Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
J Lipid Res ; 58(6): 1044-1054, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28420706

RESUMEN

Cholesterol-protein interactions are essential for the architectural organization of cell membranes and for lipid metabolism. While cholesterol-sensing motifs in transmembrane proteins have been identified, little is known about cholesterol recognition by soluble proteins. We reviewed the structural characteristics of binding sites for cholesterol and cholesterol sulfate from crystallographic structures available in the Protein Data Bank. This analysis unveiled key features of cholesterol-binding sites that are present in either all or the majority of sites: i) the cholesterol molecule is generally positioned between protein domains that have an organized secondary structure; ii) the cholesterol hydroxyl/sulfo group is often partnered by Asn, Gln, and/or Tyr, while the hydrophobic part of cholesterol interacts with Leu, Ile, Val, and/or Phe; iii) cholesterol hydrogen-bonding partners are often found on α-helices, while amino acids that interact with cholesterol's hydrophobic core have a slight preference for ß-strands and secondary structure-lacking protein areas; iv) the steroid's C21 and C26 constitute the "hot spots" most often seen for steroid-protein hydrophobic interactions; v) common "cold spots" are C8-C10, C13, and C17, at which contacts with the proteins were not detected. Several common features we identified for soluble protein-steroid interaction appear evolutionarily conserved.


Asunto(s)
Colesterol/metabolismo , Proteínas/química , Proteínas/metabolismo , Animales , Sitios de Unión , Colesterol/química , Cristalografía por Rayos X , Humanos , Solubilidad , Sulfatos/química
2.
Mol Neurodegener ; 10: 24, 2015 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-26104027

RESUMEN

BACKGROUND: Cerebral infarction due to thrombosis leads to the most common type of stroke and a likely cause of age-related cognitive decline and dementia. Endothelial nitric oxide synthase (eNOS) generates NO, which plays a crucial role in maintaining vascular function and exerting an antithrombotic action. Reduced eNOS expression and eNOS polymorphisms have been associated with stroke and Alzheimer's disease (AD), the most common type of dementia associated with neurovascular dysfunction. However, direct proof of such association is lacking. Since there are no reports of complete eNOS deficiency in humans, we used heterozygous eNOS(+/-) mice to mimic partial deficiency of eNOS, and determine its impact on cerebrovascular pathology and perfusion of cerebral vessels. RESULTS: Combining cerebral angiography with immunohistochemistry, we found thrombotic cerebral infarctions in eNOS(+/-) mice as early as 3-6 months of age but not in eNOS(+/+) mice at any age. Remarkably, vascular occlusions in eNOS(+/-) mice were found almost exclusively in three areas: temporoparietal and retrosplenial granular cortexes, and hippocampus this distribution precisely matching the hypoperfused areas identified in preclinical AD patients. Moreover, progressive cerebral amyloid angiopaphy (CAA), blood brain barrier (BBB) breakdown, and cognitive impairment were also detected in aged eNOS(+/-) mice. CONCLUSIONS: These data provide for the first time the evidence that partial eNOS deficiency results in spontaneous thrombotic cerebral infarctions that increase with age, leading to progressive CAA and cognitive impairments. We thus conclude that eNOS(+/-) mouse may represent an ideal model of ischemic stroke to address early and progressive damage in spontaneously-evolving chronic cerebral ischemia and thus, study vascular mechanisms contributing to vascular dementia and AD.


Asunto(s)
Barrera Hematoencefálica/fisiología , Angiopatía Amiloide Cerebral/genética , Infarto Cerebral/genética , Trastornos del Conocimiento/genética , Óxido Nítrico Sintasa de Tipo III/fisiología , Envejecimiento , Animales , Angiopatía Amiloide Cerebral/patología , Infarto Cerebral/enzimología , Infarto Cerebral/patología , Infarto Cerebral/psicología , Circulación Cerebrovascular , Trastornos del Conocimiento/enzimología , Trastornos del Conocimiento/patología , Progresión de la Enfermedad , Endotelio Vascular/fisiopatología , Regulación de la Expresión Génica , Heterocigoto , Hipocampo/irrigación sanguínea , Hipocampo/enzimología , Hipocampo/patología , Aprendizaje por Laberinto , Trastornos de la Memoria/enzimología , Trastornos de la Memoria/genética , Trastornos de la Memoria/patología , Ratones , Ratones Mutantes Neurológicos , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Óxido Nítrico Sintasa de Tipo III/deficiencia , Óxido Nítrico Sintasa de Tipo III/genética , Especificidad de Órganos , Lóbulo Parietal/irrigación sanguínea , Lóbulo Parietal/enzimología , Lóbulo Parietal/patología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Lóbulo Temporal/irrigación sanguínea , Lóbulo Temporal/enzimología , Lóbulo Temporal/patología
3.
Alcohol Clin Exp Res ; 38(5): 1216-26, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24588122

RESUMEN

BACKGROUND: Binge drinking represents the major form of excessive alcohol (ethanol [EtOH]) consumption in the United States. Episodic (such as binge) drinking results in blood alcohol levels (BAL) of 18 to 80 mM and leads to alcohol-induced cerebral artery constriction (AICAC). AICAC was shown to arise from EtOH-induced inhibition of large-conductance, calcium/voltage-gated potassium (BK) channels in the vascular smooth muscle. Factors that modulate BK channel-mediated AICAC remain largely unknown. METHODS: Male Sprague Dawley rats were placed on high-cholesterol (2% of cholesterol) diet for 18 to 23 weeks. Their littermates were placed on control iso-caloric diet. AICAC was evaluated both in vivo and in vitro, by means of pial arteriole diameter monitoring through a closed cranial window and diameter measurements of isolated, pressurized cerebral arteries. Cholesterol level in the cerebral artery tissue was manipulated by methyl-ß-cyclodextrin to reverse dietary-induced accumulation of cholesterol. BK channel surface presence on the plasma membrane of cerebral artery myocytes was evaluated by immunofluorescence staining. BK channel function in pressurized cerebral artery was assessed using selective BK channel blocker paxilline. RESULTS: Within 5 minutes of 50 mM EtOH injection into carotid artery in vivo, arteriole diameter decreased by 20% in control group. Pial arteriole constriction was significantly reduced in rats on high-cholesterol diet, resulting in only 10% reduction in diameter. BAL in both groups, however, remained the same. Significant reduction in AICAC in group on high-cholesterol diet compared to control was also observed after middle cerebral artery dissection and in vitro pressurization at 60 mmHg, this reduction remaining after endothelium removal. Cholesterol level in de-endothelialized cerebral arteries was significantly increased in rats on high-cholesterol diet. Removal of excessive cholesterol content restored AICAC to the level observed in cerebral arteries of rats on normal diet. Immunofluorescence staining of BK channel-forming and accessory, smooth muscle-specific ß1 subunit in freshly isolated cerebral artery myocyte showed that high-cholesterol diet did not down-regulate surface presence of BK protein. However, paxilline-induced cerebral artery constriction was diminished in arteries from rats on high-cholesterol diet. CONCLUSIONS: Our data indicate that dietary cholesterol protects against AICAC. This protection is caused by cholesterol buildup in the arterial tissue and diminished function (but not surface presence) of EtOH target-BK channel.


Asunto(s)
Arterias Cerebrales/efectos de los fármacos , Colesterol en la Dieta/farmacología , Etanol/farmacología , Vasoconstricción/efectos de los fármacos , Animales , HDL-Colesterol/sangre , LDL-Colesterol/sangre , Canales de Potasio de Gran Conductancia Activados por el Calcio/efectos de los fármacos , Masculino , Ratas Sprague-Dawley
4.
Pharmacol Ther ; 135(2): 133-50, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22584144

RESUMEN

Cholesterol (CLR) is an essential component of eukaryotic plasma membranes. CLR regulates the membrane physical state, microdomain formation and the activity of membrane-spanning proteins, including ion channels. Large conductance, voltage- and Ca²âº-gated K⁺ (BK) channels link membrane potential to cell Ca²âº homeostasis. Thus, they control many physiological processes and participate in pathophysiological mechanisms leading to human disease. Because plasmalemma BK channels cluster in CLR-rich membrane microdomains, a major driving force for studying BK channel-CLR interactions is determining how membrane CLR controls the BK current phenotype, including its pharmacology, channel sorting, distribution, and role in cell physiology. Since both BK channels and CLR tissue levels play a pathophysiological role in human disease, identifying functional and structural aspects of the CLR-BK channel interaction may open new avenues for therapeutic intervention. Here, we review the studies documenting membrane CLR-BK channel interactions, dissecting out the many factors that determine the final BK current response to changes in membrane CLR content. We also summarize work in reductionist systems where recombinant BK protein is studied in artificial lipid bilayers, which documents a direct inhibition of BK channel activity by CLR and builds a strong case for a direct interaction between CLR and the BK channel-forming protein. Bilayer lipid-mediated mechanisms in CLR action are also discussed. Finally, we review studies of BK channel function during hypercholesterolemia, and underscore the many consequences that the CLR-BK channel interaction brings to cell physiology and human disease.


Asunto(s)
Colesterol/fisiología , Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Animales , Etanol/farmacología , Humanos
5.
Proc Natl Acad Sci U S A ; 108(50): 20207-12, 2011 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-22123969

RESUMEN

Large conductance, voltage- and calcium-gated potassium (BK) channels regulate several physiological processes, including myogenic tone and thus, artery diameter. Nongenomic modulation of BK activity by steroids is increasingly recognized, but the precise location of steroid action remains unknown. We have shown that artery dilation by lithocholate (LC) and related cholane steroids is caused by a 2× increase in vascular myocyte BK activity (EC(50) = 45 µM), an action that requires ß1 but not other (ß2-ß4) BK accessory subunits. Combining mutagenesis and patch-clamping under physiological conditions of calcium and voltage on BK α- (cbv1) and ß1 subunits from rat cerebral artery myocytes, we identify the steroid interaction site from two regions in BK ß1 transmembrane domain 2 proposed by computational dynamics: the outer site includes L157, L158, and T165, whereas the inner site includes T169, L172, and L173. As expected from computational modeling, cbv1+rß1T165A,T169A channels were LC-unresponsive. However, cbv1 + rß1T165A and cbv1 + rß1T165A,L157A,L158A were fully sensitive to LC. Data indicate that the transmembrane domain 2 outer site does not contribute to steroid action. Cbv1 + rß1T169A was LC-insensitive, with rß1T169S being unable to rescue responsiveness to LC. Moreover, cbv1 + rß1L172A, and cbv1 + rß1L173A channels were LC-insensitive. These data and computational modeling indicate that tight hydrogen bonding between T169 and the steroid α-hydroxyl, and hydrophobic interactions between L172,L173 and the steroid rings are both necessary for LC action. Therefore, ß1 TM2 T169,L172,L173 provides the interaction area for cholane steroid activation of BK channels. Because this amino acid triplet is unique to BK ß1, our study provides a structural basis for advancing ß1 subunit-specific pharmacology of BK channels.


Asunto(s)
Membrana Celular/química , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/química , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Ácido Litocólico/farmacología , Alanina/genética , Sustitución de Aminoácidos/efectos de los fármacos , Animales , Sitios de Unión , Membrana Celular/efectos de los fármacos , Colanos/farmacología , Ácido Litocólico/química , Modelos Moleculares , Estructura Terciaria de Proteína , Ratas , Relación Estructura-Actividad
6.
Am J Physiol Cell Physiol ; 299(2): C264-78, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20445169

RESUMEN

Ryanodine receptors (RyRs) regulate contractility in resistance-size cerebral artery smooth muscle, yet their molecular identity, subcellular location, and phenotype in this tissue remain unknown. Following rat resistance-size cerebral artery myocyte sarcoplasmic reticulum (SR) purification and incorporation into POPE-POPS-POPC (5:3:2; wt/wt) bilayers, unitary conductances of 110 +/- 8, 334 +/- 15, and 441 +/- 27 pS in symmetric 300 mM Cs(+) were usually detected. The most frequent (34/40 bilayers) conductance (334 pS) decreased to

Asunto(s)
Arterias Cerebrales/citología , Arterias Cerebrales/fisiología , Células Musculares/fisiología , Subunidades de Proteína/fisiología , Canal Liberador de Calcio Receptor de Rianodina/fisiología , Animales , Células Cultivadas , Arterias Cerebrales/química , Femenino , Masculino , Células Musculares/química , Subunidades de Proteína/análisis , Ratas , Ratas Sprague-Dawley , Canal Liberador de Calcio Receptor de Rianodina/análisis
7.
Biochem Biophys Res Commun ; 390(3): 995-1000, 2009 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-19852931

RESUMEN

Large conductance, calcium- and voltage-gated potassium (BK) channels regulate numerous physiological processes. While most basic functional characteristics of native BK channels are reproduced by BK alpha (slo1) subunit homotetramers, key biophysical and pharmacological properties are drastically modified by the presence of auxiliary beta subunits (encoded by KCNMB1-4). Numerous physiological steroids, including sex hormones, gluco- and mineralocorticoids, activate beta subunit-containing BK channels, yet these steroids appear to be sensed by different types of beta subunits, with some steroids being sensed by homomeric slo1 channels as well. We recently showed that beta1 sensitizes the BK channel to microM concentrations of lithocholate (LC). Following expression of rat cerebral artery myocyte slo1 subunits ("cbv1") with beta1, beta2, beta3 or beta4 in Xenopus laevis oocytes we now demonstrate that BK beta2, beta3 and beta4 subunits fail to substitute for beta1 in providing LC-sensitivity (150 microM) to the BK channel. These findings document for the first time a rather selective steroid activation of BK channels via a particular channel accessory subunit. In addition, LC routinely activated native BK channels in myocytes freshly isolated from rat cerebral artery smooth muscle, where BK beta1 is highly expressed, while failing to do so in skeletal (flexor digitorum brevis) muscle, where BK beta1 expression is negligible. This indicates that the native environment of the BK channel sustains the LC-sensitivity distinctly provided to the BK channel by beta1 subunits. Our study indicates that LC represents a unique tool to probe the presence of functional beta1-subunits and selectively activate BK channels in tissues that highly express KCNMB1.


Asunto(s)
Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/agonistas , Ácido Litocólico/farmacología , Esteroides/farmacología , Animales , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/genética , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Oocitos , Ratas , Proteínas Recombinantes/agonistas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Xenopus laevis
8.
FEBS Lett ; 583(17): 2779-84, 2009 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-19616547

RESUMEN

Ethanol-induced inhibition of myocyte large conductance, calcium- and voltage-gated potassium (BK) current causes cerebrovascular constriction, yet the molecular targets mediating EtOH action remain unknown. Using BK channel-forming (cbv1) subunits from cerebral artery myocytes, we demonstrate that EtOH potentiates and inhibits current at Ca(i)(2+) lower and higher than approximately 15 microM, respectively. By increasing cbv1's apparent Ca(i)(2+)-sensitivity, accessory BK beta(1) subunits shift the activation-to-inhibition crossover of EtOH action to <3 microM Ca(i)(2+), with consequent inhibition of current under conditions found during myocyte contraction. Knocking-down KCNMB1 suppresses EtOH-reduction of arterial myocyte BK current and vessel diameter. Therefore, BK beta(1) is the molecular effector of alcohol-induced BK current inhibition and cerebrovascular constriction.


Asunto(s)
Circulación Cerebrovascular , Etanol/farmacología , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Vasoconstricción/efectos de los fármacos , Animales , Calcio/metabolismo , Arterias Cerebrales/citología , Arterias Cerebrales/efectos de los fármacos , Circulación Cerebrovascular/efectos de los fármacos , Circulación Cerebrovascular/fisiología , Activación del Canal Iónico/efectos de los fármacos , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Musculares/citología , Células Musculares/efectos de los fármacos , Células Musculares/fisiología , Músculo Liso Vascular/citología , Oocitos/citología , Oocitos/fisiología , Técnicas de Placa-Clamp , Fenilefrina/farmacología , Ratas , Vasoconstrictores/farmacología , Xenopus laevis
9.
Am J Physiol Heart Circ Physiol ; 297(2): H680-8, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19502562

RESUMEN

Voltage-dependent calcium (Ca(2+), Ca(V)1.2) channels are the primary Ca(2+) entry pathway in smooth muscle cells of resistance-size (myogenic) arteries, but their molecular identity remains unclear. Here we identified and quantified Ca(V)1.2 alpha(1)-subunit splice variation in myocytes of rat resistance-size (100-200 microm diameter) cerebral arteries. Full-length clones containing either exon 1b or the recently identified exon 1c exhibited additional primary splice variation at exons 9*, 21/22, 31/32, and +/- 33. Real-time PCR confirmed the findings from full-length clones and indicated that the major Ca(V)1.2 variant contained exons 1c, 8, 21, and 32+33, with approximately 57% containing 9*. Exon 9* was more prevalent in clones containing 1c (72%) than in those containing 1b (33%), suggesting exon-selective combinatorial splicing. To examine the functional significance of this splicing profile, membrane currents produced by each of the four exon 1b/c/ +/- 9* variants were characterized following transfection in HEK293 cells. Exon 1c and 9* caused similar hyperpolarizing shifts in both current-voltage relationships and voltage-dependent activation of currents. Furthermore, exon 9* induced a hyperpolarizing shift only in the voltage-dependent activation of channels containing exon 1b, but not in those containing exon 1c. In contrast, exon 1b, 1c, or +9* did not alter voltage-dependent inactivation. In summary, we have identified the Ca(V)1.2 alpha(1)-subunit splice variant population that is expressed in myocytes of resistance-size arteries and the unique electrophysiological properties of recombinant channels formed by exon 1 and 9* variation. The predominance of exon 1c and 9* in smooth muscle cell Ca(V)1.2 channels causes a hyperpolarizing shift in the voltage sensitivity of currents toward the physiological arterial voltage range.


Asunto(s)
Empalme Alternativo/fisiología , Canales de Calcio Tipo L/genética , Arterias Cerebrales/citología , Circulación Cerebrovascular/fisiología , Músculo Liso Vascular/fisiología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Canales de Calcio Tipo L/química , Canales de Calcio Tipo L/fisiología , Células Cultivadas , Exones/genética , Datos de Secuencia Molecular , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/fisiología , Técnicas de Placa-Clamp , Estructura Terciaria de Proteína , Ratas , Ratas Sprague-Dawley , Resistencia Vascular/fisiología
10.
Alcohol Clin Exp Res ; 33(7): 1125-35, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19389201

RESUMEN

Small (SK) and large conductance (BK) Ca(2+)-activated K(+) channels contribute to action potential repolarization, shape dendritic Ca(2+)spikes and postsynaptic responses, modulate the release of hormones and neurotransmitters, and contribute to hippocampal-dependent synaptic plasticity. Over the last decade, SK and BK channels have emerged as important targets for the development of acute ethanol tolerance and for altering neuronal excitability following chronic ethanol consumption. In this mini-review, we discuss new evidence implicating SK and BK channels in ethanol tolerance and ethanol-associated homeostatic plasticity. Findings from recent reports demonstrate that chronic ethanol produces a reduction in the function of SK channels in VTA dopaminergic and CA1 pyramidal neurons. It is hypothesized that the reduction in SK channel function increases the propensity for burst firing in VTA neurons and increases the likelihood for aberrant hyperexcitability during ethanol withdrawal in hippocampus. There is also increasing evidence supporting the idea that ethanol sensitivity of native BK channel results from differences in BK subunit composition, the proteolipid microenvironment, and molecular determinants of the channel-forming subunit itself. Moreover, these molecular entities play a substantial role in controlling the temporal component of ethanol-associated neuroadaptations in BK channels. Taken together, these studies suggest that SK and BK channels contribute to ethanol tolerance and adaptive plasticity.


Asunto(s)
Alcoholismo/metabolismo , Alcoholismo/patología , Etanol/farmacología , Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Plasticidad Neuronal/fisiología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/fisiología , Animales , Etanol/efectos adversos , Humanos , Plasticidad Neuronal/efectos de los fármacos
11.
J Lipid Res ; 49(11): 2441-51, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18650555

RESUMEN

Lithocholate (LC) (10-300 microM) in physiological solution is sensed by vascular myocyte large conductance, calcium- and voltage-gated potassium (BK) channel beta(1) accessory subunits, leading to channel activation and arterial dilation. However, the structural features in steroid and target that determine LC action are unknown. We tested LC and close analogs on BK channel (pore-forming cbv1+beta(1) subunits) activity using the product of the number of functional ion channels in the membrane patch (N) and the open channel probability (Po). LC (5beta-cholanic acid-3alpha-ol), 5alpha-cholanic acid-3alpha-ol, and 5beta-cholanic acid-3beta-ol increased NPo (EC(50) approximately 45 microM). At maximal increase in NPo, LC increased NPo by 180%, whereas 5alpha-cholanic acid-3alpha-ol and 5beta-cholanic acid-3beta-ol raised NPo by 40%. Thus, the alpha-hydroxyl and the cis A-B ring junction are both required for robust channel potentiation. Lacking both features, 5alpha-cholanic acid-3beta-ol and 5-cholenic acid-3beta-ol were inactive. Three-dimensional structures show that only LC displays a bean shape with clear-cut convex and concave hemispheres; 5alpha-cholanic acid-3alpha-ol and 5beta-cholanic acid-3beta-ol partially matched LC shape, and 5alpha-cholanic acid-3beta-ol and 5-cholenic acid-3beta-ol did not. Increasing polarity in steroid rings (5beta-cholanic acid-3alpha-sulfate) or reducing polarity in lateral chain (5beta-cholanic acid 3alpha-ol methyl ester) rendered poorly active compounds, consistent with steroid insertion between beta(1) and bilayer lipids, with the steroid-charged tail near the aqueous phase. Molecular dynamics identified two regions in beta(1) transmembrane domain 2 that meet unique requirements for bonding with the LC concave hemisphere, where the steroid functional groups are located.


Asunto(s)
Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/química , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Ácido Litocólico/química , Ácido Litocólico/fisiología , Animales , Simulación por Computador , Femenino , Vectores Genéticos , Hidroxilación , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/genética , Ácido Litocólico/genética , Modelos Moleculares , Músculo Liso Vascular/metabolismo , Técnicas de Placa-Clamp , Estructura Terciaria de Proteína , Transporte de Proteínas/genética , Transporte de Proteínas/fisiología , Ratas , Xenopus laevis/genética
12.
Mol Pharmacol ; 74(3): 628-40, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18552122

RESUMEN

Ethanol modulation of calcium- and voltage-gated potassium (slo1) channels alters neuronal excitability, cerebrovascular tone, brain function, and behavior, yet the mechanism of this modulation remains unknown. Using patch-clamp electrophysiology on recombinant BK(Ca) channels cloned from mouse brain and expressed in Xenopus laevis oocytes, we demonstrate that ethanol, even at concentrations maximally effective to modulate BK(Ca) channel function (100 mM), fails to gate the channel in absence of activating calcium. Moreover, ethanol does not modify intrinsic, voltage- or physiological magnesium-driven gating. The alcohol works as an adjuvant of calcium by selectively facilitating calcium-driven gating. This facilitation, however, renders differential ethanol effects on channel activity: potentiation at low (<10 microM) and inhibition at high (>10 microM) calcium, this dual pattern remaining largely unmodified by coexpression of brain slo1 channels with the neuronally abundant BK(Ca) channel beta(4) subunit. Calcium recognition by either of the slo1 high-affinity sensors (calcium bowl and RCK1 Asp362/Asp367) is required for ethanol to amplify channel activation by calcium. The Asp362/Asp367 site, however, is necessary and sufficient to sustain ethanol inhibition. This inhibition also results from ethanol facilitation of calcium action; in this case, ethanol favors channel dwelling in a calcium-driven, low-activity mode. The agonist-adjuvant mechanism that we advance from the calcium-ethanol interaction on slo1 might be applicable to data of ethanol action on a wide variety of ligand-gated channels.


Asunto(s)
Calcio/farmacología , Etanol/farmacología , Activación del Canal Iónico/efectos de los fármacos , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/metabolismo , Animales , Señalización del Calcio/efectos de los fármacos , Cinética , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/química , Ratones , Modelos Biológicos , Oocitos/efectos de los fármacos , Estructura Terciaria de Proteína , Xenopus
13.
Bioorg Med Chem Lett ; 18(11): 3427-30, 2008 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-18462939

RESUMEN

Physiological and pharmacological agents that activate large conductance, voltage-, and calcium-gated potassium (BK) channels located in the smooth muscle are effective vasodilators. Thus, activators of smooth muscle BK channels may be potential therapeutic tools to treat cardiovascular disease associated with vasoconstriction and/or impaired dilation, such as cerebrovascular spasm and constriction. We previously showed that lithocholic acid (LC) and other cholane derivatives activated smooth muscle BK channels and, thus, caused endothelium-independent cerebral artery dilation. However, clinical use of these cholane derivatives could be limited by the actions of these steroids, such as elevation of intracellular calcium and induction of apoptosis. Using LC as template, we designed and synthesized a series of hydroxy-alkynoic acids and corresponding methyl esters, as putative, non-steroid BK channel activators. Indeed, the newly synthesized compounds effectively and reversibly activated rat cerebrovascular myocyte BK channel at concentrations similar to those found effective with LC. Among all the novel compounds tested, C-10 hydroxy-alkynoic acid methyl ester appears to be the most effective activator of vascular myocyte BK channels.


Asunto(s)
Hidroxiácidos/síntesis química , Hidroxiácidos/farmacología , Canales de Potasio de Gran Conductancia Activados por el Calcio/agonistas , Ácido Litocólico/farmacología , Animales , Hidroxiácidos/química , Conformación Molecular , Estructura Molecular , Células Musculares/efectos de los fármacos , Músculo Liso/efectos de los fármacos , Ratas
14.
FEBS Lett ; 582(5): 673-8, 2008 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-18242174

RESUMEN

Bile acids and other steroids modify large conductance, calcium- and voltage-gated potassium (BK) channel activity contributing to non-genomic modulation of myogenic tone. Accessory BK beta(1) subunits are necessary for lithocholate (LC) to activate BK channels and vasodilate. The protein regions that sense steroid action, however, remain unknown. Using recombinant channels in 1-palmitoyl-2-oleoyl-phosphatidylethanolamine/1-palmitoyl-2-oleoyl-phosphatidylserine bilayers we now demonstrate that complex proteolipid domains and cytoarchitecture are unnecessary for beta(1) to mediate LC action; beta(1) and a simple phospholipid microenvironment suffice. Since beta(1) senses LC but beta(4) does not, we made chimeras swapping regions between these subunits and, following channel heterologous expression, demonstrate that beta(1) TM2 is a bile acid-recognizing sensor.


Asunto(s)
Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/química , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Ácido Litocólico/farmacología , Animales , Calcio/metabolismo , Línea Celular , Humanos , Activación del Canal Iónico/efectos de los fármacos , Membrana Dobles de Lípidos/metabolismo , Estructura Terciaria de Proteína , Ratas , Xenopus
15.
Alcohol Clin Exp Res ; 31(10): 1625-32, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17850640

RESUMEN

In most neurons and other excitable cells, calcium-activated potassium channels of small (SK) and large conductance (BK; MaxiK) control excitability and neurotransmitter release. The spontaneous activity of dopamine neurons of the ventral tegmental area is increased by ethanol. This ethanol excitation is potentiated by selective blockade of SK, indicating that SK channels modulate ethanol stimulation of neurons that are critical in reward and reinforcement. On the other hand, ethanol directly modulates BK channel activity in a variety of systems, including rat neurohypophysial nerve endings, primary sensory dorsal root ganglia, nucleus accumbens neurons, Caenorhabditis elegans type-IV dopaminergic CEP neurons, and nonneuronal preparations, such as rat pituitary cells, cerebrovascular myocytes and human umbilical vein endothelial cells. Ethanol action on BK channels can modify neuropeptide and growth hormone release, nociception, cerebrovascular tone, and endothelial proliferation. Ethanol modulates BK channels even when the drug is evaluated using recombinant BK channel-forming alpha (slo) subunits or channel reconstitution in artificial, binary lipid bilayers, indicating that the slo subunit and its immediate lipid microenvironment are the essential targets of ethanol. Consistent with this, single amino acid slo channel mutants display altered ethanol sensitivity. Furthermore, C. elegans slo1 null mutants are resistant to ethanol-induced motor incoordination. On the other hand, Drosophila melanogaster slo null mutants fail to acquire acute tolerance to ethanol sedation. Ethanol action on slo channels, however, may be tuned by a variety of factors, including posttranslational modification of slo subunits, coexpression of channel accessory subunits, and the lipid microenvironment, resulting in increase, refractoriness, or even decrease in channel activity. In brief, both SK and BK channels are important targets of ethanol throughout the body, and interference with ethanol effects on these channels could form the basis for novel pharmacotherapies to ameliorate the actions or consequences of alcohol abuse.


Asunto(s)
Depresores del Sistema Nervioso Central/farmacología , Etanol/farmacología , Canales de Potasio Calcio-Activados/efectos de los fármacos , Alcoholismo/tratamiento farmacológico , Animales , Caenorhabditis elegans , Drosophila melanogaster , Humanos , Neuronas/efectos de los fármacos
16.
J Biol Chem ; 282(40): 29211-21, 2007 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-17699517

RESUMEN

Voltage-dependent L-type Ca(2+) (Ca(V)1.2) channels are the principal Ca(2+) entry pathway in arterial myocytes. Ca(V)1.2 channels regulate multiple vascular functions and are implicated in the pathogenesis of human disease, including hypertension. However, the molecular identity of Ca(V)1.2 channels expressed in myocytes of myogenic arteries that regulate vascular pressure and blood flow is unknown. Here, we cloned Ca(V)1.2 subunits from resistance size cerebral arteries and demonstrate that myocytes contain a novel, cysteine rich N terminus that is derived from exon 1 (termed "exon 1c"), which is located within CACNA1C, the Ca(V)1.2 gene. Quantitative PCR revealed that exon 1c was predominant in arterial myocytes, but rare in cardiac myocytes, where exon 1a prevailed. When co-expressed with alpha(2)delta subunits, Ca(V)1.2 channels containing the novel exon 1c-derived N terminus exhibited: 1) smaller whole cell current density, 2) more negative voltages of half activation (V(1/2,act)) and half-inactivation (V(1/2,inact)), and 3) reduced plasma membrane insertion, when compared with channels containing exon 1b. beta(1b) and beta(2a) subunits caused negative shifts in the V(1/2,act) and V(1/2,inact) of exon 1b-containing Ca(V)1.2alpha(1)/alpha(2)delta currents that were larger than those in exon 1c-containing Ca(V)1.2alpha(1)/alpha(2)delta currents. In contrast, beta(3) similarly shifted V(1/2,act) and V(1/2,inact) of currents generated by exon 1b- and exon 1c-containing channels. beta subunits isoform-dependent differences in current inactivation rates were also detected between N-terminal variants. Data indicate that through novel alternative splicing at exon 1, the Ca(V)1.2 N terminus modifies regulation by auxiliary subunits. The novel exon 1c should generate distinct voltage-dependent Ca(2+) entry in arterial myocytes, resulting in tissue-specific Ca(2+) signaling.


Asunto(s)
Arterias/metabolismo , Canales de Calcio Tipo L/química , Animales , Secuencia de Bases , Células COS , Canales de Calcio Tipo L/fisiología , Chlorocebus aethiops , Electrofisiología/métodos , Humanos , Datos de Secuencia Molecular , Células Musculares/metabolismo , Miocardio/metabolismo , Estructura Terciaria de Proteína , Ratas , Ratas Sprague-Dawley , Homología de Secuencia de Ácido Nucleico , Transducción de Señal
17.
Mol Pharmacol ; 72(2): 359-69, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17468198

RESUMEN

Among the nongenomic effects of steroids, control of vasomotion has received increasing attention. Lithocholate (LC) and other physiologically relevant cholane-derived steroids cause vasodilation, yet the molecular targets and mechanisms underlying this action remain largely unknown. We demonstrate that LC (45 microM) reversibly increases the diameter of pressurized resistance cerebral arteries by approximately 10%, which would result in approximately 30% increase in cerebral blood flow. LC action is independent of endothelial integrity, prevented by 55 nM iberiotoxin, and unmodified by 0.8 mM 4-aminopyridine, indicating that LC causes vasodilation via myocyte BK channels. Indeed, LC activates BK channels in isolated myocytes through a destabilization of channel long-closed states without modifying unitary conductance. LC channel activation occurs within a wide voltage range and at Ca2+ concentrations reached in the myocyte at rest and during contraction. Channel accessory beta1 subunits, which are predominant in smooth muscle, are necessary for LC to modify channel activity. In contrast, beta4 subunits, which are predominant in neuronal tissues, fail to evoke LC sensitivity. LC activation of cbv1+beta1 and native BK channels display identical characteristics, including EC50 (46 microM) and Emax (approximately 300 microM) values, strongly suggesting that the cbv1+beta1 complex is necessary and sufficient to evoke LC action. Finally, intact arteries from beta1 subunit knockout mice fail to relax in response to LC, although they are able to respond to other vasodilators. This study pinpoints the BK beta1 subunit as the molecule that senses LC, which results in myocyte BK channel activation and, thus, endothelial-independent relaxation of small, resistance-size arteries.


Asunto(s)
Canales de Potasio de Gran Conductancia Activados por el Calcio/efectos de los fármacos , Ácido Litocólico/farmacología , Vasodilatación/efectos de los fármacos , Animales , Calcio/metabolismo , Arterias Cerebrales/efectos de los fármacos , Arterias Cerebrales/fisiología , Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ácido Oleanólico/análogos & derivados , Ácido Oleanólico/farmacología , Subunidades de Proteína , Ratas , Ratas Sprague-Dawley , Saponinas/farmacología , Tamoxifeno/farmacología , Resistencia Vascular
18.
Brain Res ; 1087(1): 75-82, 2006 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-16626657

RESUMEN

Toluene (methylbenzene) is representative of a class of industrial solvents that are voluntarily inhaled as drugs of abuse. Previous data from this laboratory and others have shown that these compounds alter the function of a variety of ion channels including ligand-gated channels activated by ATP, acetylcholine, GABA, glutamate and serotonin, as well as voltage-dependent sodium and calcium channels. It is less clear what effects toluene may have on potassium channels that act to reduce the excitability of most cells. Previous studies have shown that ethanol potentiates the function of both the large conductance, calcium-activated potassium channel (BK) and specific members of the G-protein-coupled inwardly rectifying potassium channels (GirKs). Since toluene and other abused inhalants share many behavioral effects with ethanol, it was hypothesized that toluene would also enhance the function of these channels. This hypothesis was tested using two-electrode voltage-clamp electrophysiology to measure the activity of BK and GirK potassium channel currents expressed in Xenopus laevis oocytes. As reported previously, ethanol potentiated currents in oocytes expressing either BK or GirK2 channels. In contrast, toluene caused a concentration-dependent inhibition of BK channel currents with 3 mM producing approximately 50% inhibition of control currents. Currents in oocytes injected with GirK2 mRNA were also inhibited by toluene while those expressing GirK1/2 and 1/4 channels were minimally affected. In oocytes co-injected with mRNA for GirK2 and the mGluR1a metabotropic receptor, exposure to glutamate potentiated currents evoked by a high-potassium solution. Toluene inhibited these glutamate-activated currents to approximately the same degree as those induced under basal conditions. The results of these studies show that toluene has effects on BK and GirK channels that are opposite to those of ethanol, suggesting that these channels are unlikely to underlie behaviors that these two drugs of abuse share.


Asunto(s)
Etanol/farmacología , Oocitos/efectos de los fármacos , Canales de Potasio Calcio-Activados/fisiología , Tolueno/farmacología , Animales , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Conductividad Eléctrica , Estimulación Eléctrica , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/fisiología , Concentración 50 Inhibidora , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Potenciales de la Membrana/efectos de la radiación , Microinyecciones/métodos , Biología Molecular/métodos , Oocitos/fisiología , Técnicas de Placa-Clamp/métodos , Péptidos/farmacología , Receptores de Glutamato Metabotrópico/fisiología , Solventes/farmacología , Xenopus laevis
19.
Nat Neurosci ; 9(1): 41-9, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16341213

RESUMEN

High-conductance, Ca(2+)-activated and voltage-gated (BK) channels set neuronal firing. They are almost universally activated by alcohol, leading to reduced neuronal excitability and neuropeptide release and to motor intoxication. However, several BK channels are inhibited by alcohol, and most other voltage-gated K(+) channels are refractory to drug action. BK channels are homotetramers (encoded by Slo1) that possess a unique transmembrane segment (S0), leading to a cytosolic S0-S1 loop. We identified Thr107 of bovine slo (bslo) in this loop as a critical residue that determines BK channel responses to alcohol. In addition, the activity of Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) in the cell controlled channel activity and alcohol modulation. Incremental CaMKII-mediated phosphorylation of Thr107 in the BK tetramer progressively increased channel activity and gradually switched the channel alcohol responses from robust activation to inhibition. Thus, CaMKII phosphorylation of slo Thr107 works as a 'molecular dimmer switch' that could mediate tolerance to alcohol, a form of neuronal plasticity.


Asunto(s)
Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Depresores del Sistema Nervioso Central/farmacología , Etanol/farmacología , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/efectos de los fármacos , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/fisiología , Canales de Potasio de Gran Conductancia Activados por el Calcio/efectos de los fármacos , Treonina/metabolismo , Fosfatasa Alcalina/química , Secuencia de Aminoácidos , Animales , Bencilaminas/farmacología , Bradiquinina/fisiología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina , Proteínas Quinasas Dependientes de Calcio-Calmodulina/genética , Electrofisiología , Canales de Potasio de Gran Conductancia Activados por el Calcio/genética , Ratones , Datos de Secuencia Molecular , Mutagénesis , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Técnicas de Placa-Clamp , Fosfoproteínas Fosfatasas/metabolismo , Fosforilación , ARN Complementario/biosíntesis , ARN Complementario/genética , Sulfonamidas/farmacología , Treonina/genética , Xenopus
20.
Circ Res ; 97(8): 805-12, 2005 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-16166559

RESUMEN

Carbon monoxide (CO) is an endogenous paracrine and autocrine gaseous messenger that regulates physiological functions in a wide variety of tissues. CO induces vasodilation by activating arterial smooth muscle large-conductance Ca2+-activated potassium (BK(Ca)) channels. However, the mechanism by which CO activates BK(Ca) channels remains unclear. Here, we tested the hypothesis that CO activates BK(Ca) channels by binding to channel-bound heme, a BK(Ca) channel inhibitor, and altering the interaction between heme and the conserved heme-binding domain (HBD) of the channel alpha subunit C terminus. Data obtained using thin-layer chromatography, spectrophotometry, mass spectrometry (MS), and MS-MS indicate that CO modifies the binding of reduced heme to the alpha subunit HBD. In contrast, CO does not alter the interaction between the HBD and oxidized heme (hemin), to which CO cannot bind. Consistent with these findings, electrophysiological measurements of native and cloned (cbv) cerebral artery smooth muscle BK(Ca) channels show that CO reverses BK(Ca) channel inhibition by heme but not by hemin. Site-directed mutagenesis of the cbv HBD from CKACH to CKASR abolished both heme-induced channel inhibition and CO-induced activation. Furthermore, on binding CO, heme switches from being a channel inhibitor to an activator. These findings indicate that reduced heme is a functional CO receptor for BK(Ca) channels, introduce a unique mechanism by which CO regulates the activity of a target protein, and reveal a novel process by which a gaseous messenger regulates ion channel activity.


Asunto(s)
Monóxido de Carbono/farmacología , Hemo/metabolismo , Canales de Potasio Calcio-Activados/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Sitios de Unión , Células COS , Monóxido de Carbono/metabolismo , Proteínas Portadoras/metabolismo , Chlorocebus aethiops , Hemo/farmacología , Proteínas de Unión al Hemo , Hemoproteínas/metabolismo , Hemina/farmacología , Datos de Secuencia Molecular , Oxígeno/farmacología , Canales de Potasio Calcio-Activados/química , Canales de Potasio Calcio-Activados/metabolismo , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...