Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Angiogenesis ; 24(3): 677-693, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33770321

RESUMEN

Endothelial barrier disruption and vascular leak importantly contribute to organ dysfunction and mortality during inflammatory conditions like sepsis and acute respiratory distress syndrome. We identified the kinase Arg/Abl2 as a mediator of endothelial barrier disruption, but the role of Arg in endothelial monolayer regulation and its relevance in vivo remain poorly understood. Here we show that depletion of Arg in endothelial cells results in the activation of both RhoA and Rac1, increased cell spreading and elongation, redistribution of integrin-dependent cell-matrix adhesions to the cell periphery, and improved adhesion to the extracellular matrix. We further show that Arg is activated in the endothelium during inflammation, both in murine lungs exposed to barrier-disruptive agents, and in pulmonary microvessels of septic patients. Importantly, Arg-depleted endothelial cells were less sensitive to barrier-disruptive agents. Despite the formation of F-actin stress fibers and myosin light chain phosphorylation, Arg depletion diminished adherens junction disruption and intercellular gap formation, by reducing the disassembly of cell-matrix adhesions and cell retraction. In vivo, genetic deletion of Arg diminished vascular leak in the skin and lungs, in the presence of a normal immune response. Together, our data indicate that Arg is a central and non-redundant regulator of endothelial barrier integrity, which contributes to cell retraction and gap formation by increasing the dynamics of adherens junctions and cell-matrix adhesions in a Rho GTPase-dependent fashion. Therapeutic inhibition of Arg may provide a suitable strategy for the treatment of a variety of clinical conditions characterized by vascular leak.


Asunto(s)
Matriz Extracelular/metabolismo , Uniones Comunicantes/enzimología , Células Endoteliales de la Vena Umbilical Humana/enzimología , Proteínas Tirosina Quinasas/metabolismo , Alveolos Pulmonares/enzimología , Animales , Adhesión Celular/genética , Activación Enzimática , Matriz Extracelular/genética , Uniones Comunicantes/genética , Humanos , Inflamación/enzimología , Inflamación/genética , Ratones , Ratones Noqueados , Proteínas Tirosina Quinasas/genética
2.
Clin Sci (Lond) ; 135(20): 2409-2422, 2021 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-34386810

RESUMEN

Oxidative stress could be a possible mechanism and a therapeutic target of atrial fibrillation (AF). However, the effects of the xanthine oxidase (XO) inhibition for AF remain to be fully elucidated. We investigated the effects of a novel XO inhibitor febuxostat on AF compared with allopurinol in hypertension rat model. Five-week-old Dahl salt-sensitive rats were fed either low-salt (LS) (0.3% NaCl) or high-salt (HS) (8% NaCl) diet. After 4 weeks of diet, HS diet rats were divided into three groups: orally administered to vehicle (HS-C), febuxostat (5 mg/kg/day) (HS-F), or allopurinol (50 mg/kg/day) (HS-A). After 4 weeks of treatment, systolic blood pressure (SBP) was significantly higher in HS-C than LS, and it was slightly but significantly decreased by treatment with each XO inhibitor. AF duration was significantly prolonged in HS-C compared with LS, and significantly suppressed in both HS-F and HS-A (LS; 5.8 ± 3.5 s, HS-C; 33.9 ± 23.7 s, HS-F; 15.0 ± 14.1 s, HS-A; 20.1 ± 11.9 s: P<0.05). Ca2+ spark frequency was obviously increased in HS-C rats and reduced in the XO inhibitor-treated rats, especially in HS-F group. Western blotting revealed that the atrial expression levels of Met281/282-oxidized Ca2+/Calmodulin-dependent kinase II (CaMKII) and Ser2814-phosphorylated ryanodine receptor 2 were significantly increased in HS-C, and those were suppressed in HS-F and HS-A. Decreased expression of gap junction protein connexin 40 in HS-C was partially restored by treatment with each XO inhibitor. In conclusion, XO inhibitor febuxostat, as well as allopurinol, could reduce hypertension-related increase in AF perpetuation by restoring Ca2+ handling and gap junction.


Asunto(s)
Fibrilación Atrial/prevención & control , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Inhibidores Enzimáticos/farmacología , Febuxostat/farmacología , Hipertensión/tratamiento farmacológico , Miocitos Cardíacos/efectos de los fármacos , Xantina Oxidasa/antagonistas & inhibidores , Alopurinol/farmacología , Animales , Fibrilación Atrial/enzimología , Fibrilación Atrial/genética , Fibrilación Atrial/fisiopatología , Señalización del Calcio , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Conexinas/genética , Conexinas/metabolismo , Modelos Animales de Enfermedad , Fibrosis , Uniones Comunicantes/efectos de los fármacos , Uniones Comunicantes/enzimología , Uniones Comunicantes/patología , Hipertensión/enzimología , Hipertensión/genética , Hipertensión/fisiopatología , Masculino , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/patología , Oxidación-Reducción , Fosforilación , Ratas Endogámicas Dahl , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Cloruro de Sodio Dietético , Xantina Oxidasa/metabolismo , Proteína alfa-5 de Unión Comunicante
3.
Basic Res Cardiol ; 115(6): 62, 2020 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-32975669

RESUMEN

Neuraminidase (NEU)1 forms a multienzyme complex with beta-galactosidase (ß-GAL) and protective-protein/cathepsin (PPC) A, which cleaves sialic-acids from cell surface glycoconjugates. We investigated the role of NEU1 in the myocardium after ischemia/reperfusion (I/R). Three days after inducing I/R, left ventricles (LV) of male mice (3 months-old) displayed upregulated neuraminidase activity and increased NEU1, ß-GAL and PPCA expression. Mice hypomorphic for neu1 (hNEU1) had less neuraminidase activity, fewer pro-inflammatory (Lin-CD11b+F4/80+Ly-6Chigh), and more anti-inflammatory macrophages (Lin-CD11b+F4/80+Ly-6Clow) 3 days after I/R, and less LV dysfunction 14 days after I/R. WT mice transplanted with hNEU1-bone marrow (BM) and hNEU1 mice with WT-BM showed significantly better LV function 14 days after I/R compared with WT mice with WT-BM. Mice with a cardiomyocyte-specific NEU1 overexpression displayed no difference in inflammation 3 days after I/R, but showed increased cardiomyocyte hypertrophy, reduced expression and mislocalization of Connexin-43 in gap junctions, and LV dysfunction despite a similar infarct scar size to WT mice 14 days after I/R. The upregulation of NEU1 after I/R contributes to heart failure by promoting inflammation in invading monocytes/macrophages, enhancing cardiomyocyte hypertrophy, and impairing gap junction function, suggesting that systemic NEU1 inhibition may reduce heart failure after I/R.


Asunto(s)
Insuficiencia Cardíaca/etiología , Hipertrofia Ventricular Izquierda/etiología , Macrófagos/enzimología , Monocitos/enzimología , Infarto del Miocardio/complicaciones , Daño por Reperfusión Miocárdica/complicaciones , Miocitos Cardíacos/enzimología , Neuraminidasa/deficiencia , Disfunción Ventricular Izquierda/etiología , Animales , Catepsina A/metabolismo , Conexina 43/metabolismo , Modelos Animales de Enfermedad , Femenino , Uniones Comunicantes/enzimología , Uniones Comunicantes/patología , Insuficiencia Cardíaca/enzimología , Insuficiencia Cardíaca/inmunología , Insuficiencia Cardíaca/fisiopatología , Hipertrofia Ventricular Izquierda/enzimología , Hipertrofia Ventricular Izquierda/inmunología , Hipertrofia Ventricular Izquierda/fisiopatología , Macrófagos/inmunología , Masculino , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Monocitos/inmunología , Infarto del Miocardio/enzimología , Infarto del Miocardio/inmunología , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/enzimología , Daño por Reperfusión Miocárdica/inmunología , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/patología , Neuraminidasa/genética , Disfunción Ventricular Izquierda/enzimología , Disfunción Ventricular Izquierda/inmunología , Disfunción Ventricular Izquierda/fisiopatología , Función Ventricular Izquierda , Remodelación Ventricular , beta-Galactosidasa/metabolismo
4.
Arterioscler Thromb Vasc Biol ; 37(12): 2280-2290, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29025706

RESUMEN

OBJECTIVE: Because of its strategic position between endothelial and smooth muscle cells in microvessels, Cx37 (Connexin 37) plays an important role in myoendothelial gap junctional intercellular communication. We have shown before that NO inhibits gap junctional intercellular communication through gap junctions containing Cx37. However, the underlying mechanism is not yet identified. APPROACH AND RESULTS: Using channel-forming Cx37 mutants exhibiting partial deletions or amino acid exchanges in their C-terminal loops, we now show that the phosphorylation state of a tyrosine residue at position 332 (Y332) in the C-terminus of Cx37 controls the gap junction-dependent spread of calcium signals. Mass spectra revealed that NO protects Cx37 from dephosphorylation at Y332 by inhibition of the protein tyrosine phosphatase SHP-2. Functionally, the inhibition of gap junctional intercellular communication by NO decreased the spread of the calcium signal (induced by mechanical stimulation of individual endothelial cells) from endothelial to smooth muscle cells in intact vessels, while, at the same time, augmenting the calcium signal spreading within the endothelium. Consequently, preincubation of small resistance arteries with exogenous NO enhanced the endothelium-dependent dilator response to acetylcholine in spite of a pharmacological blockade of NO-dependent cGMP formation by the soluable guanylyl cyclase inhibitor ODQ (1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one). CONCLUSIONS: Our results identify a novel mechanism by which NO can increase the efficacy of calcium, rising vasoactive agonists in the microvascular endothelium.


Asunto(s)
Señalización del Calcio/efectos de los fármacos , Comunicación Celular/efectos de los fármacos , Conexinas/metabolismo , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Extremidad Inferior/irrigación sanguínea , Músculo Liso Vascular/efectos de los fármacos , Donantes de Óxido Nítrico/farmacología , Óxido Nítrico/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Animales , Arterias/efectos de los fármacos , Arterias/enzimología , Conexinas/genética , Relación Dosis-Respuesta a Droga , Uniones Comunicantes/efectos de los fármacos , Uniones Comunicantes/enzimología , Células HeLa , Células Endoteliales de la Vena Umbilical Humana/enzimología , Humanos , Masculino , Ratones Endogámicos C57BL , Músculo Liso Vascular/enzimología , Óxido Nítrico/farmacología , Donantes de Óxido Nítrico/metabolismo , Fosforilación , Dominios Proteicos , Interferencia de ARN , Proteínas Recombinantes de Fusión/metabolismo , Transfección , Tirosina , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología , Proteína alfa-4 de Unión Comunicante
5.
J Biol Chem ; 291(30): 15867-80, 2016 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-27235399

RESUMEN

Connexin43 (Cx43) assembly and degradation, the regulation of electrical and metabolic coupling, as well as modulating the interaction with other proteins, involve phosphorylation. Here, we identified and characterized the biological significance of a novel tyrosine kinase that phosphorylates Cx43, tyrosine kinase 2 (Tyk2). Activation of Tyk2 led to a decrease in Cx43 gap junction communication by increasing the turnover rate of Cx43 from the plasma membrane. Tyk2 directly phosphorylated Cx43 residues Tyr-247 and Tyr-265, leading to indirect phosphorylation on residues Ser-279/Ser-282 (MAPK) and Ser-368 (PKC). Although this phosphorylation pattern is similar to what has been observed following Src activation, the response caused by Tyk2 occurred when Src was inactive in NRK cells. Knockdown of Tyk2 at the permissive temperature (active v-Src) in LA-25 cells decreased Cx43 phosphorylation, indicating that although activation of Tyk2 and v-Src leads to phosphorylation of the same Cx43CT residues, they are not identical in level at each site. Additionally, angiotensin II activation of Tyk2 increased the intracellular protein level of Cx43 via STAT3. These findings indicate that, like Src, Tyk2 can also inhibit gap junction communication by phosphorylating Cx43.


Asunto(s)
Conexina 43/biosíntesis , Uniones Comunicantes/enzimología , Regulación de la Expresión Génica , TYK2 Quinasa/metabolismo , Animales , Línea Celular , Conexina 43/genética , Uniones Comunicantes/genética , Proteína Oncogénica pp60(v-src)/genética , Proteína Oncogénica pp60(v-src)/metabolismo , Fosforilación/genética , Ratas , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , TYK2 Quinasa/genética
6.
Diabetes Obes Metab ; 19 Suppl 1: 30-41, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28466490

RESUMEN

Ca2+ is the key intracellular regulator of insulin secretion, acting in the ß-cell as the ultimate trigger for exocytosis. In response to high glucose, ATP-sensitive K+ channel closure and plasma membrane depolarization engage a sophisticated machinery to drive pulsatile cytosolic Ca2+ changes. Voltage-gated Ca2+ channels, Ca2+ -activated K+ channels and Na+ /Ca2+ exchange all play important roles. The use of targeted Ca2+ probes has revealed that during each cytosolic Ca2+ pulse, uptake of Ca2+ by mitochondria, endoplasmic reticulum (ER), secretory granules and lysosomes fine-tune cytosolic Ca2+ dynamics and control organellar function. For example, changes in the expression of the Ca2+ -binding protein Sorcin appear to provide a link between ER Ca2+ levels and ER stress, affecting ß-cell function and survival. Across the islet, intercellular communication between highly interconnected "hubs," which act as pacemaker ß-cells, and subservient "followers," ensures efficient insulin secretion. Loss of connectivity is seen after the deletion of genes associated with type 2 diabetes (T2D) and follows metabolic and inflammatory insults that characterize this disease. Hubs, which typically comprise ~1%-10% of total ß-cells, are repurposed for their specialized role by expression of high glucokinase (Gck) but lower Pdx1 and Nkx6.1 levels. Single cell-omics are poised to provide a deeper understanding of the nature of these cells and of the networks through which they communicate. New insights into the control of both the intra- and intercellular Ca2+ dynamics may thus shed light on T2D pathology and provide novel opportunities for therapy.


Asunto(s)
Señalización del Calcio , Comunicación Celular , Islotes Pancreáticos/metabolismo , Modelos Biológicos , Animales , Membrana Celular/enzimología , Membrana Celular/metabolismo , Diabetes Mellitus Tipo 2/enzimología , Diabetes Mellitus Tipo 2/metabolismo , Retículo Endoplásmico/enzimología , Retículo Endoplásmico/metabolismo , Exocitosis , Uniones Comunicantes/enzimología , Uniones Comunicantes/metabolismo , Regulación de la Expresión Génica , Glucosa/metabolismo , Humanos , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/enzimología , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/enzimología , Mitocondrias/enzimología , Mitocondrias/metabolismo , Vías Secretoras
7.
Circulation ; 124(10): 1138-50, 2011 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-21844074

RESUMEN

BACKGROUND: Ischemia/reperfusion injury may have deleterious short- and long-term consequences for cardiac allografts. The underlying mechanisms involve microvascular dysfunction that may culminate in primary graft failure or untreatable chronic rejection. METHODS AND RESULTS: Here, we report that rat cardiac allograft ischemia/reperfusion injury resulted in profound microvascular dysfunction that was prevented by donor treatment with peroral single-dose simvastatin, a 3-hydroxy-3-methylglutaryl coenzyme A reductase and Rho GTPase inhibitor, 2 hours before graft procurement. During allograft preservation, donor simvastatin treatment inhibited microvascular endothelial cell and pericyte RhoA/Rho-associated protein kinase activation and endothelial cell-endothelial cell gap formation; decreased intragraft mRNA levels of hypoxia-inducible factor-1α, inducible nitric oxide synthase, and endothelin-1; and increased heme oxygenase-1. Donor, but not recipient, simvastatin treatment prevented ischemia/reperfusion injury-induced vascular leakage, leukocyte infiltration, the no-reflow phenomenon, and myocardial injury. The beneficial effects of simvastatin on vascular stability and the no-reflow phenomenon were abolished by concomitant nitric oxide synthase inhibition with N-nitro-l-arginine methyl ester and RhoA activation by geranylgeranyl pyrophosphate supplementation, respectively. In the chronic rejection model, donor simvastatin treatment inhibited cardiac allograft inflammation, transforming growth factor-ß1 signaling, and myocardial fibrosis. In vitro, simvastatin inhibited transforming growth factor-ß1-induced microvascular endothelial-to-mesenchymal transition. CONCLUSIONS: Our results demonstrate that donor simvastatin treatment prevents microvascular endothelial cell and pericyte dysfunction, ischemia/reperfusion injury, and chronic rejection and suggest a novel, clinically feasible strategy to protect cardiac allografts.


Asunto(s)
Inhibidores Enzimáticos/uso terapéutico , Rechazo de Injerto/prevención & control , Trasplante de Corazón , Microvasos/efectos de los fármacos , Disfunción Primaria del Injerto/prevención & control , Daño por Reperfusión/prevención & control , Simvastatina/uso terapéutico , Animales , Células Endoteliales/efectos de los fármacos , Células Endoteliales/enzimología , Endotelina-1/biosíntesis , Uniones Comunicantes/efectos de los fármacos , Uniones Comunicantes/enzimología , Hemo-Oxigenasa 1/biosíntesis , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Complejo Mayor de Histocompatibilidad/efectos de los fármacos , Masculino , Microvasos/enzimología , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Fenómeno de no Reflujo/prevención & control , Fosfatos de Poliisoprenilo/farmacología , Disfunción Primaria del Injerto/enzimología , Ratas , Ratas Endogámicas WF , Quinasas Asociadas a rho/antagonistas & inhibidores
8.
Glia ; 59(6): 973-80, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21446052

RESUMEN

Aquaporin-4 (AQP4) is the main water channel in the brain and primarily localized to astrocytes where the channels are thought to contribute to water and K(+) homeostasis. The close apposition of AQP4 and inward rectifier K(+) channels (Kir4.1) led to the hypothesis of direct functional interactions between both channels. We investigated the impact of AQP4 on stimulus-induced alterations of the extracellular K(+) concentration ([K(+)](o)) in murine hippocampal slices. Recordings with K(+)-selective microelectrodes combined with field potential analyses were compared in wild type (wt) and AQP4 knockout (AQP4(-/-)) mice. Astrocyte gap junction coupling was assessed with tracer filling during patch clamp recording. Antidromic fiber stimulation in the alveus evoked smaller increases and slower recovery of [K(+)](o) in the stratum pyramidale of AQP4(-/-) mice indicating reduced glial swelling and a larger extracellular space when compared with control tissue. Moreover, the data hint at an impairment of the glial Na(+)/K(+) ATPase in AQP4-deficient astrocytes. In a next step, we investigated the laminar profile of [K(+)](o) by moving the recording electrode from the stratum pyramidale toward the hippocampal fissure. At distances beyond 300 µm from the pyramidal layer, the stimulation-induced, normalized increases of [K(+)](o) in AQP4(-/-) mice exceeded the corresponding values of wt mice, indicating facilitated spatial buffering. Astrocytes in AQP4(-/-) mice also displayed enhanced tracer coupling, which might underlie the improved spatial re- distribution of [K(+)](o) in the hippocampus. These findings highlight the role of AQP4 channels in the regulation of K(+) homeostasis.


Asunto(s)
Acuaporina 4/fisiología , Uniones Comunicantes/metabolismo , Hipocampo/metabolismo , Canales de Potasio de Rectificación Interna/fisiología , Potasio/metabolismo , Animales , Acuaporina 4/deficiencia , Tampones (Química) , Comunicación Celular/genética , Uniones Comunicantes/enzimología , Uniones Comunicantes/genética , Hipocampo/enzimología , Homeostasis/genética , Potenciales de la Membrana/genética , Ratones , Ratones Noqueados , Técnicas de Cultivo de Órganos , Técnicas de Placa-Clamp/métodos , Potasio/fisiología , ATPasa Intercambiadora de Sodio-Potasio/metabolismo
9.
Circ Res ; 104(9): 1103-12, 2009 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-19342602

RESUMEN

Lateralization of the ventricular gap junction protein connexin 43 (Cx43) occurs in epicardial border zone myocytes following myocardial infarction (MI) and is arrhythmogenic. Alterations in Cx43 protein partners have been hypothesized to play a role in lateralization although mechanisms by which this occurs are unknown. To examine potential mechanisms we did nuclear magnetic resonance, yeast 2-hybrid, and surface plasmon resonance studies and found that the SH3 domain of the tyrosine kinase c-Src binds to the Cx43 scaffolding protein zonula occludens-1 (ZO-1) with a higher affinity than does Cx43. This suggests c-Src outcompetes Cx43 for binding to ZO-1, thus acting as a chaperone for ZO-1 and causing unhooking from Cx43. To determine whether c-Src/ZO-1 interactions affect Cx43 lateralization within the epicardial border zone, we performed Western blot, immunoprecipitation, and immunolocalization for active c-Src (p-cSrc) post-MI using a canine model of coronary occlusion. We found that post-MI p-cSrc interacts with ZO-1 as Cx43 begins to decrease its interaction with ZO-1 and undergo initial loss of intercalated disk localization. This indicates that the molecular mechanisms by which Cx43 is lost from the intercalated disk following MI includes an interaction of p-cSrc with ZO-1 and subsequent loss of scaffolding of Cx43 leaving Cx43 free to diffuse in myocyte membranes from areas of high Cx43, as at the intercalated disk, to regions of lower Cx43 content, the lateral myocyte membrane. Therefore shifts in Cx43 protein partners may underlie, in part, arrhythmogenesis in the post-MI heart.


Asunto(s)
Conexina 43/metabolismo , Uniones Comunicantes/metabolismo , Proteínas de la Membrana/metabolismo , Infarto del Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Pericardio/metabolismo , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Secuencia de Aminoácidos , Animales , Unión Competitiva , Conexina 43/química , Modelos Animales de Enfermedad , Perros , Uniones Comunicantes/enzimología , Uniones Comunicantes/patología , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Datos de Secuencia Molecular , Infarto del Miocardio/enzimología , Infarto del Miocardio/patología , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/patología , Dominios PDZ , Pericardio/enzimología , Pericardio/patología , Fosforilación , Unión Proteica , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Mapeo de Interacción de Proteínas , Transporte de Proteínas , Proteínas Proto-Oncogénicas pp60(c-src)/química , Resonancia por Plasmón de Superficie , Técnicas del Sistema de Dos Híbridos , Proteína de la Zonula Occludens-1 , Dominios Homologos src
10.
BMC Cell Biol ; 11: 3, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-20078861

RESUMEN

BACKGROUND: We investigated the effects of the signaling molecules, cyclic AMP (cAMP) and protein-kinase C (PKC), on gap junctional intercellular communication (GJIC) between thymic epithelial cells (TEC). RESULTS: Treatment with 8-Br-cAMP, a cAMP analog; or forskolin, which stimulates cAMP production, resulted in an increase in dye transfer between adjacent TEC, inducing a three-fold enhancement in the mean fluorescence of coupled cells, ascertained by flow cytometry after calcein transfer. These treatments also increased Cx43 mRNA expression, and stimulated Cx43 protein accumulation in regions of intercellular contacts. VIP, adenosine, and epinephrine which may also signal through cyclic nucleotides were tested. The first two molecules did not mimic the effects of 8-Br-cAMP, however epinephrine was able to increase GJIC suggesting that this molecule functions as an endogenous inter-TEC GJIC modulators. Stimulation of PKC by phorbol-myristate-acetate inhibited inter-TEC GJIC. Importantly, both the enhancing and the decreasing effects, respectively induced by cAMP and PKC, were observed in both mouse and human TEC preparations. Lastly, experiments using mouse thymocyte/TEC heterocellular co-cultures suggested that the presence of thymocytes does not affect the degree of inter-TEC GJIC. CONCLUSIONS: Overall, our data indicate that cAMP and PKC intracellular pathways are involved in the homeostatic control of the gap junction-mediated communication in the thymic epithelium, exerting respectively a positive and negative role upon cell coupling. This control is phylogenetically conserved in the thymus, since it was seen in both mouse and human TEC preparations. Lastly, our work provides new clues for a better understanding of how the thymic epithelial network can work as a physiological syncytium.


Asunto(s)
Comunicación Celular/fisiología , AMP Cíclico/metabolismo , Células Epiteliales/metabolismo , Proteína Quinasa C/metabolismo , Timo/citología , 8-Bromo Monofosfato de Adenosina Cíclica/farmacología , Animales , Colforsina/farmacología , Conexina 43/genética , Conexina 43/metabolismo , Regulación hacia Abajo , Uniones Comunicantes/enzimología , Uniones Comunicantes/metabolismo , Humanos , Ratones , Ésteres del Forbol/farmacología
11.
Biochem Biophys Res Commun ; 400(2): 230-5, 2010 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-20727856

RESUMEN

Gap junctional communication, which is mediated by the connexin protein family, is essential for the maintenance of normal tissue function and homeostasis. Loss of intercellular communication results in a failure to coordinately regulate cellular functions, and it can facilitate tumorigenesis. Expression of oncogenes and stimulation with cytokines has been shown to suppress intercellular communication; however, the exact mechanism by which intercellular communication is disrupted by these factors remains uncertain. In this report, we show that Akt is essential for the disruption of gap junctional communication in v-Src-transformed cells. In addition, inhibition of Akt restores gap junctional communication after it is suppressed by TNF-α signaling. Furthermore, we demonstrate that the expression of a constitutively active form of Akt1, but not of Akt2 or Akt3, is sufficient to suppress gap junctional communication. Our results clearly define Akt1 as one of the critical regulators of gap junctional communication.


Asunto(s)
Comunicación Celular , Uniones Comunicantes/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Línea Celular Transformada , Uniones Comunicantes/enzimología , Ratones , Proteína Oncogénica pp60(v-src)/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Ratas , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo
12.
Cell Tissue Res ; 340(2): 229-42, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20336469

RESUMEN

Gap junction intercellular communication and cell-cell adhesion are essential for maintaining a normal cellular phenotype, including the control of growth and proliferation. Loss of either cell-cell adhesion or communication is common in cancers, while restoration of function is associated with tumor suppression. Protein kinase C (PKC) isozymes regulate a broad spectrum of cellular functions including growth and proliferation, and their overexpression has been correlated with carcinogenesis. Consequently, PKC inhibitors are currently undergoing clinical trials as an anti-cancer agents although the precise cellular alterations induced by PKC inhibitors remain to be elucidated. In the current study, the effects of PKC inhibitors on cell interactions were investigated using human neuroblastoma (IMR32, SKNMC, and SHSY-5Y) cell lines. An analysis of intercellular communication revealed an increase in gap junctional coupling with PKC inhibition. The observed increase in coupling was not associated with a change in Connexin 43 distribution or an alteration of phosphorylation status of the protein. There was also an increase in cell-cell adhesion with PKC inhibitor treatment as indicated by a cell aggregation assay. Therefore, the growth suppressive abilities of PKC inhibition on tumors may be due to the cancer suppressive effects of increased gap junction intercellular communication and cell-cell adhesion.


Asunto(s)
Comunicación Celular , Uniones Comunicantes/enzimología , Neuroblastoma/enzimología , Neuroblastoma/patología , Proteína Quinasa C/antagonistas & inhibidores , Bioensayo , Adhesión Celular/efectos de los fármacos , Agregación Celular/efectos de los fármacos , Comunicación Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Conexina 43/metabolismo , Activación Enzimática/efectos de los fármacos , Uniones Comunicantes/efectos de los fármacos , Humanos , Immunoblotting , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Transporte de Proteínas/efectos de los fármacos
13.
J Cell Physiol ; 220(3): 600-10, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19373869

RESUMEN

This manuscript describes how the permeability of pulmonary artery microvascular endothelial cell (RPMEC) monolayer is elevated by hypoxia and the role played by HSP27 phosphorylation. p38 MAP kinase activation leading to HSP27 phosphorylation was previously shown by our laboratory to alter the actin cytoskeleton and tethering properties of RPMEC. This effect was independent of hypoxia-induced contractility which was ROCK-dependent rather than HSP27-dependent. Results described here show that increased HSP27 phosphorylation not only does not underlie hypoxia-induced permeability, but may actually augment the endothelial barrier. Hypoxia causes gap formation between RPMEC and increases MLC2 phosphorylation. The phosphorylation of MYPT1, which inhibits MLC2 phosphatase, is also increased in hypoxia. In addition, FAK phosphorylation, which alters focal adhesion signaling, is increased in hypoxia. Overexpressing phosphomimicking HSP27 (pmHSP27), which induces significant actin stress fiber formation, surprisingly renders RPMEC resistant to hypoxia- or TGFbeta-induced permeability. siRNA against pmHSP27 reverses the increased actin stress fiber formation in pmHSP27-overexpressing cells, and disrupting actin stress fibers in pmHSP27-overexpressing RPMEC renders them more susceptible to hypoxia. Finally, hypoxia-induced gap formation, as well as phosphorylation of MLC2, MYPT1 and FAK are almost abolished by overexpressing pmHSP27 in RPMEC. These effects of pmHSP27 overexpression might represent decreased cytoskeletal plasticity and increased tethering which counteracts permeability-inducing contractility. Thus hypoxia activates two pathways one leading to contractility and increased permeability, the other leading to actin stress fibers, stronger adhesion, and reduced permeability. Altering HSP27 phosphorylation, which tips the balance towards decreased permeability, might be targeted in managing endothelial barrier dysfunction.


Asunto(s)
Permeabilidad Capilar , Células Endoteliales/metabolismo , Uniones Comunicantes/metabolismo , Proteínas de Choque Térmico HSP27/metabolismo , Transducción de Señal , Animales , Miosinas Cardíacas/metabolismo , Hipoxia de la Célula , Células Cultivadas , Células Endoteliales/enzimología , Quinasa 1 de Adhesión Focal/metabolismo , Uniones Comunicantes/enzimología , Proteínas de Choque Térmico HSP27/genética , Proteínas de Choque Térmico , Chaperonas Moleculares , Mutación , Cadenas Ligeras de Miosina/metabolismo , Fosforilación , Proteína Fosfatasa 1/metabolismo , Interferencia de ARN , Ratas , Fibras de Estrés/metabolismo , Factores de Tiempo , Transfección , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Quinasas Asociadas a rho/metabolismo
14.
J Cell Physiol ; 220(3): 716-26, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19472214

RESUMEN

Barrier stabilizing effects of cAMP as well as of the small GTPase Rac 1 are well established. Moreover, it is generally believed that permeability-increasing mediators such as thrombin disrupt endothelial barrier functions primarily via activation of Rho A. In this study, we provide evidence that decrease of both cAMP levels and of Rac 1 activity contribute to thrombin-mediated barrier breakdown. Treatment of human dermal microvascular endothelial cells (HDMEC) with Rac 1-inhibitor NSC-23766 decreased transendothelial electrical resistance (TER) and caused intercellular gap formation. These effects were reversed by addition of forskolin/rolipram (F/R) to increase intracellular cAMP but not by the cAMP analogue 8-pCPT-2'-O-Methyl-cAMP (O-Me-cAMP) which primarily stimulates protein kinase A (PKA)-independent signaling via Epac/Rap 1. However, both F/R and O-Me-cAMP did not increase TER above control levels in the presence of NSC-23766 in contrast to experiments without Rac 1 inhibition. Because Rac 1 was required for maintenance of barrier functions as well as for cAMP-mediated barrier stabilization, we tested the role of Rac 1 and cAMP in thrombin-induced barrier breakdown. Thrombin-induced drop of TER and intercellular gap formation were paralleled by a rapid decrease of cAMP as revealed by fluorescence resonance energy transfer (FRET). The efficacy of F/R or O-Me-cAMP to block barrier-destabilizing effects of thrombin was comparable to Y27632-induced inhibition of Rho kinase but was blunted when Rac 1 was inactivated by NSC-23766. Taken together, these data indicate that decrease of cAMP and Rac 1 activity may be an important step in inflammatory barrier disruption.


Asunto(s)
Permeabilidad Capilar , AMP Cíclico/metabolismo , Células Endoteliales/enzimología , Uniones Comunicantes/enzimología , Transducción de Señal , Trombina/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Aminoquinolinas/farmacología , Antígenos CD/metabolismo , Técnicas Biosensibles , Cadherinas/metabolismo , Calcio/metabolismo , Permeabilidad Capilar/efectos de los fármacos , Células Cultivadas , Colforsina/farmacología , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacología , Impedancia Eléctrica , Células Endoteliales/efectos de los fármacos , Activadores de Enzimas/farmacología , Inhibidores Enzimáticos/farmacología , Transferencia Resonante de Energía de Fluorescencia , Uniones Comunicantes/efectos de los fármacos , Humanos , Microscopía Fluorescente , Pirimidinas/farmacología , Rolipram/farmacología , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Proteína de Unión al GTP cdc42/metabolismo , Proteína de Unión al GTP rac1/antagonistas & inhibidores
15.
J Cell Physiol ; 217(3): 759-68, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18668519

RESUMEN

One of the shared physiological roles between TGF-beta and connexin family members is to inhibit epithelial cell cycle progression and consequently, to provide protection against malignant transformation. Herein, we demonstrated that TGF-beta1 induces the expression of connexin43 (Cx43) in normal murine mammary gland (NMuMG) cell lines at the protein and mRNA levels, and transcriptionally. Using overexpression of a truncated dominant-negative form of Cx43, we determined that the modulation of gap junctional communication by TGF-beta1 plays a key role in the control of NMuMG cells proliferation by TGF-beta1. In addition, using overexpression of truncated dominant-negative forms of either Smad2 or Smad3, and MDA-MB-468 human breast carcinoma cells deficient for Smad4, we determined that the Smad cascade is not implicated in TGF-beta1 effect on Cx43 expression. Using specific pharmacologic inhibitors for JNK, ERK, p38, and PI3K/AKT signaling pathways, we demonstrated the cooperative role of p38 and PI3K/AKT signaling in TGF-beta1-induced Cx43 expression and gap junctional communication. Furthermore, transfection of a c-jun antisense expression vector significantly prevented TGF-beta1-induced Cx43 gene expression demonstrating the involvement of c-Jun/AP-1 pathway together with p38 and PI3K/AKT pathways in mediating TGF-beta1-induced Cx43 gene expression.


Asunto(s)
Conexina 43/genética , Células Epiteliales/enzimología , Glándulas Mamarias Animales/citología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Comunicación Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Femenino , Uniones Comunicantes/efectos de los fármacos , Uniones Comunicantes/enzimología , Humanos , Glándulas Mamarias Animales/enzimología , Ratones , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas Smad/metabolismo , Transcripción Genética/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos , Xenopus
16.
Biochem Biophys Res Commun ; 373(4): 597-601, 2008 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-18601906

RESUMEN

Gap junctions are plasma membrane domains containing channels that directly connect the cytosols of neighbouring cells. Gap junction channels are made of a family of transmembrane proteins called connexins, of which the best studied is Connexin43 (Cx43). MAP kinase-induced phosphorylation of Cx43 has previously been shown to cause inhibition of gap junction channel permeability and increased Cx43 endocytosis. As Cx43 assembles into gap junction plaques, Cx43 acquires detergent resistance. Here we report that the detergent resistance is lost after activation of MAP kinase. Treatment of IAR20 rat liver epithelial cells with 12-O-tetradecanoylphorbol 13-acetate (TPA) or epidermal growth factor (EGF) caused a rapid increase in the solubility of Cx43 in Triton X-100. This process was mediated by MAP kinase and was initiated at the plasma membrane. The data suggest that loss of the detergent resistance of Cx43 is an early step in TPA- and EGF-induced endocytosis of gap junctions.


Asunto(s)
Conexina 43/química , Conexina 43/metabolismo , Endocitosis , Uniones Comunicantes/metabolismo , Animales , Línea Celular , Activación Enzimática , Factor de Crecimiento Epidérmico/farmacología , Uniones Comunicantes/enzimología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Octoxinol/química , Fosforilación , Proteína Quinasa C/metabolismo , Ratas , Solubilidad , Acetato de Tetradecanoilforbol/farmacología
17.
Cancer Res ; 62(17): 4922-8, 2002 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-12208742

RESUMEN

2-Methyl-1,4-naphthoquinone, vitamin K(3) (menadione), which is frequently used as a model quinone in cell culture and in vivo studies, was tested for its effects on gap-junctional intercellular communication (GJC). Exposure of WB-F344 rat liver epithelial cells to menadione (50-100 micro M) led to a 50-75% decrease in GJIC. Different from the phorbol ester 12-O-tetradecanoylphorbol 13-acetate, menadione did not induce internalization of gap junctions. Rather, the decreased GJIC was found to be because of phosphorylation of connexin 43, the major connexin in the used cell line, which was mediated by MAPK/ERK kinase (MEK) 1 and MEK 2 as well as by activation of their direct substrates, extracellular signal-regulated kinase (ERK) 1 and ERK 2. Activation of ERK 1/2 was demonstrated to be independent of NAD(P)H:quinone oxidoreductase using the inhibitor dicoumarol, thus excluding redox cycling as the major mechanism causing these menadione effects. A substantial increase in tyrosine phosphorylation was detected in the cell membrane immunocytochemically upon exposure to menadione, consistent with arylation by menadione bearing the responsibility for the signaling events induced and consistent with the fact that protein tyrosine phosphatases are known targets of arylation reactions. ERK activation was attenuated using specific inhibitors of the epidermal growth factor receptor tyrosine kinase. Similarly, these inhibitors as well as inhibitors of MEK 1/2 counteracted the loss in gap-junctional communication elicited by menadione. This is of interest for chemotherapeutic approaches exploiting the bystander-effect, which is based upon intact GJIC.


Asunto(s)
Comunicación Celular/efectos de los fármacos , Receptores ErbB/fisiología , Uniones Comunicantes/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Vitamina K 3/farmacología , Animales , Comunicación Celular/fisiología , Células Cultivadas , Conexina 43/metabolismo , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/enzimología , Uniones Comunicantes/enzimología , Uniones Comunicantes/fisiología , Hígado/citología , Hígado/efectos de los fármacos , Hígado/enzimología , MAP Quinasa Quinasa 1 , MAP Quinasa Quinasa 2 , Sistema de Señalización de MAP Quinasas/fisiología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Ratas , Ratas Endogámicas F344
18.
Oncol Rep ; 36(6): 3251-3258, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27748887

RESUMEN

Chemotherapeutic drug-induced apoptosis is enhanced by gap junction intercellular communication (GJIC) in a variety of tumor cells. Oxaliplatin and gefitinib are the most widely used chemotherapeutic drugs. However, the synergistic influence remains unknown in testicular cancer chemotherapy. The aim of the present study was to investigate the apoptosis induced by oxaliplatin combined with gefitinib and the potential mechanisms in I-10 testicular cancer cells. The results showed that gefitinib significantly enhanced oxaliplatin-induced apoptosis. Furthermore, the ratio of Bcl-2/Bax and the cleavage of caspase-3 and -9 were increased by gefitinib during oxaliplatin-induced apoptosis. The oxaliplatin-induced apoptosis was enhanced through the upregulation of gap junction (GJ) channels composed of connexin 43 (Cx43) by gefitinib. The mechanism of GJIC enhancement involved the suppression by gefitinib of the expression levels of Src and PKC, which phosphorylate Cx43 and reduce GJIC. PP2 (Src inhibitor) and GF109203X (PKC inhibitor) also enhanced GJIC function. Our findings demonstrated that gefitinib enhanced oxaliplatin-induced apoptosis in I-10 cells and gefitinib upregulated the GJIC by inhibiting Src and PKC-modulated Cx43 phosphorylation.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Uniones Comunicantes/enzimología , Compuestos Organoplatinos/farmacología , Quinazolinas/farmacología , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Línea Celular Tumoral , Conexina 43/metabolismo , Resistencia a Antineoplásicos , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Gefitinib , Humanos , Oxaliplatino , Fosforilación , Proteína Quinasa C/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína X Asociada a bcl-2/metabolismo , Familia-src Quinasas/metabolismo
19.
J Comp Neurol ; 491(1): 46-55, 2005 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-16127697

RESUMEN

The role of auditory circuitry is to decipher relevant information from acoustic signals. Acoustic parameters used by different insect species vary widely. All these auditory systems, however, share a common transducer: tympanal organs as well as the Drosophila flagellar ears use chordotonal organs as the auditory mechanoreceptors. We here describe the central neural projections of the Drosophila Johnston's organ (JO). These neurons, which represent the antennal auditory organ, terminate in the antennomechanosensory center. To ensure correct identification of these terminals we made use of a beta-galactosidase-expressing transgene that labels JO neurons specifically. Analysis of these projection pathways shows that parallel JO fibers display extensive contacts, including putative gap junctions. We find that the synaptic boutons show both chemical synaptic structures as well as putative gap junctions, indicating mixed synapses, and belong largely to the divergent type, with multiple small postsynaptic processes. The ultrastructure of JO fibers and synapses may indicate an ability to process temporally discretized acoustic information.


Asunto(s)
Vías Auditivas/ultraestructura , Drosophila/ultraestructura , Ganglios de Invertebrados/ultraestructura , Mecanorreceptores/ultraestructura , Órganos de los Sentidos/ultraestructura , Sinapsis/ultraestructura , Animales , Vías Auditivas/enzimología , Drosophila/enzimología , Oído , Ganglios de Invertebrados/enzimología , Uniones Comunicantes/enzimología , Uniones Comunicantes/ultraestructura , Audición/fisiología , Mecanorreceptores/enzimología , Nervios Periféricos/enzimología , Nervios Periféricos/ultraestructura , Órganos de los Sentidos/enzimología , Sinapsis/enzimología , beta-Galactosidasa/metabolismo
20.
Brain Res ; 1039(1-2): 189-98, 2005 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-15781061

RESUMEN

We have previously shown that the enhancement of glucose uptake caused by the inhibition of gap junctional communication is a consequence of the increase in astrocyte proliferation. Since C6 glioma cells are highly proliferative and are poorly coupled through gap junctions, we used these cells to investigate the effect of increasing gap junctional communication on the rate of glucose uptake. Previous work by us had shown that tolbutamide increases gap junctional communication in C6 glioma cells, as does dbcAMP, a classical activator of gap junctional communication. In this work, our results show that both tolbutamide and dbcAMP reduce the rate of glucose uptake in C6 glioma cells and that their effects are additive. The main glucose transporters expressed in C6 glioma cells are GLUT-1 and GLUT-3. Neither the expression nor the cellular localization of either GLUT-1 or GLUT-3 were modified by increasing gap junctional communication. The estimation of glucose uptake with 2-deoxyglucose includes not only glucose transport but also glucose phosphorylation, which in C6 glioma cells is mainly catalyzed by type I and type II hexokinase. Our results reveal that the increase in gap junctional communication caused by tolbutamide and dbcAMP is associated with a decrease in the activity of hexokinase. In agreement with this, tolbutamide and dbcAMP caused a rapid change in the localization of both type I and type II hexokinase, which were detached from the mitochondria to the cytosol.


Asunto(s)
Uniones Comunicantes/enzimología , Glioma/enzimología , Glucosa/metabolismo , Hexoquinasa/metabolismo , Mitocondrias/metabolismo , Proteínas de Transporte de Monosacáridos/metabolismo , Animales , Bucladesina/farmacología , Comunicación Celular/fisiología , Proliferación Celular , Transportador de Glucosa de Tipo 1 , Transportador de Glucosa de Tipo 3 , Hipoglucemiantes/farmacología , Proteínas de Transporte de Monosacáridos/efectos de los fármacos , Proteínas del Tejido Nervioso/efectos de los fármacos , Proteínas del Tejido Nervioso/metabolismo , Ratas , Tolbutamida/farmacología , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA