Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Proc Natl Acad Sci U S A ; 111(6): 2361-6, 2014 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-24464482

RESUMEN

Ion channels composed of pore-forming and auxiliary subunits control physiological functions in virtually all cell types. A conventional view is that channels assemble with their auxiliary subunits before anterograde plasma membrane trafficking of the protein complex. Whether the multisubunit composition of surface channels is fixed following protein synthesis or flexible and open to acute and, potentially, rapid modulation to control activity and cellular excitability is unclear. Arterial smooth muscle cells (myocytes) express large-conductance Ca(2+)-activated potassium (BK) channel α and auxiliary ß1 subunits that are functionally significant modulators of arterial contractility. Here, we show that native BKα subunits are primarily (∼95%) plasma membrane-localized in human and rat arterial myocytes. In contrast, only a small fraction (∼10%) of total ß1 subunits are located at the cell surface. Immunofluorescence resonance energy transfer microscopy demonstrated that intracellular ß1 subunits are stored within Rab11A-postive recycling endosomes. Nitric oxide (NO), acting via cGMP-dependent protein kinase, and cAMP-dependent pathways stimulated rapid (≤1 min) anterograde trafficking of ß1 subunit-containing recycling endosomes, which increased surface ß1 almost threefold. These ß1 subunits associated with surface-resident BKα proteins, elevating channel Ca(2+) sensitivity and activity. Our data also show that rapid ß1 subunit anterograde trafficking is the primary mechanism by which NO activates myocyte BK channels and induces vasodilation. In summary, we show that rapid ß1 subunit surface trafficking controls functional BK channel activity in arterial myocytes and vascular contractility. Conceivably, regulated auxiliary subunit trafficking may control ion channel activity in a wide variety of cell types.


Asunto(s)
Vasos Sanguíneos/fisiología , Canales de Potasio Calcio-Activados/fisiología , Animales , Transferencia Resonante de Energía de Fluorescencia , Transporte Iónico , Masculino , Técnicas de Placa-Clamp , Canales de Potasio Calcio-Activados/química , Ratas , Ratas Sprague-Dawley
2.
Am J Physiol Cell Physiol ; 310(11): C885-93, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27076616

RESUMEN

Plasma membrane-localized CaV1.2 channels are the primary calcium (Ca(2+)) influx pathway in arterial smooth muscle cells (myocytes). CaV1.2 channels regulate several cellular functions, including contractility and gene expression, but the trafficking pathways that control the surface expression of these proteins are unclear. Similarly, expression and physiological functions of small Rab GTPases, proteins that control vesicular trafficking in arterial myocytes, are poorly understood. Here, we investigated Rab proteins that control functional surface abundance of CaV1.2 channels in cerebral artery myocytes. Western blotting indicated that Rab25, a GTPase previously associated with apical recycling endosomes, is expressed in cerebral artery myocytes. Immunofluorescence Förster resonance energy transfer (immunoFRET) microscopy demonstrated that Rab25 locates in close spatial proximity to CaV1.2 channels in myocytes. Rab25 knockdown using siRNA reduced CaV1.2 surface and intracellular abundance in arteries, as determined using arterial biotinylation. In contrast, CaV1.2 was not located nearby Rab11A or Rab4 and CaV1.2 protein was unaltered by Rab11A or Rab4A knockdown. Rab25 knockdown resulted in CaV1.2 degradation by a mechanism involving both lysosomal and proteasomal pathways and reduced whole cell CaV1.2 current density but did not alter voltage dependence of current activation or inactivation in isolated myocytes. Rab25 knockdown also inhibited depolarization (20-60 mM K(+)) and pressure-induced vasoconstriction (myogenic tone) in cerebral arteries. These data indicate that Rab25 is expressed in arterial myocytes where it promotes surface expression of CaV1.2 channels to control pressure- and depolarization-induced vasoconstriction.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/enzimología , Proteínas de Unión al GTP rab/metabolismo , Animales , Células Cultivadas , Arterias Cerebrales/enzimología , Lisosomas/metabolismo , Masculino , Potenciales de la Membrana , Complejo de la Endopetidasa Proteasomal/metabolismo , Transporte de Proteínas , Proteolisis , Interferencia de ARN , Ratas Sprague-Dawley , Transducción de Señal , Transfección , Vasoconstricción , Proteínas de Unión al GTP rab/genética
3.
Circ Res ; 115(4): 423-31, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24906643

RESUMEN

RATIONALE: Smooth muscle cell (myocyte) large-conductance calcium (Ca)(2+)-activated potassium (BK) channels are functionally significant modulators of arterial contractility. Arterial myocytes express both pore-forming BKα and auxiliary ß1 subunits, which increase channel Ca(2+) sensitivity. Recently, several leucine-rich repeat containing (LRRC) proteins have been identified as auxiliary γ subunits that elevate the voltage sensitivity of recombinant and prostate adenocarcinoma BK channels. LRRC expression and physiological functions in native cell types are unclear. OBJECTIVE: Investigate the expression and physiological functions of leucine-rich repeat containing protein 26 (LRRC26) in arterial myocytes. METHODS AND RESULTS: Reverse transcription polymerase chain reaction and Western blotting detected LRRC26 mRNA and protein in cerebral artery myocytes. Biotinylation, immunofluorescence resonance energy transfer microscopy, and coimmunoprecipitation indicated that LRRC26 was located in close spatial proximity to, and associated with, plasma membrane BKα subunits. LRRC26 knockdown (RNAi) reduced total and surface LRRC26, but did not alter BKα or ß1, proteins in arteries. LRRC26 knockdown did not alter Ca(2+) sparks but reduced BK channel voltage sensitivity, which decreased channel apparent Ca(2+) sensitivity and transient BK current frequency and amplitude in myocytes. LRRC26 knockdown also increased myogenic tone over a range (40-100 mm Hg) of intravascular pressures, and reduced vasoconstriction to iberiotoxin and vasodilation to NS1619, BK channel inhibitors and activators, respectively. In contrast, LRRC26 knockdown did not alter depolarization (60 mmol/L K(+))-induced vasoconstriction. CONCLUSIONS: LRRC26 is expressed, associates with BKα subunits, and elevates channel voltage- and apparent Ca(2+) sensitivity in arterial myocytes to induce vasodilation. This study indicates that arterial myocytes express a functional BK channel γ subunit.


Asunto(s)
Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Proteínas de Neoplasias/metabolismo , Animales , Western Blotting , Señalización del Calcio , Arterias Cerebrales/metabolismo , Regulación de la Expresión Génica , Inmunoprecipitación , Canales de Potasio de Gran Conductancia Activados por el Calcio/genética , Masculino , Potenciales de la Membrana , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Proteínas de Neoplasias/genética , Subunidades de Proteína , Interferencia de ARN , ARN Mensajero/metabolismo , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Técnicas de Cultivo de Tejidos , Transfección , Vasoconstricción , Vasoconstrictores/farmacología , Vasodilatación , Vasodilatadores/farmacología
4.
Am J Physiol Cell Physiol ; 309(6): C392-402, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26179602

RESUMEN

Arterial smooth muscle cells (myocytes) express large-conductance Ca(2+)-activated K(+) (BK) channel α and auxiliary ß1 subunits that modulate arterial contractility. In arterial myocytes, ß1 subunits are stored within highly mobile rab11A-positive recycling endosomes. In contrast, BKα subunits are primarily plasma membrane-localized. Trafficking pathways for BKα and whether physiological stimuli that regulate arterial contractility alter BKα localization in arterial myocytes are unclear. Here, using biotinylation, immunofluorescence resonance energy transfer (immunoFRET) microscopy, and RNAi-mediated knockdown, we demonstrate that rab4A-positive early endosomes traffic BKα to the plasma membrane in myocytes of resistance-size cerebral arteries. Angiotensin II (ANG II), a vasoconstrictor, reduced both surface and total BKα, an effect blocked by bisindolylmaleimide-II, concanavalin A, and dynasore, protein kinase C (PKC), internalization, and endocytosis inhibitors, respectively. In contrast, ANG II did not reduce BKα mRNA, and sodium nitroprusside, a nitric oxide donor, did not alter surface BKα protein over the same time course. MG132 and bafilomycin A, proteasomal and lysosomal inhibitors, respectively, also inhibited the ANG II-induced reduction in surface and total BKα, resulting in intracellular BKα accumulation. ANG II-mediated BK channel degradation reduced BK currents in isolated myocytes and functional responses to iberiotoxin, a BK channel blocker, and NS1619, a BK activator, in pressurized (60 mmHg) cerebral arteries. These data indicate that rab4A-positive early endosomes traffic BKα to the plasma membrane in arterial myocytes. We also show that ANG II stimulates PKC-dependent BKα internalization and degradation. These data describe a unique mechanism by which ANG II inhibits arterial myocyte BK currents, by reducing surface channel number, to induce vasoconstriction.


Asunto(s)
Angiotensina II/farmacología , Membrana Celular/metabolismo , Arterias Cerebrales/metabolismo , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Células Musculares/metabolismo , Proteolisis/efectos de los fármacos , Vasoconstricción/efectos de los fármacos , Animales , Arterias Cerebrales/efectos de los fármacos , Endosomas/efectos de los fármacos , Endosomas/metabolismo , Masculino , Potenciales de la Membrana/efectos de los fármacos , Células Musculares/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Ratas , Ratas Sprague-Dawley , Vasoconstrictores/farmacología
5.
Nat Genet ; 38(4): 447-51, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16501573

RESUMEN

Potassium channel mutations have been described in episodic neurological diseases. We report that K+ channel mutations cause disease phenotypes with neurodevelopmental and neurodegenerative features. In a Filipino adult-onset ataxia pedigree, the causative gene maps to 19q13, overlapping the SCA13 disease locus described in a French pedigree with childhood-onset ataxia and cognitive delay. This region contains KCNC3 (also known as Kv3.3), encoding a voltage-gated Shaw channel with enriched cerebellar expression. Sequencing revealed two missense mutations, both of which alter KCNC3 function in Xenopus laevis expression systems. KCNC3(R420H), located in the voltage-sensing domain, had no channel activity when expressed alone and had a dominant-negative effect when co-expressed with the wild-type channel. KCNC3(F448L) shifted the activation curve in the negative direction and slowed channel closing. Thus, KCNC3(R420H) and KCNC3(F448L) are expected to change the output characteristics of fast-spiking cerebellar neurons, in which KCNC channels confer capacity for high-frequency firing. Our results establish a role for KCNC3 in phenotypes ranging from developmental disorders to adult-onset neurodegeneration and suggest voltage-gated K+ channels as candidates for additional neurodegenerative diseases.


Asunto(s)
Ataxia Cerebelosa/genética , Activación del Canal Iónico , Mutación Missense , Mutación , Canales de Potasio Shaw/genética , Secuencia de Aminoácidos , Animales , Humanos , Datos de Secuencia Molecular , Fenotipo , Canales de Potasio Shaw/química , Xenopus laevis
6.
Circ Res ; 111(8): 1027-36, 2012 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-22872152

RESUMEN

RATIONALE: Pressure-induced arterial depolarization and constriction (the myogenic response) is a smooth muscle cell (myocyte)-specific mechanism that controls regional organ blood flow and systemic blood pressure. Several different nonselective cation channels contribute to pressure-induced depolarization, but signaling mechanisms involved are unclear. Similarly uncertain is the contribution of anion channels to the myogenic response and physiological functions and mechanisms of regulation of recently discovered transmembrane 16A (TMEM16A), also termed Anoctamin 1, chloride (Cl(-)) channels in arterial myocytes. OBJECTIVE: To investigate the hypothesis that myocyte TMEM16A channels control membrane potential and contractility and contribute to the myogenic response in cerebral arteries. METHODS AND RESULTS: Cell swelling induced by hyposmotic bath solution stimulated Cl(-) currents in arterial myocytes that were blocked by TMEM16A channel inhibitory antibodies, RNAi-mediated selective TMEM16A channel knockdown, removal of extracellular calcium (Ca(2+)), replacement of intracellular EGTA with BAPTA, a fast Ca(2+) chelator, and Gd(3+) and SKF-96365, nonselective cation channel blockers. In contrast, nimodipine, a voltage-dependent Ca(2+) channel inhibitor, or thapsigargin, which depletes intracellular Ca(2+) stores, did not alter swelling-activated TMEM16A currents. Pressure-induced (-40 mm Hg) membrane stretch activated ion channels in arterial myocyte cell-attached patches that were inhibited by TMEM16A antibodies and were of similar amplitude to recombinant TMEM16A channels. TMEM16A knockdown reduced intravascular pressure-induced depolarization and vasoconstriction but did not alter depolarization-induced (60 mmol/L K(+)) vasoconstriction. CONCLUSIONS: Membrane stretch activates arterial myocyte TMEM16A channels, leading to membrane depolarization and vasoconstriction. Data also provide a mechanism by which a local Ca(2+) signal generated by nonselective cation channels stimulates TMEM16A channels to induce myogenic constriction.


Asunto(s)
Presión Sanguínea/fisiología , Señalización del Calcio/fisiología , Arterias Cerebrales/fisiología , Canales de Cloruro/fisiología , Miocitos del Músculo Liso/fisiología , Animales , Anoctamina-1 , Bloqueadores de los Canales de Calcio/farmacología , Señalización del Calcio/efectos de los fármacos , Arterias Cerebrales/citología , Circulación Cerebrovascular/fisiología , Canales de Cloruro/genética , Cloruros/metabolismo , Células HEK293 , Humanos , Imidazoles/farmacología , Masculino , Miocitos del Músculo Liso/citología , Nimodipina/farmacología , Técnicas de Placa-Clamp , ARN Interferente Pequeño/genética , Ratas , Ratas Sprague-Dawley , Tapsigargina/farmacología , Vasoconstricción/fisiología
7.
J Physiol ; 591(12): 2987-98, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23568894

RESUMEN

Voltage-dependent L-type Ca(2+) channels (CaV1.2) are the primary Ca(2+) entry pathway in vascular smooth muscle cells (myocytes). CaV1.2 channels control systemic blood pressure and organ blood flow and are pathologically altered in vascular diseases, which modifies vessel contractility. The CaV1.2 distal C-terminus is susceptible to proteolytic cleavage, which yields a truncated CaV1.2 subunit and a cleaved C-terminal fragment (CCt). Previous studies in cardiac myocytes and neurons have identified CCt as both a transcription factor and CaV1.2 channel inhibitor, with different signalling mechanisms proposed to underlie some of these effects. CCt existence and physiological functions in arterial myocytes are unclear, but important to study given the functional significance of CaV1.2 channels. Here, we show that CCt exists in myocytes of both rat and human resistance-size cerebral arteries, where it locates to both the nucleus and plasma membrane. Recombinant CCt expression in arterial myocytes inhibited CaV1.2 transcription and reduced CaV1.2 protein. CCt induced a depolarizing shift in the voltage dependence of both CaV1.2 current activation and inactivation, and reduced non-inactivating current in myocytes. Recombinant truncated CCt lacking a putative nuclear localization sequence (92CCt) did not locate to the nucleus and had no effect on arterial CaV1.2 transcription or protein. However, 92CCt shifted the voltage dependence of CaV1.2 activation and inactivation similarly to CCt. CCt and 92CCt both inhibited pressure- and depolarization-induced vasoconstriction, although CCt was a far more effective vasodilator. These data demonstrate that endogenous CCt exists and reduces both CaV1.2 channel expression and voltage sensitivity in arterial myocytes. Thus, CCt is a bi-modal vasodilator.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Músculo Liso Vascular/metabolismo , Vasodilatación , Potenciales de Acción , Transporte Activo de Núcleo Celular , Adolescente , Animales , Canales de Calcio Tipo L/química , Canales de Calcio Tipo L/genética , Núcleo Celular/metabolismo , Arterias Cerebrales/citología , Arterias Cerebrales/fisiología , Humanos , Masculino , Músculo Liso Vascular/fisiología , Mutación , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/fisiología , Señales de Localización Nuclear , Estructura Terciaria de Proteína , Ratas , Ratas Sprague-Dawley , Transcripción Genética
8.
Mol Membr Biol ; 29(8): 321-32, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22881396

RESUMEN

Voltage-gated ion (K(+), Na(+), Ca(2+)) channels contain a pore domain (PD) surrounded by four voltage sensing domains (VSD). Each VSD is made up of four transmembrane helices, S1-S4. S4 contains 6-7 positively charged residues (arginine/lysine) separated two hydrophobic residues, whereas S1-S3 contribute to two negatively charged clusters. These structures are conserved among all members of the voltage-gated ion channel family and play essential roles in voltage gating. The role of S4 charged residues in voltage gating is well established: During depolarization, they move out of the membrane electric field, exerting a mechanical force on channel gates, causing them to open. However, the role of the intervening hydrophobic residues in voltage sensing is unclear. Here we studied the role of these residues in the prototypical Shaker potassium channel. We have altered the physicochemical properties of both charged and hydrophobic positions of S4 and examined the effect of these modifications on the gating properties of the channel. For this, we have introduced cysteines at each of these positions, expressed the mutants in Xenopus oocytes, and examined the effect of in situ addition of charge, via Cd(2+), on channel gating by two-electrode voltage clamp. Our results reveal a face of the S4 helix (comprising residues L358, L361, R365 and R368) where introduction of charge at hydrophobic positions destabilises the closed state and removal of charges from charged positions has an opposite effect. We propose that hydrophobic residues play a crucial role in limiting gating to a physiological voltage range.


Asunto(s)
Interacciones Hidrofóbicas e Hidrofílicas , Canales de Potasio de la Superfamilia Shaker/química , Canales de Potasio de la Superfamilia Shaker/metabolismo , Secuencia de Aminoácidos , Animales , Cadmio/farmacología , Cisteína/genética , Interacciones Hidrofóbicas e Hidrofílicas/efectos de los fármacos , Activación del Canal Iónico/efectos de los fármacos , Iones , Cinética , Datos de Secuencia Molecular , Proteínas Mutantes/metabolismo , Mutación/genética , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Subunidades de Proteína/química , Transporte de Proteínas/efectos de los fármacos , Xenopus
9.
J Biol Chem ; 286(17): 15058-66, 2011 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-21357696

RESUMEN

Voltage-dependent Ca(2+) (Ca(V)1.2) channels are the primary Ca(2+) influx pathway in arterial smooth muscle cells and are essential for contractility regulation by a variety of stimuli, including intravascular pressure. Arterial smooth muscle cell Ca(V)1.2 mRNA is alternatively spliced at exon 1 (e1), generating e1b or e1c variants, with e1c exhibiting relatively smooth muscle-specific expression in the cardiovascular system. Here, we examined physiological functions of Ca(V)1.2e1 variants and tested the hypothesis that targeting Ca(V)1.2e1 modulates resistance size cerebral artery contractility. Custom antibodies that selectively recognize Ca(V)1.2 channel proteins containing sequences encoded by either e1b (Ca(V)1.2e1b) or e1c (Ca(V)1.2e1c) both detected Ca(V)1.2 in rat and human cerebral arteries. shRNA targeting e1b or e1c reduced expression of that Ca(V)1.2 variant, induced compensatory up-regulation of the other variant, decreased total Ca(V)1.2, and reduced intravascular pressure- and depolarization-induced vasoconstriction. Ca(V)1.2e1b and Ca(V)1.2e1c knockdown reduced whole cell Ca(V)1.2 currents, with Ca(V)1.2e1c knockdown most effectively reducing total Ca(V)1.2 and inducing the largest vasodilation. Knockdown of α(2)δ-1, a Ca(V)1.2 auxiliary subunit, reduced surface expression of both Ca(V)1.2e1 variants, inhibiting Ca(V)1.2e1c more than Ca(V)1.2e1b. e1b or e1c overexpression reduced Ca(V)1.2 surface expression and whole cell currents, leading to vasodilation, with e1c overexpression inducing the largest effect. In summary, data indicate that arterial smooth muscle cells express Ca(V)1.2 channels containing e1b or e1c-encoded N termini that contribute to Ca(V)1.2 surface expression, α(2)δ-1 preferentially traffics the Ca(V)1.2e1c variant to the plasma membrane, and targeting of Ca(V)1.2e1 message or the Ca(V)1.2 channel proximal N terminus induces vasodilation.


Asunto(s)
Canales de Calcio Tipo L/genética , Arterias Cerebrales/fisiología , Regulación de la Expresión Génica/fisiología , Miocitos del Músculo Liso/metabolismo , Vasoconstricción , Animales , Calcio/metabolismo , Canales de Calcio Tipo L/fisiología , Membrana Celular/metabolismo , Arterias Cerebrales/citología , Pollos , Cobayas , Humanos , Músculo Liso Vascular/citología , Isoformas de Proteínas/fisiología , Ratas
10.
Circ Res ; 106(10): 1603-12, 2010 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-20378853

RESUMEN

RATIONALE: Inositol 1,4,5-trisphosphate (IP(3))-induced vasoconstriction can occur independently of intracellular Ca(2+) release and via IP(3) receptor (IP(3)R) and canonical transient receptor potential (TRPC) channel activation, but functional signaling mechanisms mediating this effect are unclear. OBJECTIVES: Study mechanisms by which IP(3)Rs stimulate TRPC channels in myocytes of resistance-size cerebral arteries. METHODS AND RESULTS: Immunofluorescence resonance energy transfer (immuno-FRET) microscopy using isoform-selective antibodies indicated that endogenous type 1 IP(3)Rs (IP(3)R1) are in close spatial proximity to TRPC3, but distant from TRPC6 or TRPM4 channels in arterial myocytes. Endothelin-1 (ET-1), a phospholipase C-coupled receptor agonist, elevated immuno-FRET between IP(3)R1 and TRPC3, but not between IP(3)R1 and TRPC6 or TRPM4. TRPC3, but not TRPC6, coimmunoprecipitated with IP(3)R1. TRPC3 and TRPC6 antibodies selectively inhibited recombinant channels, but only the TRPC3 antibody blocked IP(3)-induced nonselective cation current (I(Cat)) in myocytes. TRPC3 knockdown attenuated immuno-FRET between IP(3)R1 and TRPC3, IP(3)-induced I(Cat) activation, and ET-1 and IP(3)-induced vasoconstriction, whereas TRPC6 channel knockdown had no effect. ET-1 did not alter total or plasma membrane-localized TRPC3, as determined using surface biotinylation. RT-PCR demonstrated that C-terminal calmodulin and IP(3)R binding (CIRB) domains are present in myocyte TRPC3 and TRPC6 channels. A peptide corresponding to the IP(3)R N-terminal region that can interact with TRPC channels activated I(Cat). A TRPC3 CIRB domain peptide attenuated IP(3)- and ET-1-induced I(Cat) activation and vasoconstriction. CONCLUSIONS: IP(3) stimulates direct coupling between IP(3)R1 and membrane-resident TRPC3 channels in arterial myocytes, leading to I(Cat) activation and vasoconstriction. Close spatial proximity between IP(3)R1 and TRPC3 establishes this isoform-selective functional interaction.


Asunto(s)
Arterias Cerebrales/fisiología , Receptores de Inositol 1,4,5-Trifosfato/fisiología , Células Musculares/fisiología , Músculo Liso Vascular/fisiología , Canales Catiónicos TRPC/fisiología , Vasoconstricción/fisiología , Animales , Línea Celular , Arterias Cerebrales/citología , Silenciador del Gen , Homeostasis , Humanos , Riñón , Microscopía Confocal , Células Musculares/citología , Ratas , Ratas Sprague-Dawley , Canales Catiónicos TRPC/deficiencia , Canales Catiónicos TRPC/genética , Transfección
11.
Am J Physiol Heart Circ Physiol ; 301(5): H1819-27, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21856902

RESUMEN

Transmembrane protein (TMEM)16A channels are recently discovered membrane proteins that display electrophysiological properties similar to classic Ca(2+)-activated Cl(-) (Cl(Ca)) channels in native cells. The molecular identity of proteins that generate Cl(Ca) currents in smooth muscle cells (SMCs) of resistance-size arteries is unclear. Similarly, whether cerebral artery SMCs generate Cl(Ca) currents is controversial. Here, using molecular biology and patch-clamp electrophysiology, we examined TMEM16A channel expression and characterized Cl(-) currents in arterial SMCs of resistance-size rat cerebral arteries. RT-PCR amplified transcripts for TMEM16A but not TMEM16B-TMEM16H, TMEM16J, or TMEM16K family members in isolated pure cerebral artery SMCs. Western blot analysis using an antibody that recognized recombinant (r)TMEM16A channels detected TMEM16A protein in cerebral artery lysates. Arterial surface biotinylation and immunofluorescence indicated that TMEM16A channels are located primarily within the arterial SMC plasma membrane. Whole cell Cl(Ca) currents in arterial SMCs displayed properties similar to those generated by rTMEM16A channels, including Ca(2+) dependence, current-voltage relationship linearization by an elevation in intracellular Ca(2+) concentration, a Nerstian shift in reversal potential induced by reducing the extracellular Cl(-) concentration, and a negative reversal potential shift when substituting extracellular I(-) for Cl(-). A pore-targeting TMEM16A antibody similarly inhibited both arterial SMC Cl(Ca) and rTMEM16A currents. TMEM16A knockdown using small interfering RNA also inhibited arterial SMC Cl(Ca) currents. In summary, these data indicate that TMEM16A channels are expressed, insert into the plasma membrane, and generate Cl(Ca) currents in cerebral artery SMCs.


Asunto(s)
Calcio/metabolismo , Canales de Cloruro/metabolismo , Cloruros/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Animales , Anoctamina-1 , Western Blotting , Membrana Celular/metabolismo , Canales de Cloruro/genética , Técnica del Anticuerpo Fluorescente , Células HEK293 , Humanos , Masculino , Potenciales de la Membrana , Microscopía Confocal , Arteria Cerebral Media/metabolismo , Técnicas de Placa-Clamp , Reacción en Cadena de la Polimerasa , Interferencia de ARN , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Transfección
12.
Circ Res ; 105(10): 948-55, 2009 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-19797702

RESUMEN

RATIONALE: Voltage-dependent L-type (Ca(V)1.2) Ca(2+) channels are a heteromeric complex formed from pore-forming alpha(1) and auxiliary alpha(2)delta and beta subunits. Ca(V)1.2 channels are the principal Ca(2+) influx pathway in arterial myocytes and regulate multiple physiological functions, including contraction. The macromolecular composition of arterial myocyte Ca(V)1.2 channels remains poorly understood, with no studies having examined the molecular identity or physiological functions of alpha(2)delta subunits. OBJECTIVE: We investigated the functional significance of alpha(2)delta subunits in myocytes of resistance-size (100 to 200 mum diameter) cerebral arteries. METHODS AND RESULTS: alpha(2)delta-1 was the only alpha(2)delta isoform expressed in cerebral artery myocytes. Pregabalin, an alpha(2)delta-1/-2 ligand, and an alpha(2)delta-1 antibody, inhibited Ca(V)1.2 currents in isolated myocytes. Acute pregabalin application reversibly dilated pressurized arteries. Using a novel application of surface biotinylation, data indicated that >95% of Ca(V)1.2 alpha(1) and alpha(2)delta-1 subunits were present in the arterial myocyte plasma membrane. Alpha(2)delta-1 knockdown using short hairpin RNA reduced plasma membrane-localized Ca(V)1.2 alpha(1) subunits, caused a corresponding elevation in cytosolic Ca(V)1.2 alpha(1) subunits, decreased intracellular Ca(2+) concentration, inhibited pressure-induced vasoconstriction ("myogenic tone"), and attenuated pregabalin-induced vasodilation. Prolonged (24-hour) pregabalin exposure did not alter total alpha(2)delta-1 or Ca(V)1.2 alpha(1) proteins but decreased plasma membrane expression of each subunit, which reduced myogenic tone. CONCLUSIONS: alpha(2)delta-1 is essential for plasma membrane expression of arterial myocyte Ca(V)1.2 alpha(1) subunits. alpha(2)delta-1 targeting can block Ca(V)1.2 channels directly and inhibit surface expression of Ca(V)1.2 alpha(1) subunits, leading to vasodilation. These data identify alpha(2)delta-1 as a novel molecular target in arterial myocytes, the manipulation of which regulates contractility.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Arterias Cerebrales/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Vasoconstricción/fisiología , Vasodilatación/fisiología , Animales , Anticonvulsivantes/farmacología , Calcio/metabolismo , Células Cultivadas , Arterias Cerebrales/citología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Técnicas de Silenciamiento del Gen , Masculino , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología , Pregabalina , Isoformas de Proteínas/metabolismo , Subunidades de Proteína/metabolismo , Ratas , Ratas Sprague-Dawley , Vasoconstricción/efectos de los fármacos , Vasodilatación/efectos de los fármacos , Ácido gamma-Aminobutírico/análogos & derivados , Ácido gamma-Aminobutírico/farmacología
13.
Neuron ; 39(3): 467-81, 2003 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-12895421

RESUMEN

A recently proposed model for voltage-dependent activation in K+ channels, largely influenced by the KvAP X-ray structure, suggests that S4 is located at the periphery of the channel and moves through the lipid bilayer upon depolarization. To investigate the physical distance between S4 and the pore domain in functional channels in a native membrane environment, we engineered pairs of cysteines, one each in S4 and the pore of Shaker channels, and identified two instances of spontaneous intersubunit disulfide bond formation, between R362C/A419C and R362C/F416C. After reduction, these cysteine pairs bound Cd2+ with high affinity, verifying that the residues are in atomic proximity. Molecular modeling based on the MthK structure revealed a single position for S4 that was consistent with our results and many other experimental constraints. The model predicts that S4 is located in the groove between pore domains from different subunits, rather than at the periphery of the protein.


Asunto(s)
Modelos Moleculares , Canales de Potasio/química , Canales de Potasio/fisiología , Animales , Femenino , Potenciales de la Membrana/fisiología , Mutación , Oocitos , Canales de Potasio de la Superfamilia Shaker , Xenopus
14.
Sci Signal ; 8(390): ra83, 2015 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-26286025

RESUMEN

Voltage-dependent potassium (K(v)) channels are present in various cell types, including smooth muscle cells (myocytes) of resistance-sized arteries that control systemic blood pressure and regional organ blood flow. Intravascular pressure depolarizes arterial myocytes, stimulating calcium (Ca(2+)) influx through voltage-dependent Ca(2+) (Ca(v)) channels that results in vasoconstriction and also K(+) efflux through K(v) channels that oppose vasoconstriction. We hypothesized that pressure-induced depolarization may not only increase the open probability of plasma membrane-resident K(v) channels but also increase the abundance of these channels at the surface of arterial myocytes to limit vasoconstriction. We found that K(v)1.5 and K(v)2.1 proteins were abundant in the myocytes of resistance-sized mesenteric arteries. K(v)1.5, but not K(v)2.1, continuously recycled between the intracellular compartment and the plasma membrane in contractile arterial myocytes. Using ex vivo preparations of intact arteries, we showed that physiological intravascular pressure through membrane depolarization or membrane depolarization in the absence of pressure inhibited the degradation of internalized K(v)1.5 and increased recycling of K(v)1.5 to the plasma membrane. Accordingly, by stimulating the activity of Ca(v)1.2, membrane depolarization increased whole-cell K(v)1.5 current density in myocytes and K(v)1.5 channel activity in pressurized arteries. In contrast, the total amount and cell surface abundance of K(v)2.1 were independent of intravascular pressure or membrane potential. Thus, our data indicate that intravascular pressure-induced membrane depolarization selectively increased K(v)1.5 surface abundance to increase K(v) currents in arterial myocytes, which would limit vasoconstriction.


Asunto(s)
Membrana Celular/fisiología , Canal de Potasio Kv1.5/fisiología , Arterias Mesentéricas/fisiología , Miocitos del Músculo Liso/fisiología , Vasoconstricción/fisiología , Animales , Western Blotting , Células Cultivadas , Células HEK293 , Humanos , Técnicas In Vitro , Canal de Potasio Kv1.5/genética , Canal de Potasio Kv1.5/metabolismo , Masculino , Potenciales de la Membrana/fisiología , Arterias Mesentéricas/citología , Arterias Mesentéricas/metabolismo , Miocitos del Músculo Liso/metabolismo , Técnicas de Placa-Clamp , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/fisiología
15.
Hypertension ; 60(4): 1006-15, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22949532

RESUMEN

A hallmark of hypertension is an increase in arterial myocyte voltage-dependent Ca2+ (CaV1.2) currents that induces pathological vasoconstriction. CaV1.2 channels are heteromeric complexes composed of a pore-forming CaV1.2α1 with auxiliary α2δ and ß subunits. Molecular mechanisms that elevate CaV1.2 currents during hypertension and the potential contribution of CaV1.2 auxiliary subunits are unclear. Here, we investigated the pathological significance of α2δ subunits in vasoconstriction associated with hypertension. Age-dependent development of hypertension in spontaneously hypertensive rats was associated with an unequal elevation in α2δ-1 and CaV1.2α1 mRNA and protein in cerebral artery myocytes, with α2δ-1 increasing more than CaV1.2α1. Other α2δ isoforms did not emerge in hypertension. Myocytes and arteries of hypertensive spontaneously hypertensive rats displayed higher surface-localized α2δ-1 and CaV1.2α1 proteins, surface α2δ-1:CaV1.2α1 ratio, CaV1.2 current density and noninactivating current, and pressure- and depolarization-induced vasoconstriction than those of Wistar-Kyoto controls. Pregabalin, an α2δ-1 ligand, did not alter α2δ-1 or CaV1.2α1 total protein but normalized α2δ-1 and CaV1.2α1 surface expression, surface α2δ-1:CaV1.2α1, CaV1.2 current density and inactivation, and vasoconstriction in myocytes and arteries of hypertensive rats to control levels. Genetic hypertension is associated with an elevation in α2δ-1 expression that promotes surface trafficking of CaV1.2 channels in cerebral artery myocytes. This leads to an increase in CaV1.2 current-density and a reduction in current inactivation that induces vasoconstriction. Data also suggest that α2δ-1 targeting is a novel strategy that may be used to reverse pathological CaV1.2 channel trafficking to induce cerebrovascular dilation in hypertension.


Asunto(s)
Canales de Calcio/genética , Hipertensión/genética , Músculo Liso Vascular/metabolismo , Regulación hacia Arriba/genética , Vasoconstricción/genética , Animales , Canales de Calcio/metabolismo , Canales de Calcio Tipo L/genética , Canales de Calcio Tipo L/metabolismo , Arterias Cerebrales/efectos de los fármacos , Arterias Cerebrales/metabolismo , Hipertensión/metabolismo , Hipertensión/fisiopatología , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Pregabalina , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/genética , Regulación hacia Arriba/efectos de los fármacos , Vasoconstricción/efectos de los fármacos , Ácido gamma-Aminobutírico/análogos & derivados , Ácido gamma-Aminobutírico/farmacología
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.
Proc Natl Acad Sci U S A ; 102(51): 18718-23, 2005 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-16339906

RESUMEN

In voltage-dependent ether-à-go-go (eag) K+ channels, the process of activation is modulated by Mg2+ and other divalent cations, which bind to a site in the voltage sensor and slow channel opening. Previous analysis of eag ionic and gating currents indicated that Mg2+ has a much larger effect on ionic than gating current kinetics. From this, we hypothesized that ion binding modulates voltage sensor conformational changes that are poorly represented in gating current recordings. We have now tested this proposal by using a combined electrophysiological and optical approach. We find that a fluorescent probe attached near S4 in the voltage sensor reports on two phases of the activation process. One component of the optical signal corresponds to the main charge-moving conformational changes of the voltage sensor. This is the phase of activation that is well represented in gating current recordings. Another component of the optical signal reflects voltage sensor conformational changes that occur at more hyperpolarized potentials. These transitions, which are rate-determining for activation and highly modulated by Mg2+, have not been detected in gating current recordings. Our results demonstrate that the eag voltage sensor undergoes conformational changes that have gone undetected in electrical measurements. These transitions account for the time course of eag activation in the presence and absence of extracellular Mg2+.


Asunto(s)
Canales de Potasio Éter-A-Go-Go/química , Canales de Potasio Éter-A-Go-Go/metabolismo , Activación del Canal Iónico/efectos de los fármacos , Magnesio/farmacología , Animales , Cationes Bivalentes/farmacología , Electrofisiología , Canales de Potasio Éter-A-Go-Go/genética , Fluorescencia , Cinética , Lisina/genética , Lisina/metabolismo , Oocitos/metabolismo , Técnicas de Placa-Clamp , Conformación Proteica , Espectrometría de Fluorescencia , Xenopus laevis
18.
Biophys J ; 87(5): 3110-21, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15347589

RESUMEN

In ether-a-go-go K+ channels, voltage-dependent activation is modulated by ion binding to a site located in an extracellular-facing crevice between transmembrane segments S2 and S3 in the voltage sensor. We find that acidic residues D278 in S2 and D327 in S3 are able to coordinate a variety of divalent cations, including Mg2+, Mn2+, and Ni2+, which have qualitatively similar functional effects, but different half-maximal effective concentrations. Our data indicate that ions binding to individual voltage sensors in the tetrameric channel act without cooperativity to modulate activation gating. We have taken advantage of the unique phenotype of Ni2+ in the D274A channel, which contains a mutation of a nonbinding site residue, to demonstrate that ions can access the binding site from the extracellular solution when the voltage sensor is in the resting conformation. Our results are difficult to reconcile with the x-ray structure of the KvAP K+ channel, in which the binding site residues are widely separated, and with the hydrophobic paddle model for voltage-dependent activation, in which the voltage sensor domain, including the S3-S4 loop, is near the cytoplasmic side of the membrane in the closed channel.


Asunto(s)
Activación del Canal Iónico/fisiología , Potenciales de la Membrana/fisiología , Metales/farmacología , Oocitos/fisiología , Canales de Potasio/fisiología , Sustitución de Aminoácidos , Animales , Sitios de Unión , Células Cultivadas , Relación Dosis-Respuesta a Droga , Canales de Potasio Éter-A-Go-Go , Activación del Canal Iónico/efectos de los fármacos , Iones , Magnesio/farmacología , Manganeso/farmacología , Potenciales de la Membrana/efectos de los fármacos , Mutagénesis Sitio-Dirigida , Níquel/farmacología , Canales de Potasio/efectos de los fármacos , Unión Proteica , Estructura Terciaria de Proteína/efectos de los fármacos , Proteínas Recombinantes/metabolismo , Relación Estructura-Actividad , Xenopus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA