Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Am J Physiol Lung Cell Mol Physiol ; 319(6): L997-L1009, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32936026

RESUMEN

The gasotransmitter carbon monoxide (CO) regulates fluid and electrolyte movements across epithelial tissues. However, its action on anion channels is incompletely understood. Here, we investigate the direct action of CO on the cystic fibrosis transmembrane conductance regulator (CFTR) by applying CO-releasing molecules (CO-RMs) to the intracellular side of excised inside-out membrane patches from cells heterologously expressing wild-type human CFTR. Addition of increasing concentrations of tricarbonyldichlororuthenium(II) dimer (CORM-2) (1-300 µM) inhibited CFTR channel activity, whereas the control RuCl3 (100 µM) was without effect. CORM-2 predominantly inhibited CFTR by decreasing the frequency of channel openings and, hence, open probability (Po). But, it also reduced current flow through open channels with very fast kinetics, particularly at elevated concentrations. By contrast, the chemically distinct CO-releasing molecule CORM-3 inhibited CFTR by decreasing Po without altering current flow through open channels. Neither depolarizing the membrane voltage nor raising the ATP concentration on the intracellular side of the membrane affected CFTR inhibition by CORM-2. Interestingly, CFTR inhibition by CORM-2, but not by CFTRinh-172, was prevented by prior enhancement of channel activity by the clinically approved CFTR potentiator ivacaftor. Similarly, when added after CORM-2, ivacaftor completely relieved CFTR inhibition. In conclusion, CORM-2 has complex effects on wild-type human CFTR consistent with allosteric inhibition and open-channel blockade. Inhibition of CFTR by CO-releasing molecules suggests that CO regulates CFTR activity and that the gasotransmitter has tissue-specific effects on epithelial ion transport. The action of ivacaftor on CFTR Cl- channels inhibited by CO potentially expands the drug's clinical utility.


Asunto(s)
Monóxido de Carbono/farmacología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Activación del Canal Iónico/efectos de los fármacos , Transporte Iónico/efectos de los fármacos , Adenosina Trifosfato/metabolismo , Animales , Monóxido de Carbono/metabolismo , Humanos
2.
Sci Rep ; 6: 21975, 2016 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-26911344

RESUMEN

Optimal fetal lung growth requires anion-driven fluid secretion into the lumen of the developing organ. The fetus is hypercalcemic compared to the mother and here we show that in the developing human lung this hypercalcaemia acts on the extracellular calcium-sensing receptor, CaSR, to promote fluid-driven lung expansion through activation of the cystic fibrosis transmembrane conductance regulator, CFTR. Several chloride channels including TMEM16, bestrophin, CFTR, CLCN2 and CLCA1, are also expressed in the developing human fetal lung at gestational stages when CaSR expression is maximal. Measurements of Cl(-)-driven fluid secretion in organ explant cultures show that pharmacological CaSR activation by calcimimetics stimulates lung fluid secretion through CFTR, an effect which in humans, but not mice, was also mimicked by fetal hypercalcemic conditions, demonstrating that the physiological relevance of such a mechanism appears to be species-specific. Calcimimetics promote CFTR opening by activating adenylate cyclase and we show that Ca(2+)-stimulated type I adenylate cyclase is expressed in the developing human lung. Together, these observations suggest that physiological fetal hypercalcemia, acting on the CaSR, promotes human fetal lung development via cAMP-dependent opening of CFTR. Disturbances in this process would be expected to permanently impact lung structure and might predispose to certain postnatal respiratory diseases.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Madurez de los Órganos Fetales , Pulmón/embriología , Pulmón/metabolismo , Organogénesis , Receptores Sensibles al Calcio/metabolismo , Adenilil Ciclasas/metabolismo , Animales , Anoctamina-1 , Bestrofinas , Canales de Cloruro/genética , Canales de Cloruro/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Espacio Extracelular , Proteínas del Ojo/metabolismo , Feto , Regulación del Desarrollo de la Expresión Génica , Humanos , Hipercalcemia/genética , Hipercalcemia/metabolismo , Inmunohistoquímica , Activación del Canal Iónico , Canales Iónicos/metabolismo , Ratones , Modelos Biológicos
3.
Pflugers Arch ; 462(2): 267-79, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21559843

RESUMEN

Impairment of lung liquid absorption can lead to severe respiratory symptoms, such as those observed in pulmonary oedema. In the adult lung, liquid absorption is driven by cation transport through two pathways: a well-established amiloride-sensitive Na(+) channel (ENaC) and, more controversially, an amiloride-insensitive channel that may belong to the cyclic nucleotide-gated (CNG) channel family. Here, we show robust CNGA1 (but not CNGA2 or CNGA3) channel expression principally in rat alveolar type I cells; CNGA3 was expressed in ciliated airway epithelial cells. Using a rat in situ lung liquid clearance assay, CNG channel activation with 1 mM 8Br-cGMP resulted in an approximate 1.8-fold stimulation of lung liquid absorption. There was no stimulation by 8Br-cGMP when applied in the presence of either 100 µM L: -cis-diltiazem or 100 nM pseudechetoxin (PsTx), a specific inhibitor of CNGA1 channels. Channel specificity of PsTx and amiloride was confirmed by patch clamp experiments showing that CNGA1 channels in HEK 293 cells were not inhibited by 100 µM amiloride and that recombinant αßγ-ENaC were not inhibited by 100 nM PsTx. Importantly, 8Br-cGMP stimulated lung liquid absorption in situ, even in the presence of 50 µM amiloride. Furthermore, neither L: -cis-diltiazem nor PsTx affected the ß(2)-adrenoceptor agonist-stimulated lung liquid absorption, but, as expected, amiloride completely ablated it. Thus, transport through alveolar CNGA1 channels, located in type I cells, underlies the amiloride-insensitive component of lung liquid reabsorption. Furthermore, our in situ data highlight the potential of CNGA1 as a novel therapeutic target for the treatment of diseases characterised by lung liquid overload.


Asunto(s)
GMP Cíclico/metabolismo , Canales Catiónicos Regulados por Nucleótidos Cíclicos/metabolismo , Células Epiteliales/metabolismo , Pulmón/metabolismo , Isoformas de Proteínas/metabolismo , Alveolos Pulmonares/metabolismo , Absorción , Amilorida/metabolismo , Animales , Acuaporina 5/metabolismo , Transporte Biológico/fisiología , GMP Cíclico/análogos & derivados , Canales Catiónicos Regulados por Nucleótidos Cíclicos/antagonistas & inhibidores , Canales Catiónicos Regulados por Nucleótidos Cíclicos/genética , Diuréticos/metabolismo , Venenos Elapídicos/metabolismo , Femenino , Células HEK293 , Humanos , Activación del Canal Iónico/fisiología , Pulmón/citología , Masculino , Técnicas de Placa-Clamp , Isoformas de Proteínas/genética , Ratas , Ratas Wistar
4.
J Physiol ; 589(Pt 13): 3055-62, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21521759

RESUMEN

Carbon monoxide is rapidly emerging as an important cellular messenger, regulating a wide range of physiological processes. Crucial to its role in both physiology and disease is its ability differentially to regulate several classes of ion channels, including examples from calcium-activated K(+) (BK(Ca)), voltage-activated K(+) (K(v)) and Ca(2+) channel (L-type) families, ligand-gated P2X receptors (P2X2 and P2X4), tandem P domain K(+) channels (TREK1) and the epithelial Na(+) channel (ENaC). The mechanisms by which CO regulates these ion channels are still unclear and remain somewhat controversial. However, available structure-function studies suggest that a limited range of amino acid residues confer CO sensitivity, either directly or indirectly, to particular ion channels and that cellular redox state appears to be important to the final integrated response. Whatever the molecular mechanism by which CO regulates ion channels, endogenous production of this gasotransmitter has physiologically important roles and is currently being explored as a potential therapeutic.


Asunto(s)
Canales de Calcio/metabolismo , Monóxido de Carbono/fisiología , Activación del Canal Iónico/fisiología , Canales Iónicos/fisiología , Animales , Canales de Calcio/química , Canales de Calcio/fisiología , Señalización del Calcio/fisiología , Monóxido de Carbono/química , Humanos , Canales Iónicos/química , Relación Estructura-Actividad
5.
Exp Lung Res ; 37(5): 269-78, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21352089

RESUMEN

The authors have recently demonstrated that, in the developing mouse lung, fetal plasma Ca(2+) suppresses branching morphogenesis and cell proliferation while promoting fluid secretion via activation of the extracellular Ca(2+)-sensing receptor (CaSR). The aim of the current study was to further elucidate the role of Ca(2+) in lung development by studying the effects of extracellular Ca(2+) on fetal lung development in mice lacking the CaSR. These mice were produced by exon 5 deletion in the CaSR gene. Since such a maneuver has been known to induce the expression of an exon 5-less splice variant of the CaSR in some tissues, the molecular and functional expression of this splice variant in the developing mouse lung was also investigated. Whereas there was a mild in vivo phenotype observed in these mice, in vitro sensitivity of Casr(-/-) lung explants to specific activators of the CaSR was unaffected. These results imply that compensatory expression of an exon 5-less splice variant rescues CaSR function in this mouse model and therefore a lung-specific, complete CaSR knockout model must be developed to fully appreciate the role for this receptor in lung development and the contribution of its ablation to postnatal respiratory disease.


Asunto(s)
Calcio/metabolismo , Exones , Pulmón/crecimiento & desarrollo , Pulmón/metabolismo , Receptores Sensibles al Calcio/genética , Receptores Sensibles al Calcio/metabolismo , Animales , Señalización del Calcio/fisiología , Procesos de Crecimiento Celular/fisiología , Madurez de los Órganos Fetales/genética , Madurez de los Órganos Fetales/fisiología , Pulmón/citología , Pulmón/embriología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Técnicas de Cultivo de Órganos , Fenotipo
6.
Patient Educ Couns ; 85(3): 375-82, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21295433

RESUMEN

OBJECTIVE: The purpose of this study was to develop and evaluate a 12-week weight management intervention involving computerized self-monitoring and technology-assisted feedback with and without an enhanced behavioral component. METHODS: 120 overweight (30.5±2.6kg/m(2)) adults (45.0±10.3 years) were randomized to one of three groups: computerized self-monitoring with Basic feedback (n=45), Enhanced behavioral feedback (n=45), or wait-list control (n=30). Intervention participants used a computer software program to record dietary and physical activity information. Weekly e-mail feedback was based on computer-generated reports, and participants attended monthly measurement visits. RESULTS: The Basic and Enhanced groups experienced significant weight reduction (-2.7±3.3kg and -2.5±3.1kg) in comparison to the Control group (0.3±2.2; p<0.05). Waist circumference and systolic blood pressure also decreased in intervention groups compared to Control (p<0.01). CONCLUSIONS: A program using computerized self-monitoring, technology-assisted feedback, and monthly measurement visits produced significant weight loss after 12 weeks. However, the addition of an enhanced behavioral component did not improve the effectiveness of the program. PRACTICE IMPLICATIONS: This study suggests that healthcare professionals can effectively deliver a weight management intervention using technology-assisted strategies in a format that may complement and reduce face-to-face sessions.


Asunto(s)
Terapia Conductista , Correo Electrónico , Internet , Pérdida de Peso , Adulto , Índice de Masa Corporal , Dieta , Ejercicio Físico , Retroalimentación , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Autocuidado , Factores Socioeconómicos , Texas , Resultado del Tratamiento
7.
Nat Neurosci ; 14(1): 17-8, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21170052

RESUMEN

In the closed structure of the P2X cation channel, three α-helical transmembrane domains cross the membrane obliquely. In rat P2X2 receptors, these intersect at Thr(339). Replacing Thr(339) by lysine in one, two or three subunits progressively increased chloride permeability and reduced unitary conductance. This implies that the closed-open transition involves a symmetrical separation of the three subunits and that Thr(339) from each subunit contributes symmetrically to the open channel permeation pathway.


Asunto(s)
Permeabilidad de la Membrana Celular/fisiología , Potenciales de la Membrana/fisiología , Receptores Purinérgicos P2X2/fisiología , Adenosina Trifosfato/fisiología , Sustitución de Aminoácidos/fisiología , Animales , Cloruros/fisiología , Ratas
8.
Respir Physiol Neurobiol ; 172(3): 169-78, 2010 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-20576528

RESUMEN

Recent evidence suggests that H(2)S contributes to activation of the carotid body by hypoxia by inhibiting K(+) channels. Here, we determine both the molecular identity of the K(+) channel target within the carotid body and the biophysical characteristics of the H(2)S-evoked inhibition by analyzing native rat and human recombinant BK(Ca) channel activity in voltage-clamped, inside-out membrane patches. Rat glomus cells express the enzymes necessary for the endogenous generation of H(2)S, cystathionine-beta-synthase and cystathionine-gamma-lyase. H(2)S inhibits native carotid body and human recombinant BK(Ca) channels with IC(50) values of around 275 microM. Inhibition by H(2)S is rapid and reversible, works by a mechanism which is distinct from that suggested for CO gas regulation of this channel and does not involve an interaction with either the "Ca bowl" or residues distal to this Ca(2+)-sensing domain. These data show that BK(Ca) is a K(+) channel target of H(2)S, and suggest a mechanism to explain the H(2)S-dependent component of O(2) sensing in the carotid body.


Asunto(s)
Contaminantes Atmosféricos/farmacología , Sulfuro de Hidrógeno/farmacología , Canales de Potasio de Gran Conductancia Activados por el Calcio/antagonistas & inhibidores , Bloqueadores de los Canales de Potasio , Animales , Cuerpo Carotídeo/efectos de los fármacos , Cuerpo Carotídeo/metabolismo , Línea Celular , Cistationina betasintasa/metabolismo , Cistationina gamma-Liasa/metabolismo , Electrofisiología , Humanos , Inmunohistoquímica , Canales de Potasio de Gran Conductancia Activados por el Calcio/genética , Masculino , Mutación , Técnicas de Placa-Clamp , Cianuro de Potasio/farmacología , ARN Mensajero/biosíntesis , Ratas , Ratas Wistar , Proteínas Recombinantes/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
9.
Ann N Y Acad Sci ; 1177: 112-8, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19845613

RESUMEN

The ability of ion channels to respond to an acute perturbation in oxygen tension is a widespread phenomenon, which encompasses many of the major ion channel families. Integral to the ability of several ion channels to respond to acute hypoxic challenge is modulation by upstream enzymatic reactions, suggesting that many ion channels sense oxygen via enzyme-linked processes. Several enzyme-linked oxygen sensing systems have been proposed, including nicotinamide adenine dinucleotide phosphate (NADPH) oxidase-dependent production of hydrogen peroxide, hemoxygenase-dependent generation of carbon monoxide, adenosine monophosphate (AMP) kinase-dependent channel phosphorylation, and src-Lck protein tyrosine kinase, via a currently undetermined mechanism. Each of these enzymes has been shown to endow specific ion channels with the ability to respond to changes in oxygen, with hypoxia exclusively evoking channel inhibition. This article reviews these proposed mechanisms and presents new insights into how one system, hemeoxygenase-2, confers oxygen sensitivity to large conductance, voltage- and calcium-activated potassium channels.


Asunto(s)
Oxígeno/metabolismo , Canales de Potasio/metabolismo , Adenilato Quinasa/metabolismo , Animales , Humanos , Hipoxia/metabolismo , Hipoxia/fisiopatología , NADPH Oxidasas/metabolismo , Familia-src Quinasas/metabolismo
10.
Pflugers Arch ; 458(6): 1007-22, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19484257

RESUMEN

The capacity to sense and adapt to changes in environmental cues is of paramount importance for every living organism. From yeast to man, cells must be able to match cellular activities to growth environment and nutrient availability. Key to this process is the development of membrane-bound systems that can detect modifications in the extracellular environment and to translate these into biological responses. Evidence gathered over the last 15 years has demonstrated that many of these cell surface "sensors" belong to the G protein-coupled receptor superfamily. Crucial to our understanding of nutrient sensing in mammalian species has been the identification of the extracellular Ca(2+)/cation-sensing receptor, CaR. CaR was the first ion-sensing molecule identified in man and genetic studies in humans have revealed the importance of the CaR in mineral ion metabolism. Latter, it has become apparent that the CaR also plays an important role outside the Ca(2+) homeostatic system, as an integrator of multiple environmental signals for the regulation of many vital cellular processes, from cell-to-cell communication to secretion and cell survival/cell death. Recently, novel aspects of receptor function reveal an unexpected role for the CaR in the regulation of growth and development in utero.


Asunto(s)
Receptores Sensibles al Calcio/genética , Animales , Huesos/metabolismo , Señalización del Calcio/fisiología , Sistema Cardiovascular/metabolismo , Tracto Gastrointestinal/metabolismo , Regulación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Homeostasis/fisiología , Humanos , Riñón/fisiología , Túbulos Renales/metabolismo , Pulmón/embriología , Neoplasias/fisiopatología , Sistema Nervioso/embriología , Piel/metabolismo
11.
FASEB J ; 23(4): 1153-60, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19050048

RESUMEN

Pulmonary neuroepithelial bodies (NEBs) are densely innervated groups of complex sensory airway receptors involved in the regulation of breathing. Together with their surrounding Clara-like cells, they exhibit stem cell potential through their capacity to regenerate depopulated areas of the epithelium following lung injury. We have employed confocal live cell imaging microscopy and novel electrophysiological techniques in a new ex vivo lung slice model to unravel potential purinergic signaling pathways within the NEB microenvironment. Quinacrine histochemistry indicated high amounts of vesicular ATP in NEB cells. Using a "reporter-patching" method adapted to create a uniquely sensitive and selective biosensor for the direct detection of ATP release from NEBs ex vivo, we demonstrated quantal ATP release from NEBs following their depolarization. Enhancing enzymatic extracellular ATP hydrolysis or inhibiting P2 receptors confirmed the central role of ATP in paracrine interactions between NEB cells and Clara-like cells. Combined calcium imaging, pharmacology, and immunohistochemistry showed that ligand-binding to functional P2Y(2) receptors underpins the activation of Clara-like cells. Hence, NEB cells communicate with their cellular neighbors in the NEB microenvironment by releasing ATP, which rapidly evokes purinergic activation of surrounding Clara-like cells. Besides ATP acting on the P2X(3) receptor expressing vagal sensory nerve terminals between NEB cells, local paracrine purinergic signaling within this potential stem cell niche may be important to both normal airway function, airway epithelial regeneration after injury, and/or the pathogenesis of small cell lung carcinomas.


Asunto(s)
Pulmón/metabolismo , Cuerpos Neuroepiteliales/fisiología , Receptores Purinérgicos P2/metabolismo , Mucosa Respiratoria/fisiología , Transducción de Señal , Adenosina Trifosfato/antagonistas & inhibidores , Adenosina Trifosfato/metabolismo , Compuestos de Anilina/metabolismo , Animales , Calcio/metabolismo , Colorantes Fluorescentes/metabolismo , Inmunohistoquímica , Cinética , Pulmón/citología , Ratones , Ratones Endogámicos C57BL , Cuerpos Neuroepiteliales/citología , Cuerpos Neuroepiteliales/metabolismo , Compuestos de Piridinio/metabolismo , Quinacrina/metabolismo , Mucosa Respiratoria/metabolismo , Suramina/farmacología , Xantenos/metabolismo
12.
J Physiol ; 586(24): 6007-19, 2008 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-18955379

RESUMEN

Postnatal lung function is critically dependent upon optimal embryonic lung development. As the free ionized plasma calcium concentration ([Ca(2+)](o)) of the fetus is higher than that of the adult, the process of lung development occurs in a hypercalcaemic environment. In the adult, [Ca(2+)](o) is monitored by the G-protein coupled, extracellular calcium-sensing receptor (CaR), but neither its ontogeny nor its potential role in lung development are known. Here, we demonstrate that CaR is expressed in the mouse lung epithelium, and that its expression is developmentally regulated, with a peak of expression at embryonic day 12.5 (E12.5) and a subsequent decrease by E18, after which the receptor is absent. Experiments carried out using the lung explant culture model in vitro show that lung branching morphogenesis is sensitive to [Ca(2+)](o), being maximal at physiological adult [Ca(2+)](o) (i.e. 1.0-1.3 mM) and lowest at the higher, fetal (i.e. 1.7 mM) [Ca(2+)](o). Administration of the specific CaR positive allosteric modulator, the calcimimetic R-568, mimics the suppressive effects of high [Ca(2+)](o) on branching morphogenesis while both phospholipase C and PI3 kinase inhibition reverse these effects. CaR activation suppresses cell proliferation while it enhances intracellular calcium signalling, lung distension and fluid secretion. Conditions which are restrictive either to branching or to secretion can be rescued by manipulating [Ca(2+)](o) in the culture medium. In conclusion, fetal Ca(2+)(o), acting through a developmentally regulated CaR, is an important extrinsic factor that modulates the intrinsic lung developmental programme. Our observations support a novel role for the CaR in preventing hyperplastic lung disease in utero.


Asunto(s)
Calcio/metabolismo , Embrión de Mamíferos/embriología , Pulmón/embriología , Receptores Sensibles al Calcio/fisiología , Compuestos de Anilina/farmacología , Animales , Animales Recién Nacidos , Calcio/farmacología , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Carbacol/farmacología , Proliferación Celular/efectos de los fármacos , Cromonas/farmacología , Embrión de Mamíferos/metabolismo , Inhibidores Enzimáticos/farmacología , Estrenos/farmacología , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Pulmón/citología , Pulmón/metabolismo , Masculino , Mesodermo/embriología , Mesodermo/metabolismo , Ratones , Ratones Endogámicos C57BL , Morfogénesis/efectos de los fármacos , Morfolinas/farmacología , Fenetilaminas , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Embarazo , Propilaminas , Pirrolidinonas/farmacología , Receptores Sensibles al Calcio/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Técnicas de Cultivo de Tejidos , Fosfolipasas de Tipo C/antagonistas & inhibidores , Fosfolipasas de Tipo C/metabolismo
13.
Mol Pharmacol ; 70(4): 1159-63, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16840712

RESUMEN

Lysine residues near each end of the receptor ectodomain (in rat P2X2 Lys69 and Lys308) have been implicated in ATP binding to P2X receptors. We recorded membrane currents from human embryonic kidney cells expressing P2X subunits and found that lysine-to-alanine substitutions at equivalent positions in the P2X3 receptor (Lys63 and Lys299) also prevented channel function. Heteromeric P2X2/3 receptors are formed when P2X2 and P2X3 subunits are expressed together; they can be distinguished by their relatively sustained response to alphabeta-methylene-ATP. By coexpression of wild-type P2X3 and mutated P2X2 subunit, we found that the heteromeric P2X2/3 channel functioned normally when either lysine in the P2X2 subunit was mutated to alanine (i.e., [K69A] or [K308A]) but not when both lysines were mutated to alanine (i.e., [K69A, K308A]). However, coexpression of wild-type P2X2 with a mutated P2X3 subunit ([K68A] or [K299A]) produced no functional heteromers. The rescue of the single lysine mutant P2X2 subunit by wild-type P2X3 (but not the converse) suggests that the heteromeric channel contains one P2X2 and two P2X3 subunits and that the receptor functions essentially normally as long as two subunits are not mutated. The failure to rescue function in the P2X2 subunit with both lysines mutated by wild-type P2X3 suggests that these residues from two different subunits interact in agonist binding or channel opening.


Asunto(s)
Sustitución de Aminoácidos , Lisina/metabolismo , Potenciales de la Membrana , Receptores Purinérgicos P2/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Línea Celular , Riñón/metabolismo , Modelos Biológicos , Subunidades de Proteína , Ratas , Receptores Purinérgicos P2/genética , Receptores Purinérgicos P2/fisiología , Receptores Purinérgicos P2X2 , Receptores Purinérgicos P2X3 , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...