Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Lab Invest ; 100(8): 1057-1067, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32341518

RESUMEN

Endothelial barrier disruption is a hallmark of tissue injury, edema, and inflammation. Vascular endothelial cells express the G protein-coupled receptor (GPCR) protease acctivated receptor 1 (PAR1) and the ion channel transient receptor potential vanilloid 4 (TRPV4), and these signaling proteins are known to respond to inflammatory conditions and promote edema through remodeling of cell-cell junctions and modulation of endothelial barriers. It has previously been established that signaling initiated by the related protease activated receptor 2 (PAR2) is enhanced by TRPV4 in sensory neurons and that this functional interaction plays a critical role in the development of neurogenic inflammation and nociception. Here, we investigated the PAR1-TRPV4 axis, to determine if TRPV4 plays a similar role in the control of edema mediated by thrombin-induced signaling. Using Evans Blue permeation and retention as an indication of increased vascular permeability in vivo, we showed that TRPV4 contributes to PAR1-induced vascular hyperpermeability in the airways and upper gastrointestinal tract of mice. TRPV4 contributes to sustained PAR1-induced Ca2+ signaling in recombinant cell systems and to PAR1-dependent endothelial junction remodeling in vitro. This study supports the role of GPCR-TRP channel functional interactions in inflammatory-associated changes to vascular function and indicates that TRPV4 is a signaling effector for multiple PAR family members.


Asunto(s)
Inflamación/genética , Receptor PAR-1/genética , Receptor PAR-2/genética , Transducción de Señal/genética , Canales Catiónicos TRPV/genética , Animales , Calcio/metabolismo , Permeabilidad Capilar/genética , Edema/genética , Edema/metabolismo , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Inflamación/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor PAR-1/metabolismo , Receptor PAR-2/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Canales Catiónicos TRPV/metabolismo
2.
J Biol Chem ; 288(8): 5790-802, 2013 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-23288842

RESUMEN

G protein-coupled receptors of nociceptive neurons can sensitize transient receptor potential (TRP) ion channels, which amplify neurogenic inflammation and pain. Protease-activated receptor 2 (PAR(2)), a receptor for inflammatory proteases, is a major mediator of neurogenic inflammation and pain. We investigated the signaling mechanisms by which PAR(2) regulates TRPV4 and determined the importance of tyrosine phosphorylation in this process. Human TRPV4 was expressed in HEK293 cells under control of a tetracycline-inducible promoter, allowing controlled and graded channel expression. In cells lacking TRPV4, the PAR(2) agonist stimulated a transient increase in [Ca(2+)](i). TRPV4 expression led to a markedly sustained increase in [Ca(2+)](i). Removal of extracellular Ca(2+) and treatment with the TRPV4 antagonists Ruthenium Red or HC067047 prevented the sustained response. Inhibitors of phospholipase A(2) and cytochrome P450 epoxygenase attenuated the sustained response, suggesting that PAR(2) generates arachidonic acid-derived lipid mediators, such as 5',6'-EET, that activate TRPV4. Src inhibitor 1 suppressed PAR(2)-induced activation of TRPV4, indicating the importance of tyrosine phosphorylation. The TRPV4 tyrosine mutants Y110F, Y805F, and Y110F/Y805F were expressed normally at the cell surface. However, PAR(2) was unable to activate TRPV4 with the Y110F mutation. TRPV4 antagonism suppressed PAR(2) signaling to primary nociceptive neurons, and TRPV4 deletion attenuated PAR(2)-stimulated neurogenic inflammation. Thus, PAR(2) activation generates a signal that induces sustained activation of TRPV4, which requires a key tyrosine residue (TRPV4-Tyr-110). This mechanism partly mediates the proinflammatory actions of PAR(2).


Asunto(s)
Receptor PAR-2/metabolismo , Canales Catiónicos TRPV/metabolismo , Animales , Calcio/metabolismo , Citocromo P-450 CYP2J2 , Inhibidores Enzimáticos del Citocromo P-450 , Sistema Enzimático del Citocromo P-450 , Células HEK293 , Humanos , Inflamación , Masculino , Ratones , Modelos Biológicos , Mutagénesis Sitio-Dirigida , Dolor , Inhibidores de Fosfolipasa A2 , Fosforilación , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Tirosina/química , Tirosina/metabolismo
3.
J Biol Chem ; 287(26): 21765-72, 2012 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-22570472

RESUMEN

The balance of glycosylation and deglycosylation of ion channels can markedly influence their function and regulation. However, the functional importance of glycosylation of the TRPV1 receptor, a key sensor of pain-sensing nerves, is not well understood, and whether TRPV1 is glycosylated in neurons is unclear. We report that TRPV1 is N-glycosylated and that N-glycosylation is a major determinant of capsaicin-evoked desensitization and ionic permeability. Both N-glycosylated and unglycosylated TRPV1 was detected in extracts of peripheral sensory nerves by Western blotting. TRPV1 expressed in HEK-293 cells exhibited various degrees of glycosylation. A mutant of asparagine 604 (N604T) was not glycosylated but did not alter plasma membrane expression of TRPV1. Capsaicin-evoked increases in intracellular calcium ([Ca(2+)](i)) were sustained in wild-type TRPV1 HEK-293 cells but were rapidly desensitized in N604T TRPV1 cells. There was marked cell-to-cell variability in capsaicin responses and desensitization between individual cells expressing wild-type TRPV1 but highly uniform responses in cells expressing N604T TRPV1, consistent with variable levels of glycosylation of the wild-type channel. These differences were also apparent when wild-type or N604T TRPV1-GFP fusion proteins were expressed in neurons from trpv1(-/-) mice. Capsaicin evoked a marked, concentration-dependent increase in uptake of the large cationic dye YO-PRO-1 in cells expressing wild-type TRPV1, indicative of loss of ion selectivity, that was completely absent in cells expressing N604T TRPV1. Thus, TRPV1 is variably N-glycosylated and glycosylation is a key determinant of capsaicin regulation of TRPV1 desensitization and permeability. Our findings suggest that physiological or pathological alterations in TRPV1 glycosylation would affect TRPV1 function and pain transmission.


Asunto(s)
Canales Catiónicos TRPV/química , Animales , Biotinilación , Membrana Celular/metabolismo , Colorantes/farmacología , Relación Dosis-Respuesta a Droga , Vectores Genéticos , Glicosilación , Células HEK293 , Humanos , Iones , Masculino , Ratones , Ratones Transgénicos , Neuronas/metabolismo , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/química , Permeabilidad , Unión Proteica , Estructura Terciaria de Proteína , Ratas , Canales Catiónicos TRPV/metabolismo
4.
Am J Physiol Heart Circ Physiol ; 303(8): H1011-21, 2012 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-22886413

RESUMEN

Circulating ghrelin reduces blood pressure, but the mechanism for this action is unknown. This study investigated whether ghrelin has direct vasodilator effects mediated through the growth hormone secretagogue receptor 1a (GHSR1a) and whether ghrelin reduces sympathetic nerve activity. Mice expressing enhanced green fluorescent protein under control of the promoter for growth hormone secretagogue receptor (GHSR) and RT-PCR were used to locate sites of receptor expression. Effects of ghrelin and the nonpeptide GHSR1a agonist capromorelin on rat arteries and on transmission in sympathetic ganglia were measured in vitro. In addition, rat blood pressure and sympathetic nerve activity responses to ghrelin were determined in vivo. In reporter mice, expression of GHSR was revealed at sites where it has been previously demonstrated (hypothalamic neurons, renal tubules, sympathetic preganglionic neurons) but not in any artery studied, including mesenteric, cerebral, and coronary arteries. In rat, RT-PCR detected GHSR1a mRNA expression in spinal cord and kidney but not in the aorta or in mesenteric arteries. Moreover, the aorta and mesenteric arteries from rats were not dilated by ghrelin or capromorelin at concentrations >100 times their EC(50) determined in cells transfected with human or rat GHSR1a. These agonists did not affect transmission from preganglionic sympathetic neurons that express GHSR1a. Intravenous application of ghrelin lowered blood pressure and decreased splanchnic nerve activity. It is concluded that the blood pressure reduction to ghrelin occurs concomitantly with a decrease in sympathetic nerve activity and is not caused by direct actions on blood vessels or by inhibition of transmission in sympathetic ganglia.


Asunto(s)
Presión Sanguínea/fisiología , Sistema Cardiovascular/inervación , Ganglios Simpáticos/fisiología , Ghrelina/metabolismo , Receptores de Ghrelina/metabolismo , Animales , Aorta Torácica/inervación , Aorta Torácica/fisiología , Presión Sanguínea/efectos de los fármacos , Ganglios Simpáticos/efectos de los fármacos , Ghrelina/farmacología , Proteínas Fluorescentes Verdes/genética , Células HEK293 , Humanos , Ligandos , Masculino , Arterias Mesentéricas/inervación , Arterias Mesentéricas/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Piperidinas/farmacología , Pirazoles/farmacología , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Ghrelina/agonistas , Receptores de Ghrelina/genética , Vasodilatación/efectos de los fármacos , Vasodilatación/fisiología
5.
Neuron ; 34(2): 209-20, 2002 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-11970863

RESUMEN

M(1) muscarinic (M(1)AChRs) and B(2) bradykinin (B(2)Rs) receptors are two PLCbeta-coupled receptors that mobilize Ca(2+) in nonexcitable cells. In many neurons, however, B(2)Rs but not M(1)AChRs mobilize intracellular Ca(2+). We have studied the membrane organization and dynamics underlying this coupling specificity by using Trp channels as biosensors for real-time detection of PLCbeta products. We found that, in sympathetic neurons, although both receptors rapidly produced DAG and InsP(3) as messengers, only InsP(3) formed by B(2)Rs has the ability to activate IP(3)Rs. This exclusive coupling results from spatially restricted complexes linking B(2)Rs to IP(3)Rs, a missing partnership for M(1)AChRs. These complexes allow fast and localized rises of InsP(3), necessary to activate the low-affinity neuronal IP(3)R. Thus, these signaling microdomains are of critical importance for the induction of selective responses, discriminating proinflammatory information associated with B(2)Rs from cholinergic neurotransmission.


Asunto(s)
Canales de Calcio/fisiología , Inositol 1,4,5-Trifosfato/metabolismo , Estructura Terciaria de Proteína/fisiología , Receptores de Bradiquinina/fisiología , Receptores Citoplasmáticos y Nucleares/fisiología , Receptores Muscarínicos/fisiología , Transducción de Señal/fisiología , Actinas/fisiología , Animales , Técnicas Biosensibles , Calcio/metabolismo , Canales de Calcio/metabolismo , Calmodulina/fisiología , Citoesqueleto/fisiología , Diglicéridos/biosíntesis , Receptores de Inositol 1,4,5-Trifosfato , Isoenzimas/genética , Isoenzimas/metabolismo , Cinética , Fosfolipasa C beta , Proteína Quinasa C/genética , Proteína Quinasa C/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor de Bradiquinina B2 , Receptor Muscarínico M1 , Canales Catiónicos TRPC , Fosfolipasas de Tipo C/metabolismo
6.
J Neurosci ; 26(5): 1588-95, 2006 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-16452682

RESUMEN

Basal forebrain (BF) neurons provide the principal cholinergic drive to the hippocampus and cortex. Their degeneration is associated with the cognitive defects of Alzheimer's disease. Immunohistochemical studies suggest that some of these neurons contain glutamate, so might also release it. To test this, we made microisland cultures of single BF neurons from 12- to 14-d-old rats. Over 1-8 weeks in culture, neuronal processes made autaptic connections onto the neuron. In 34 of 36 cells tested, a somatically generated action potential was followed by a short-latency EPSC that was blocked by 1 mM kynurenic acid, showing that they released glutamate. To test whether the same neuron also released acetylcholine, we placed a voltage-clamped rat myoball expressing nicotinic receptors in contact with a neurite. In six of six neurons tested, the glutamatergic EPSC was accompanied by a nicotinic (hexamethonium-sensitive) myoball current. Stimulation of the M2-muscarinic presynaptic receptors (characterized using tripitramine and pirenzepine) produced a parallel inhibition of autaptic glutamatergic and myoball nicotinic responses; metabotropic glutamate receptor stimulation produced similar but less consistent and weaker effects. Atropine enhanced the glutamatergic EPSCs during repetitive stimulation by 25 +/- 6%; the anti-cholinesterase neostigmine reduced the train EPSCs by 37 +/- 6%. Hence, synaptically released acetylcholine exerted a negative-feedback inhibition of coreleased glutamate. We conclude that most cholinergic basal forebrain neurons are capable of releasing glutamate as a cotransmitter and that the release of both transmitters is subject to simultaneous feedback inhibition by synaptically released acetylcholine. This has implications for BF neuron function and for the use of cholinesterase inhibitors in Alzheimer's disease.


Asunto(s)
Acetilcolina/metabolismo , Ácido Glutámico/metabolismo , Neuronas/metabolismo , Prosencéfalo/metabolismo , Potenciales de Acción , Animales , Células Cultivadas , Inhibición Neural , Neuronas/citología , Neuronas/fisiología , Técnicas de Placa-Clamp , Prosencéfalo/citología , Ratas , Ratas Sprague-Dawley , Receptor Muscarínico M2/fisiología , Receptores de Glutamato/metabolismo , Receptores Presinapticos/fisiología , Sinapsis/metabolismo
7.
J Neurosci ; 25(13): 3400-13, 2005 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-15800195

RESUMEN

The relationship between receptor-induced membrane phosphatidylinositol-4'5'-bisphosphate (PIP2) hydrolysis and M-current inhibition was assessed in single-dissociated rat sympathetic neurons by simultaneous or parallel recording of membrane current and membrane-to-cytosol translocation of the fluorescent PIP2/inositol 1,4,5-trisphosphate (IP3)-binding peptide green fluorescent protein-tagged pleckstrin homology domain of phospholipase C (GFP-PLCdelta-PH). The muscarinic receptor agonist oxotremorine-M produced parallel time- and concentration-dependent M-current inhibition and GFP-PLCdelta-PH translocation; bradykinin also produced parallel time-dependent inhibition and translocation. Phosphatidylinositol-4-phosphate-5-kinase (PI5-K) overexpression reduced both M-current inhibition and GFP-PLCdelta-PH translocation by both oxotremorine-M and bradykinin. These effects were partly reversed by wortmannin, which inhibits phosphatidylinositol-4-kinase (PI4-K). PI5-K overexpression also reduced the inhibitory action of oxotremorine-M on PIP2-gated G-protein-gated inward rectifier (Kir3.1/3.2) channels; bradykinin did not inhibit these channels. Overexpression of neuronal calcium sensor-1 protein (NCS-1), which increases PI4-K activity, did not affect responses to oxotremorine-M but reduced both fluorescence translocation and M-current inhibition by bradykinin. Using an intracellular IP3 membrane fluorescence-displacement assay, initial mean concentrations of membrane [PIP2] were estimated at 261 microm (95% confidence limit; 192-381 microm), rising to 693 microm (417-1153 microm) in neurons overexpressing PI5-K. Changes in membrane [PIP2] during application of oxotremorine-M were calculated from fluorescence data. The results, taken in conjunction with previous data for KCNQ2/3 (Kv7.2/Kv7.3) channel gating by PIP2 (Zhang et al., 2003), accorded with the hypothesis that the inhibitory action of oxotremorine-M on M current resulted from depletion of PIP2. The effects of bradykinin require additional components of action, which might involve IP3-induced Ca2+ release and consequent M-channel inhibition (as proposed previously) and stimulation of PIP2 synthesis by Ca2+-dependent activation of NCS-1.


Asunto(s)
Membrana Celular/fisiología , Inhibición Neural/fisiología , Neuronas/fisiología , Fosfatidilinositol 4,5-Difosfato/fisiología , Receptores Muscarínicos/metabolismo , Animales , Animales Recién Nacidos , Bradiquinina/farmacología , Calcio/metabolismo , Proteínas de Unión al Calcio/metabolismo , Membrana Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Estimulación Eléctrica/métodos , Estrenos/farmacología , Femenino , Colorantes Fluorescentes/metabolismo , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/genética , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Expresión Génica/fisiología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Inmunohistoquímica/métodos , Isoenzimas/genética , Isoenzimas/metabolismo , Masculino , Microscopía Confocal/métodos , Agonistas Muscarínicos/farmacología , Mutación/fisiología , Inhibición Neural/efectos de los fármacos , Proteínas Sensoras del Calcio Neuronal , Neuronas/citología , Neuronas/efectos de los fármacos , Neuropéptidos/metabolismo , Oxotremorina/análogos & derivados , Oxotremorina/farmacología , Técnicas de Placa-Clamp/métodos , Inhibidores de Fosfodiesterasa/farmacología , Fosfolipasa C delta , Pirrolidinonas/farmacología , Ratas , Ratas Sprague-Dawley , Receptores Muscarínicos/efectos de los fármacos , Receptores Muscarínicos/genética , Ganglio Cervical Superior/citología , Transfección/métodos , Fosfolipasas de Tipo C/genética , Fosfolipasas de Tipo C/metabolismo
8.
PLoS One ; 8(8): e71809, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23977150

RESUMEN

The KCNQ2 gene product, Kv7.2, is a subunit of the M-channel, a low-threshold voltage-gated K(+) channel that regulates mammalian and human neuronal excitability. Spontaneous mutations one of the KCNQ2 genes cause disorders of neural excitability such as Benign Familial Neonatal Seizures. However there appear to be no reports in which both human KCNQ2 genes are mutated. We therefore asked what happens to M-channel function when both KCNQ2 genes are disrupted. We addressed this using sympathetic neurons isolated from mice in which the KCNQ2 gene was truncated at a position corresponding to the second transmembrane domain of the Kv7.2 protein. Since homozygote KCNQ2-/- mice die postnatally, experiments were largely restricted to neurons from late embryos. Quantitative PCR revealed an absence of KCNQ2 mRNA in ganglia from KCNQ2-/- embryos but 100-120% increase of KCNQ3 and KCNQ5 mRNAs; KCNQ2+/- ganglia showed ∼30% less KCNQ2 mRNA than wild-type (+/+) ganglia but 40-50% more KCNQ3 and KCNQ5 mRNA. Neurons from KCNQ2-/- embryos showed a complete absence of M-current, even after applying the Kv7 channel enhancer, retigabine. Neurons from heterozygote KCNQ2+/- embryos had ∼60% reduced M-current. In contrast, M-currents in neurons from adult KCNQ2+/- mice were no smaller than those in neurons from wild-type mice. Measurements of tetraethylammonium block did not indicate an increased expression of Kv7.5-containing subunits, implying a compensatory increase in Kv7.2 expression from the remaining KCNQ2 gene. We conclude that mouse embryonic M-channels have an absolute requirement for Kv7.2 subunits for functionality, that the reduced M-channel activity in heterozygote KCNQ2+/- mouse embryos results primarily from a gene-dosage effect, and that there is a compensatory increase in Kv7.2 expression in adult mice.


Asunto(s)
Potenciales de Acción , Canal de Potasio KCNQ2/genética , Proteínas del Tejido Nervioso/genética , Neuronas/fisiología , Animales , Carbamatos/farmacología , Células Cultivadas , Femenino , Expresión Génica , Humanos , Canal de Potasio KCNQ2/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de los fármacos , Técnicas de Placa-Clamp , Fenilendiaminas/farmacología , Bloqueadores de los Canales de Potasio/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ganglio Cervical Superior/citología , Tetraetilamonio/farmacología
9.
J Neurochem ; 85(3): 610-21, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12694387

RESUMEN

A new class of Conus peptides, the I-superfamily of conotoxins, has been characterized using biochemical, electrophysiological and molecular genetic methods. Peptides in this superfamily have a novel pattern of eight Cys residues. Five peptides that elicited excitatory symptomatology, r11a, r11b, r11c, r11d and r11e, were purified from Conus radiatus venom; four were tested on amphibian peripheral axons and shown to elicit repetitive action potentials, consistent with being members of the 'lightning-strike cabal' of toxins that effect instant immobilization of fish prey. A parallel analysis of Conus cDNA clones revealed a new class of conotoxin genes that was particularly enriched (with 18 identified paralogues) in a Conus radiatus venom duct library; several C. radiatus clones encoded the excitatory peptides directly characterized from venom. The remarkable diversity of related I-superfamily peptides within a single Conus species is unprecedented. When combined with the excitatory effects observed on peripheral circuitry, this unexpected diversity suggests a corresponding molecular complexity of the targeted signaling components in peripheral axons; the I-conotoxin superfamily should provide a rich lode of pharmacological tools for dissecting and understanding these. Thus, the I-superfamily conotoxins promise to provide a significant new technology platform for dissecting the molecular components of axons.


Asunto(s)
Conotoxinas/genética , Conotoxinas/aislamiento & purificación , Familia de Multigenes , Péptidos/genética , Péptidos/aislamiento & purificación , Potenciales de Acción/efectos de los fármacos , Animales , Bioensayo , Conotoxinas/farmacología , Electrofisiología , Biblioteca de Genes , Carpa Dorada , Técnicas In Vitro , Ratones , Venenos de Moluscos/química , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/inervación , Músculo Esquelético/fisiología , Neurotoxinas/genética , Neurotoxinas/aislamiento & purificación , Neurotoxinas/farmacología , Péptidos/farmacología , Nervios Periféricos/efectos de los fármacos , Nervios Periféricos/fisiología , Rana pipiens , Análisis de Secuencia de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA