induced PVN Oxt neuron activation was also attenuated by OTR antagonist. Seven chemical components (rutin, ursolic acid, (Z )-butylidenephtalide, p-cymene, senkunolide, [6]-shogaol, [8]-shogaol) of three ingredients (Zizyphi Fructus, Angelicae Acutilobae Radix, Zingiberis Rhizoma) from KKT had potential to activate OTR. KKT can directly activate PVN Oxt neurons by interacting with OTR. The interaction of seven chemical components from KKT may contribute to activate OTR. Effect of KKT on Oxt neurons and OTR may contribute to the treatment of Oxt related disorders.


Asunto(s)
Oxitocina , Receptores de Oxitocina , Animales , Células HEK293 , Humanos , Japón , Medicina Tradicional de Asia Oriental , Oxitocina/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Receptores de Oxitocina/genética , Receptores de Oxitocina/metabolismo
9.
J Gen Physiol ; 153(8)2021 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-34032848

RESUMEN

The TREK subfamily of two-pore domain K+ (K2P) channels are inhibited by fluoxetine and its metabolite, norfluoxetine (NFx). Although not the principal targets of this antidepressant, TREK channel inhibition by NFx has provided important insights into the conformational changes associated with channel gating and highlighted the role of the selectivity filter in this process. However, despite the availability of TREK-2 crystal structures with NFx bound, the precise mechanisms underlying NFx inhibition remain elusive. NFx has previously been proposed to be a state-dependent inhibitor, but its binding site suggests many possible ways in which this positively charged drug might inhibit channel activity. Here we show that NFx exerts multiple effects on single-channel behavior that influence both the open and closed states of the channel and that the channel can become highly activated by 2-APB while remaining in the down conformation. We also show that the inhibitory effects of NFx are unrelated to its positive charge but can be influenced by agonists which alter filter stability, such as ML335, as well as by an intrinsic voltage-dependent gating process within the filter. NFx therefore not only inhibits channel activity by altering the equilibrium between up and down conformations but also can directly influence filter gating. These results provide further insight into the complex allosteric mechanisms that modulate filter gating in TREK K2P channels and highlight the different ways in which filter gating can be regulated to permit polymodal regulation.


Asunto(s)
Canales de Potasio de Dominio Poro en Tándem , Sitios de Unión , Fluoxetina/análogos & derivados , Fluoxetina/farmacología , Activación del Canal Iónico , Canales de Potasio de Dominio Poro en Tándem/metabolismo
10.
Diabetes ; 70(5): 1145-1156, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33568422

RESUMEN

The ATP-sensitive K+ (KATP) channel controls blood glucose levels by coupling glucose metabolism to insulin secretion in pancreatic ß-cells. E23K, a common polymorphism in the pore-forming KATP channel subunit (KCNJ11) gene, has been linked to increased risk of type 2 diabetes. Understanding the risk-allele-specific pathogenesis has the potential to improve personalized diabetes treatment, but the underlying mechanism has remained elusive. Using a genetically engineered mouse model, we now show that the K23 variant impairs glucose-induced insulin secretion and increases diabetes risk when combined with a high-fat diet (HFD) and obesity. KATP-channels in ß-cells with two K23 risk alleles (KK) showed decreased ATP inhibition, and the threshold for glucose-stimulated insulin secretion from KK islets was increased. Consequently, the insulin response to glucose and glycemic control was impaired in KK mice fed a standard diet. On an HFD, the effects of the KK genotype were exacerbated, accelerating diet-induced diabetes progression and causing ß-cell failure. We conclude that the K23 variant increases diabetes risk by impairing insulin secretion at threshold glucose levels, thus accelerating loss of ß-cell function in the early stages of diabetes progression.


Asunto(s)
Glucosa/farmacología , Insulina/metabolismo , Adenosina Trifosfato/genética , Adenosina Trifosfato/metabolismo , Animales , Predisposición Genética a la Enfermedad/genética , Variación Genética/genética , Variación Genética/fisiología , Humanos , Secreción de Insulina/efectos de los fármacos , Canales de Potasio de Rectificación Interna/genética , Canales de Potasio de Rectificación Interna/metabolismo
11.
Prog Biophys Mol Biol ; 99(1): 7-19, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18983870

RESUMEN

ATP-sensitive potassium (K(ATP)) channels couple cell metabolism to plasmalemmal potassium fluxes in a variety of cell types. The activity of these channels is primarily determined by intracellular adenosine nucleotides, which have both inhibitory and stimulatory effects. The role of K(ATP) channels has been studied most extensively in pancreatic beta-cells, where they link glucose metabolism to insulin secretion. Many mutations in K(ATP) channel subunits (Kir6.2, SUR1) have been identified that cause either neonatal diabetes or congenital hyperinsulinism. Thus, a mechanistic understanding of K(ATP) channel behavior is necessary for modeling beta-cell electrical activity and insulin release in both health and disease. Here, we review recent advances in the K(ATP) channel structure and function. We focus on the molecular mechanisms of K(ATP) channel gating by adenosine nucleotides, phospholipids and sulphonylureas and consider the advantages and limitations of various mathematical models of macroscopic and single-channel K(ATP) currents. Finally, we outline future directions for the development of more realistic models of K(ATP) channel gating.


Asunto(s)
Adenosina Trifosfato/metabolismo , Membrana Celular/metabolismo , Simulación por Computador , Activación del Canal Iónico/fisiología , Canales KATP/fisiología , Potenciales de la Membrana/fisiología , Modelos Biológicos
13.
EMBO Rep ; 9(7): 648-54, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18497752

RESUMEN

Activating mutations in the pore-forming Kir6.2 (KCNJ11) and regulatory sulphonylurea receptor SUR1 (ABCC8) subunits of the K(ATP) channel are a common cause of transient neonatal diabetes mellitus (TNDM). We identified a new TNDM mutation (R826W) in the first nucleotide-binding domain (NBD1) of SUR1. The mutation was found in a region that heterodimerizes with NBD2 to form catalytic site 2. Functional analysis showed that this mutation decreases MgATP hydrolysis by purified maltose-binding protein MBP-NBD1 fusion proteins. Inhibition of ATP hydrolysis by MgADP or BeF was not changed. The results indicate that the ATPase cycle lingers in the post-hydrolytic MgADP.P(i)-bound state, which is associated with channel activation. The extent of MgADP-dependent activation of K(ATP) channel activity was unaffected by the R826W mutation, but the time course of deactivation was slowed. Channel inhibition by MgATP was reduced, leading to an increase in resting whole-cell currents. In pancreatic beta cells, this would lead to less insulin secretion and thereby diabetes.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/química , Transportadoras de Casetes de Unión a ATP/genética , Adenosina Trifosfatasas/metabolismo , Diabetes Mellitus/enzimología , Diabetes Mellitus/genética , Enfermedades del Recién Nacido/genética , Mutación/genética , Canales de Potasio de Rectificación Interna/química , Canales de Potasio de Rectificación Interna/genética , Receptores de Droga/química , Receptores de Droga/genética , Adenosina Difosfato/farmacología , Adenosina Trifosfatasas/química , Adenosina Trifosfato/farmacología , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Arginina/genética , Niño , Humanos , Recién Nacido , Enfermedades del Recién Nacido/enzimología , Activación del Canal Iónico/efectos de los fármacos , Cinética , Masculino , Datos de Secuencia Molecular , Proteínas Mutantes/química , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Receptores de Sulfonilureas , Triptófano/genética
14.
Adv Exp Med Biol ; 654: 165-92, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20217498

RESUMEN

The ATP-sensitive potassium (K(ATP)) channel plays a crucial role in insulin secretion and thus glucose homeostasis. K(ATP) channel activity in the pancreatic beta-cell is finely balanced; increased activity prevents insulin secretion, whereas reduced activity stimulates insulin release. The beta-cell metabolism tightly regulates K(ATP) channel gating, and if this coupling is perturbed, two distinct disease states can result. Diabetes occurs when the K(ATP) channel fails to close in response to increased metabolism, whereas congenital hyperinsulinism results when K(ATP) channels remain closed even at very low blood glucose levels. In general there is a good correlation between the magnitude of K(ATP) current and disease severity. Mutations that cause a complete loss of K(ATP) channels in the beta-cell plasma membrane produce a severe form of congenital hyperinsulinism, whereas mutations that partially impair channel function produce a milder phenotype. Similarly mutations that greatly reduce the ATP sensitivity of the K(ATP) channel lead to a severe form of neonatal diabetes with associated neurological complications, whilst mutations that cause smaller shifts in ATP sensitivity cause neonatal diabetes alone. This chapter reviews our current understanding of the pancreatic beta-cell K(ATP) channel and highlights recent structural, functional and clinical advances.


Asunto(s)
Adenosina Trifosfato/metabolismo , Canales de Potasio/metabolismo , Adenosina Trifosfato/química , Animales , Glucemia/metabolismo , Modelos Animales de Enfermedad , Heterocigoto , Humanos , Hiperinsulinismo/genética , Hiperinsulinismo/metabolismo , Recién Nacido , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/citología , Ratones , Modelos Biológicos , Mutación , Páncreas/metabolismo
15.
Commun Chem ; 3(1): 147, 2020 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-36703430

RESUMEN

Membrane proteins are frequently modulated by specific protein-lipid interactions. The activation of human inward rectifying potassium (hKir) channels by phosphoinositides (PI) has been well characterised. Here, we apply a coarse-grained molecular dynamics free-energy perturbation (CG-FEP) protocol to capture the energetics of binding of PI lipids to hKir channels. By using either a single- or multi-step approach, we establish a consistent value for the binding of PIP2 to hKir channels, relative to the binding of the bulk phosphatidylcholine phospholipid. Furthermore, by perturbing amino acid side chains on hKir6.2, we show that the neonatal diabetes mutation E179K increases PIP2 affinity, while the congenital hyperinsulinism mutation K67N results in a reduced affinity. We show good agreement with electrophysiological data where E179K exhibits a reduction in neomycin sensitivity, implying that PIP2 binds more tightly E179K channels. This illustrates the application of CG-FEP to compare affinities between lipid species, and for annotating amino acid residues.

16.
Wellcome Open Res ; 5: 15, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-34368464

RESUMEN

Background: The K ATP channel plays a key role in glucose homeostasis by coupling metabolically generated changes in ATP to insulin secretion from pancreatic beta-cells.  Gain-of-function mutations in either the pore-forming (Kir6.2) or regulatory (SUR1) subunit of this channel are a common cause of transient neonatal diabetes mellitus (TNDM), in which diabetes presents shortly after birth but remits within the first few years of life, only to return in later life. The reasons behind this time dependence are unclear. Methods: In an attempt to understand the mechanism behind diabetes remission and relapse, we generated mice expressing the common TNDM mutation SUR1-R1183W. We employed Cre/LoxP technology for both inducible and constitutive expression of SUR1-R1183W specifically in mouse beta-cells, followed by investigation of their phenotype using glucose tolerance tests and insulin secretion from isolated islets.  Results: We found that the R1183W mutation impaired inhibition of K ATP channels by ATP when heterologously expressed in human embryonic kidney cells. However, neither induced nor constitutive expression of SUR1-R1183W in mice resulted in changes in blood glucose homeostasis, compared to littermate controls. When challenged with a high fat diet, female mice expressing SUR1-R1183W showed increased weight gain, elevated blood glucose and impaired glycaemic control, but glucose-stimulated insulin secretion from pancreatic islets appeared unchanged. Conclusions: The mouse model of TNDM did not recapitulate the human phenotype. We discuss multiple potential reasons why this might be the case. Based on our findings, we recommend future TNDM mouse models employing a gain-of-function SUR1 mutation should be created using the minimally invasive CRISPR/Cas technology, which avoids many potential pitfalls associated with the Cre/LoxP system.

17.
Endocr J ; 56(9): 1079-82, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19734693

RESUMEN

We performed a receiver operator characteristic (ROC) curve analysis of 3915 men and 2032 women. Subjects who were diagnosed with two or more factors among high blood pressure, hyperglycaemia or high triglyceride and/or low HDL were classified as the metabolic syndrome group. By performing a ROC curve analysis, we have determined the cut-off point of waist circumference (WC) and BMI to define metabolic syndrome and further calculated the sensitivity and specificity of these two factors for the diagnosis. Cut-off point for the diagnosis of metabolic syndrome was 85 cm (men) and 80 cm (women) in WC and 24 (men) and 23 (women) in BMI. By combining these two factors, the sensitivity for the diagnosis increased to more than 80%. We conclude that it is beneficial to combine both WC and BMI for diagnosis of metabolic syndrome.


Asunto(s)
Índice de Masa Corporal , Síndrome Metabólico/diagnóstico , Obesidad Abdominal/diagnóstico , Circunferencia de la Cintura , Adulto , Técnicas de Diagnóstico Endocrino , Femenino , Humanos , Japón , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Curva ROC , Valores de Referencia , Factores de Riesgo , Sensibilidad y Especificidad , Caracteres Sexuales
18.
Methods Mol Biol ; 491: 127-39, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18998089

RESUMEN

Oocytes from the Xenopus laevis represent one of the most widely used expression systems for functional characterization of ion channels. Their large size facilitates both injection of heterologous cRNA and subsequent electrophysiological recordings of ion channel currents. Furthermore, Xenopus oocytes translate cRNA very efficiently, resulting in the generation of a large number of ion channels in the plasma membrane. In this chapter, we outline methods for oocyte preparation and maintenance and describe procedures for patch-clamping of oocytes, with a special focus on the macropatch technique. We discuss some common problems associated with patch-clamping of oocytes and their use as an expression system for ion channels.


Asunto(s)
Canales de Cloruro/fisiología , Canales Iónicos/fisiología , Canales KATP/fisiología , Oocitos/fisiología , Animales , Canales de Cloruro/aislamiento & purificación , Electrofisiología/métodos , Femenino , Canales Iónicos/genética , Canales KATP/aislamiento & purificación , Técnicas de Placa-Clamp , ARN Complementario/genética , Xenopus laevis
19.
PLoS One ; 13(5): e0197634, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29772022

RESUMEN

Sulphonylurea drugs stimulate insulin secretion from pancreatic ß-cells primarily by inhibiting ATP sensitive potassium (KATP) channels in the ß-cell membrane. The effective sulphonylurea concentration at its site of action is significantly attenuated by binding to serum albumin, which makes it difficult to compare in vitro and in vivo data. We therefore measured the ability of gliclazide and glibenclamide to inhibit KATP channels and stimulate insulin secretion in the presence of serum albumin. We used this data, together with estimates of free drug concentrations from binding studies, to predict the extent of sulphonylurea inhibition of KATP channels at therapeutic concentrations in vivo. KATP currents from mouse pancreatic ß-cells and Xenopus oocytes were measured using the patch-clamp technique. Gliclazide and glibenclamide binding to human plasma were determined in spiked plasma samples using an ultrafiltration-mass spectrometry approach. Bovine serum albumin (60g/l) produced a mild, non-significant reduction of gliclazide block of KATP currents in pancreatic ß-cells and Xenopus oocytes. In contrast, glibenclamide inhibition of recombinant KATP channels was dramatically suppressed by albumin (predicted free drug concentration <0.1%). Insulin secretion was also reduced. Free concentrations of gliclazide and glibenclamide in the presence of human plasma measured in binding experiments were 15% and 0.05%, respectively. Our data suggest the free concentration of glibenclamide in plasma is too low to account for the drug's therapeutic effect. In contrast, the free gliclazide concentration in plasma is high enough to close KATP channels and stimulate insulin secretion.


Asunto(s)
Gliclazida/farmacología , Gliburida/farmacología , Hipoglucemiantes/farmacología , Canales KATP/antagonistas & inhibidores , Albúmina Sérica/farmacología , Animales , Bovinos , Células Cultivadas , Gliclazida/sangre , Gliclazida/metabolismo , Gliclazida/farmacocinética , Gliburida/sangre , Gliburida/metabolismo , Gliburida/farmacocinética , Humanos , Hipoglucemiantes/sangre , Hipoglucemiantes/metabolismo , Hipoglucemiantes/farmacocinética , Insulina/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Ratones , Ratones Endogámicos C57BL , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Técnicas de Placa-Clamp , Unión Proteica , Proteínas Recombinantes/metabolismo , Albúmina Sérica/metabolismo , Albúmina Sérica Bovina/metabolismo , Albúmina Sérica Bovina/farmacología , Xenopus laevis
20.
Diabetes ; 55(6): 1731-7, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16731836

RESUMEN

Heterozygous mutations in the human Kir6.2 gene (KCNJ11), the pore-forming subunit of the ATP-sensitive K(+) channel (K(ATP) channel), cause neonatal diabetes. To date, all mutations increase whole-cell K(ATP) channel currents by reducing channel inhibition by MgATP. Here, we provide functional characterization of two mutations (F35L and F35V) at residue F35 of Kir6.2, which lies within the NH(2)-terminus. We further show that the F35V patient can be successfully transferred from insulin to sulfonylurea therapy. The patient has been off insulin for 24 months and shows improved metabolic control (mean HbA(1c) 7.58 before and 6.18% after sulfonylurea treatment; P < 0.007). Wild-type and mutant Kir6.2 were heterologously coexpressed with SUR1 in Xenopus oocytes. Whole-cell K(ATP) channel currents through homomeric and heterozygous F35V and F35L channels were increased due to a reduced sensitivity to inhibition by MgATP. The mutation also increased the open probability (P(O)) of homomeric F35 mutant channels in the absence of ATP. These effects on P(O) and ATP sensitivity were abolished in the absence of SUR1. Our results suggest that mutations at F35 cause permanent neonatal diabetes by affecting K(ATP) channel gating and thereby, indirectly, ATP inhibition. Heterozygous F35V channels were markedly inhibited by the sulfonylurea tolbutamide, accounting for the efficacy of sulfonylurea therapy in the patient.


Asunto(s)
Diabetes Mellitus/genética , Mutación , Fenilalanina/genética , Canales de Potasio de Rectificación Interna/fisiología , Compuestos de Sulfonilurea/farmacología , Adenosina Trifosfato/metabolismo , Adenosina Trifosfato/farmacología , Animales , Diabetes Mellitus/tratamiento farmacológico , Electrofisiología , Femenino , Heterocigoto , Humanos , Hipoglucemiantes/farmacología , Hipoglucemiantes/uso terapéutico , Recién Nacido , Insulina/farmacología , Insulina/uso terapéutico , Masculino , Potenciales de la Membrana/efectos de los fármacos , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Oocitos/fisiología , Canales de Potasio de Rectificación Interna/genética , Compuestos de Sulfonilurea/uso terapéutico , Xenopus laevis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA