Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Curr Hypertens Rep ; 22(9): 69, 2020 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-32852643

RESUMEN

PURPOSE OF REVIEW: The main goal of this article is to discuss the role of the epithelial sodium channel (ENaC) in extracellular fluid and blood pressure regulation. RECENT FINDINGS: Besides its role in sodium handling in the kidney, recent studies have found that ENaC expressed in other cells including immune cells can influence blood pressure via extra-renal mechanisms. Dendritic cells (DCs) are activated and contribute to salt-sensitive hypertension in an ENaC-dependent manner. We discuss recent studies on how ENaC is regulated in both the kidney and other sites including the vascular smooth muscles, endothelial cells, and immune cells. We also discuss how this extra-renal ENaC can play a role in salt-sensitive hypertension and its promise as a novel therapeutic target. The role of ENaC in blood pressure regulation in the kidney has been well studied. Recent human gene sequencing efforts have identified thousands of variants among the genes encoding ENaC, and research efforts to determine if these variants and their expression in extra-renal tissue play a role in hypertension will advance our understanding of the pathogenesis of ENaC-mediated cardiovascular disease and lead to novel therapeutic targets.


Asunto(s)
Hipertensión , Presión Sanguínea , Células Endoteliales/metabolismo , Canales Epiteliales de Sodio , Humanos , Riñón/metabolismo , Cloruro de Sodio Dietético/efectos adversos , Cloruro de Sodio Dietético/metabolismo
3.
J Mol Med (Berl) ; 94(12): 1335-1347, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27783111

RESUMEN

Recent studies indicate that inflammasomes serve as intracellular machinery to initiate classical cytokine-mediated inflammatory responses and play a crucial role in the pathogenesis of cardiovascular diseases. However, whether or not the activation of endothelial inflammasomes directly causes cell dysfunction or tissue injury without recruitment of inflammatory cells is unknown. We explored the role of endothelial cell inflammasome activation in mediating tight junction disruption, a hallmark event of endothelial barrier dysfunction leading to endothelial hyperpermeability in diabetes. We used confocal microscopy to study the formation and activation of NOD-like receptor family pyrin domain containing-3 (Nlrp3) inflammasomes and expression of tight junction proteins in coronary arteries of streptozotocin-treated diabetic wild type and Nlrp3 gene-deleted mice. We found that Nlrp3 ablation prevented inflammasome activation and tight junction disassembly in the coronary arterial endothelium of diabetic mice. Similarly, Nlrp3 gene silencing prevented high glucose-induced down-regulation of tight junction proteins in cultured mouse vascular endothelial cells (MVECs). The high glucose-induced tight junction disruption and consequent endothelial permeability were attributed to increased release of the high mobility group box protein-1 (HMGB1), which is dependent on enhanced Nlrp3 inflammasome activity. Mechanistically, reducing reactive oxygen species (ROS) production abolished high glucose-induced inflammasome activation, tight junction disruption, and endothelial hyperpermeability in MVECs. Collectively, the ROS-dependent activation of endothelial Nlrp3 inflammasomes by hyperglycemia may be an important initiating mechanism to cause endothelial dysfunction. These effects could contribute to the early onset of endothelial injury in diabetes. KEY MESSAGE: Endothelial tight junction disruption in diabetes requires Nlrp3 inflammasomes. High glucose activates Nlrp3 inflammasome in endothelial cells via ROS production. Activation of endothelial inflammasome by high glucose triggers release of HMGB1. Blockade of Nlrp3/HMGB1 axis inhibits high glucose-induced endothelial permeability.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Endotelio Vascular/metabolismo , Hiperglucemia/metabolismo , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Uniones Estrechas/metabolismo , Animales , Línea Celular , Vasos Coronarios/metabolismo , Vasos Coronarios/patología , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patología , Células Endoteliales/metabolismo , Células Endoteliales/patología , Endotelio Vascular/patología , Regulación de la Expresión Génica , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Humanos , Hiperglucemia/genética , Hiperglucemia/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Proteína con Dominio Pirina 3 de la Familia NLR/deficiencia , Oxidación-Reducción , Permeabilidad , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Estreptozocina , Uniones Estrechas/ultraestructura
4.
J Cell Mol Med ; 19(12): 2715-27, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26293846

RESUMEN

Recent studies have indicated that the inflammasome plays a critical role in the pathogenesis of vascular diseases. However, the pathological relevance of this inflammasome activation, particularly in vascular cells, remains largely unknown. Here, we investigated the role of endothelial (Nucleotide-binding Oligomerization Domain) NOD-like receptor family pyrin domain containing three (Nlrp3) inflammasomes in modulating inter-endothelial junction proteins, which are associated with endothelial barrier dysfunction, an early onset of obesity-associated endothelial injury. Our findings demonstrate that the activation of Nlrp3 inflammasome by visfatin markedly decreased the expression of inter-endothelial junction proteins including tight junction proteins ZO-1, ZO-2 and occludin, and adherens junction protein VE-cadherin in cultured mouse vascular endothelial (VE) cell monolayers. Such visfatin-induced down-regulation of junction proteins in endothelial cells was attributed to high mobility group box protein 1 (HMGB1) release derived from endothelial inflammasome-dependent caspase-1 activity. Similarly, in the coronary arteries of wild-type mice, high-fat diet (HFD) treatment caused a down-regulation of inter-endothelial junction proteins ZO-1, ZO-2, occludin and VE-cadherin, which was accompanied with enhanced inflammasome activation and HMGB1 expression in the endothelium as well as transmigration of CD43(+) T cells into the coronary arterial wall. In contrast, all these HFD-induced alterations in coronary arteries were prevented in mice with Nlrp3 gene deletion. Taken together, these data strongly suggest that the activation of endothelial Nlrp3 inflammasomes as a result of the increased actions of injurious adipokines such as visfatin produces HMGB1, which act in paracrine or autocrine fashion to disrupt inter-endothelial junctions and increase paracellular permeability of the endothelium contributing to the early onset of endothelial injury during metabolic disorders such as obesity or high-fat/cholesterol diet.


Asunto(s)
Adipoquinas/farmacología , Células Endoteliales/efectos de los fármacos , Proteína HMGB1/metabolismo , Inflamasomas/efectos de los fármacos , Uniones Intercelulares/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Animales , Línea Celular , Movimiento Celular/genética , Vasos Coronarios/metabolismo , Dieta Alta en Grasa , Células Endoteliales/metabolismo , Proteína HMGB1/genética , Immunoblotting , Inflamasomas/metabolismo , Uniones Intercelulares/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Nicotinamida Fosforribosiltransferasa/farmacología , Interferencia de ARN , Receptor para Productos Finales de Glicación Avanzada/genética , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Proteínas de Uniones Estrechas/genética , Proteínas de Uniones Estrechas/metabolismo , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
5.
Antioxid Redox Signal ; 22(13): 1084-96, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25739025

RESUMEN

AIMS: This study hypothesized that activation of endothelial nucleotide oligomerization domain-like receptor protein with pyrin domain containing 3 (Nlrp3) inflammasomes directly produces endothelial dysfunction during hypercholesterolemia, which is distinct from its canonical roles in inflammation. RESULTS: Acute hypercholesterolemia in mice was induced by intraperitoneal administration of poloxamer 407 (0.5 g/kg) for 24 h. Endothelial dysfunction was assessed by evaluating endothelium-dependent vasodilation in isolated, perfused, and pressurized coronary arteries in response to bradykinin (10(-10)-10(-6) M) and acetylcholine (10(-9)-10(-5) M). Impaired endothelium-dependent vasodilation was observed in Nlrp3(+/+) mice with acute hypercholesterolemia, which was markedly ameliorated in Nlrp3(-/-) mice. Treatment of mice with inhibitors for caspase-1 or high mobility group box 1 (HMGB1) significantly restored endothelium-dependent vasodilation in Nlrp3(+/+) mice with acute hypercholesterolemia. Confocal microscopic analysis demonstrated that hypercholesterolemia markedly increased caspase-1 activity and HMGB1 expression in coronary arterial endothelium of Nlrp3(+/+) mice, which was absent in Nlrp3-deficient mice. Further, recombinant HMGB1 directly induced endothelial dysfunction in normal Nlrp3(+/+) coronary arteries. In vitro, Nlrp3 inflammasome formation and its activity were instigated in cultured endothelial cells by cholesterol crystal, a danger factor associated with hypercholesterolemia. Moreover, cholesterol crystals directly induced endothelial dysfunction in coronary arteries from Nlrp3(+/+) mice, which was attenuated in Nlrp3(-/-) arteries. Such cholesterol crystal-induced impairment was associated with enhanced superoxide production, downregulation of endothelial nitric oxide synthase activity, and pyroptosis. INNOVATION AND CONCLUSION: Our data provide the first evidence that activation of endothelial Nlrp3 inflammasome directly impairs endothelial function beyond its canonical inflammatory actions. This novel non-canonical action of Nlrp3 inflammasomes may initiate or exacerbate vascular injury during hypercholesterolemia.


Asunto(s)
Vasos Coronarios/patología , Células Endoteliales/metabolismo , Hipercolesterolemia/metabolismo , Inflamasomas/metabolismo , Inflamación/metabolismo , Receptores de Superficie Celular/metabolismo , Acetilcolina/efectos adversos , Animales , Bradiquinina/efectos adversos , Proteínas Portadoras/metabolismo , Caspasa 1/metabolismo , Colesterol/farmacología , Vasos Coronarios/efectos de los fármacos , Vasos Coronarios/metabolismo , Proteínas del Citoesqueleto/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Ácido Glicirrínico/farmacología , Proteína HMGB1/metabolismo , Hipercolesterolemia/inducido químicamente , Hipercolesterolemia/patología , Masculino , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR , Óxido Nítrico/metabolismo , Nucleótidos/metabolismo , Poloxámero , Pirina , Piroptosis/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Serpinas/farmacología , Superóxidos/metabolismo , Vasodilatación/efectos de los fármacos , Proteínas Virales/farmacología
6.
Biochim Biophys Acta ; 1853(2): 396-408, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25450976

RESUMEN

Inflammasomes play a critical role in the development of vascular diseases. However, the molecular mechanisms activating the inflammasome in endothelial cells and the relevance of this inflammasome activation is far from clear. Here, we investigated the mechanisms by which an Nlrp3 inflammasome is activated to result in endothelial dysfunction during coronary arteritis by Lactobacillus casei (L. casei) cell wall fragments (LCWE) in a mouse model for Kawasaki disease. Endothelial dysfunction associated with increased vascular cell adhesion protein 1 (VCAM-1) expression and endothelial-leukocyte adhesion was observed during coronary arteritis in mice treated with LCWE. Accompanied with these changes, the inflammasome activation was also shown in coronary arterial endothelium, which was characterized by a marked increase in caspase-1 activity and IL-1ß production. In cultured endothelial cells, LCWE induced Nlrp3 inflammasome formation, caspase-1 activation and IL-1ß production, which were blocked by Nlrp3 gene silencing or lysosome membrane stabilizing agents such as colchicine, dexamethasone, and ceramide. However, a potassium channel blocker glibenclamide or an oxygen free radical scavenger N-acetyl-l-cysteine had no effects on LCWE-induced inflammasome activation. LCWE also increased endothelial cell lysosomal membrane permeability and triggered lysosomal cathepsin B release into cytosol. Silencing cathepsin B blocked LCWE-induced Nlrp3 inflammasome formation and activation in endothelial cells. In vivo, treatment of mice with cathepsin B inhibitor also abolished LCWE-induced inflammasome activation in coronary arterial endothelium. It is concluded that LCWE enhanced lysosomal membrane permeabilization and consequent release of lysosomal cathepsin B, resulting in activation of the endothelial Nlrp3 inflammasome, which may contribute to the development of coronary arteritis.


Asunto(s)
Arteritis/patología , Proteínas Portadoras/metabolismo , Vasos Coronarios/metabolismo , Vasos Coronarios/patología , Células Endoteliales/metabolismo , Inflamasomas/metabolismo , Lisosomas/metabolismo , Animales , Arteritis/metabolismo , Catepsina B/antagonistas & inhibidores , Catepsina B/metabolismo , Pared Celular/química , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Depuradores de Radicales Libres/metabolismo , Silenciador del Gen/efectos de los fármacos , Inflamación/patología , Lacticaseibacillus casei , Lisosomas/efectos de los fármacos , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR , Bloqueadores de los Canales de Potasio/farmacología , Especies Reactivas de Oxígeno/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo
7.
J Cell Mol Med ; 18(11): 2165-75, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24912985

RESUMEN

Dynein-mediated autophagosome (AP) trafficking was recently demonstrated to contribute to the formation of autophagolysosomes (APLs) and autophagic flux process in coronary arterial myocytes (CAMs). However, it remains unknown how the function of dynein as a motor protein for AP trafficking is regulated under physiological and pathological conditions. The present study tested whether the dynein-mediated autophagy maturation is regulated by a redox signalling associated with lysosomal Ca(2+) release machinery. In primary cultures of CAMs, reactive oxygen species (ROS) including H2 O2 and O2 (-.) (generated by xanthine/xanthine oxidase) significantly increased dynein ATPase activity and AP movement, which were accompanied by increased lysosomal fusion with AP and APL formation. Inhibition of dynein activity by (erythro-9-(2-hydroxy-3-nonyl)adenine) (EHNA) or disruption of the dynein complex by dynamitin (DCTN2) overexpression blocked ROS-induced dynein activation, AP movement and APL formation, and resulted in an accumulation of AP along with a failed breakdown of AP. Antagonism of nicotinic acid adenine dinucleotide phosphate (NAADP)-mediated Ca(2+) signalling with NED-19 and PPADS abolished ROS-enhanced lysosomal Ca(2+) release and dynein activation in CAMs. In parallel, all these changes were also enhanced by overexpression of NADPH oxidase-1 (Nox1) gene in CAMs. Incubation with high glucose led to a marked O2 (-.) production compared with normoglycaemic CAMs, while Nox1 inhibitor ML117 abrogated this effect. Moreover, ML117 and NED-19 and PPADS significantly suppressed dynein activity and APL formation caused by high glucose. Taken together, these data suggest that ROS function as important players to regulate dynein-dependent AP trafficking leading to efficient autophagic maturation in CAMs.


Asunto(s)
Autofagia/genética , Vasos Coronarios/metabolismo , Dineínas/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Calcio/metabolismo , Dineínas/genética , Humanos , Lisosomas/metabolismo , Ratones , Miocitos Cardíacos/citología , Fagosomas/genética , Fagosomas/metabolismo , Transporte de Proteínas/genética , Especies Reactivas de Oxígeno/metabolismo
8.
Biochim Biophys Acta ; 1843(5): 836-45, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24508291

RESUMEN

Inflammasome, an intracellular inflammatory machinery, has been reported to be involved in a variety of chronic degenerative diseases such as atherosclerosis, autoinflammatory diseases and Alzheimer's disease. The present study hypothesized that the formation and activation of inflammasomes associated with apoptosis associated speck-like protein (ASC) are an important initiating mechanism resulting in obesity-associated podocyte injury and consequent glomerular sclerosis. To test this hypothesis, Asc gene knockout (Asc(-/-)), wild type (Asc(+/+)) and intrarenal Asc shRNA-transfected wild type (Asc shRNA) mice were fed a high fat diet (HFD) or normal diet (ND) for 12 weeks to produce obesity and associated glomerular injury. Western blot and RT-PCR analyses demonstrated that renal tissue Asc expression was lacking in Asc(-/-) mice or substantially reduced in Asc shRNA transfected mice compared to Asc(+/+) mice. Confocal microscopic and co-immunoprecipitation analysis showed that the HFD enhanced the formation of inflammasome associated with Asc in podocytes as shown by colocalization of Asc with Nod-like receptor protein 3 (Nalp3). This inflammasome complex aggregation was not observed in Asc(-/-) and local Asc shRNA-transfected mice. The caspase-1 activity, IL-1ß production and glomerular damage index (GDI) were also significantly attenuated in Asc(-/-) and Asc shRNA-transfected mice fed the HFD. This decreased GDI in Asc(-/-) and Asc shRNA transfected mice on the HFD was accompanied by attenuated proteinuria, albuminuria, foot process effacement of podocytes and loss of podocyte slit diaphragm molecules. In conclusion, activation and formation of inflammasomes in podocytes are importantly implicated in the development of obesity-associated glomerular injury.


Asunto(s)
Proteínas del Citoesqueleto/genética , Dieta Alta en Grasa , Eliminación de Gen , Silenciador del Gen , Inflamasomas/inmunología , Podocitos/patología , Animales , Proteínas Reguladoras de la Apoptosis , Western Blotting , Proteínas Adaptadoras de Señalización CARD , Masculino , Ratones , Ratones Noqueados , Obesidad/patología , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
9.
J Mol Med (Berl) ; 92(5): 473-85, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24463558

RESUMEN

UNLABELLED: Recent studies have indicated a protective role of autophagy in regulating vascular smooth muscle cells homeostasis in atherogenesis, but the mechanisms controlling autophagy, particularly autophagy maturation, are poorly understood. Here, we investigated whether acid sphingomyelinase (ASM)-regulated lysosome function is involved in autophagy maturation in coronary arterial smooth muscle cells (CASMCs) in the pathogenesis of atherosclerosis. In coronary arterial wall of ASM-deficient (Smpd1⁻/⁻) mice on Western diet, there were high expression levels of both LC3B, a robust marker of autophagosomes (APs), and p62, a selective autophagy substrate, compared with those in wild-type (Smpd1⁺/⁺) mice. By Western blotting and flow cytometry, atherogenic stimulation of Smpd1⁺/⁺ CASMCs with 7-ketocholesterol was found to significantly enhance LC3B expression and increase the content of both APs and autophagolysosomes (APLs). In Smpd1⁻/⁻ CASMCs, such 7-ketocholesterol-induced increases in LC3B and p62 expression and APs were further augmented, but APLs formation was abolished. Analysis of fluorescence resonance energy transfer between fluorescence-labeled LC3B and Lamp1 (lysosome marker) showed that 7-ketocholesterol markedly induced fusion of APs with lysosomes in Smpd1⁺/⁺ CASMCs, which was abolished in Smpd1⁻/⁻ CASMCs. Moreover, 7-ketocholesterol-induced expression of cell dedifferentiation marker vimentin and proliferation was enhanced in Smpd1⁻/⁻ CASMCs compared with those in Smpd1⁺/⁺ CASMCs. Lastly, overexpression of ASM further increased APLs formation in Smpd1⁺/⁺ CASMCs and restored APLs formation in Smpd1⁻/⁻ CASMCs indicating that increased ASM expression is highly correlated with enhanced APLs formation. Taken together, our data suggest that the control of lysosome trafficking and fusion by ASM is essential to a normal autophagic flux in CASMCs, which implicates that the deficiency of ASM-mediated regulation of autophagy maturation may result in imbalance of arterial smooth muscle cell homeostasis and thus serve as an important atherogenic mechanism in coronary arteries. KEY MESSAGES: Acid sphingomyelinase (ASM) controls autophagy maturation in smooth muscle cells. ASM maintains smooth muscle cell homeostasis and its contractile phenotype. ASM plays a protective role in smooth muscle dysfunction and atherosclerosis.


Asunto(s)
Aterosclerosis/enzimología , Autofagia , Vasos Coronarios/enzimología , Miocitos del Músculo Liso/enzimología , Esfingomielina Fosfodiesterasa/metabolismo , Animales , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Catepsinas/metabolismo , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Vasos Coronarios/metabolismo , Vasos Coronarios/patología , Eliminación de Gen , Lisosomas/enzimología , Lisosomas/metabolismo , Lisosomas/patología , Ratones , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Esfingomielina Fosfodiesterasa/genética , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...