Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
FASEB J ; 34(9): 12805-12819, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32772419

RESUMEN

Increased endothelial permeability leads to excessive exudation of plasma proteins and leukocytes in the interstitium, which characterizes several vascular diseases including acute lung injury. The myosin light chain kinase long (MYLK-L) isoform is canonically known to regulate the endothelial permeability by phosphorylating myosin light chain (MLC-P). Compared to the short MYLK isoform, MYLK-L contains an additional stretch of ~919 amino acid at the N-terminus of unknown function. We show that thapsigargin and thrombin-induced SOCE was markedly reduced in Mylk-L-/- endothelial cells (EC) or MYLK-L-depleted human EC. These agonists also failed to increase endothelial permeability in MYLK-L-depleted EC and Mylk-L-/- lungs, thus demonstrating the novel role of MYLK-L-induced SOCE in increasing vascular permeability. MYLK-L augmented SOCE by increasing endoplasmic reticulum (ER)-plasma membrane (PM) junctions and STIM1 translocation to these junctions. Transduction of N-MYLK domain (amino acids 1-919 devoid of catalytic activity) into Mylk-L-/- EC rescued SOCE to the level seen in control EC in a STIM1-dependent manner. N-MYLK-induced SOCE augmented endothelial permeability without MLC-P via an actin-binding motif, DVRGLL. Liposomal-mediated delivery of N-MYLK mutant but not ∆DVRGLL-N-MYLK mutant in Mylk-L-/- mice rescued vascular permeability increase in response to endotoxin, indicating that targeting of DVRGLL motif within MYLK-L may limit SOCE-induced vascular hyperpermeability.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Permeabilidad Capilar , Membrana Celular/enzimología , Retículo Endoplásmico/enzimología , Quinasa de Cadena Ligera de Miosina/metabolismo , Lesión Pulmonar Aguda/metabolismo , Animales , Células Endoteliales de la Vena Umbilical Humana , Humanos , Isoenzimas/metabolismo , Pulmón/metabolismo , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Neoplasias/metabolismo , Molécula de Interacción Estromal 1/metabolismo
2.
Am J Respir Cell Mol Biol ; 61(2): 257-265, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30811958

RESUMEN

Dysfunctional endothelial cell (EC) barrier and increased lung vascular permeability is a cardinal feature of acute lung injury and sepsis that may result in a pathophysiological condition characterized by alveolar flooding, pulmonary edema, and subsequent hypoxemia. In lung ECs, activation of Rho-associated kinase-1 (ROCK1) phosphorylates myosin light chain (MLC)-associated phosphatase at its inhibitory site, which favors phosphorylation of MLC, stress fiber formation, and hyperpermeability during acute lung injury. The role of microRNA-144 (miR-144) has been well investigated in many human diseases, including cardiac ischemia/reperfusion-induced injury, lung cancer, and lung viral infection; however, its role in pulmonary EC barrier regulation remains obscure. Here, we investigated the miR-144-mediated mechanism in the protection of endothelial barrier function in an LPS-induced lung injury model. By using transendothelial electrical resistance and transwell permeability assay to examine in vitro permeability and immunofluorescence microscopy to determine barrier integrity, we showed that ectopic expression of miR-144 effectively blocked lung EC barrier disruption and hyperpermeability in response to proinflammatory agents. Furthermore, using a gain-and-loss-of-function strategy, overexpression of miR-144 significantly decreased ROCK1 expression. Concomitantly, miR-144 inhibits ROCK1-mediated phosphorylation of MLC phosphataseThr853 and thus phosphorylation of MLCThr18/Ser19 to counteract stress fiber formation in LPS-activated EC. Finally, in LPS-challenged mice, intranasal delivery of miR-144 mimic via liposomes attenuated endotoxemia-induced increases in lung wet/dry ratio, vascular permeability, and inflammation. In conclusion, these data suggest that miR-144-attenuated activation of inflammatory ROCK1/MLC pathway in vascular ECs is a promising therapeutic strategy to counter inflammatory lung injury.


Asunto(s)
Células Endoteliales/metabolismo , Pulmón/metabolismo , MicroARNs/metabolismo , Quinasas Asociadas a rho/metabolismo , Animales , Impedancia Eléctrica , Células Endoteliales/efectos de los fármacos , Humanos , Inflamación , Lipopolisacáridos , Liposomas/metabolismo , Pulmón/irrigación sanguínea , Pulmón/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Microcirculación , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Permeabilidad , Daño por Reperfusión , Transducción de Señal
3.
FASEB J ; 30(1): 102-10, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26316271

RESUMEN

Stability of endothelial cell (EC) adherens junctions (AJs) is central for prevention of tissue edema, the hallmark of chronic inflammatory diseases including acute respiratory distress syndrome. Here, we demonstrate a previously unsuspected role of sphingosine kinase 1 (SPHK1) in the mechanism by which transient receptor potential channel 1 (Trpc1)-mediated Ca(2+) entry destabilizes AJs. Trpc1(-/-) monolayers showed a 2.2-fold increase in vascular endothelial (VE)-cadherin cell-surface expression above wild-type (WT) monolayers. Thrombin increased endothelial permeability (evident by a 5-fold increase in interendothelial gap area and 60% decrease in transendothelial electrical resistance) in WT but not Trpc1(-/-) ECs. Trpc1(-/-) mice resisted the hyperpermeability effects of the edemagenic agonists used and exhibited 60% less endotoxin-induced mortality. Because sphingosine-1-phosphate (S1P) strengthens AJs, we determined if TRPC1 functioned by inhibiting SPHK1 activity, which generates S1P. Intriguingly, Trpc1(-/-) ECs or ECs transducing a TRPC1-inactive mutant showed a 1.5-fold increase in basal SPHK1 expression compared with WT ECs, resulting in a 2-fold higher S1P level. SPHK1 inhibitor SK1-I decreased basal transendothelial electrical resistance more in WT ECs (48 and 72% reduction at 20 and 50 µM, respectively) than in Trpc1(-/-) ECs. However, SK1-I pretreatment rescued thrombin-induced EC permeability in Trpc1(-/-) ECs. Thus, TRPC1 suppression of basal SPHK1 activity enables EC-barrier destabilization by edemagenic agonists.


Asunto(s)
Uniones Adherentes/metabolismo , Células Endoteliales/metabolismo , Endotelio/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Canales Catiónicos TRPC/metabolismo , Animales , Cadherinas/metabolismo , Calcio/metabolismo , Permeabilidad de la Membrana Celular , Ratones Noqueados , Transducción de Señal/fisiología , Canales Catiónicos TRPC/genética
4.
Arterioscler Thromb Vasc Biol ; 36(2): 380-8, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26743170

RESUMEN

OBJECTIVE: Increased vascular permeability is a hallmark of sepsis and acute respiratory distress syndrome. Angiopoietin (Ang2) induces vascular leak, and excess Ang2 generation is associated with patient mortality from these diseases. However, mechanisms dampening Ang2 generation during injury remain unclear. Interestingly, microRNA (miR)-150 levels were decreased in septic patients. miR regulate signaling networks by silencing mRNAs containing complementary sequences. Thus, we hypothesized that miR-150 suppresses Ang2 generation and thereby resolves vascular injury. APPROACH AND RESULTS: Wild-type or miR-150(-/-) mice or endothelial cells were exposed to lipopolysaccharide or sepsis, and Ang2 levels, adherens junction reannealing, endothelial barrier function, and mortality were determined. Although Ang2 transiently increased during lipopolysaccharide-induced injury in wild-type endothelial cells and lungs, miR-150 expression was elevated only during recovery from injury. Deletion of miR-150 caused a persistent increase in Ang2 levels and impaired adherens junctions reannealing after injury, resulting thereby in an irreversible increase in vascular permeability. Also, miR-150(-/-) mice died rapidly after sepsis. Rescuing miR-150 expression in endothelial cells prevented Ang2 generation, thereby restoring vascular barrier function in miR-150(-/-) mice. miR-150 terminated Ang2 generation by targeting the transcription factor, early growth response 2. Thus, early growth response 2 or Ang2 depletion in miR-150(-/-) endothelial cells restored junctional reannealing and reinstated barrier function. Importantly, upregulating miR-150 expression by injecting a chemically synthesized miR-150 mimic into wild-type mice vasculature decreased early growth response 2 and Ang2 levels and hence mortality from sepsis. CONCLUSIONS: miR-150 is a novel suppressor of Ang2 generation with a key role in resolving vascular injury and reducing mortality resulting from sepsis.


Asunto(s)
Angiopoyetina 2/metabolismo , Células Endoteliales/metabolismo , MicroARNs/metabolismo , Arteria Pulmonar/metabolismo , Enfermedades Vasculares/metabolismo , Remodelación Vascular , Uniones Adherentes/metabolismo , Uniones Adherentes/patología , Angiopoyetina 2/genética , Animales , Permeabilidad Capilar , Células Cultivadas , Modelos Animales de Enfermedad , Proteína 2 de la Respuesta de Crecimiento Precoz/metabolismo , Células Endoteliales/patología , Regulación de la Expresión Génica , Genotipo , Lipopolisacáridos , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/genética , Fenotipo , Arteria Pulmonar/patología , Sepsis/complicaciones , Transducción de Señal , Factores de Tiempo , Transfección , Enfermedades Vasculares/etiología , Enfermedades Vasculares/genética , Enfermedades Vasculares/patología , Enfermedades Vasculares/terapia
5.
Am J Respir Cell Mol Biol ; 53(3): 355-67, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25569851

RESUMEN

Hypoxic pulmonary vasoconstriction (HPV) is an important physiological response that optimizes the ventilation/perfusion ratio. Chronic hypoxia causes vascular remodeling, which is central to the pathogenesis of hypoxia-induced pulmonary hypertension (HPH). We have previously shown that Notch3 is up-regulated in HPH and that activation of Notch signaling enhances store-operated Ca(2+) entry (SOCE), an important mechanism that contributes to pulmonary arterial smooth muscle cell (PASMC) proliferation and contraction. Here, we investigate the role of Notch signaling in HPV and hypoxia-induced enhancement of SOCE. We examined SOCE in human PASMCs exposed to hypoxia and pulmonary arterial pressure in mice using the isolated perfused/ventilated lung method. Wild-type and canonical transient receptor potential (TRPC) 6(-/-) mice were exposed to chronic hypoxia to induce HPH. Inhibition of Notch signaling with a γ-secretase inhibitor attenuates hypoxia-enhanced SOCE in PASMCs and hypoxia-induced increase in pulmonary arterial pressure. Our results demonstrate that hypoxia activates Notch signaling and up-regulates TRPC6 channels. Additionally, treatment with a Notch ligand can mimic hypoxic responses. Finally, inhibition of TRPC6, either pharmacologically or genetically, attenuates HPV, hypoxia-enhanced SOCE, and the development of HPH. These results demonstrate that hypoxia-induced activation of Notch signaling mediates HPV and the development of HPH via functional activation and up-regulation of TRPC6 channels. Understanding the molecular mechanisms that regulate cytosolic free Ca(2+) concentration and PASMC proliferation is critical to elucidation of the pathogenesis of HPH. Targeting Notch regulation of TRPC6 will be beneficial in the development of novel therapies for pulmonary hypertension associated with hypoxia.


Asunto(s)
Señalización del Calcio , Hipertensión Pulmonar/metabolismo , Receptor Notch1/metabolismo , Vasoconstricción , Animales , Proteínas de Unión al Calcio/metabolismo , Hipoxia de la Célula , Células Cultivadas , Humanos , Hipertensión Pulmonar/fisiopatología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiopatología , Miocitos del Músculo Liso/metabolismo , Arteria Pulmonar/metabolismo , Arteria Pulmonar/fisiopatología , Proteínas Serrate-Jagged , Canales Catiónicos TRPC/antagonistas & inhibidores , Canales Catiónicos TRPC/genética , Canales Catiónicos TRPC/metabolismo , Canal Catiónico TRPC6
6.
Am J Physiol Cell Physiol ; 308(8): C581-93, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25673771

RESUMEN

Pulmonary arterial hypertension (PAH) is a progressive disease that, if left untreated, eventually leads to right heart failure and death. Elevated pulmonary arterial pressure (PAP) in patients with PAH is mainly caused by an increase in pulmonary vascular resistance (PVR). Sustained vasoconstriction and excessive pulmonary vascular remodeling are two major causes for elevated PVR in patients with PAH. Excessive pulmonary vascular remodeling is mediated by increased proliferation of pulmonary arterial smooth muscle cells (PASMC) due to PASMC dedifferentiation from a contractile or quiescent phenotype to a proliferative or synthetic phenotype. Increased cytosolic Ca(2+) concentration ([Ca(2+)]cyt) in PASMC is a key stimulus for cell proliferation and this phenotypic transition. Voltage-dependent Ca(2+) entry (VDCE) and store-operated Ca(2+) entry (SOCE) are important mechanisms for controlling [Ca(2+)]cyt. Stromal interacting molecule proteins (e.g., STIM2) and Orai2 both contribute to SOCE and we have previously shown that STIM2 and Orai2, specifically, are upregulated in PASMC from patients with idiopathic PAH and from animals with experimental pulmonary hypertension in comparison to normal controls. In this study, we show that STIM2 and Orai2 are upregulated in proliferating PASMC compared with contractile phenotype of PASMC. Additionally, a switch in Ca(2+) regulation is observed in correlation with a phenotypic transition from contractile PASMC to proliferative PASMC. PASMC in a contractile phenotype or state have increased VDCE, while in the proliferative phenotype or state PASMC have increased SOCE. The data from this study indicate that upregulation of STIM2 and Orai2 is involved in the phenotypic transition of PASMC from a contractile state to a proliferative state; the enhanced SOCE due to upregulation of STIM2 and Orai2 plays an important role in PASMC proliferation.


Asunto(s)
Canales de Calcio/biosíntesis , Hipertensión Pulmonar/metabolismo , Glicoproteínas de Membrana/biosíntesis , Miocitos del Músculo Liso/citología , Canales Catiónicos TRPC/biosíntesis , Remodelación Vascular/fisiología , Animales , Calcio/metabolismo , Calcio/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio/genética , Canales de Calcio Tipo L/metabolismo , Señalización del Calcio/fisiología , Desdiferenciación Celular , Proliferación Celular , Células Cultivadas , Masculino , Ratones , Ratones Endogámicos C57BL , Contracción Muscular/fisiología , Músculo Liso Vascular/citología , Nifedipino/farmacología , Proteína ORAI2 , Arteria Pulmonar/citología , Interferencia de ARN , ARN Interferente Pequeño , Ratas , Ratas Sprague-Dawley , Molécula de Interacción Estromal 2 , Canales Catiónicos TRPC/genética , Canal Catiónico TRPC6 , Factor de Crecimiento Transformador beta/farmacología , Resistencia Vascular , Vasoconstricción
7.
Blood ; 119(1): 308-19, 2012 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-22049513

RESUMEN

Increased endothelial permeability contributes to the morbidity and mortality associated with chronic inflammatory diseases, including acute lung injury. Cyclic AMP response element-binding protein (CREB) transcriptional factor induces genes that regulate inflammation and vascular remodeling. However, the role of CREB in regulating endothelial barrier function is unknown. Here, we demonstrate that CREB maintains basal endothelial barrier function and suppresses endothelial permeability increase by diverse agonists such as thrombin, lipopolysaccharide, histamine, and VEGF. We show that CREB transcriptionally controls the expression of p190RhoGAP-A, a GTPase-activating protein that inhibits small GTPase RhoA. Impairing CREB function using small interfering RNA or dominant-negative (dn)-CREB mutant (dn-CREB) markedly suppressed p190RhoGAP-A expression, increased RhoA activity, induced actin stress fiber formation, and produced an amplified and protracted increase in endothelial permeability in response to thrombin. Rescuing p190RhoGAP-A expression restored the permeability defect in dn-CREB-transducing endothelial cells. These findings were recapitulated in vivo because dn-CREB expression in mice vasculature increased basal lung microvessel permeability and exaggerated permeability increase induced by thrombin and lipopolysaccharide. Inhibiting RhoA signaling restored endothelial barrier dysfunction in the dn-CREB-expressing lung microvasculature. These results uncover a pivotal role of CREB in regulating endothelial barrier function by restricting RhoA signaling through controlling p190RhoGAP-A expression.


Asunto(s)
Permeabilidad Capilar/fisiología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Endotelio Vascular/metabolismo , Regulación de la Expresión Génica , Factores de Intercambio de Guanina Nucleótido/genética , Regiones Promotoras Genéticas/genética , Proteínas Represoras/genética , Animales , Western Blotting , Permeabilidad Capilar/efectos de los fármacos , Células Cultivadas , Inmunoprecipitación de Cromatina , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/antagonistas & inhibidores , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Ensayo de Cambio de Movilidad Electroforética , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Citometría de Flujo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Hemostáticos/farmacología , Histamina/metabolismo , Humanos , Lipopolisacáridos/farmacología , Luciferasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Microvasos/efectos de los fármacos , Arteria Pulmonar/citología , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/metabolismo , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Represoras/metabolismo , Transducción de Señal , Trombina/farmacología
8.
Circ Res ; 111(6): 739-49, 2012 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-22798526

RESUMEN

RATIONALE: Adherens junctions (AJs) are the primary intercellular junctions in microvessels responsible for endothelial barrier function. Homophilic adhesion of vascular endothelial (VE) cadherin forms AJs, which are stabilized by binding of p120-catenin (p120). p120 dissociation from VE-cadherin results in loss of VE-cadherin homotypic interaction and AJ disassembly; however, the signaling mechanisms regulating p120 dissociation from VE-cadherin are not understood. OBJECTIVE: To address the mechanism of protein kinase C (PKC)-α function in increasing endothelial permeability, we determined the role of PKCα phosphorylation of p120 in mediating disruption of AJ integrity. METHODS AND RESULTS: We showed that PKCα phosphorylation of p120 at serine (S)879 in response to thrombin or lipopolysaccharide challenge reduced p120 binding affinity for VE-cadherin and mediated AJ disassembly secondary to VE-cadherin internalization. In studies in mouse lung vessels, expression of the phosphodeficient S879A-p120 mutant prevented the increase in vascular permeability induced by activation of the thrombin receptor PAR-1. CONCLUSIONS: PKCα phosphorylation of p120 at S879 is a critical phospho-switch mediating disassociation of p120 from VE-cadherin that results in AJ disassembly. Therefore, blocking PKCα-mediated p120 phosphorylation represents a novel targeted anti-inflammatory strategy to prevent disruption of vascular endothelial barrier function.


Asunto(s)
Uniones Adherentes/metabolismo , Antígenos CD/metabolismo , Cadherinas/metabolismo , Cateninas/metabolismo , Proteína Quinasa C-alfa/metabolismo , Uniones Adherentes/efectos de los fármacos , Animales , Permeabilidad Capilar/efectos de los fármacos , Permeabilidad Capilar/fisiología , Cateninas/genética , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Humanos , Immunoblotting , Técnicas In Vitro , Lipopolisacáridos/farmacología , Pulmón/irrigación sanguínea , Pulmón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microvasos/metabolismo , Microvasos/fisiología , Mutación , Fosforilación , Unión Proteica/efectos de los fármacos , Proteína Quinasa C-alfa/genética , Interferencia de ARN , Receptor PAR-1/metabolismo , Serina/genética , Serina/metabolismo , Trombina/farmacología , Catenina delta
9.
Cell Rep ; 43(6): 114297, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38824643

RESUMEN

The mechanical environment generated through the adhesive interaction of endothelial cells (ECs) with the matrix controls nuclear tension, preventing aberrant gene synthesis and the transition from restrictive to leaky endothelium, a hallmark of acute lung injury (ALI). However, the mechanisms controlling tension transmission to the nucleus and EC-restrictive fate remain elusive. Here, we demonstrate that, in a kinase-independent manner, focal adhesion kinase (FAK) safeguards tension transmission to the nucleus to maintain EC-restrictive fate. In FAK-depleted ECs, robust activation of the RhoA-Rho-kinase pathway increased EC tension and phosphorylation of the nuclear envelope protein, emerin, activating DNMT3a. Activated DNMT3a methylates the KLF2 promoter, impairing the synthesis of KLF2 and its target S1PR1 to induce the leaky EC transcriptome. Repleting FAK (wild type or kinase dead) or inhibiting RhoA-emerin-DNMT3a activities in damaged lung ECs restored KLF2 transcription of the restrictive EC transcriptome. Thus, FAK sensing and control of tension transmission to the nucleus govern restrictive endothelium to maintain lung homeostasis.


Asunto(s)
Núcleo Celular , Células Endoteliales , Factores de Transcripción de Tipo Kruppel , Transcriptoma , Proteína de Unión al GTP rhoA , Humanos , Núcleo Celular/metabolismo , Transcriptoma/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Células Endoteliales/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Proteína de Unión al GTP rhoA/genética , ADN Metiltransferasa 3A , Quinasa 1 de Adhesión Focal/metabolismo , Quinasa 1 de Adhesión Focal/genética , Animales , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Ratones , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Quinasas Asociadas a rho/metabolismo , Quinasas Asociadas a rho/genética , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/genética , Regiones Promotoras Genéticas/genética , Fosforilación
10.
Am J Physiol Lung Cell Mol Physiol ; 305(4): L291-300, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23771883

RESUMEN

Loss of lung-fluid homeostasis is the hallmark of acute lung injury (ALI). Association of catenins and actin cytoskeleton with vascular endothelial (VE)-cadherin is generally considered the main mechanism for stabilizing adherens junctions (AJs), thereby preventing disruption of lung vascular barrier function. The present study identifies endothelial focal adhesion kinase (FAK), a nonreceptor tyrosine kinase that canonically regulates focal adhesion turnover, as a novel AJ-stabilizing mechanism. In wild-type mice, induction of ALI by intraperitoneal administration of lipopolysaccharide or cecal ligation and puncture markedly decreased FAK expression in lungs. Using a mouse model in which FAK was conditionally deleted only in endothelial cells (ECs), we show that loss of EC-FAK mimicked key features of ALI (diffuse lung hemorrhage, increased transvascular albumin influx, edema, and neutrophil accumulation in the lung). EC-FAK deletion disrupted AJs due to impairment of the fine balance between the activities of RhoA and Rac1 GTPases. Deletion of EC-FAK facilitated RhoA's interaction with p115-RhoA guanine exchange factor, leading to activation of RhoA. Activated RhoA antagonized Rac1 activity, destabilizing AJs. Inhibition of Rho kinase, a downstream effector of RhoA, reinstated normal endothelial barrier function in FAK-/- ECs and lung vascular integrity in EC-FAK-/- mice. Our findings demonstrate that EC-FAK plays an essential role in maintaining AJs and thereby lung vascular barrier function by establishing the normal balance between RhoA and Rac1 activities.


Asunto(s)
Endotelio/enzimología , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Eliminación de Gen , Pulmón/irrigación sanguínea , Pulmón/fisiopatología , Neuropéptidos/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Animales , Líquidos Corporales/metabolismo , Células Endoteliales/enzimología , Células Endoteliales/patología , Endotelio/patología , Endotelio/fisiopatología , Estabilidad de Enzimas , Proteína-Tirosina Quinasas de Adhesión Focal/deficiencia , Humanos , Integrasas/metabolismo , Pulmón/enzimología , Pulmón/patología , Lesión Pulmonar/enzimología , Lesión Pulmonar/patología , Lesión Pulmonar/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Especificidad de Órganos
11.
Expert Opin Ther Pat ; 31(11): 1059-1074, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34082658

RESUMEN

INTRODUCTION: Mucormycosis is an uncommon but life-threatening infection with nonspecific clinical manifestations that make its diagnosis/treatment difficult. The current literature indicates that mucormycosis case incidences are on the rise in developing and developed countries, and, unfortunately, there are only a few treatments available. Accordingly, it is essential to provide more treatment options for mucormycosis. AREA COVERED: This patent review focuses on the granted patents and patent applications related to medication for mucormycosis treatment from the publication year of the amphotericin-B patent application (1958) till 30 January 2021. EXPERT OPINION: Mucormycosis has few available treatments, including amphotericin-B, isavuconazonium sulfate, posaconazole, or their combination. A few anti-mucormycosis medicines are under clinical development. The exact burden of mucormycosis is unknown, but it is expected to be higher than the reported cases because of mucormycosis epidemiological changes. This patent review has shown that scientists are progressing toward developing a new treatment for mucormycosis in the form of new chemical compounds, new drug combinations, and dosage forms, vaccines, plant products, drug repurposing, and derivatives of the biomolecules. This progress is encouraging to fight this devastating illness.


Asunto(s)
Antifúngicos/farmacología , Desarrollo de Medicamentos , Mucormicosis/tratamiento farmacológico , Animales , Reposicionamiento de Medicamentos , Humanos , Mucormicosis/epidemiología , Mucormicosis/microbiología , Patentes como Asunto
12.
Pharmaceuticals (Basel) ; 14(8)2021 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-34451807

RESUMEN

Protein kinase inhibitors (PKIs) are important therapeutic agents. As of 31 May 2021, the United States Food and Drug Administration (USFDA) has approved 70 PKIs. Most of the PKIs are employed to treat cancer and inflammatory diseases. Imatinib was the first PKI approved by USFDA in 2001. This review summarizes the compound patents and the essential polymorph patents of the PKIs approved by the USFDA from 2001 to 31 May 2021. The dates on the generic drug availability of the PKIs in the USA market have also been forecasted. It is expected that 19 and 48 PKIs will be genericized by 2025 and 2030, respectively, due to their compound patent expiry. This may reduce the financial toxicity associated with the existing PKIs. There are nearly 535 reported PKs. However, the USFDA approved PKIs target only about 10-15% of the total said PKs. As a result, there are still a large number of unexplored PKs. As the field advances during the next 20 years, one can anticipate that PKIs with many scaffolds, chemotypes, and pharmacophores will be developed.

13.
Circ Res ; 103(10): 1164-72, 2008 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-18849324

RESUMEN

The lipid mediator sphingosine-1-phosphate (S1P), the product of sphingosine kinase (SPHK)-induced phosphorylation of sphingosine, is known to stabilize interendothelial junctions and prevent microvessel leakiness. Here, we investigated the role of SPHK1 activation in regulating the increase in pulmonary microvessel permeability induced by challenge of mice with lipopolysaccharide or thrombin ligation of protease-activating receptor (PAR)-1. Both lipopolysaccharide and thrombin increased mouse lung microvascular permeability and resulted in a delayed activation of SPHK1 that was coupled to the onset of restoration of permeability. In contrast to wild-type mice, Sphk1(-/-) mice showed markedly enhanced pulmonary edema formation in response to lipopolysaccharide and PAR-1 activation. Using endothelial cells challenged with thrombin concentration (50 nmol/L) that elicited a transient but reversible increase in endothelial permeability, we observed that increased SPHK1 activity and decreased intracellular S1P concentration preceded the onset of barrier recovery. Thus, we tested the hypothesis that released S1P in a paracrine manner activates its receptor S1P1 to restore the endothelial barrier. Knockdown of SPHK1 decreased basal S1P production and Rac1 activity but increased basal endothelial permeability. In SPHK1-depleted cells, PAR-1 activation failed to induce Rac1 activation but augmented RhoA activation and endothelial hyperpermeability response. Knockdown of S1P1 receptor in endothelial cells also enhanced the increase in endothelial permeability following PAR-1 activation. S1P treatment of Sphk1(-/-) lungs or SPHK1-deficient endothelial cells restored endothelial barrier function. Our results suggest the crucial role of activation of the SPHK1-->S1P-->S1P1 signaling pathway in response to inflammatory mediators in endothelial cells in regulating endothelial barrier homeostasis.


Asunto(s)
Permeabilidad Capilar , Mediadores de Inflamación/metabolismo , Uniones Intercelulares/metabolismo , Pulmón/metabolismo , Lisofosfolípidos/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/análogos & derivados , Animales , Permeabilidad Capilar/efectos de los fármacos , Permeabilidad Capilar/genética , Células Cultivadas , Activación Enzimática/efectos de los fármacos , Activación Enzimática/genética , Hemostáticos/efectos adversos , Hemostáticos/farmacología , Humanos , Mediadores de Inflamación/farmacología , Uniones Intercelulares/genética , Lipopolisacáridos/toxicidad , Lisofosfolípidos/genética , Lisofosfolípidos/farmacología , Ratones , Ratones Noqueados , Neuropéptidos/genética , Neuropéptidos/metabolismo , Comunicación Paracrina/efectos de los fármacos , Comunicación Paracrina/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Edema Pulmonar/inducido químicamente , Edema Pulmonar/genética , Edema Pulmonar/metabolismo , Receptor PAR-1/genética , Receptor PAR-1/metabolismo , Receptores de Lisoesfingolípidos/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Esfingosina/genética , Esfingosina/metabolismo , Esfingosina/farmacología , Receptores de Esfingosina-1-Fosfato , Trombina/efectos adversos , Trombina/farmacología , Proteínas de Unión al GTP rac/genética , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rac1/metabolismo
14.
Cell Rep ; 30(12): 4096-4109.e5, 2020 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-32209471

RESUMEN

Acute lung injury (ALI) is a lethal inflammatory lung disorder whose incidence is on the rise. Alveolar macrophages normally act to resolve inflammation, but when dysregulated they can provoke ALI. We demonstrate that monocyte-derived macrophages (CD11b+ macrophages) recruited into the airspace upregulate the anti-inflammatory function of alveolar macrophages by suppressing their stimulator of type 1 interferon gene (STING) signaling. Depletion of CD11b+ macrophages in mice (macrophagedep mice) after endotoxin or after Pseudomonas aeruginosa causes expansion of the inflammatory alveolar macrophage population, leading to neutrophil accumulation, irreversible loss of lung vascular barrier function, and lethality. We show that CD11b+ macrophages suppress alveolar macrophage-STING signaling via sphingosine kinase-2 (SPHK2) generation of sphingosine-1-phosphate (S1P). Thus, adoptive transfer of wild-type (WT) or STING-/-, but not SPHK2-/-, CD11b monocytes from murine bone marrow into injured macrophagedep mice rescue anti-inflammatory alveolar macrophages and reverse lung vascular injury. SPHK2-induced S1P generation in CD11b+ macrophages has the potential to educate alveolar macrophages to resolve ALI.


Asunto(s)
Antígeno CD11b/metabolismo , Inflamación/patología , Lisofosfolípidos/metabolismo , Macrófagos Alveolares/metabolismo , Proteínas de la Membrana/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Esfingosina/análogos & derivados , Traslado Adoptivo , Animales , Citocinas/metabolismo , Humanos , Mediadores de Inflamación/metabolismo , Pulmón/irrigación sanguínea , Pulmón/patología , Macrófagos Alveolares/microbiología , Ratones Endogámicos C57BL , Nucleótidos Cíclicos/metabolismo , Pseudomonas aeruginosa/fisiología , Transducción de Señal , Esfingosina/metabolismo , Células U937
15.
Cell Rep ; 27(3): 793-805.e4, 2019 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-30995477

RESUMEN

Alveolar macrophages (AMs), upon sensing pathogens, trigger host defense by activating toll-like receptor 4 (TLR4), but the counterbalancing mechanisms that deactivate AM inflammatory signaling and prevent lethal edema, the hallmark of acute lung injury (ALI), remain unknown. Here, we demonstrate the essential role of AM protease-activating receptor 2 (PAR2) in rapidly suppressing inflammation to prevent long-lasting injury. We show that thrombin, released during TLR4-induced lung injury, directly activates PAR2 to generate cAMP, which abolishes Ca2+ entry through the TRPV4 channel. Deletion of PAR2 and thus the accompanying cAMP generation augments Ca2+ entry via TRPV4, causing sustained activation of the transcription factor NFAT to produce long-lasting TLR4-mediated inflammatory lung injury. Rescuing thrombin-sensitive PAR2 expression or blocking TRPV4 activity in PAR2-null AMs restores their capacity to resolve inflammation and reverse lung injury. Thus, activation of the thrombin-induced PAR2-cAMP cascade in AMs suppresses TLR4 inflammatory signaling to reinstate tissue integrity.


Asunto(s)
Señalización del Calcio , AMP Cíclico/metabolismo , Inflamación/prevención & control , Macrófagos Alveolares/metabolismo , Receptor PAR-2/metabolismo , Canales Catiónicos TRPV/metabolismo , Receptor Toll-Like 4/metabolismo , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/patología , Animales , Calcio/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Inflamación/metabolismo , Lipopolisacáridos/toxicidad , Macrófagos Alveolares/citología , Macrófagos Alveolares/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Transcripción NFATC/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Receptor PAR-2/deficiencia , Receptor PAR-2/genética , Canales Catiónicos TRPV/antagonistas & inhibidores , Canales Catiónicos TRPV/genética , Trombina/metabolismo
16.
J Pharm Pharmacol ; 60(5): 615-23, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18416938

RESUMEN

The aims of this investigation were to determine the distribution in the gastrointestinal (GI) tract of Eudragit S-100 encapsulated colon-specific sodium alginate microspheres containing 5-fluorouracil (5-FU) in rats, and to perform pharmacokinetic and pharmacodynamic studies. Comparisons were with a control immediate-release (IR) formulation of 5-FU. 5-FU was distributed predominantly in the upper GI tract from the IR formulation but was distributed primarily to the lower part of the GI tract from the microsphere formulation. No drug was released in the stomach and intestinal regions from the colon-specific microspheres. Significantly, a high concentration of the active drug was achieved in colonic tissues from the colon-specific microspheres (P < 0.001), which was higher than the IC50 required to halt the growth of and/or kill colon cancer cells. Colon cancer was induced in rats by subcutaneous injection of 1,2-dimethylhydrazine (40 mg kg (-1)) for 10 weeks. The tumours induced were non-invasive adenocarcinomas and were in Duke's stage A. The 5-FU formulations were administered for 4 weeks after tumour induction. Non-significant reductions in tumour volume and multiplicity were observed in animals given the colon-specific microspheres. Enhanced levels of liver enzymes (SGOT, SGPT and alkaline phosphatase) were found in animals given the IR formulation of 5-FU, and values differed significantly (P < 0.001) from those in animals treated with the colon-specific microspheres. Elevated levels of serum albumin and creatinine, and leucocytopenia and thrombocytopenia were observed in the animals given the IR formulation. In summary, Eudragit S-100 coated alginate microspheres delivered 5-FU to colonic tissues, with reduced systemic side-effects. A long-term dosing study is required to ascertain the therapeutic benefits.


Asunto(s)
Alginatos/administración & dosificación , Antimetabolitos Antineoplásicos/administración & dosificación , Colon/metabolismo , Neoplasias del Colon/metabolismo , Fluorouracilo/administración & dosificación , Ácidos Polimetacrílicos/administración & dosificación , 1,2-Dimetilhidrazina , Alginatos/farmacocinética , Animales , Antimetabolitos Antineoplásicos/sangre , Antimetabolitos Antineoplásicos/farmacocinética , Ciego/metabolismo , Colon/efectos de los fármacos , Colon/patología , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/patología , Fluorouracilo/sangre , Fluorouracilo/farmacocinética , Mucosa Gástrica/metabolismo , Ácido Glucurónico/administración & dosificación , Ácido Glucurónico/farmacocinética , Ácidos Hexurónicos/administración & dosificación , Ácidos Hexurónicos/farmacocinética , Intestino Delgado/metabolismo , Masculino , Microesferas , Ácidos Polimetacrílicos/farmacocinética , Ratas , Ratas Wistar , Carga Tumoral
17.
Int J Biol Macromol ; 119: 23-31, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30053390

RESUMEN

Venom peptides anoplin, cupiennin 1a, latarcin 1, latarcin 3a, latarcin 5, melittin, and pandinin 2 are known to have antibacterial properties. In the current study, we examined whether the antimicrobial properties of these venom peptides have any connection to the binding and inhibition of bacterial ATP synthase. Venom peptides inhibited Escherichia coli wild type and mutant membrane-bound F1Fo ATP synthase to varying degrees. Although significant loss of oxidative phosphorylation was observed for wild type, very little loss occurred for null and mutant E. coli strains in the presence of venom peptides. This study also reaffirms that ßDELSEED-motif residues of ATP synthase are required for peptide binding. Modified venom peptides with C-terminal amide (NH2) groups caused augmented inhibition of ATP synthase and E. coli cell death. Growth patterns of wild type, null, and mutant strains in the presence of melittin, anoplin, cupiennin 1a, latarcin 1, latarcin 3a, latarcin 5, pandinin 2, and their modified variants suggested the possibility of additional molecular targets. Our results demonstrate that the antimicrobial properties of venom peptides are connected to the binding and inhibition of bacterial ATP synthase. Moreover, selective inhibition of ATP synthase by venom peptides suggests a viable alternative to combat antibiotic-resistant microbial infections.


Asunto(s)
Adenosina Trifosfatasas/antagonistas & inhibidores , Adenosina Trifosfatasas/química , Péptidos Catiónicos Antimicrobianos/química , Péptidos Catiónicos Antimicrobianos/farmacología , Ponzoñas/química , Ponzoñas/farmacología , Adenosina Trifosfatasas/genética , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Escherichia coli/enzimología , Modelos Moleculares , Conformación Molecular , Mutación , Unión Proteica , Relación Estructura-Actividad
18.
Eur J Pharmacol ; 556(1-3): 136-43, 2007 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-17187777

RESUMEN

Besides its well-known effects on platelet aggregation, aspirin has been suggested to be an antioxidant and is also known to improve the lipid profile. In the present study we tested the hypothesis that aspirin by its antioxidant effect, improves haemodynamic profile and baroreflex sensitivity in rat model of hypercholesterolemia. Hypercholesterolemia was induced in Wistar rats by feeding 1% cholesterol rich diet for 10 weeks. Lipid profile, lipid peroxidation and reduced glutathione were estimated in serum. Haemodynamic changes and baroreflex were measured in anaesthetized rats. Hypercholesterolemic rats showed significant increase in total cholesterol, low-density lipoprotein-cholesterol (LDL-C), very low-density lipoprotein-cholesterol (VLDL-C) and atherogenic index and significant decrease in high-density lipoprotein-cholesterol (HDL-C). Significant rise in blood pressure, heart rate and attenuation of baroreflex sensitivity were also found in hypercholesterolemic rat. Aspirin in the dose of 100 mg/kg showed significant decrease in total cholesterol, LDL-C, VLDL-C and atherogenic index and significant increase in HDL-C. Aspirin treatment prevented the rise in blood pressure, heart rate and significantly improved baroreflex sensitivity in hypercholesterolemic rats. Hypercholesterolemic rats showed free radical generation, evident by a significant increase in serum lipid peroxidation and significant reduction in serum reduced glutathione content. Aspirin treatment significantly decreased lipid peroxidation and significantly increased reduced glutathione content. We have demonstrated that aspirin improves baroreflex response and prevents the rise in blood pressure and heart rate possibly by reducing sympathetic activity due to its antioxidant effect in experimentally induced hypercholesterolemic rats.


Asunto(s)
Antioxidantes/uso terapéutico , Aspirina/uso terapéutico , Barorreflejo/efectos de los fármacos , Hipercolesterolemia/fisiopatología , Animales , Presión Sanguínea/efectos de los fármacos , Colesterol/sangre , Colesterol en la Dieta , Frecuencia Cardíaca/efectos de los fármacos , Hipercolesterolemia/tratamiento farmacológico , Peroxidación de Lípido/efectos de los fármacos , Lípidos/sangre , Masculino , Ratas , Ratas Wistar
19.
Sci Rep ; 7: 42758, 2017 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-28218251

RESUMEN

Store-operated Ca2+ entry (SOCE) mediates the increase in intracellular calcium (Ca2+) in endothelial cells (ECs) that regulates several EC functions including tissue-fluid homeostasis. Stromal-interaction molecule 1 (STIM1), upon sensing the depletion of (Ca2+) from the endoplasmic reticulum (ER) store, organizes as puncta that trigger store-operated Ca2+ entry (SOCE) via plasmalemmal Ca2+-selective Orai1 channels. While the STIM1 and Orai1 binding interfaces have been mapped, signaling mechanisms activating STIM1 recruitment of Orai1 and STIM1-Orai1 interaction remains enigmatic. Here, we show that ER Ca2+-store depletion rapidly induces STIM1 phosphorylation at Y361 via proline-rich kinase 2 (Pyk2) in ECs. Surprisingly, the phospho-defective STIM1-Y361F mutant formed puncta but failed to recruit Orai1, thereby preventing. SOCE Furthermore, studies in mouse lungs, expression of phosphodefective STIM1-Y361F mutant in ECs prevented the increase in vascular permeability induced by the thrombin receptor, protease activated receptor 1 (PAR1). Hence, Pyk2-dependent phosphorylation of STIM1 at Y361 is a critical phospho-switch enabling recruitment of Orai1 into STIM1 puncta leading to SOCE. Therefore, Y361 in STIM1 represents a novel target for limiting SOCE-associated vascular leak.


Asunto(s)
Calcio/metabolismo , Quinasa 2 de Adhesión Focal/metabolismo , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Proteína ORAI1/metabolismo , Molécula de Interacción Estromal 1/química , Molécula de Interacción Estromal 1/metabolismo , Tirosina/metabolismo , Animales , Sitios de Unión , Células Cultivadas , Retículo Endoplásmico/metabolismo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Humanos , Ratones , Mutación , Proteínas de Neoplasias/genética , Fosforilación , Unión Proteica , Molécula de Interacción Estromal 1/genética
20.
J Exp Med ; 212(11): 1883-99, 2015 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-26392222

RESUMEN

Leukocyte transendothelial migration (TEM) is a tightly regulated, multistep process that is critical to the inflammatory response. A transient increase in endothelial cytosolic free calcium ion concentration (↑[Ca(2+)]i) is required for TEM. However, the mechanism by which endothelial ↑[Ca(2+)]i regulates TEM and the channels mediating this ↑[Ca(2+)]i are unknown. Buffering ↑[Ca(2+)]i in endothelial cells does not affect leukocyte adhesion or locomotion but selectively blocks TEM, suggesting a role for ↑[Ca(2+)]i specifically for this step. Transient receptor potential canonical 6 (TRPC6), a Ca(2+) channel expressed in endothelial cells, colocalizes with platelet/endothelial cell adhesion molecule-1 (PECAM) to surround leukocytes during TEM and clusters when endothelial PECAM is engaged. Expression of dominant-negative TRPC6 or shRNA knockdown in endothelial cells arrests neutrophils apically over the junction, similar to when PECAM is blocked. Selectively activating endothelial TRPC6 rescues TEM during an ongoing PECAM blockade, indicating that TRPC6 functions downstream of PECAM. Furthermore, endothelial TRPC6 is required for trafficking of lateral border recycling compartment membrane, which facilitates TEM. Finally, mice lacking TRPC6 in the nonmyeloid compartment (i.e., endothelium) exhibit a profound defect in neutrophil TEM with no effect on leukocyte trafficking. Our findings identify endothelial TRPC6 as the calcium channel mediating the ↑[Ca(2+)]i required for TEM at a step downstream of PECAM homophilic interactions.


Asunto(s)
Movimiento Celular , Células Endoteliales/fisiología , Leucocitos/fisiología , Canales Catiónicos TRPC/fisiología , Animales , Calcio/metabolismo , Citosol/metabolismo , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Humanos , Inflamación/metabolismo , Ratones , Ratones Endogámicos C57BL , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/fisiología , Canal Catiónico TRPC6
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA