Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 12(1): 4482, 2021 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-34301959

RESUMEN

Activation of Pannexin 1 (PANX1) ion channels causes release of intercellular signaling molecules in a variety of (patho)physiological contexts. PANX1 can be activated by G protein-coupled receptors (GPCRs), including α1-adrenergic receptors (α1-ARs), but how receptor engagement leads to channel opening remains unclear. Here, we show that GPCR-mediated PANX1 activation can occur via channel deacetylation. We find that α1-AR-mediated activation of PANX1 channels requires Gαq but is independent of phospholipase C or intracellular calcium. Instead, α1-AR-mediated PANX1 activation involves RhoA, mammalian diaphanous (mDia)-related formin, and a cytosolic lysine deacetylase activated by mDia - histone deacetylase 6. HDAC6 associates with PANX1 and activates PANX1 channels, even in excised membrane patches, suggesting direct deacetylation of PANX1. Substitution of basally-acetylated intracellular lysine residues identified on PANX1 by mass spectrometry either prevents HDAC6-mediated activation (K140/409Q) or renders the channels constitutively active (K140R). These data define a non-canonical RhoA-mDia-HDAC6 signaling pathway for GαqPCR activation of PANX1 channels and uncover lysine acetylation-deacetylation as an ion channel silencing-activation mechanism.


Asunto(s)
Conexinas/metabolismo , Histona Desacetilasa 6/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores Adrenérgicos alfa 1/metabolismo , Acetilación , Animales , Células Cultivadas , Conexinas/genética , Conexinas/fisiología , Células HEK293 , Histona Desacetilasa 6/genética , Humanos , Células Jurkat , Lisina/genética , Lisina/metabolismo , Potenciales de la Membrana/fisiología , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/fisiología , Técnicas de Placa-Clamp , Receptores Adrenérgicos alfa 1/genética , Transducción de Señal/genética , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoA/metabolismo
2.
Mol Metab ; 4(9): 610-8, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26413467

RESUMEN

OBJECTIVE: Defective glucose uptake in adipocytes leads to impaired metabolic homeostasis and insulin resistance, hallmarks of type 2 diabetes. Extracellular ATP-derived nucleotides and nucleosides are important regulators of adipocyte function, but the pathway for controlled ATP release from adipocytes is unknown. Here, we investigated whether Pannexin 1 (Panx1) channels control ATP release from adipocytes and contribute to metabolic homeostasis. METHODS: We assessed Panx1 functionality in cultured 3T3-L1 adipocytes and in adipocytes isolated from murine white adipose tissue by measuring ATP release in response to known activators of Panx1 channels. Glucose uptake in cultured 3T3-L1 adipocytes was measured in the presence of Panx1 pharmacologic inhibitors and in adipocytes isolated from white adipose tissue from wildtype (WT) or adipocyte-specific Panx1 knockout (AdipPanx1 KO) mice generated in our laboratory. We performed in vivo glucose uptake studies in chow fed WT and AdipPanx1 KO mice and assessed insulin resistance in WT and AdipPanx1 KO mice fed a high fat diet for 12 weeks. Panx1 channel function was assessed in response to insulin by performing electrophysiologic recordings in a heterologous expression system. Finally, we measured Panx1 mRNA in human visceral adipose tissue samples by qRT-PCR and compared expression levels with glucose levels and HOMA-IR measurements in patients. RESULTS: Our data show that adipocytes express functional Pannexin 1 (Panx1) channels that can be activated to release ATP. Pharmacologic inhibition or selective genetic deletion of Panx1 from adipocytes decreased insulin-induced glucose uptake in vitro and in vivo and exacerbated diet-induced insulin resistance in mice. Further, we identify insulin as a novel activator of Panx1 channels. In obese humans Panx1 expression in adipose tissue is increased and correlates with the degree of insulin resistance. CONCLUSIONS: We show that Panx1 channel activity regulates insulin-stimulated glucose uptake in adipocytes and thus contributes to control of metabolic homeostasis.

3.
Sci Signal ; 8(364): ra17, 2015 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-25690012

RESUMEN

Both purinergic signaling through nucleotides such as ATP (adenosine 5'-triphosphate) and noradrenergic signaling through molecules such as norepinephrine regulate vascular tone and blood pressure. Pannexin1 (Panx1), which forms large-pore, ATP-releasing channels, is present in vascular smooth muscle cells in peripheral blood vessels and participates in noradrenergic responses. Using pharmacological approaches and mice conditionally lacking Panx1 in smooth muscle cells, we found that Panx1 contributed to vasoconstriction mediated by the α1 adrenoreceptor (α1AR), whereas vasoconstriction in response to serotonin or endothelin-1 was independent of Panx1. Analysis of the Panx1-deficient mice showed that Panx1 contributed to blood pressure regulation especially during the night cycle when sympathetic nervous activity is highest. Using mimetic peptides and site-directed mutagenesis, we identified a specific amino acid sequence in the Panx1 intracellular loop that is essential for activation by α1AR signaling. Collectively, these data describe a specific link between noradrenergic and purinergic signaling in blood pressure homeostasis.


Asunto(s)
Presión Sanguínea/fisiología , Conexinas/metabolismo , Músculo Liso Vascular/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores Adrenérgicos alfa 1/metabolismo , Transducción de Señal/fisiología , Vasoconstricción/fisiología , Adenosina Trifosfato/metabolismo , Secuencia de Aminoácidos , Análisis de Varianza , Animales , Western Blotting , Conexinas/genética , Endotelina-1/metabolismo , Técnica del Anticuerpo Fluorescente , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Músculo Liso Vascular/citología , Mutagénesis Sitio-Dirigida , Proteínas del Tejido Nervioso/genética , Técnicas de Placa-Clamp , Serotonina/metabolismo , Telemetría
4.
J Biol Chem ; 287(47): 39602-12, 2012 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-23033481

RESUMEN

S-nitrosylation is a post-translational modification on cysteine(s) that can regulate protein function, and pannexin 1 (Panx1) channels are present in the vasculature, a tissue rich in nitric oxide (NO) species. Therefore, we investigated whether Panx1 can be S-nitrosylated and whether this modification can affect channel activity. Using the biotin switch assay, we found that application of the NO donor S-nitrosoglutathione (GSNO) or diethylammonium (Z)-1-1(N,N-diethylamino)diazen-1-ium-1,2-diolate (DEA NONOate) to human embryonic kidney (HEK) 293T cells expressing wild type (WT) Panx1 and mouse aortic endothelial cells induced Panx1 S-nitrosylation. Functionally, GSNO and DEA NONOate attenuated Panx1 currents; consistent with a role for S-nitrosylation, current inhibition was reversed by the reducing agent dithiothreitol and unaffected by 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one, a blocker of guanylate cyclase activity. In addition, ATP release was significantly inhibited by treatment with both NO donors. To identify which cysteine residue(s) was S-nitrosylated, we made single cysteine-to-alanine substitutions in Panx1 (Panx1(C40A), Panx1(C346A), and Panx1(C426A)). Mutation of these single cysteines did not prevent Panx1 S-nitrosylation; however, mutation of either Cys-40 or Cys-346 prevented Panx1 current inhibition and ATP release by GSNO. This observation suggested that multiple cysteines may be S-nitrosylated to regulate Panx1 channel function. Indeed, we found that mutation of both Cys-40 and Cys-346 (Panx1(C40A/C346A)) prevented Panx1 S-nitrosylation by GSNO as well as the GSNO-mediated inhibition of Panx1 current and ATP release. Taken together, these results indicate that S-nitrosylation of Panx1 at Cys-40 and Cys-346 inhibits Panx1 channel currents and ATP release.


Asunto(s)
Conexinas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Óxido Nítrico/metabolismo , Adenosina Trifosfato/genética , Adenosina Trifosfato/metabolismo , Sustitución de Aminoácidos , Animales , Conexinas/genética , Ditiotreitol/farmacología , Glutatión/análogos & derivados , Glutatión/farmacología , Células HEK293 , Humanos , Ratones , Mutación , Mutación Missense , Proteínas del Tejido Nervioso/genética , Donantes de Óxido Nítrico/farmacología , Nitrocompuestos/farmacología , Oxadiazoles/farmacología , Oxidación-Reducción/efectos de los fármacos , Compuestos de Amonio Cuaternario/farmacología , Quinoxalinas/farmacología
5.
J Physiol ; 590(24): 6257-66, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23070703

RESUMEN

It is widely recognized that ATP, along with other nucleotides, subserves important intercellular signalling processes. Among various nucleotide release mechanisms, the relatively recently identified pannexin 1 (Panx1) channel is gaining prominence by virtue of its ability to support nucleotide permeation and release in a variety of different tissues. Here, we review recent advances in our understanding of the factors that control Panx1 channel activity. By using electrophysiological and biochemical approaches, diverse mechanisms that dynamically regulate Panx1 channel function have been identified in various settings; these include, among others, activation by caspase-mediated channel cleavage in apoptotic immune cells, by G protein-coupled receptors in vascular smooth muscle, by low oxygen tension in erythrocytes and neurons, by high extracellular K(+) in various cell types and by stretch/strain in airway epithelia. Delineating the distinct mechanisms of Panx1 modulation that prevail in different physiological contexts provides the possibility that these channels, and ATP release, could ultimately be targeted in a context-dependent manner.


Asunto(s)
Adenosina Trifosfato/metabolismo , Conexinas/metabolismo , Activación del Canal Iónico , Proteínas del Tejido Nervioso/metabolismo , Animales , Conexinas/química , Humanos , Mecanotransducción Celular , Potenciales de la Membrana , Proteínas del Tejido Nervioso/química , Péptido Hidrolasas/metabolismo , Permeabilidad , Transporte de Proteínas , Receptores Purinérgicos/metabolismo
6.
Trends Cardiovasc Med ; 22(3): 68-72, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22841835

RESUMEN

Pannexins are a recently discovered protein family with the isoform Panx1 ubiquitously expressed and therefore extensively studied. Panx1 proteins form membrane channels known to release purines such as ATP. Because ATP and, more generally, purinergic signaling plays an important role in the vasculature, it became evident that Panx1 could have a key role in vascular functions. This article reviews recent findings on the pivotal role of Panx1 in smooth muscle cells in the contraction of arteries as well as recent insights into Panx1 channel regulation.


Asunto(s)
Arterias/fisiología , Conexinas/biosíntesis , Miocitos del Músculo Liso/fisiología , Proteínas del Tejido Nervioso/biosíntesis , Adenosina Trifosfato , Humanos , Receptores Adrenérgicos alfa 1/biosíntesis , Receptores Adrenérgicos alfa 1/fisiología , Transducción de Señal
7.
J Biol Chem ; 287(14): 11303-11, 2012 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-22311983

RESUMEN

Pannexin 1 (PANX1) channels mediate release of ATP, a "find-me" signal that recruits macrophages to apoptotic cells; PANX1 activation during apoptosis requires caspase-mediated cleavage of PANX1 at its C terminus, but how the C terminus inhibits basal channel activity is not understood. Here, we provide evidence suggesting that the C terminus interacts with the human PANX1 (hPANX1) pore and that cleavage-mediated channel activation requires disruption of this inhibitory interaction. Basally silent hPANX1 channels localized on the cell membrane could be activated directly by protease-mediated C-terminal cleavage, without additional apoptotic effectors. By serial deletion, we identified a C-terminal region just distal to the caspase cleavage site that is required for inhibition of hPANX1; point mutations within this small region resulted in partial activation of full-length hPANX1. Consistent with the C-terminal tail functioning as a pore blocker, we found that truncated and constitutively active hPANX1 channels could be inhibited, in trans, by the isolated hPANX1 C terminus either in cells or when applied directly as a purified peptide in inside-out patch recordings. Furthermore, using a cysteine cross-linking approach, we showed that relief of inhibition following cleavage requires dissociation of the C terminus from the channel pore. Collectively, these data suggest a mechanism of hPANX1 channel regulation whereby the intact, pore-associated C terminus inhibits the full-length hPANX1 channel and a remarkably well placed caspase cleavage site allows effective removal of key inhibitory C-terminal determinants to activate hPANX1.


Asunto(s)
Adenosina Trifosfato/metabolismo , Caspasas/metabolismo , Conexinas/química , Conexinas/metabolismo , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Proteolisis , Secuencia de Aminoácidos , Animales , Sitios de Unión , Membrana Celular/metabolismo , Células HEK293 , Humanos , Ratones , Datos de Secuencia Molecular , Porosidad
8.
Nature ; 467(7317): 863-7, 2010 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-20944749

RESUMEN

Apoptotic cells release 'find-me' signals at the earliest stages of death to recruit phagocytes. The nucleotides ATP and UTP represent one class of find-me signals, but their mechanism of release is not known. Here, we identify the plasma membrane channel pannexin 1 (PANX1) as a mediator of find-me signal/nucleotide release from apoptotic cells. Pharmacological inhibition and siRNA-mediated knockdown of PANX1 led to decreased nucleotide release and monocyte recruitment by apoptotic cells. Conversely, PANX1 overexpression enhanced nucleotide release from apoptotic cells and phagocyte recruitment. Patch-clamp recordings showed that PANX1 was basally inactive, and that induction of PANX1 currents occurred only during apoptosis. Mechanistically, PANX1 itself was a target of effector caspases (caspases 3 and 7), and a specific caspase-cleavage site within PANX1 was essential for PANX1 function during apoptosis. Expression of truncated PANX1 (at the putative caspase cleavage site) resulted in a constitutively open channel. PANX1 was also important for the 'selective' plasma membrane permeability of early apoptotic cells to specific dyes. Collectively, these data identify PANX1 as a plasma membrane channel mediating the regulated release of find-me signals and selective plasma membrane permeability during apoptosis, and a new mechanism of PANX1 activation by caspases.


Asunto(s)
Apoptosis , Permeabilidad de la Membrana Celular/fisiología , Conexinas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Fagocitosis , Adenosina Trifosfato/metabolismo , Apoptosis/efectos de los fármacos , Carbenoxolona/farmacología , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Quimiotaxis/efectos de los fármacos , Conexinas/antagonistas & inhibidores , Conexinas/deficiencia , Conexinas/genética , Conductividad Eléctrica , Humanos , Células Jurkat , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Técnicas de Placa-Clamp , Fagocitos/citología , Fagocitos/fisiología , Fagocitosis/efectos de los fármacos , Uridina Trifosfato/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...