Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Cell Calcium ; 55(1): 38-47, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24342753

RESUMEN

The importance of the TRPV4 channel for human physiology has been highlighted in recent years with the identification of an increasing number of hereditary diseases associated with mutations of this channel. However, the functional understanding of TRPV4 associated pathologies remains a puzzle due to incomplete understanding of the polymodal regulation of TRPV4 channels and lack of insight into the structure-function relationship of the channel. In this work, we identified a series of highly conserved aromatic residues in transmembrane (TM) helices 5-6 with profound importance for TRPV4 activity. Substituting F617, Y621 or F624 in TM5 with leucine reduced channel sensitivity to the agonist 4α-PDD and heat, yet two of these mutants - F617L and Y621L - showed increased activation in response to cell swelling. In TM6, a Y702L mutation significantly reduced sensitivity to all of the above stimuli. In conclusion, we have identified residues in TM5-6 which differentially affect heat and agonist activation, and we have demonstrated distinct activation pathways for 4α-PDD and osmolarity.


Asunto(s)
Soluciones Hipotónicas/farmacología , Ésteres del Forbol/farmacología , Mutación Puntual/genética , Porinas/fisiología , Canales Catiónicos TRPV/efectos de los fármacos , Canales Catiónicos TRPV/genética , Secuencia de Aminoácidos , Calcio/fisiología , Carcinógenos/farmacología , Fenómenos Electrofisiológicos , Células HEK293 , Humanos , Datos de Secuencia Molecular , Concentración Osmolar , Estructura Secundaria de Proteína/efectos de los fármacos , Estructura Secundaria de Proteína/genética , Canales Catiónicos TRPV/química
2.
PLoS One ; 8(8): e73424, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23977387

RESUMEN

OBJECTIVE: Regulation of apical calcium entry is important for the function of principal cells of the collecting duct. However, the molecular identity and the regulators of the transporter/channel, which is responsible for apical calcium entry and what factors regulate the calcium conduction remain unclear. METHODS AND RESULTS: We report that endogenous TRPP2 and TRPV4 assemble to form a 23-pS divalent cation-permeable non-selective ion channel at the apical membrane of renal principal cells of the collecting duct. TRPP2\TRPV4 channel complex was identified by patch-clamp, immunofluorescence and co-immunprecipitation studies in both principal cells that either possess normal cilia (cilia (+)) or in which cilia are absent (cilia (-)). This channel has distinct biophysical and pharmacological and regulatory profiles compared to either TRPP2 or TRPV4 channels. The rate of occurrence detected by patch clamp was higher in cilia (-) compared to cilia (+) cells. In addition, shRNA knockdown of TRPP2 increased the prevalence of TRPV4 channel activity while knockdown of TRPV4 resulted in TRPP2 activity and knockdown of both proteins vastly decreased the 23-pS channel activity. Epidermal growth factor (EGF) stimulated TRPP2\TRPV4 channel through the EGF receptor (EGFR) tyrosine kinase-dependent signaling. With loss of cilia, apical EGF treatment resulted in 64-fold increase in channel activity in cilia (-) but not cilia (+) cells. In addition EGF increased cell proliferation in cilia (-) cell that was dependent upon TRPP2\TRPV4 channel mediated increase in intracellular calcium. CONCLUSION: We conclude that in the absence of cilia, an EGF activated TRPP2\TRPV4 channel may play an important role in increased cell proliferation and cystogenesis.


Asunto(s)
Agonistas de los Canales de Calcio/farmacología , Calcio/metabolismo , Permeabilidad de la Membrana Celular/efectos de los fármacos , Polaridad Celular/efectos de los fármacos , Factor de Crecimiento Epidérmico/farmacología , Túbulos Renales Colectores/metabolismo , Canales Catiónicos TRPP/metabolismo , Canales Catiónicos TRPV/metabolismo , Animales , Cationes Bivalentes/metabolismo , Proliferación Celular/efectos de los fármacos , Cilios/metabolismo , Receptores ErbB/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Técnica del Anticuerpo Fluorescente , Silenciador del Gen/efectos de los fármacos , Inmunoprecipitación , Activación del Canal Iónico/efectos de los fármacos , Túbulos Renales Colectores/citología , Túbulos Renales Colectores/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Fosforilación/efectos de los fármacos
3.
Mol Pharmacol ; 84(3): 325-34, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23757176

RESUMEN

Allyl isothiocyanate (AITC; aka, mustard oil) is a powerful irritant produced by Brassica plants as a defensive trait against herbivores and confers pungency to mustard and wasabi. AITC is widely used experimentally as an inducer of acute pain and neurogenic inflammation, which are largely mediated by the activation of nociceptive cation channels transient receptor potential ankyrin 1 and transient receptor potential vanilloid 1 (TRPV1). Although it is generally accepted that electrophilic agents activate these channels through covalent modification of cytosolic cysteine residues, the mechanism underlying TRPV1 activation by AITC remains unknown. Here we show that, surprisingly, AITC-induced activation of TRPV1 does not require interaction with cysteine residues, but is largely dependent on S513, a residue that is involved in capsaicin binding. Furthermore, AITC acts in a membrane-delimited manner and induces a shift of the voltage dependence of activation toward negative voltages, which is reminiscent of capsaicin effects. These data indicate that AITC acts through reversible interactions with the capsaicin binding site. In addition, we show that TRPV1 is a locus for cross-sensitization between AITC and acidosis in nociceptive neurons. Furthermore, we show that residue F660, which is known to determine the stimulation by low pH in human TRPV1, is also essential for the cross-sensitization of the effects of AITC and low pH. Taken together, these findings demonstrate that not all reactive electrophiles stimulate TRPV1 via cysteine modification and help understanding the molecular bases underlying the surprisingly large role of this channel as mediator of the algesic properties of AITC.


Asunto(s)
Isotiocianatos/farmacología , Canales Catiónicos TRPV/metabolismo , Animales , Sitios de Unión , Capsaicina/farmacología , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Células Cultivadas , Ganglios Espinales/citología , Humanos , Concentración de Iones de Hidrógeno , Ratones , Ratones Endogámicos C57BL , Mutación , Técnicas de Placa-Clamp , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/metabolismo , Canales Catiónicos TRPV/agonistas , Canales Catiónicos TRPV/genética
4.
Pflugers Arch ; 464(5): 425-58, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23001121

RESUMEN

The Transient Receptor Potential Ankyrin 1 channel (TRPA1), is a member of the large TRP family of ion channels, and functions as a Ca(2+) permeable non-selective cation channel in many different cell processes, ranging from sensory to homeostatic tasks. TRPA1 is highly conserved across the animal kingdom. The only mammalian TRPA subfamily member, TRPA1, is widely expressed in neuronal (e.g. sensory dorsal root and trigeminal ganglia neurons)- and in non-neuronal cells (e.g. epithelial cells, hair cells). It exhibits 14-19 amino-(N-)terminal ankyrin repeats, an unusual structural feature. The TRPA1 channel is activated by noxious cold (<17 °C) as well as by a plethora of chemical compounds that includes not only electrophilic compounds and oxidants that can modify, in an alkylative or oxidative fashion, nucleophilic cysteine residues in the channel's N-terminus, but also compounds that do not covalently bind to the channel proteins (e.g. menthol, nifedipin). Based on localization and functional properties, TRPA1 is considered a key player in acute and chronic (neuropathic) pain and inflammation. Moreover, its role in the (patho)physiology of nearly all organ systems is anticipated, and will be discussed along with the potential of TRPA1 as a drug target for the management of various pathological conditions.


Asunto(s)
Canales de Potencial de Receptor Transitorio/genética , Canales de Potencial de Receptor Transitorio/fisiología , Animales , Agonistas de los Canales de Calcio/metabolismo , Canalopatías/genética , Humanos , Inflamación/metabolismo , Activación del Canal Iónico , Nocicepción , Dolor/metabolismo , Filogenia , Estructura Terciaria de Proteína , Canal Catiónico TRPA1 , Canales de Potencial de Receptor Transitorio/química , Canales de Potencial de Receptor Transitorio/metabolismo
5.
Cell Calcium ; 49(6): 387-94, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21511334

RESUMEN

Gonadotropin-releasing hormone (GnRH)-secreting neurons are key regulators of the reproductive behaviour in vertebrates. These neurons show a peculiar migratory pattern during embryonic development, and its perturbations have profound impact on fertility and other related functional aspects. Changes in the intracellular calcium concentration, [Ca(2+)](i), induced by different extracellular signals, play a central role in the control of neuronal migration, but the available knowledge regarding GnRH neurons is still limited. Our goal was to investigate mechanisms that may be involved in the Ca(2+) dependence of the migratory behaviour in these neurons. We focused on the "classical" Transient Receptor Potential (TRPC) subfamily of Ca(2+)-permeable cation channels, recently shown to be involved in other aspects of neuronal development. Using GN11 cells, immortalized early stage GnRH neurons, we set to investigate Ca(2+) signals under basal conditions and in the presence of a well-established motogen, fetal calf serum (FCS), and the effect of pharmacological TRPC agonists and antagonists on Ca(2+) oscillations, cell motility and proliferation. We have found that a subpopulation of GN11 cells shows spontaneous Ca(2+) transients and that this activity is increased in the presence of serum. Quantitative real-time PCR showed that transcripts of some TRPC members are expressed in GN11 cells. Interestingly, pharmacological experiments with inhibitors, SKF-96365, lanthanum, anti-TRPC1 antibody, and activators, 1-oleil 2-acetyl-sn-glycerol, of TRPCs suggested that the activation of these channels can account for both the basal Ca(2+) oscillations and the increased activity in the presence of FCS. Moreover, functional studies using the same pharmacological tools supported their involvement in the control of motility and proliferation. Thus, our data provide evidence for the involvement of Ca(2+)-permeable channels of the TRPC subfamily in the control of functional properties of neurosecretory cells and neuronal motility.


Asunto(s)
Calcio/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Neuronas/citología , Canales Catiónicos TRPC/metabolismo , Animales , Anticuerpos/inmunología , Señalización del Calcio , Línea Celular , Movimiento Celular , Proliferación Celular , Diglicéridos/farmacología , Imidazoles/farmacología , Lantano/farmacología , Ratones , Canales Catiónicos TRPC/agonistas , Canales Catiónicos TRPC/antagonistas & inhibidores
6.
Genome Biol ; 12(3): 218, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21401968

RESUMEN

The transient receptor potential (TRP) multigene superfamily encodes integral membrane proteins that function as ion channels. Members of this family are conserved in yeast, invertebrates and vertebrates. The TRP family is subdivided into seven subfamilies: TRPC (canonical), TRPV (vanilloid), TRPM (melastatin), TRPP (polycystin), TRPML (mucolipin), TRPA (ankyrin) and TRPN (NOMPC-like); the latter is found only in invertebrates and fish. TRP ion channels are widely expressed in many different tissues and cell types, where they are involved in diverse physiological processes, such as sensation of different stimuli or ion homeostasis. Most TRPs are non-selective cation channels, only few are highly Ca2+ selective, some are even permeable for highly hydrated Mg2+ ions. This channel family shows a variety of gating mechanisms, with modes of activation ranging from ligand binding, voltage and changes in temperature to covalent modifications of nucleophilic residues. Activated TRP channels cause depolarization of the cellular membrane, which in turn activates voltage-dependent ion channels, resulting in a change of intracellular Ca2+ concentration; they serve as gatekeeper for transcellular transport of several cations (such as Ca2+ and Mg2+), and are required for the function of intracellular organelles (such as endosomes and lysosomes). Because of their function as intracellular Ca2+ release channels, they have an important regulatory role in cellular organelles. Mutations in several TRP genes have been implicated in diverse pathological states, including neurodegenerative disorders, skeletal dysplasia, kidney disorders and pain, and ongoing research may help find new therapies for treatments of related diseases.


Asunto(s)
Canales de Potencial de Receptor Transitorio/genética , Canales de Potencial de Receptor Transitorio/metabolismo , Animales , Evolución Molecular , Predisposición Genética a la Enfermedad , Humanos , Mutación , Filogenia , Conformación Proteica , Transporte de Proteínas , Canales de Potencial de Receptor Transitorio/química , Canales de Potencial de Receptor Transitorio/clasificación
7.
J Physiol ; 589(Pt 7): 1543-9, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21078588

RESUMEN

Transient receptor potential (TRP) channels have been extensively studied over the past years. Yet, in most cases, the gating mechanisms of these polymodal cation channels still remain a puzzle. Using the nociceptive channel TRPA1 as an example, we discuss the role of dynamic regulation of the pore size (pore dilatation) on channel gating. Additionally, we critically revise current knowledge of the role of intracellular domains, such as ankyrin repeats and EF hand motifs, in channel activation and function. Finally, we assess some problems inherent to activation of TRPA1 by the reaction of electrophilic compounds with the nucleophilic thiol sink of N-terminal reactive cysteines.


Asunto(s)
Canales de Calcio/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Canales de Potencial de Receptor Transitorio/metabolismo , Secuencia de Aminoácidos , Animales , Repetición de Anquirina , Calcio/metabolismo , Canales de Calcio/química , Canales de Calcio/genética , Cisteína/química , Humanos , Activación del Canal Iónico/fisiología , Ratones , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Permeabilidad , Estructura Terciaria de Proteína , Canal Catiónico TRPA1 , Canales de Potencial de Receptor Transitorio/química , Canales de Potencial de Receptor Transitorio/genética
8.
Proc Natl Acad Sci U S A ; 107(44): 19084-9, 2010 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-20956320

RESUMEN

Reduced functional bladder capacity and concomitant increased micturition frequency (pollakisuria) are common lower urinary tract symptoms associated with conditions such as cystitis, prostatic hyperplasia, neurological disease, and overactive bladder syndrome. These symptoms can profoundly affect the quality of life of afflicted individuals, but available pharmacological treatments are often unsatisfactory. Recent work has demonstrated that the cation channel TRPV4 is highly expressed in urothelial cells and plays a role in sensing the normal filling state of the bladder. In this article, we show that the development of cystitis-induced bladder dysfunction is strongly impaired in Trpv4(-/-) mice. Moreover, we describe HC-067047, a previously uncharacterized, potent, and selective TRPV4 antagonist that increases functional bladder capacity and reduces micturition frequency in WT mice and rats with cystitis. HC-067047 did not affect bladder function in Trpv4(-/-) mice, demonstrating that its in vivo effects are on target. These results indicate that TRPV4 antagonists may provide a promising means of treating bladder dysfunction.


Asunto(s)
Antineoplásicos Alquilantes/efectos adversos , Ciclofosfamida/efectos adversos , Cistitis , Moduladores del Transporte de Membrana/farmacología , Morfolinas/farmacología , Pirroles/farmacología , Canales Catiónicos TRPV/antagonistas & inhibidores , Vejiga Urinaria/fisiopatología , Urotelio/fisiopatología , Animales , Antineoplásicos Alquilantes/farmacología , Ciclofosfamida/farmacología , Cistitis/inducido químicamente , Cistitis/tratamiento farmacológico , Cistitis/metabolismo , Cistitis/fisiopatología , Humanos , Ratones , Ratones Noqueados , Ratas , Ratas Wistar , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/metabolismo , Vejiga Urinaria/metabolismo , Micción/efectos de los fármacos , Urotelio/metabolismo
9.
Cell Calcium ; 45(3): 226-32, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19046767

RESUMEN

We demonstrate here that the transient receptor potential melastatin subfamily channel, TRPM4, controls migration of bone marrow-derived mast cells (BMMCs), triggered by dinitrophenylated human serum albumin (DNP-HSA) or stem cell factor (SCF). Wild-type BMMCs migrate after stimulation with DNP-HSA or SCF whereas both stimuli do not induce migration in BMMCs derived from TRPM4 knockout mice (trpm4(-/-)). Mast cell migration is a Ca(2+)-dependent process, and TRPM4 likely controls this process by setting the intracellular Ca(2+) level upon cell stimulation. Cell migration depends on filamentous actin (F-actin) rearrangement, since pretreatment with cytochalasin B, an inhibitor of F-actin formation, prevented both DNP-HSA- and SCF-induced migration in wild-type BMMC. Immunocytochemical experiments using fluorescence-conjugated phalloidin demonstrate a reduced level of F-actin formation in DNP-HSA-stimulated BMMCs from trpm4(-/-) mice. Thus, our results suggest that TRPM4 is critically involved in migration of BMMCs by regulation of Ca(2+)-dependent actin cytoskeleton rearrangements.


Asunto(s)
Movimiento Celular , Mastocitos/citología , Mastocitos/metabolismo , Canales Catiónicos TRPM/metabolismo , Actinas/metabolismo , Animales , Antígenos/farmacología , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Calcio/metabolismo , Quimiotaxis/efectos de los fármacos , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Dinitrofenoles/metabolismo , Humanos , Inmunoglobulina E/inmunología , Mastocitos/efectos de los fármacos , Ratones , Albúmina Sérica/metabolismo , Factor de Células Madre/farmacología
10.
Nat Genet ; 40(8): 999-1003, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18587396

RESUMEN

The brachyolmias constitute a clinically and genetically heterogeneous group of skeletal dysplasias characterized by a short trunk, scoliosis and mild short stature. Here, we identify a locus for an autosomal dominant form of brachyolmia on chromosome 12q24.1-12q24.2. Among the genes in the genetic interval, we selected TRPV4, which encodes a calcium permeable cation channel of the transient receptor potential (TRP) vanilloid family, as a candidate gene because of its cartilage-selective gene expression pattern. In two families with the phenotype, we identified point mutations in TRPV4 that encoded R616Q and V620I substitutions, respectively. Patch clamp studies of transfected HEK cells showed that both mutations resulted in a dramatic gain of function characterized by increased constitutive activity and elevated channel activation by either mechano-stimulation or agonist stimulation by arachidonic acid or the TRPV4-specific agonist 4alpha-phorbol 12,13-didecanoate (4alphaPDD). This study thus defines a previously unknown mechanism, activation of a calcium-permeable TRP ion channel, in skeletal dysplasia pathogenesis.


Asunto(s)
Enfermedades del Desarrollo Óseo/genética , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/metabolismo , Enfermedades del Desarrollo Óseo/diagnóstico por imagen , Línea Celular , Femenino , Humanos , Masculino , Mutación Missense , Linaje , Radiografía
11.
Biochem Biophys Res Commun ; 372(1): 210-5, 2008 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-18485891

RESUMEN

This study explored the role of transient receptor potential melastatin 8 ion channels (TRPM8) in mechanisms of human glioblastoma (DBTRG) cell migration. Menthol stimulated influx of Ca(2+), membrane current, and migration of DBTRG cells. Effects on Ca(2+) and migration were enhanced by pre-treatment with hepatocyte growth factor/scatter factor (HGF/SF). Effects on Ca(2+) also were greater in migrating cells compared with non-migrating cells. 2-Aminoethoxydiphenyl borate (2-APB) inhibited all menthol stimulations. RT-PCR and immunoblot analysis showed that DBTRG cells expressed both mRNA and protein for TRPM8 ion channels. Two proteins were evident: one (130-140 kDa) in a plasma membrane-enriched fraction, and a variant (95-100 kDa) in microsome- and plasma membrane-enriched fractions. Thus, TRPM8 plays a role in mechanisms that increase [Ca(2+)](i) needed for DBTRG cell migration.


Asunto(s)
Calcio/metabolismo , Movimiento Celular , Neoplasias del Sistema Nervioso Central/patología , Glioblastoma/patología , Factor de Crecimiento de Hepatocito/metabolismo , Mentol/toxicidad , Canales Catiónicos TRPM/metabolismo , Compuestos de Boro/farmacología , Movimiento Celular/efectos de los fármacos , Neoplasias del Sistema Nervioso Central/metabolismo , Glioblastoma/metabolismo , Factor de Crecimiento de Hepatocito/farmacología , Humanos , Invasividad Neoplásica , ARN Mensajero/análisis , ARN Mensajero/metabolismo , Canales Catiónicos TRPM/agonistas , Canales Catiónicos TRPM/genética
12.
J Biol Chem ; 283(10): 6272-80, 2008 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-18174177

RESUMEN

TRPV4, a member of the vanilloid subfamily of the transient receptor potential (TRP) channels, is activated by a variety of stimuli, including cell swelling, moderate heat, and chemical compounds such as synthetic 4alpha-phorbol esters. TRPV4 displays a widespread expression in various cells and tissues and has been implicated in diverse physiological processes, including osmotic homeostasis, thermo- and mechanosensation, vasorelaxation, tuning of neuronal excitability, and bladder voiding. The mechanisms that regulate TRPV4 in these different physiological settings are currently poorly understood. We have recently shown that the relative amount of TRPV4 in the plasma membrane is enhanced by interaction with the SH3 domain of PACSIN 3, a member of the PACSIN family of proteins involved in synaptic vesicular membrane trafficking and endocytosis. Here we demonstrate that PACSIN 3 strongly inhibits the basal activity of TRPV4 and its activation by cell swelling and heat, while leaving channel gating induced by the synthetic ligand 4alpha-phorbol 12,13-didecanoate unaffected. A single proline mutation in the SH3 domain of PACSIN 3 abolishes its inhibitory effect on TRPV4, indicating that PACSIN 3 must bind to the channel to modulate its function. In line herewith, mutations at specific proline residues in the N terminus of TRPV4 abolish binding of PACSIN 3 and render the channel insensitive to PACSIN 3-induced inhibition. Taken together, these data suggest that PACSIN 3 acts as an auxiliary protein of TRPV4 channel that not only affects the channel's subcellular localization but also modulates its function in a stimulus-specific manner.


Asunto(s)
Membrana Celular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Fosfoproteínas/metabolismo , Canales Catiónicos TRPV/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Carcinógenos/farmacología , Línea Celular , Membrana Celular/genética , Proteínas del Citoesqueleto , Homeostasis/efectos de los fármacos , Homeostasis/fisiología , Calor , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Mecanotransducción Celular/efectos de los fármacos , Mecanotransducción Celular/fisiología , Ratones , Neuronas/metabolismo , Especificidad de Órganos/fisiología , Ésteres del Forbol/farmacología , Fosfoproteínas/genética , Unión Proteica/fisiología , Estructura Terciaria de Proteína/fisiología , Canales Catiónicos TRPV/genética
13.
J Clin Invest ; 117(11): 3453-62, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17948126

RESUMEN

Here we provide evidence for a critical role of the transient receptor potential cation channel, subfamily V, member 4 (TRPV4) in normal bladder function. Immunofluorescence demonstrated TRPV4 expression in mouse and rat urothelium and vascular endothelium, but not in other cell types of the bladder. Intracellular Ca2+ measurements on urothelial cells isolated from mice revealed a TRPV4-dependent response to the selective TRPV4 agonist 4alpha-phorbol 12,13-didecanoate and to hypotonic cell swelling. Behavioral studies demonstrated that TRPV4-/- mice manifest an incontinent phenotype but show normal exploratory activity and anxiety-related behavior. Cystometric experiments revealed that TRPV4-/- mice exhibit a lower frequency of voiding contractions as well as a higher frequency of nonvoiding contractions. Additionally, the amplitude of the spontaneous contractions in explanted bladder strips from TRPV4-/- mice was significantly reduced. Finally, a decreased intravesical stretch-evoked ATP release was found in isolated whole bladders from TRPV4-/- mice. These data demonstrate a previously unrecognized role for TRPV4 in voiding behavior, raising the possibility that TRPV4 plays a critical role in urothelium-mediated transduction of intravesical mechanical pressure.


Asunto(s)
Canales Catiónicos TRPV/metabolismo , Vejiga Urinaria/metabolismo , Micción/fisiología , Adenosina Trifosfato/metabolismo , Animales , Conducta Animal/fisiología , Eliminación de Gen , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/fisiología , Ratas , Canales Catiónicos TRPV/genética , Vejiga Urinaria/anatomía & histología , Urodinámica , Urotelio/citología , Urotelio/metabolismo
14.
J Biol Chem ; 282(17): 12796-803, 2007 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-17341586

RESUMEN

TRPV4, a Ca(2+)-permeable member of the vanilloid subgroup of the transient receptor potential (TRP) channels, is activated by cell swelling and moderate heat (>27 degrees C) as well as by diverse chemical compounds including synthetic 4 alpha-phorbol esters, the plant extract bisandrographolide A, and endogenous epoxyeicosatrienoic acids (EETs; 5,6-EET and 8,9-EET). Previous work identified a tyrosine residue located in the first half of putative transmembrane segment 3 (TM3) as a crucial determinant for the activation of TRPV4 by its most specific agonist 4 alpha-phorbol 12,13-didecanoate (4 alpha-PDD), suggesting that 4 alpha-PDD interacts with the channel through its transmembrane segments. To obtain insight in the 4 alpha-PDD-binding site and in the mechanism of ligand-dependent TRPV4 activation, we investigated the consequences of specific point mutations in TM4 on the sensitivity of the channel to different chemical and physical stimuli. Mutations of two hydrophobic residues in the central part of TM4 (Leu(584) and Trp(586)) caused a severe reduction of the sensitivity of the channel to 4 alpha-PDD, bisandrographolide A, and heat, whereas responses to cell swelling, arachidonic acid, and 5,6-EET remained unaffected. In contrast, mutations of two residues in the C-terminal part of TM4 (Tyr(591) and Arg(594)) affected channel activation of TRPV4 by all stimuli, suggesting an involvement in channel gating rather than in interaction with agonists. Based on a comparison of the responses of WT and mutant TRPV4 to 4 alpha-PDD and different 4 alpha-phorbol esters, we conclude that the length of the fatty acid moiety determines the ligand binding affinity and propose a model for the interaction between 4 alpha-phorbol esters and the TM3/4 region of TRPV4.


Asunto(s)
Carcinógenos/farmacología , Activación del Canal Iónico/efectos de los fármacos , Modelos Moleculares , Ésteres del Forbol/farmacología , Canales Catiónicos TRPV/agonistas , Vasodilatadores/farmacología , Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Ácido 8,11,14-Eicosatrienoico/farmacología , Sustitución de Aminoácidos , Animales , Línea Celular , Humanos , Activación del Canal Iónico/genética , Ratones , Mutación Puntual , Unión Proteica , Estructura Terciaria de Proteína/genética , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/metabolismo
15.
J Biol Chem ; 282(5): 3325-36, 2007 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-17142461

RESUMEN

Menthol, a secondary alcohol produced by the peppermint herb, Mentha piperita, is widely used in the food and pharmaceutical industries as a cooling/soothing compound and odorant. It induces Ca2+ influx in a subset of sensory neurons from dorsal root and trigeminal ganglia, due to activation of TRPM8, a Ca2+-permeable, cold-activated member of the TRP superfamily of cation channels. Menthol also induces Ca2+ release from intracellular stores in several TRPM8-expressing cell types, which has led to the suggestion that TRPM8 can function as an intracellular Ca2+-release channel. Here we show that menthol induces Ca2+ release from intracellular stores in four widely used cell lines (HEK293, lymph node carcinoma of the prostate (LNCaP), Chinese hamster ovary (CHO), and COS), and provide several lines of evidence indicating that this release pathway is TRPM8-independent: 1) menthol-induced Ca2+ release was potentiated at higher temperatures, which contrasts to the cold activation of TRPM8; 2) overexpression of TRPM8 did not enhance the menthol-induced Ca2+) release; 3) menthol-induced Ca2+ release was mimicked by geraniol and linalool, which are structurally related to menthol, but not by the more potent TRPM8 agonists icilin or eucalyptol; and 4) TRPM8 expression in HEK293 cells was undetectable at the protein and mRNA levels. Moreover, using a novel TRPM8-specific antibody we demonstrate that both heterologously expressed TRPM8 (in HEK293 cells) and endogenous TRPM8 (in LNCaP cells) are mainly localized in the plasma membrane, which contrast to previous localization studies using commercial anti-TRPM8 antibodies. Finally, aequorin-based measurements demonstrate that the TRPM8-independent menthol-induced Ca2+ release originates from both endoplasmic reticulum and Golgi compartments.


Asunto(s)
Calcio/fisiología , Retículo Endoplásmico/fisiología , Aparato de Golgi/fisiología , Mentol/farmacología , Canales Catiónicos TRPM/fisiología , Animales , Células COS , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Línea Celular , Chlorocebus aethiops , Retículo Endoplásmico/efectos de los fármacos , Aparato de Golgi/efectos de los fármacos , Humanos , Riñón , ARN/genética , ARN/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Canales Catiónicos TRPM/genética , Transfección
16.
FEBS Lett ; 579(16): 3392-6, 2005 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-15936019

RESUMEN

A direct interaction of the regulatory domain (R domain) of the cystic fibrosis transmembrane conductance regulator protein (CFTR) with PR65, a regulatory subunit of the protein phosphatase 2A (PP2A), was shown in yeast two hybrid, pull-down and co-immunoprecipitation experiments. The R domain could be dephosphorylated by PP2A in vitro. Overexpression of the interacting domain of PR65 in Caco-2 cells, as well as treatment with okadaic acid, showed a prolonged deactivation of the chloride channel. Taken together our results show a direct and functional interaction between CFTR and PP2A.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Secuencia de Aminoácidos , Animales , Células CACO-2 , Humanos , Inmunoprecipitación , Datos de Secuencia Molecular , Proteína Fosfatasa 2 , Estructura Terciaria de Proteína , Subunidades de Proteína/metabolismo , Técnicas del Sistema de Dos Híbridos
17.
Biochem Biophys Res Commun ; 331(2): 503-11, 2005 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-15850788

RESUMEN

Using the whole-cell patch-clamp technique, we identified an amiloride (AMI)-sensitive Na(+) current in cystic fibrosis cells, JME/CF15, growing in standard medium. The reversal potential of this current depended on Na(+) concentrations and the cation selectivity was much higher for Na(+) than for K(+), indicating that the current is through ENaC channels. In contrast, cells from EGF-containing medium lacked AMI-sensitive Na(+) currents. In permeabilized cells growing in EGF-containing medium, alphaENaC was mainly detected in a perinuclear region, while in cells from standard medium it was distributed over the cell body. Western-blot analysis showed that in standard medium cells expressed fast-migrating EndoH-insensitive and slow-migrating EndoH-sensitive alphaENaC fractions, while in cells growing in the presence of EGF, alphaENaC was only detected as the fast-migrating EndoH-insensitive fraction. Long-term incubation of cells with EGF resulted in an increased basal Ca(2+) level, [Ca(2+)](i). A similar increase of [Ca(2+)](i) was also observed in the presence of 2muM thapsigargin, resulting in inhibition of ENaC function. Thus, in JME/CF15 cells inhibition of the ENaC function by chronic incubation with EGF is a Ca(2+)-mediated process that affects trafficking and surface expression of ENaC channels.


Asunto(s)
Fibrosis Quística/metabolismo , Fibrosis Quística/patología , Factor de Crecimiento Epidérmico/farmacología , Canales de Sodio/metabolismo , Calcio/metabolismo , Células Cultivadas , Conductividad Eléctrica , Canales Epiteliales de Sodio , Glicosilación , Humanos , Activación del Canal Iónico/efectos de los fármacos , Transporte Iónico/efectos de los fármacos , Técnicas de Placa-Clamp , Transporte de Proteínas/efectos de los fármacos , Sodio/metabolismo , Canales de Sodio/química , Tapsigargina/farmacología , Factores de Tiempo
18.
J Biol Chem ; 280(8): 6423-33, 2005 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-15590641

RESUMEN

TRPM4, a Ca(2+)-activated cation channel of the transient receptor potential superfamily, undergoes a fast desensitization to Ca(2+). The mechanisms underlying the alterations in Ca(2+) sensitivity are unknown. Here we show that cytoplasmic ATP reversed Ca(2+) sensitivity after desensitization, whereas mutations to putative ATP binding sites resulted in faster and more complete desensitization. Phorbol ester-induced activation of protein kinase C (PKC) increased the Ca(2+) sensitivity of wild-type TRPM4 but not of two mutants mutated at putative PKC phosphorylation sites. Overexpression of a calmodulin mutant unable to bind Ca(2+) dramatically reduced TRPM4 activation. We identified five Ca(2+)-calmodulin binding sites in TRPM4 and showed that deletion of any of the three C-terminal sites strongly impaired current activation by reducing Ca(2+) sensitivity and shifting the voltage dependence of activation to very positive potentials. Thus, the Ca(2+) sensitivity of TRPM4 is regulated by ATP, PKC-dependent phosphorylation, and calmodulin binding at the C terminus.


Asunto(s)
Canales de Calcio/metabolismo , Calcio/farmacología , Proteínas de Transporte de Catión/metabolismo , Adenosina Trifosfato/farmacología , Sitios de Unión , Canales de Calcio/genética , Canales de Calcio/fisiología , Calmodulina/metabolismo , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/fisiología , Línea Celular , Electrofisiología , Humanos , Mutación , Fosforilación , Proteína Quinasa C/metabolismo , Canales Catiónicos TRPM
19.
Mol Biol Cell ; 15(5): 2049-60, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-14978214

RESUMEN

All organisms are equipped with systems for detoxification of the metalloids arsenic and antimony. Here, we show that two parallel pathways involving the AP-1-like proteins Yap1p and Yap8p are required for acquisition of metalloid tolerance in the budding yeast S. cerevisiae. Yap8p is demonstrated to reside in the nucleus where it mediates enhanced expression of the arsenic detoxification genes ACR2 and ACR3. Using chromatin immunoprecipitation assays, we show that Yap8p is associated with the ACR3 promoter in untreated as well as arsenic-exposed cells. Like for Yap1p, specific cysteine residues are critical for Yap8p function. We further show that metalloid exposure triggers nuclear accumulation of Yap1p and stimulates expression of antioxidant genes. Yap1p mutants that are unable to accumulate in the nucleus during H(2)O(2) treatment showed nearly normal nuclear retention in response to metalloid exposure. Thus, our data are the first to demonstrate that Yap1p is being regulated by metalloid stress and to indicate that this activation of Yap1p operates in a manner distinct from stress caused by chemical oxidants. We conclude that Yap1p and Yap8p mediate tolerance by controlling separate subsets of detoxification genes and propose that the two AP-1-like proteins respond to metalloids through distinct mechanisms.


Asunto(s)
Antimonio/farmacología , Arsénico/farmacología , Proteínas de Saccharomyces cerevisiae/fisiología , Saccharomyces cerevisiae/genética , Transactivadores/fisiología , Factores de Transcripción/fisiología , Activación Transcripcional , Arseniato Reductasas , ATPasas Transportadoras de Arsenitos , Secuencia de Bases , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico , Sitios de Unión , Núcleo Celular/ultraestructura , Cisteína/genética , Cisteína/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Farmacorresistencia Fúngica , Regulación Fúngica de la Expresión Génica , Peróxido de Hidrógeno/farmacología , Bombas Iónicas/genética , Bombas Iónicas/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas de Transporte de Membrana , Modelos Genéticos , Datos de Secuencia Molecular , Complejos Multienzimáticos/genética , Complejos Multienzimáticos/metabolismo , Estrés Oxidativo/efectos de los fármacos , Elementos de Respuesta/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Reductasa de Tiorredoxina-Disulfuro/genética , Reductasa de Tiorredoxina-Disulfuro/metabolismo , Tiorredoxinas/genética , Tiorredoxinas/metabolismo , Transactivadores/genética , Transactivadores/metabolismo , Factor de Transcripción AP-1/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
20.
FEBS Lett ; 554(1-2): 173-8, 2003 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-14596935

RESUMEN

The most frequent mutant variant of the cystic fibrosis transmembrane conductance regulator (CFTR), DeltaF508-CFTR, is misprocessed and subsequently degraded in the endoplasmic reticulum. Using the patch-clamp technique, we showed that co-expressions of DeltaF508-CFTR with the N-terminal CFTR truncates containing bi-arginine (RXR) retention/retrieval motifs result in a functional rescue of the DeltaF508-CFTR mutant channel in COS-1 cells. This DeltaF508-CFTR rescue process was strongly impaired when truncated CFTR constructs possessed either the DeltaF508 mutation or arginine-to-lysine mutations in RXRs. In conclusions, our data demonstrated that expression of truncated CFTR constructs could be a novel promising approach to improve maturation of DeltaF508-CFTR channels.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Terapia Genética/métodos , Animales , Células COS , Cloruros/metabolismo , AMP Cíclico/farmacología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/fisiología , Electrofisiología , Retículo Endoplásmico/metabolismo , Vectores Genéticos , Mutación , Técnicas de Placa-Clamp , Ingeniería de Proteínas , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA