Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Curr Opin Nephrol Hypertens ; 33(2): 161-169, 2024 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-38193301

RESUMEN

PURPOSE OF REVIEW: The transmembrane protein 16A (TMEM16A) Ca 2+ -activated Cl - channel constitutes a key depolarising mechanism in vascular smooth muscle and contractile pericytes, while in endothelial cells the channel is implicated in angiogenesis and in the response to vasoactive stimuli. Here, we offer a critical analysis of recent physiological investigations and consider the potential for targeting TMEM16A channels in vascular disease. RECENT FINDINGS: Genetic deletion or pharmacological inhibition of TMEM16A channels in vascular smooth muscle decreases artery tone and lowers systemic blood pressure in rodent models. Inhibition of TMEM16A channels in cerebral cortical pericytes protects against ischemia-induced tissue damage and improves microvascular blood flow in rodent stroke models. In endothelial cells, the TMEM16A channel plays varied roles including modulation of cell division and control of vessel tone through spread of hyperpolarisation to the smooth muscle cells. Genetic studies implicate TMEM16A channels in human disease including systemic and pulmonary hypertension, stroke and Moyamoya disease. SUMMARY: The TMEM16A channel regulates vascular function by controlling artery tone and capillary diameter as well as vessel formation and histology. Preclinical and clinical investigations are highlighting the potential for therapeutic exploitation of the channel in a range of maladaptive states of the (micro)circulation.


Asunto(s)
Hipertensión Pulmonar , Accidente Cerebrovascular , Humanos , Canales de Cloruro/genética , Canales de Cloruro/metabolismo , Células Endoteliales/metabolismo , Miocitos del Músculo Liso/fisiología
2.
Proc Natl Acad Sci U S A ; 118(34)2021 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-34413188

RESUMEN

TMEM16A Ca2+-activated chloride channels are involved in multiple cellular functions and are proposed targets for diseases such as hypertension, stroke, and cystic fibrosis. This therapeutic endeavor, however, suffers from paucity of selective and potent modulators. Here, exploiting a synthetic small molecule with a biphasic effect on the TMEM16A channel, anthracene-9-carboxylic acid (A9C), we shed light on sites of the channel amenable for pharmacological intervention. Mutant channels with the intracellular gate constitutively open were generated. These channels were entirely insensitive to extracellular A9C when intracellular Ca2+ was omitted. However, when physiological Ca2+ levels were reestablished, the mutants regained sensitivity to A9C. Thus, intracellular Ca2+ is mandatory for the channel response to an extracellular modulator. The underlying mechanism is a conformational change in the outer pore that enables A9C to enter the pore to reach its binding site. The explanation of this structural rearrangement highlights a critical site for pharmacological intervention and reveals an aspect of Ca2+ gating in the TMEM16A channel.


Asunto(s)
Anoctamina-1/metabolismo , Antracenos/farmacología , Calcio/farmacología , Cloruros/farmacología , Animales , Anoctamina-1/genética , Estimulación Eléctrica , Fenómenos Electrofisiológicos , Regulación de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Ratones , Simulación de Dinámica Molecular , Farmacología en Red , Técnicas de Placa-Clamp , Mutación Puntual
3.
Int J Mol Sci ; 23(3)2022 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-35163502

RESUMEN

The TMEM16A/anoctamin-1 calcium-activated chloride channel (CaCC) contributes to a range of vital functions, such as the control of vascular tone and epithelial ion transport. The channel is a founding member of a family of 10 proteins (TMEM16x) with varied functions; some members (i.e., TMEM16A and TMEM16B) serve as CaCCs, while others are lipid scramblases, combine channel and scramblase function, or perform additional cellular roles. TMEM16x proteins are typically activated by agonist-induced Ca2+ release evoked by Gq-protein-coupled receptor (GqPCR) activation; thus, TMEM16x proteins link Ca2+-signalling with cell electrical activity and/or lipid transport. Recent studies demonstrate that a range of other cellular factors-including plasmalemmal lipids, pH, hypoxia, ATP and auxiliary proteins-also control the activity of the TMEM16A channel and its paralogues, suggesting that the TMEM16x proteins are effectively polymodal sensors of cellular homeostasis. Here, we review the molecular pathophysiology, structural biology, and mechanisms of regulation of TMEM16x proteins by multiple cellular factors.


Asunto(s)
Anoctamina-1/metabolismo , Anoctaminas/metabolismo , Canales de Cloruro/metabolismo , Animales , Anoctaminas/fisiología , Transporte Biológico/fisiología , Membrana Celular/metabolismo , Humanos , Transporte Iónico/fisiología , Proteínas de Transferencia de Fosfolípidos/metabolismo
4.
Monaldi Arch Chest Dis ; 93(4)2022 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-36412132

RESUMEN

Long term dual antiplatelet therapy (LTDAPT), with ticagrelor 60 mg and low-dose aspirin, is indicated after acute coronary syndrome (ACS) for the secondary prevention of atherothrombotic events in high-risk patients with a history of ACS of at least 1 year. LTDAPT had a good tolerability and safety profile, but the risk of TIMI major bleeding was increased. However, even non-significant bleeding may be important because it has an effect on the quality of life and therefore may lead to treatment discontinuation. We, therefore, evaluated patients' experiences with LTDAPT and the impact of nuisance bleeding on quality of life and treatment adherence. We retrospectively reviewed 225 patients in follow-up after ACS with at least one high-risk condition, treated with ticagrelor 60 mg twice daily (after 90 mg twice daily for 12 months). The outpatient follow-up program after hospitalization provides a visit on day 30 after discharge, then after 3 months, continuing with six-monthly checks. We assessed the presence and intensity of bleeding, as well as health-related quality of life (HRQoL), at each visit. The TIMI score was used to determine the severity of the bleeding. Any overt bleeding event that did not meet the major and minor criteria was labeled "minimal" and could be framed as "nuisance bleeding." The HRQoL was assessed by the EuroQol-5 and Dimension (EQ-5D) visual analog scale (VAS) score. Minimal bleedings were present in 49 patients (21%), but only in one case (by decision of the patient) there was a cause for discontinuation of therapy. However, 39 (79%) subjects had asked for opinions on stopping the therapy during the telephone consultation. Factors influencing LTDAPT knowledge included access to medication counselling, engaging with information communicated during medication counselling, and access to timely, relevant and expert information and advice after discharge from the hospital. All adverse events, judged to be "not serious" in trials, may have an effect on the quality of life and therefore may lead to treatment discontinuation. The authors underline the importance of careful outpatient follow-up and ongoing counselling, to check out compliance and possible adverse effect of LTDAPT.


Asunto(s)
Síndrome Coronario Agudo , Intervención Coronaria Percutánea , Humanos , Inhibidores de Agregación Plaquetaria/efectos adversos , Ticagrelor/efectos adversos , Calidad de Vida , Derivación y Consulta , Estudios Retrospectivos , Teléfono , Hemorragia/inducido químicamente , Hemorragia/epidemiología , Hemorragia/tratamiento farmacológico , Síndrome Coronario Agudo/tratamiento farmacológico , Síndrome Coronario Agudo/etiología , Resultado del Tratamiento
5.
Biochem Soc Trans ; 49(4): 1855-1865, 2021 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-34346486

RESUMEN

Pulmonary arterial hypertension (PAH) is a fatal disease of the cardiopulmonary system that lacks curative treatments. The main pathological event in PAH is elevated vascular resistance in the pulmonary circulation, caused by abnormal vasoconstriction and vascular remodelling. Ion channels are key determinants of vascular smooth muscle tone and homeostasis, and four PAH channelopathies (KCNK3, ABCC8, KCNA5, TRPC6) have been identified so far. However, the contribution of ion channels in other forms of PAH, which account for the majority of PAH patients, has been less well characterised. Here we reason that a variety of triggers of PAH (e.g. BMPR2 mutations, hypoxia, anorectic drugs) that impact channel function may contribute to the onset of the disease. We review the molecular mechanisms by which these 'extrinsic' factors converge on ion channels and provoke their dysregulation to promote the development of PAH. Ion channels of the pulmonary vasculature are therefore promising therapeutic targets because of the modulation they provide to both vasomotor tone and proliferation of arterial smooth muscle cells.


Asunto(s)
Canales Iónicos/metabolismo , Hipertensión Arterial Pulmonar/metabolismo , Animales , Homeostasis , Humanos , Tono Muscular , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Hipertensión Arterial Pulmonar/patología
6.
Monaldi Arch Chest Dis ; 92(1)2021 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-34526727

RESUMEN

Long term treatment with ticagrelor 60 mg and low-dose aspirin are indicated after acute coronary syndrome (ACS). We retrospectively reviewed aggregate data of 187 patients (155 M and 38 F) (mean age 63.8±9 years) in follow up after ACS with at least one high risk condition (Multivessel disease, diabetes, GFR<60 mL/min, history of prior myocardial infarction, age >65 years) treated with ticagrelor 60 mg twice daily (after 90 mg twice daily for 12 months). The results were compared with findings (characteristics of the patients at baseline, outcomes, bleeding) of PEGASUS-TIMI 54 trial and Eu Label. The highrisk groups were represented as follows: multivessel disease 105 pts (82%), diabetes 63 pts (33%), GFR< 60 mL/min 27 pts (14%), history of prior MI 33 pts (17%), >65 year aged 85 pts (45%). Treatment was withdrawn in 7 patients: 3 cases showed atrial fibrillation and were placed on oral anticoagulant drugs, one developed intracranial bleeding, in three patients a temporary withdrawal was due to surgery (1 colon polyposis and 2 cases of bladder papilloma). Chest pain without myocardial infarction occurred in 16 patients (revascularization was required in 9 patients). Dyspnea was present in 15 patients, but was not a cause for discontinuation of therapy. Long term treatment with ticagrelor 60 mg twice daily plus aspirin 100 mg/day showed a favourable benefit/risk profile after ACS.  In this study all patients had been given ticagrelor 90 mg twice daily for 12 months and the 60 mg twice daily dosage was started immediately thereafter, unlike PEGASUS-TIMI 54 trial in which it was prescribed within a period ranging from 1 day to 1 year after discontinuation of the 90 mg dose. This makes our results more consistent with current clinical practice. However, a careful outpatient follow-up and constant counseling are mandatory to check out compliance to therapy and adverse side effects.


Asunto(s)
Síndrome Coronario Agudo , Infarto del Miocardio , Inhibidores de Agregación Plaquetaria , Síndrome Coronario Agudo/tratamiento farmacológico , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Masculino , Persona de Mediana Edad , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/epidemiología , Pacientes Ambulatorios , Inhibidores de Agregación Plaquetaria/efectos adversos , Estudios Retrospectivos , Resultado del Tratamiento
7.
Exp Eye Res ; 121: 74-85, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24560797

RESUMEN

BEST1 encodes Bestrophin-1 (Best1), a homo-oligomeric, integral membrane protein localized to the basolateral plasma membrane of the retinal pigment epithelium. Mutations in BEST1 cause five distinct retinal degenerative diseases, including adult vitelliform macular dystrophy (AVMD), autosomal recessive bestrophinopathy (ARB), autosomal dominant vitreoretinochoroidopathy (ADVIRC), and retinitis pigmentosa (RP). The mechanisms underlying these diseases and why mutations cause one disease over another are, for the most part, unknown. To gain insights into these four diseases, we expressed 28 Best1 mutants fused to YFP in polarized MDCK monolayers and, via confocal microscopy and immunofluorescence, live-cell FRET, and reciprocal co-immunoprecipitation experiments, screened these mutants for defects in localization and oligomerization. All 28 mutants exhibited comparable FRET efficiencies to and co-immunoprecipitated with WT Best1, indicating unimpaired oligomerization. RP- and ADVIRC-associated mutants were properly localized to the basolateral plasma membrane of cells, while two AVMD and most ARB mutants were mislocalized. When co-expressed, all mislocalized mutants caused mislocalization of WT Best1 to intracellular compartments. Our current and past results indicate that mislocalization of Best1 is not an absolute feature of any individual bestrophinopathy, occurring in AVMD, BVMD, and ARB. Furthermore, some ARB mutants that do not also cause dominant disease cause mislocalization of Best1, indicating that mislocalization is not a cause of disease, and that absence of Best1 activity from the plasma membrane is tolerated. Lastly, we find that the ARB truncation mutants L174Qfs*57 and R200X can form oligomers with WT Best1, indicating that the first ∼174 amino acids of Best1 are sufficient for oligomerization to occur.


Asunto(s)
Canales de Cloruro/genética , Canales de Cloruro/metabolismo , Enfermedades Hereditarias del Ojo/genética , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Mutación Missense , Multimerización de Proteína/fisiología , Enfermedades de la Retina/genética , Adenoviridae/genética , Animales , Proteínas Bacterianas/metabolismo , Bestrofinas , Western Blotting , Enfermedades de la Coroides/genética , Enfermedades de la Coroides/metabolismo , Perros , Electrofisiología , Enfermedades Hereditarias del Ojo/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Colorantes Fluorescentes/metabolismo , Expresión Génica , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Proteínas Luminiscentes/metabolismo , Células de Riñón Canino Madin Darby/metabolismo , Microscopía Confocal , Técnicas de Placa-Clamp , Degeneración Retiniana/genética , Degeneración Retiniana/metabolismo , Enfermedades de la Retina/metabolismo , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/metabolismo , Transfección , Distrofia Macular Viteliforme/genética , Distrofia Macular Viteliforme/metabolismo
8.
Biophys Chem ; 308: 107194, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38401241

RESUMEN

The TMEM16/Anoctamin protein family (TMEM16x) is composed of members with different functions; some members form Ca2+-activated chloride channels, while others are lipid scramblases or combine the two functions. TMEM16x proteins are typically activated in response to agonist-induced rises of intracellular Ca2+; thus, they couple Ca2+-signalling with cell electrical activity or plasmalemmal lipid homeostasis. The structural domains underlying these functions are not fully defined. We used a Naïve Bayes classifier to gain insights into these domains. The method enabled identification of regions involved in either ion or lipid transport, and suggested domains for possible pharmacological exploitation. The method allowed the prediction of the transport property of any given TMEM16x. We envisage this strategy could be exploited to illuminate the structure-function relationship of any protein family composed of members playing different molecular roles.


Asunto(s)
Anoctaminas , Lípidos , Anoctaminas/metabolismo , Teorema de Bayes , Anoctamina-1/metabolismo , Transporte Iónico , Calcio/metabolismo
9.
G Ital Cardiol (Rome) ; 25(5): 309-317, 2024 May.
Artículo en Italiano | MEDLINE | ID: mdl-38639121

RESUMEN

Ischemia with non-obstructive coronary arteries (INOCA) is defined by the coexistence of anginal symptoms and demonstrable ischemia, with no evidence of obstructive coronary arteries. The underlying mechanism of INOCA is coronary microvascular dysfunction with or without associated vasospasm. INOCA patients have recurrent symptoms, functional limitations, repeated access to the emergency department, impaired quality of life and a higher incidence of cardiovascular events than the general population. Although well described in chronic coronary syndrome guidelines, INOCA remains underdiagnosed in clinical practice because of insufficient awareness, lack of accurate diagnostic tools, and poorly standardized and consistent definitions to diagnose, both invasively and non-invasively, coronary microvascular dysfunction.To disseminate current scientific evidence on INOCA as a distinct clinical entity, during 2022 we conducted at 30 cardiology units all over the country a clinical practice improvement initiative, with the aim of developing uniform and shared management pathways for INOCA patients across different operational settings. The present document highlights the outcomes of this multidisciplinary initiative.


Asunto(s)
Enfermedad de la Arteria Coronaria , Isquemia Miocárdica , Humanos , Vasos Coronarios , Calidad de Vida , Isquemia , Isquemia Miocárdica/terapia , Corazón
10.
J Physiol ; 591(14): 3487-505, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23613533

RESUMEN

The recently identified TMEM16/anoctamin protein family includes Ca(2+)-activated anion channels (TMEM16A, TMEM16B), a cation channel (TMEM16F) and proteins with unclear function. TMEM16 channels consist of eight putative transmembrane domains (TMs) with TM5-TM6 flanking a re-entrant loop thought to form the pore. In TMEM16A this region has also been suggested to contain residues involved in Ca(2+) binding. The role of the putative pore-loop of TMEM16 channels was investigated using a chimeric approach. Heterologous expression of either TMEM16A or TMEM16B resulted in whole-cell anion currents with very similar conduction properties but distinct kinetics and degrees of sensitivity to Ca(2+). Furthermore, whole-cell currents mediated by TMEM16A channels were ∼six times larger than TMEM16B-mediated currents. Replacement of the putative pore-loop of TMEM16A with that of TMEM16B (TMEM16A-B channels) reduced the currents by ∼six-fold, while the opposite modification (TMEM16B-A channels) produced a ∼six-fold increase in the currents. Unexpectedly, these changes were not secondary to variations in channel gating by Ca(2+) or voltage, nor were they due to changes in single-channel conductance. Instead, they depended on the number of functional channels present on the plasma membrane. Generation of additional, smaller chimeras within the putative pore-loop of TMEM16A and TMEM16B led to the identification of a region containing a non-canonical trafficking motif. Chimeras composed of the putative pore-loop of TMEM16F transplanted into the TMEM16A protein scaffold did not conduct anions or cations. These data suggest that the putative pore-loop does not form a complete, transferable pore domain. Furthermore, our data reveal an unexpected role for the putative pore-loop of TMEM16A and TMEM16B channels in the control of the whole-cell Ca(2+)-activated Cl(-) conductance.


Asunto(s)
Canales de Cloruro/química , Proteínas de Neoplasias/química , Animales , Calcio/fisiología , Membrana Celular/fisiología , Canales de Cloruro/fisiología , Células HEK293 , Humanos , Ratones , Proteínas de Neoplasias/fisiología , Estructura Terciaria de Proteína , Transfección
11.
Sci Signal ; 16(811): eadk5661, 2023 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-37963193

RESUMEN

The TMEM16A channel represents a key depolarizing mechanism in arterial smooth muscle and contractile pericytes, where it is activated by several endogenous contractile agonists. In this issue of Science Signaling, Mata-Daboin et al. demonstrate a previously unidentified role for TMEM16A in endothelial cells for acetylcholine-mediated vasorelaxation. Collectively, TMEM16A serves as a transducer of vasoactive stimuli to enable fine modulation of vessel tone.


Asunto(s)
Canales de Cloruro , Células Endoteliales , Canales de Cloruro/genética , Músculo Liso , Arterias , Aniones , Anoctamina-1/genética
12.
Trends Pharmacol Sci ; 43(9): 712-725, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35811176

RESUMEN

The TMEM16A Ca2+-gated Cl- channel is involved in a variety of vital physiological functions and may be targeted pharmacologically for therapeutic benefit in diseases such as hypertension, stroke, and cystic fibrosis (CF). The determination of the TMEM16A structure and high-throughput screening efforts, alongside ex vivo and in vivo animal studies and clinical investigations, are hastening our understanding of the physiology and pharmacology of this channel. Here, we offer a critical analysis of recent developments in TMEM16A pharmacology and reflect on the therapeutic opportunities provided by this target.


Asunto(s)
Canales de Cloruro , Fibrosis Quística , Animales , Anoctamina-1 , Calcio/metabolismo
13.
J Clin Invest ; 132(9)2022 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-35316222

RESUMEN

Pericyte-mediated capillary constriction decreases cerebral blood flow in stroke after an occluded artery is unblocked. The determinants of pericyte tone are poorly understood. We show that a small rise in cytoplasmic Ca2+ concentration ([Ca2+]i) in pericytes activated chloride efflux through the Ca2+-gated anion channel TMEM16A, thus depolarizing the cell and opening voltage-gated calcium channels. This mechanism strongly amplified the pericyte [Ca2+]i rise and capillary constriction evoked by contractile agonists and ischemia. In a rodent stroke model, TMEM16A inhibition slowed the ischemia-evoked pericyte [Ca2+]i rise, capillary constriction, and pericyte death; reduced neutrophil stalling; and improved cerebrovascular reperfusion. Genetic analysis implicated altered TMEM16A expression in poor patient recovery from ischemic stroke. Thus, pericyte TMEM16A is a crucial regulator of cerebral capillary function and a potential therapeutic target for stroke and possibly other disorders of impaired microvascular flow, such as Alzheimer's disease and vascular dementia.


Asunto(s)
Pericitos , Accidente Cerebrovascular , Calcio/metabolismo , Circulación Cerebrovascular/genética , Humanos , Isquemia/metabolismo , Pericitos/metabolismo , Accidente Cerebrovascular/metabolismo
14.
J Physiol ; 588(Pt 13): 2305-14, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20421283

RESUMEN

Calcium-activated chloride channels (CaCCs) play important roles in several physiological processes. In vascular smooth muscle, activation of these ion channels by agonist-induced Ca(2+) release results in membrane depolarization and vasoconstriction. The molecular identity of vascular CaCCs is not fully defined. Here we present evidence that TMEM16A (or anoctamin 1), a member of the transmembrane 16 (TMEM16) protein family, forms CaCCs in pulmonary artery smooth muscle cells (PASMCs). Patch-clamp analysis in freshly isolated PASMCs revealed strongly outward-rectifying, slowly activating Ca(2+)-activated Cl(-) currents sharing a high degree of similarity with heterologous TMEM16A currents. TMEM16A mRNA was identified in rat and human pulmonary arteries and various other vascular smooth muscle cell types. Further analyses revealed that several TMEM16A splice variants were detected in rat PASMCs and that TMEM16F and TMEM16K were also expressed in these cells, while TMEM16B, TMEM16D and TMEM16E were all at least 50 times less abundantly expressed and the remaining TMEM16 family members were absent. Downregulation of TMEM16A gene expression in primary cultures of rat PASMCs, with small interfering RNAs, was accompanied by almost total loss of whole-cell CaCC currents. Based on these results, we propose that TMEM16A is the major constituent of the vascular calcium-activated chloride channel in rat pulmonary artery smooth muscle.


Asunto(s)
Calcio/fisiología , Canales de Cloruro/fisiología , Miocitos del Músculo Liso/fisiología , Arteria Pulmonar/fisiología , Algoritmos , Animales , Anoctamina-1 , Células Cultivadas , Canales de Cloruro/genética , Electrofisiología , Humanos , Masculino , Arteria Pulmonar/citología , ARN Interferente Pequeño/genética , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Soluciones
16.
J Physiol ; 587(Pt 8): 1649-56, 2009 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-19237428

RESUMEN

ATP-sensitive K(+) (K(ATP)) channels couple cell metabolism to cell electrical activity. Wild-type (Kir6.2/SUR1) K(ATP) channels heterologously expressed in Xenopus oocytes give rise to very small inward currents in cell-attached patches. A large increase in the current is observed on patch excision into zero ATP solution. This is presumably due to loss of intracellular ATP leading to unblock of K(ATP) channels. In contrast, channels containing Kir6.2 mutations associated with reduced ATP-sensitivity display non-zero cell-attached currents. Unexpectedly, these cell-attached currents are significantly smaller (by approximately 40%) than those observed when excised patches are exposed to physiological ATP concentrations (1-10 mm). Cramming the patch back into the oocyte cytoplasm restores mutant K(ATP) current amplitude to that measured in the cell-attached mode. This implies that the magnitude of the cell-attached current is regulated not only by intracellular ATP but also by another cytoplasmic factor/s. This factor seems to require the nucleotide-binding domains of SUR1 to be effective. Thus a mutant Kir6.2 (Kir6.2DeltaC-I296L) expressed in the absence of SUR1 exhibited currents of similar magnitude in cell-attached patches as in inside-out patches exposed to 10 mm MgATP. Similar results were found when Kir6.2-I296L was coexpressed with an SUR1 mutant that is insensitive to MgADP or MgATP activation. This suggests the oocyte contains a cytoplasmic factor that reduces nucleotide binding/hydrolysis at the NBDs of SUR1. In conclusion, our results reveal a novel regulatory mechanism for the K(ATP) channel. This was not evident for wild-type channels because of their high sensitivity to block by ATP.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/antagonistas & inhibidores , Adenosina Difosfato/antagonistas & inhibidores , Adenosina Trifosfato/antagonistas & inhibidores , Citosol/fisiología , Canales KATP/fisiología , Bloqueadores de los Canales de Potasio/química , Canales de Potasio de Rectificación Interna/antagonistas & inhibidores , Receptores de Droga/antagonistas & inhibidores , Transportadoras de Casetes de Unión a ATP/fisiología , Animales , Citosol/química , Electrofisiología , Femenino , Humanos , Canales KATP/antagonistas & inhibidores , Canales KATP/genética , Mutación/genética , Mutación/fisiología , Oocitos/metabolismo , Técnicas de Placa-Clamp , Canales de Potasio de Rectificación Interna/genética , Canales de Potasio de Rectificación Interna/fisiología , Ratas , Receptores de Droga/fisiología , Receptores de Sulfonilureas , Xenopus
17.
Nat Commun ; 10(1): 3956, 2019 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-31477691

RESUMEN

Membranes in cells have defined distributions of lipids in each leaflet, controlled by lipid scramblases and flip/floppases. However, for some intracellular membranes such as the endoplasmic reticulum (ER) the scramblases have not been identified. Members of the TMEM16 family have either lipid scramblase or chloride channel activity. Although TMEM16K is widely distributed and associated with the neurological disorder autosomal recessive spinocerebellar ataxia type 10 (SCAR10), its location in cells, function and structure are largely uncharacterised. Here we show that TMEM16K is an ER-resident lipid scramblase with a requirement for short chain lipids and calcium for robust activity. Crystal structures of TMEM16K show a scramblase fold, with an open lipid transporting groove. Additional cryo-EM structures reveal extensive conformational changes from the cytoplasmic to the ER side of the membrane, giving a state with a closed lipid permeation pathway. Molecular dynamics simulations showed that the open-groove conformation is necessary for scramblase activity.


Asunto(s)
Anoctaminas/metabolismo , Retículo Endoplásmico/metabolismo , Lípidos/química , Proteínas de Transferencia de Fosfolípidos/metabolismo , Secuencia de Aminoácidos , Animales , Anoctaminas/química , Anoctaminas/genética , Células COS , Calcio/química , Línea Celular Tumoral , Chlorocebus aethiops , Cristalografía por Rayos X , Células HEK293 , Humanos , Simulación de Dinámica Molecular , Proteínas de Transferencia de Fosfolípidos/química , Proteínas de Transferencia de Fosfolípidos/genética , Homología de Secuencia de Aminoácido , Células Sf9 , Spodoptera
18.
Methods Mol Biol ; 491: 127-39, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18998089

RESUMEN

Oocytes from the Xenopus laevis represent one of the most widely used expression systems for functional characterization of ion channels. Their large size facilitates both injection of heterologous cRNA and subsequent electrophysiological recordings of ion channel currents. Furthermore, Xenopus oocytes translate cRNA very efficiently, resulting in the generation of a large number of ion channels in the plasma membrane. In this chapter, we outline methods for oocyte preparation and maintenance and describe procedures for patch-clamping of oocytes, with a special focus on the macropatch technique. We discuss some common problems associated with patch-clamping of oocytes and their use as an expression system for ion channels.


Asunto(s)
Canales de Cloruro/fisiología , Canales Iónicos/fisiología , Canales KATP/fisiología , Oocitos/fisiología , Animales , Canales de Cloruro/aislamiento & purificación , Electrofisiología/métodos , Femenino , Canales Iónicos/genética , Canales KATP/aislamiento & purificación , Técnicas de Placa-Clamp , ARN Complementario/genética , Xenopus laevis
19.
Br J Pharmacol ; 175(11): 2028-2045, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29486056

RESUMEN

BACKGROUND AND PURPOSE: Optogenetic control of electromechanical coupling in vascular smooth muscle cells (VSMCs) is emerging as a powerful research tool with potential applications in drug discovery and therapeutics. However, the precise ionic mechanisms involved in this control remain unclear. EXPERIMENTAL APPROACH: Cell imaging, patch-clamp electrophysiology and muscle tension recordings were used to define these mechanisms over a wide range of light stimulations. KEY RESULTS: Transgenic mice expressing a channelrhodopsin-2 variant [ChR2(H134R)] selectively in VSMCs were generated. Isolated VSMCs obtained from these mice demonstrated blue light-induced depolarizing whole-cell currents. Fine control of artery tone was attained by varying the intensity of the light stimulus. This arterial response was sufficient to overcome the endogenous, melanopsin-mediated, light-evoked, arterial relaxation observed in the presence of contractile agonists. Ca2+ entry through voltage-gated Ca2+ channels, and opening of plasmalemmal depolarizing channels (TMEM16A and TRPM) and intracellular IP3 receptors were involved in the ChR2(H134R)-dependent arterial response to blue light at intensities lower than ~0.1 mW·mm-2 . Light stimuli of greater intensity evoked a significant Ca2+ influx directly through ChR2(H134R) and produced marked intracellular alkalinization of VSMCs. CONCLUSIONS AND IMPLICATIONS: We identified the range of light intensity allowing optical control of arterial tone, primarily by means of endogenous channels and without substantial alteration to intracellular pH. Within this range, mice expressing ChR2(H134R) in VSMCs are a powerful experimental model for achieving accurate and tuneable optical voltage-clamp of VSMCs and finely graded control of arterial tone, offering new approaches to the discovery of vasorelaxant drugs.


Asunto(s)
Channelrhodopsins/metabolismo , Miocitos del Músculo Liso/metabolismo , Optogenética , Animales , Electrofisiología , Concentración de Iones de Hidrógeno , Iones/metabolismo , Ratones , Ratones Transgénicos , Miocitos del Músculo Liso/citología , Imagen Óptica , Vasoconstricción
20.
Endocr Dev ; 11: 70-82, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17986828

RESUMEN

ATP-sensitive potassium (KATP) channels are inhibited by intracellular ATP and activated by MgADP. As a consequence, they couple the metabolic state of the cell to its electrical activity. In pancreatic Beta-cells KATP channels regulate glucose-dependent insulin secretion and are the target for sulphonylurea drugs clinically employed in the treatment of type 2 diabetes. This review discusses recent advances in our understanding of the role of KATP channels in permanent neonatal diabetes mellitus.


Asunto(s)
Diabetes Mellitus/congénito , Diabetes Mellitus/genética , Diabetes Mellitus/terapia , Humanos , Recién Nacido , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/fisiología , Canales KATP/genética , Canales KATP/metabolismo , Canales KATP/fisiología , Modelos Biológicos , Modelos Moleculares , Mutación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA