Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Physiol ; 598(15): 3107-3127, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32372450

RESUMEN

KEY POINTS: Spontaneous contractions are essential for normal lymph transport and these contractions are exquisitely sensitive to the KATP channel activator pinacidil. KATP channel Kir6.1 and SUR2B subunits are expressed in mouse lymphatic smooth muscle (LSM) and form functional KATP channels as verified by electrophysiological techniques. Global deletion of Kir6.1 or SUR2 subunits results in severely impaired lymphatic contractile responses to pinacidil. Smooth muscle-specific expression of Kir6.1 gain-of-function mutant (GoF) subunits results in profound lymphatic contractile dysfunction and LSM hyperpolarization that is partially rescued by the KATP inhibitor glibenclamide. In contrast, lymphatic endothelial-specific expression of Kir6.1 GoF has essentially no effect on lymphatic contractile function. The high sensitivity of LSM to KATP channel GoF offers an explanation for the lymphoedema observed in patients with Cantú syndrome, a disorder caused by gain-of-function mutations in genes encoding Kir6.1 or SUR2, and suggests that glibenclamide may be an appropriate therapeutic agent. ABSTRACT: This study aimed to understand the functional expression of KATP channel subunits in distinct lymphatic cell types, and assess the consequences of altered KATP channel activity on lymphatic pump function. KATP channel subunits Kir6.1 and SUR2B were expressed in mouse lymphatic muscle by PCR, but only Kir6.1 was expressed in lymphatic endothelium. Spontaneous contractions of popliteal lymphatics from wild-type (WT) (C57BL/6J) mice, assessed by pressure myography, were very sensitive to inhibition by the SUR2-specific KATP channel activator pinacidil, which hyperpolarized both mouse and human lymphatic smooth muscle (LSM). In vessels from mice with deletion of Kir6.1 (Kir6.1-/- ) or SUR2 (SUR2[STOP]) subunits, contractile parameters were not significantly different from those of WT vessels, suggesting that basal KATP channel activity in LSM is not an essential component of the lymphatic pacemaker, and does not exert a strong influence over contractile strength. However, these vessels were >100-fold less sensitive than WT vessels to pinacidil. Smooth muscle-specific expression of a Kir6.1 gain-of-function (GoF) subunit resulted in severely impaired lymphatic contractions and hyperpolarized LSM. Membrane potential and contractile activity was partially restored by the KATP channel inhibitor glibenclamide. In contrast, lymphatic endothelium-specific expression of Kir6.1 GoF subunits had negligible effects on lymphatic contraction frequency or amplitude. Our results demonstrate a high sensitivity of lymphatic contractility to KATP channel activators through activation of Kir6.1/SUR2-dependent channels in LSM. In addition, they offer an explanation for the lymphoedema observed in patients with Cantú syndrome, a disorder caused by gain-of-function mutations in genes encoding Kir6.1/SUR2.


Asunto(s)
Mutación con Ganancia de Función , Hipertricosis , Adenosina Trifosfato , Animales , Humanos , Canales KATP/genética , Ratones , Ratones Endogámicos C57BL , Músculo Liso , Receptores de Sulfonilureas/genética
2.
Sci Rep ; 10(1): 70, 2020 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-31919478

RESUMEN

The spontaneous contractions of collecting lymphatic vessels provide an essential propulsive force to return lymph centrally. These contractions are driven by an intrinsic electrical pacemaker, working through an unknown underlying ionic mechanism that becomes compromised in some forms of lymphedema. In previous studies, T-type voltage-gated Ca2+ channels (VGCCs) were implicated in this pacemaking mechanism, based on the effects of the reputedly selective T-type VGCC inhibitors mibefradil and Ni2+. Our goal was to test this idea in a more definitive way using genetic knock out mice. First, we demonstrated through both PCR and immunostaining that mouse lymphatic muscle cells expressed Cav3.1 and Cav3.2 and produced functional T-type VGCC currents when patch clamped. We then employed genetic deletion strategies to selectively test the roles of each T-type VGCC isoform in the regulation of lymphatic pacemaking. Surprisingly, global deletion of either, or both, isoform(s) was without significant effect on either the frequency, amplitude, or fractional pump flow of lymphatic collectors from two different regions of the mouse, studied ex vivo. Further, both WT and Cav3.1-/-; 3.2-/- double knock-out lymphatic vessels responded similarly to mibefradil and Ni2+, which substantially reduced contraction amplitudes and slightly increased frequencies at almost all pressures in both strains: a pattern consistent with inhibition of L-type rather than T-type VGCCs. Neither T-type VGCC isoform was required for ACh-induced inhibition of contraction, a mechanism by which those channels in smooth muscle are thought to be targets of endothelium-derived nitric oxide. Sharp intracellular electrode measurements in lymphatic smooth muscle revealed only subtle, but not significant, differences in the resting membrane potential and action potential characteristics between vessels from wild-type and Cav3.1-/-; 3.2-/- double knock-out mice. In contrast, smooth-muscle specific deletion of the L-type VGCC, Cav1.2, completely abolished all lymphatic spontaneous contractions. Collectively our results suggest that, although T-type VGCCs are expressed in mouse lymphatic smooth muscle, they do not play a significant role in modulating the frequency of the ionic pacemaker or the amplitude of spontaneous contractions. We conclude that the effects of mibefradil and Ni2+ in other lymphatic preparations are largely or completely explained by off-target effects on L-type VGCCs, which are essential for controlling both the frequency and strength of spontaneous contractions.


Asunto(s)
Canales de Calcio Tipo T/genética , Vasos Linfáticos/fisiología , Contracción Muscular/fisiología , Animales , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo L/química , Canales de Calcio Tipo L/genética , Canales de Calcio Tipo L/metabolismo , Canales de Calcio Tipo T/deficiencia , Canales de Calcio Tipo T/metabolismo , Masculino , Potenciales de la Membrana/efectos de los fármacos , Mibefradil/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Contracción Muscular/efectos de los fármacos , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Níquel/farmacología , Marcapaso Artificial , Ratas , Ratas Wistar
3.
J Gen Physiol ; 151(4): 532-554, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30862712

RESUMEN

Lymphatic collecting vessels exhibit spontaneous contractions with a pressure-dependent contraction frequency. The initiation of contraction has been proposed to be mediated by the activity of a Ca2+-activated Cl- channel (CaCC). Here, we show that the canonical CaCC Anoctamin 1 (Ano1, TMEM16a) plays an important role in lymphatic smooth muscle pacemaking. We find that isolated murine lymphatic muscle cells express Ano1, and demonstrate functional CaCC currents that can be inhibited by the Ano1 inhibitor benzbromarone. These currents are absent in lymphatic muscle cells from Cre transgenic mouse lines targeted for Ano1 genetic deletion in smooth muscle. We additionally show that loss of functional Ano1 in murine inguinal-axillary lymphatic vessels, whether through genetic manipulation or pharmacological inhibition, results in an impairment of the pressure-frequency relationship that is attributable to a hyperpolarized resting membrane potential and a significantly depressed diastolic depolarization rate preceding each action potential. These changes are accompanied by alterations in action potential shape and duration, and a reduced duration but increased amplitude of the action potential-induced global "Ca2+ flashes" that precede lymphatic contractions. These findings suggest that an excitatory Cl- current provided by Ano1 is critical for mediating the pressure-sensitive contractile response and is a major component of the murine lymphatic action potential.


Asunto(s)
Anoctamina-1/metabolismo , Vasos Linfáticos/fisiología , Animales , Anoctamina-1/genética , Benzbromarona/farmacología , Calcio/metabolismo , Regulación de la Expresión Génica , Vasos Linfáticos/efectos de los fármacos , Masculino , Potenciales de la Membrana , Ratones , Ratones Transgénicos , Presión , Conformación Proteica , Uricosúricos/farmacología
4.
Biophys J ; 115(5): 936-949, 2018 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-30143234

RESUMEN

Contractions of lymphangions, i.e., the segment between two one-way lymphatic valves, generate the pressure gradients that propel lymph back to the circulation. Each lymphangion is comprised of an inner sheet of lymphatic endothelial cells circumscribed by one or more layers of lymphatic muscle cells (LMCs). Each contraction is produced by an LMC action potential (AP) that propagates via gap junctions along the lymphangion. Yet, electrical coupling within and between cell layers and the impact on AP waves is poorly understood. Here, we combine studies in rat and mouse lymphatic vessels with mathematical modeling to show that initiation of AP waves depends on high input resistance (low current drain), whereas propagation depends on morphology and sufficient LMC:LMC coupling. Simulations show that 1) myoendothelial coupling is insignificant to facilitate AP generation and sustain an experimentally measured cross-junctional potential difference of 25 mV, i.e., AP waves propagate along the LMC layer only; 2) LMC:LMC resistance is estimated around 2-10 MΩ but depends on vessel structure and cell-cell coupling, e.g., some degree of LMC overlap protects AP waves against LMC decoupling; 3) the propensity of AP wave initiation is highest around the valves, where the density of LMCs is low; and 4) a single pacemaker cell embedded in the LMC layer must be able to generate very large currents to overcome the current drain from the layer. However, the required current generation to initiate an AP wave is reduced upon stimulation of multiple adjacent LMCs. With stimulation of all LMCs, AP waves can also arise from heterogeneity in the electrical activity of LMCs. The findings advance our understanding of the electrical constraints that underlie initiation of APs in the LMC layer and make testable predictions about how morphology, LMC excitability, and LMC:LMC electrical coupling interact to determine the ability to initiate and propagate AP waves in small lymphatic vessels.


Asunto(s)
Fenómenos Electrofisiológicos , Sistema Linfático/fisiología , Animales , Sistema Linfático/irrigación sanguínea , Ratones , Modelos Biológicos , Ratas
5.
Am J Physiol Cell Physiol ; 305(5): C568-77, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23804206

RESUMEN

The activity of persistent Ca²âº sparklets, which are characterized by longer and more frequent channel open events than low-activity sparklets, contributes substantially to steady-state Ca²âº entry under physiological conditions. Here, we addressed two questions related to the regulation of Ca²âº sparklets by PKC-α and c-Src, both of which increase whole cell Cav1.2 current: 1) Does c-Src activation enhance persistent Ca²âº sparklet activity? 2) Does PKC-α activate c-Src to produce persistent Ca²âº sparklets? With the use of total internal reflection fluorescence microscopy, Ca²âº sparklets were recorded from voltage-clamped tsA-201 cells coexpressing wild-type (WT) or mutant Cav1.2c (the neuronal isoform of Cav1.2) constructs ± active or inactive PKC-α/c-Src. Cells expressing Cav1.2c exhibited both low-activity and persistent Ca²âº sparklets. Persistent Ca²âº sparklet activity was significantly reduced by acute application of the c-Src inhibitor PP2 or coexpression of kinase-dead c-Src. Cav1.2c constructs mutated at one of two COOH-terminal residues (Y²¹²²F and Y²¹³9F) were used to test the effect of blocking putative phosphorylation sites for c-Src. Expression of Y²¹²²F but not Y²¹³9F Cav1.2c abrogated the potentiating effect of c-Src on Ca²âº sparklet activity. We could not detect a significant change in persistent Ca²âº sparklet activity or density in cells coexpressing Cav1.2c + PKC-α, regardless of whether WT or Y²¹²²F Cav1.2c was used, or after PP2 application, suggesting that PKC-α does not act upstream of c-Src to produce persistent Ca²âº sparklets. However, our results indicate that persistent Ca²âº sparklet activity is promoted by the action of c-Src on residue Y²¹²² of the Cav1.2c COOH terminus.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Calcio/metabolismo , Neuronas/metabolismo , Proteína Quinasa C-alfa/metabolismo , Familia-src Quinasas/metabolismo , Animales , Proteína Tirosina Quinasa CSK , Canales de Calcio Tipo L/genética , Línea Celular Transformada , Regulación de la Expresión Génica , Humanos , Microscopía Fluorescente , Mutación , Neuronas/citología , Neuronas/efectos de los fármacos , Técnicas de Placa-Clamp , Fosforilación , Proteína Quinasa C-alfa/genética , Inhibidores de Proteínas Quinasas/farmacología , Estructura Terciaria de Proteína , Pirimidinas/farmacología , Ratas , Transducción de Señal , Transfección , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/genética
6.
Am J Physiol Cell Physiol ; 300(3): C477-89, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21178109

RESUMEN

Engagement of α(5)ß(1)-integrin by fibronectin (FN) acutely enhances Cav1.2 channel (Ca(L)) current in rat arteriolar smooth muscle and human embryonic kidney cells (HEK293-T) expressing Ca(L). Using coimmunoprecipitation strategies, we show that coassociation of Ca(L) with α(5)- or ß(1)-integrin in HEK293-T cells is specific and depends on cell adhesion to FN. In rat arteriolar smooth muscle, coassociations between Ca(L) and α(5)ß(1)-integrin and between Ca(L) and phosphorylated c-Src are also revealed and enhanced by FN treatment. Using site-directed mutagenesis of Ca(L) heterologously expressed in HEK293-T cells, we identified two regions of Ca(L) required for these interactions: 1) COOH-terminal residues Ser(1901) and Tyr(2122), known to be phosphorylated by protein kinase A (PKA) and c-Src, respectively; and 2) two proline-rich domains (PRDs) near the middle of the COOH terminus. Immunofluorescence confocal imaging revealed a moderate degree of wild-type Ca(L) colocalization with ß(1)-integrin on the plasma membrane. Collectively, our results strongly suggest that 1) upon ligation by FN, Ca(L) associates with α(5)ß(1)-integrin in a macromolecular complex including PKA, c-Src, and potentially other protein kinases; 2) phosphorylation of Ca(L) at Y(2122) and/or S(1901) is required for association of Ca(L) with α(5)ß(1)-integrin; and 3) c-Src, via binding to PRDs that reside in the II-III linker region and/or the COOH terminus of Ca(L), mediates current potentiation following α(5)ß(1)-integrin engagement. These findings provide new evidence for how interactions between α(5)ß(1)-integrin and FN can modulate Ca(L) entry and consequently alter the physiological function of multiple types of excitable cells.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Señalización del Calcio/genética , Membrana Celular/metabolismo , Células Epiteliales/metabolismo , Integrina alfa5beta1/fisiología , Músculo Liso Vascular/metabolismo , Animales , Canales de Calcio Tipo L/química , Canales de Calcio Tipo L/genética , Señalización del Calcio/efectos de los fármacos , Adhesión Celular/genética , Adhesión Celular/fisiología , Membrana Celular/química , Membrana Celular/ultraestructura , Células Cultivadas , Células Epiteliales/química , Fibronectinas/genética , Fibronectinas/metabolismo , Fibronectinas/fisiología , Células HEK293 , Humanos , Integrina alfa5beta1/química , Integrina alfa5beta1/genética , Microscopía Confocal , Músculo Liso Vascular/química , Músculo Liso Vascular/citología , Mutagénesis Sitio-Dirigida , Fosforilación/genética , Fosforilación/fisiología , Ratas
7.
Adv Exp Med Biol ; 674: 69-79, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20549941

RESUMEN

A role for integrins in mechanotransduction has been suggested because these molecules form an important mechanical link between the extracellular matrix (ECM) and the cytoskeleton. An example of mechanotransduction in blood vessels is the myogenic response--the rapid and maintained constriction of arterioles in response to pressure elevation. L-type calcium channels and large-conductance, calcium-activated potassium (BK) channels are known to play important roles in the myogenic response and in the maintenance of myogenic (pressure-induced) vascular tone. Our recent studies on isolated, cannulated arterioles and freshly-dispersed arteriolar smooth muscle cells show that both L-type calcium channels (Ca(v)1.2) and BK channels are regulated by alpha5beta1 integrin activation. Alpha5beta1 integrin interacts with the ECM protein fibronectin, which is distributed in basement membrane and interstitial matrices surrounding smooth muscle cells within the arteriolar wall. Truncation and site-directed mutagenesis strategies reveal that regulation of Ca(v)1.2 by alpha5beta1 integrin requires phosphorylation of the channel alpha1C subunit at C-terminal residues Ser-1901 and Tyr-2122. Likewise, BK channel potentiation by alpha5beta1 integrin activation requires c-Src phosphorylation of the channel alpha-subunit at residue Tyr-766. Thus, both L-type calcium channels and BK channels can be regulated coordinately through integrin-linked phosphorylation cascades involving c-Src. We propose that these two channels are under constitutive control by alpha5beta1 integrin-fibronectin interactions in the vessel wall such that the balance of their activity determines myogenic tone and the vascular response to vessel wall injury/remodeling.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Fibronectinas/metabolismo , Integrina alfa5beta1/metabolismo , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Transducción de Señal , Animales , Membrana Basal/lesiones , Membrana Basal/metabolismo , Membrana Basal/patología , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Genes src , Humanos , Tono Muscular , Músculo Liso Vascular/lesiones , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Fosforilación
8.
J Biol Chem ; 285(1): 131-41, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19887442

RESUMEN

Large conductance, calcium-activated K(+) (BK) channels are important regulators of cell excitability and recognized targets of intracellular kinases. BK channel modulation by tyrosine kinases, including focal adhesion kinase and c-src, suggests their potential involvement in integrin signaling. Recently, we found that fibronectin, an endogenous alpha5beta1 integrin ligand, enhances BK channel current through both Ca(2+)- and phosphorylation-dependent mechanisms in vascular smooth muscle. Here, we show that macroscopic currents from HEK 293 cells expressing murine BK channel alpha-subunits (mSlo) are acutely potentiated following alpha5beta1 integrin activation. The effect occurs in a Ca(2+)-dependent manner, 1-3 min after integrin engagement. After integrin activation, normalized conductance-voltage relations for mSlo are left-shifted at free Ca(2+) concentrations >or=1 microm. Overexpression of human c-src with mSlo, in the absence of integrin activation, leads to similar shifts in mSlo Ca(2+) sensitivity, whereas overexpression of catalytically inactive c-src blocks integrin-induced potentiation. However, neither integrin activation nor c-src overexpression potentiates current in BK channels containing a point mutation at Tyr-766. Biochemical tests confirmed the critical importance of residue Tyr-766 in integrin-induced channel phosphorylation. Thus, BK channel activity is enhanced by alpha5beta1 integrin activation, likely through an intracellular signaling pathway involving c-src phosphorylation of the channel alpha-subunit at Tyr-766. The net result is increased current amplitude, enhanced Ca(2+) sensitivity, and rate of activation of the BK channel, which would collectively promote smooth muscle hyperpolarization in response to integrin-extracellular matrix interactions.


Asunto(s)
Calcio/farmacología , Integrina alfa5beta1/metabolismo , Activación del Canal Iónico/efectos de los fármacos , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Sustitución de Aminoácidos/genética , Animales , Línea Celular , Activación Enzimática/efectos de los fármacos , Fibronectinas/farmacología , Humanos , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/química , Ratones , Datos de Secuencia Molecular , Mutación/genética , Fosforilación/efectos de los fármacos , Fosfotirosina/metabolismo , Pirimidinas/farmacología
9.
J Physiol ; 586(6): 1699-713, 2008 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-18218680

RESUMEN

Injury/degradation of the extracellular matrix (ECM) is associated with vascular wall remodelling and impaired reactivity, a process in which altered ECM-integrin interactions play key roles. Previously, we found that peptides containing the RGD integrin-binding sequence produce sustained vasodilatation of rat skeletal muscle arterioles. Here, we tested the hypothesis that RGD ligands work through alpha5beta1 integrin to modulate the activity of large conductance, Ca(2+)-activated K(+) (BK) channels in arteriolar smooth muscle. K(+) currents were recorded in single arteriolar myocytes using whole-cell and single-channel patch clamp methods. Activation of alpha5beta1 integrin by an appropriate, insoluble alpha5beta1 antibody resulted in a 30-50% increase in the amplitude of iberiotoxin (IBTX)-sensitive, whole-cell K(+) current. Current potentiation occurred 1-8 min after bead-antibody application to the cell surface. Similarly, the endogenous alpha5beta1 integrin ligand fibronectin (FN) potentiated IBTX-sensitive K(+) current by 26%. Current potentiation was blocked by the c-Src inhibitor PP2 but not by PP3 (0.1-1 mum). In cell-attached patches, number of open channels x open probability (NP(o)) of a 230-250 pS K(+) channel was significantly increased after FN application locally to the external surface of cell-attached patches through the recording pipette. In excised, inside-out patches, the same method of FN application led to large, significant increases in NP(o) and caused a leftward shift in the NP(o)-voltage relationship at constant [Ca(2+)]. PP2 (but not PP3) nearly abolished the effect of FN on channel activity, suggesting that signalling between the integrin and channel involved an increase in Ca(2+)sensitivity of the channel via a membrane-delimited pathway. The effects of alpha5beta1 integrin activation on both whole-cell and single-channel BK currents could be reproduced in HEK 293 cells expressing the BK channel alpha-subunit. This is the first demonstration at the single-channel level that integrin signalling can regulate an ion channel. Our results show that alpha5beta1 integrin activation potentiates BK channel activity in vascular smooth muscle through both Ca(2+)- and c-Src-dependent mechanisms. This mechanism is likely to play a role in the arteriolar dilatation and impaired vascular reactivity associated with ECM degradation.


Asunto(s)
Arteriolas/fisiología , Integrina alfa5beta1/metabolismo , Activación del Canal Iónico/fisiología , Músculo Liso/fisiología , Miocitos Cardíacos/fisiología , Canales de Potasio Calcio-Activados/fisiología , Adaptación Fisiológica/fisiología , Animales , Células Cultivadas , Masculino , Ratas , Ratas Sprague-Dawley
10.
J Biol Chem ; 281(20): 14015-25, 2006 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-16554304

RESUMEN

L-type, voltage-gated Ca2+ channels (CaL) play critical roles in brain and muscle cell excitability. Here we show that currents through heterologously expressed neuronal and smooth muscle CaL channel isoforms are acutely potentiated following alpha5beta1 integrin activation. Only the alpha1C pore-forming channel subunit is critical for this process. Truncation and site-directed mutagenesis strategies reveal that regulation of Cav1.2 by alpha5beta1 integrin requires phosphorylation of alpha1C C-terminal residues Ser1901 and Tyr2122. These sites are known to be phosphorylated by protein kinase A (PKA) and c-Src, respectively, and are conserved between rat neuronal (Cav1.2c) and smooth muscle (Cav1.2b) isoforms. Kinase assays are consistent with phosphorylation of these two residues by PKA and c-Src. Following alpha5beta1 integrin activation, native CaL channels in rat arteriolar smooth muscle exhibit potentiation that is completely blocked by combined PKA and Src inhibition. Our results demonstrate that integrin-ECM interactions are a common mechanism for the acute regulation of CaL channels in brain and muscle. These findings are consistent with the growing recognition of the importance of integrin-channel interactions in cellular responses to injury and the acute control of synaptic and blood vessel function.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Integrina alfa5beta1/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Animales , Encéfalo/metabolismo , Proteína Tirosina Quinasa CSK , Datos de Secuencia Molecular , Músculos/metabolismo , Miocitos del Músculo Liso/metabolismo , Neuronas/metabolismo , Estructura Terciaria de Proteína , Ratas , Ratas Sprague-Dawley , Porcinos , Familia-src Quinasas
11.
Cell Biochem Biophys ; 36(1): 41-66, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-11939371

RESUMEN

Ion channels are regulated by protein phosphorylation and dephosphorylation of serine, threonine, and tyrosine residues. Evidence for regulation of channels by tyrosine phosphorylation comes primarily from investigations of the effects of growth factors, which act through receptor tyrosine kinases. The purpose of the present work is to summarize evidence for the regulation of ion channels by integrins, through their downstream, nonreceptor tyrosine kinases. We review both direct and indirect evidence for this regulation, with particular emphasis on Ca2+-activated K+ and voltage-gated Ca2+ channels. We then discuss the critical roles that cytoskeletal, focal-adhesion, and channel-associated scaffolding proteins may play in localizing nonreceptor tyrosine kinases to the vicinity of ion channels. We conclude by speculating on the physiological significance of these regulatory pathways.


Asunto(s)
Calcio/metabolismo , Integrinas/metabolismo , Iones , Potasio/metabolismo , Animales , División Celular , Citoesqueleto/metabolismo , Matriz Extracelular/metabolismo , Humanos , Modelos Biológicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...