Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 118
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Respir Cell Mol Biol ; 69(6): 678-688, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37639326

RESUMEN

Acute lung injury (ALI) is characterized by lung vascular endothelial cell (EC) barrier compromise resulting in increased endothelial permeability and pulmonary edema. The infection of gram-negative bacteria that produce toxins like LPS is one of the major causes of ALI. LPS activates Toll-like receptor 4, leading to cytoskeleton reorganization, resulting in lung endothelial barrier disruption and pulmonary edema in ALI. However, the signaling pathways that lead to the cytoskeleton reorganization and lung microvascular EC barrier disruption remain largely unexplored. Here we show that LPS induces calpain activation and talin cleavage into head and rod domains and that inhibition of calpain attenuates talin cleavage, RhoA activation, and pulmonary EC barrier disruption in LPS-treated human lung microvascular ECs in vitro and lung EC barrier disruption and pulmonary edema induced by LPS in ALI in vivo. Moreover, overexpression of calpain causes talin cleavage and RhoA activation, myosin light chain (MLC) phosphorylation, and increases in actin stress fiber formation. Furthermore, knockdown of talin attenuates LPS-induced RhoA activation and MLC phosphorylation and increased stress fiber formation and mitigates LPS-induced lung microvascular endothelial barrier disruption. Additionally, overexpression of talin head and rod domains increases RhoA activation, MLC phosphorylation, and stress fiber formation and enhances lung endothelial barrier disruption. Finally, overexpression of cleavage-resistant talin mutant reduces LPS-induced increases in MLC phosphorylation in human lung microvascular ECs and attenuates LPS-induced lung microvascular endothelial barrier disruption. These results provide the first evidence that calpain mediates LPS-induced lung microvascular endothelial barrier disruption in ALI via cleavage of talin.


Asunto(s)
Lesión Pulmonar Aguda , Edema Pulmonar , Humanos , Lipopolisacáridos/farmacología , Calpaína/metabolismo , Talina/metabolismo , Pulmón/metabolismo , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/metabolismo , Cadenas Ligeras de Miosina/metabolismo , Permeabilidad Capilar
2.
Am J Respir Cell Mol Biol ; 66(2): e1-e14, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35103557

RESUMEN

Advancements in methods, technology, and our understanding of the pathobiology of lung injury have created the need to update the definition of experimental acute lung injury (ALI). We queried 50 participants with expertise in ALI and acute respiratory distress syndrome using a Delphi method composed of a series of electronic surveys and a virtual workshop. We propose that ALI presents as a "multidimensional entity" characterized by four "domains" that reflect the key pathophysiologic features and underlying biology of human acute respiratory distress syndrome. These domains are 1) histological evidence of tissue injury, 2) alteration of the alveolar-capillary barrier, 3) presence of an inflammatory response, and 4) physiologic dysfunction. For each domain, we present "relevant measurements," defined as those proposed by at least 30% of respondents. We propose that experimental ALI encompasses a continuum of models ranging from those focusing on gaining specific mechanistic insights to those primarily concerned with preclinical testing of novel therapeutics or interventions. We suggest that mechanistic studies may justifiably focus on a single domain of lung injury, but models must document alterations of at least three of the four domains to qualify as "experimental ALI." Finally, we propose that a time criterion defining "acute" in ALI remains relevant, but the actual time may vary based on the specific model and the aspect of injury being modeled. The continuum concept of ALI increases the flexibility and applicability of the definition to multiple models while increasing the likelihood of translating preclinical findings to critically ill patients.


Asunto(s)
Lesión Pulmonar Aguda/patología , Inflamación/fisiopatología , Informe de Investigación/tendencias , Lesión Pulmonar Aguda/inmunología , Animales
3.
Proc Natl Acad Sci U S A ; 116(27): 13394-13403, 2019 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-31213542

RESUMEN

Increased glycolysis in the lung vasculature has been connected to the development of pulmonary hypertension (PH). We therefore investigated whether glycolytic regulator 6-phosphofructo-2-kinase/fructose-2, 6-bisphosphatase (PFKFB3)-mediated endothelial glycolysis plays a critical role in the development of PH. Heterozygous global deficiency of Pfkfb3 protected mice from developing hypoxia-induced PH, and administration of the PFKFB3 inhibitor 3PO almost completely prevented PH in rats treated with Sugen 5416/hypoxia, indicating a causative role of PFKFB3 in the development of PH. Immunostaining of lung sections and Western blot with isolated lung endothelial cells showed a dramatic increase in PFKFB3 expression and activity in pulmonary endothelial cells of rodents and humans with PH. We generated mice that were constitutively or inducibly deficient in endothelial Pfkfb3 and found that these mice were incapable of developing PH or showed slowed PH progression. Compared with control mice, endothelial Pfkfb3-knockout mice exhibited less severity of vascular smooth muscle cell proliferation, endothelial inflammation, and leukocyte recruitment in the lungs. In the absence of PFKFB3, lung endothelial cells from rodents and humans with PH produced lower levels of growth factors (such as PDGFB and FGF2) and proinflammatory factors (such as CXCL12 and IL1ß). This is mechanistically linked to decreased levels of HIF2A in lung ECs following PFKFB3 knockdown. Taken together, these results suggest that targeting PFKFB3 is a promising strategy for the treatment of PH.


Asunto(s)
Glucólisis , Hipertensión Pulmonar/etiología , Pulmón/metabolismo , Fosfofructoquinasa-2/fisiología , Animales , Modelos Animales de Enfermedad , Endotelio/metabolismo , Técnicas de Silenciamiento del Gen , Glucólisis/fisiología , Humanos , Hipertensión Pulmonar/metabolismo , Hipoxia/complicaciones , Pulmón/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Fosfofructoquinasa-2/deficiencia , Fosfofructoquinasa-2/metabolismo , Ratas , Ratas Sprague-Dawley
4.
Kidney Int ; 95(6): 1359-1372, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30905471

RESUMEN

In mice, the initial stage of nephrotoxic serum-induced nephritis (NTN) mimics antibody-mediated human glomerulonephritis. Local immune deposits generate tumor necrosis factor (TNF), which activates pro-inflammatory pathways in glomerular endothelial cells (GECs) and podocytes. Because TNF receptors mediate antibacterial defense, existing anti-TNF therapies can promote infection; however, we have previously demonstrated that different functional domains of TNF may have opposing effects. The TIP peptide mimics the lectin-like domain of TNF, and has been shown to blunt inflammation in acute lung injury without impairing TNF receptor-mediated antibacterial activity. We evaluated the impact of TIP peptide in NTN. Intraperitoneal administration of TIP peptide reduced inflammation, proteinuria, and blood urea nitrogen. The protective effect was blocked by the cyclooxygenase inhibitor indomethacin, indicating involvement of prostaglandins. Targeted glomerular delivery of TIP peptide improved pathology in moderate NTN and reduced mortality in severe NTN, indicating a local protective effect. We show that TIP peptide activates the epithelial sodium channel(ENaC), which is expressed by GEC, upon binding to the channel's α subunit. In vitro, TNF treatment of GEC activated pro-inflammatory pathways and decreased the generation of prostaglandin E2 and nitric oxide, which promote recovery from NTN. TIP peptide counteracted these effects. Despite the capacity of TIP peptide to activate ENaC, it did not increase mean arterial blood pressure in mice. In the later autologous phase of NTN, TIP peptide blunted the infiltration of Th17 cells. By countering the deleterious effects of TNF through direct actions in GEC, TIP peptide could provide a novel strategy to treat glomerular inflammation.


Asunto(s)
Canales Epiteliales de Sodio/metabolismo , Glomerulonefritis/tratamiento farmacológico , Glomérulos Renales/efectos de los fármacos , Péptidos Cíclicos/administración & dosificación , Proteinuria/tratamiento farmacológico , Animales , Nitrógeno de la Urea Sanguínea , Línea Celular , Dinoprostona/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Femenino , Glomerulonefritis/sangre , Glomerulonefritis/inmunología , Glomerulonefritis/patología , Humanos , Inyecciones Intraperitoneales , Glomérulos Renales/citología , Glomérulos Renales/patología , Ratones , Óxido Nítrico/metabolismo , Técnicas de Placa-Clamp , Cultivo Primario de Células , Proteinuria/sangre , Proteinuria/inmunología , Proteinuria/patología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Células Th17/efectos de los fármacos , Células Th17/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
5.
Proc Natl Acad Sci U S A ; 113(7): 1895-900, 2016 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-26831066

RESUMEN

Dyslipidemia associated with triglyceride-rich lipoproteins (TRLs) represents an important residual risk factor for cardiovascular and chronic kidney disease in patients with type 1 diabetes (T1D). Levels of growth hormone (GH) are elevated in T1D, which aggravates both hyperglycemia and dyslipidemia. The hypothalamic growth hormone-releasing hormone (GHRH) regulates the release of GH by the pituitary but also exerts separate actions on peripheral GHRH receptors, the functional role of which remains elusive in T1D. In a rat model of streptozotocin (STZ)-induced T1D, GHRH receptor expression was found to be up-regulated in the distal small intestine, a tissue involved in chylomicron synthesis. Treatment of T1D rats with a GHRH antagonist, MIA-602, at a dose that did not affect plasma GH levels, significantly reduced TRL, as well as markers of renal injury, and improved endothelial-dependent vasorelaxation. Glucagon-like peptide 1 (GLP-1) reduces hyperglucagonemia and postprandial TRL, the latter in part through a decreased synthesis of apolipoprotein B-48 (ApoB-48) by intestinal cells. Although plasma GLP-1 levels were elevated in diabetic animals, this was accompanied by increased rather than reduced glucagon levels, suggesting impaired GLP-1 signaling. Treatment with MIA-602 normalized GLP-1 and glucagon to control levels in T1D rats. MIA-602 also decreased secretion of ApoB-48 from rat intestinal epithelial cells in response to oleic acid stimulation in vitro, in part through a GLP-1-dependent mechanism. Our findings support the hypothesis that antagonizing the signaling of GHRH in T1D may improve GLP-1 function in the small intestine, which, in turn, diminishes TRL and reduces renal and vascular complications.


Asunto(s)
Diabetes Mellitus Tipo 1/fisiopatología , Modelos Animales de Enfermedad , Dislipidemias/fisiopatología , Hormona Liberadora de Hormona del Crecimiento/fisiología , Animales , Dislipidemias/terapia , Hormona Liberadora de Hormona del Crecimiento/antagonistas & inhibidores , Intestino Delgado/metabolismo , Masculino , Ratas , Ratas Wistar , Receptores de Neuropéptido/metabolismo , Receptores de Hormona Reguladora de Hormona Hipofisaria/metabolismo , Estreptozocina
6.
Lab Invest ; 98(10): 1300-1310, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29572498

RESUMEN

High-fat meal (HFM) consumption can produce acute lipemia and trigger myocardial infarction in patients with atherosclerosis, but the mechanisms are poorly understood. Erythrocytes (red blood cells, RBCs) intimately interact with inflammatory cells and blood vessels and play a complex role in regulating vascular function. Chronic high-fat feeding in mice induces pathological RBC remodeling, suggesting a novel link between HFM, RBCs, and vascular dysfunction. However, whether acute HFM can induce RBC remodeling in humans is unknown. Ten healthy individuals were subjected to biochemical testing and assessment of endothelial-dependent flow-mediated dilation (FMD) before and after a single HFM or iso-caloric meal (ICM). Following the HFM, triglyceride, cholesterol, and free fatty acid levels were all significantly increased, in conjunction with impaired post-prandial FMD. Additionally, peripheral blood smears demonstrated microcytes, remodeled RBCs, and fatty monocytes. Increased intracellular ROS and nitration of protein band 3 was detected in RBCs following the HFM. The HFM elevated plasma and RBC-bound myeloperoxidase (MPO), which was associated with impaired FMD and oxidation of HDL. Monocytic cells exposed to lipid in vitro released MPO, while porcine coronary arteries exposed to fatty acids ex vivo took up MPO. We demonstrate in humans that a single HFM induces pathological RBC remodeling and concurrently elevates MPO, which can potentially enter the blood vessel wall to trigger oxidative stress and destabilize vulnerable plaques. These novel findings may have implications for the short-term risk of HFM consumption and alimentary lipemia in patients with atherosclerosis.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Endotelio Vascular/fisiología , Eritrocitos/fisiología , Adulto , Animales , Sedimentación Sanguínea , Vasos Coronarios/metabolismo , Humanos , Masculino , Peroxidasa/sangre , Porcinos , Adulto Joven
7.
Kidney Int ; 94(2): 280-291, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29731111

RESUMEN

To investigate the role of protein kinase C-α (PKC-α) in glomerulonephritis, the capacity of PKC-α inhibition to reverse the course of established nephrotoxic nephritis (NTN) was evaluated. Nephritis was induced by a single injection of nephrotoxic serum and after its onset, a PKC-α inhibitor was administered either systemically or by targeted glomerular delivery. By day seven, all mice with NTN had severe nephritis, whereas mice that received PKC-α inhibitors in either form had minimal evidence of disease. To further understand the underlying mechanism, label-free shotgun proteomic analysis of the kidney cortexes were performed, using quantitative mass spectrometry. Ingenuity pathway analysis revealed 157 differentially expressed proteins and mitochondrial dysfunction as the most modulated pathway. Functional protein groups most affected by NTN were mitochondrial proteins associated with respiratory processes. These proteins were down-regulated in the mice with NTN, while their expression was restored with PKC-α inhibition. This suggests a role for proteins that regulate oxidative phosphorylation in recovery. In cultured glomerular endothelial cells, nephrotoxic serum caused a decrease in mitochondrial respiration and membrane potential, mitochondrial morphologic changes and an increase in glycolytic lactic acid production; all normalized by PKC-α inhibition. Thus, PKC-α has a critical role in NTN progression, and the results implicate mitochondrial processes through restoring oxidative phosphorylation, as an essential mechanism underlying recovery. Importantly, our study provides additional support for targeted therapy to glomeruli to reverse the course of progressive disease.


Asunto(s)
Glomerulonefritis/tratamiento farmacológico , Proteína Quinasa C-alfa/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Animales , Anticuerpos Monoclonales/inmunología , Autoantígenos/inmunología , Colágeno Tipo IV/inmunología , Modelos Animales de Enfermedad , Sistemas de Liberación de Medicamentos/métodos , Femenino , Glomerulonefritis/inmunología , Glomerulonefritis/patología , Humanos , Hibridomas , Sueros Inmunes/administración & dosificación , Sueros Inmunes/inmunología , Fragmentos de Inmunoglobulinas/inmunología , Inmunoglobulina G/inmunología , Glomérulos Renales/efectos de los fármacos , Glomérulos Renales/inmunología , Glomérulos Renales/patología , Ratones , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Mitocondrias/inmunología , Mitocondrias/metabolismo , Fosforilación Oxidativa/efectos de los fármacos , Proteína Quinasa C-alfa/inmunología , Proteína Quinasa C-alfa/metabolismo , Inhibidores de Proteínas Quinasas/inmunología , Inhibidores de Proteínas Quinasas/uso terapéutico , Resultado del Tratamiento
8.
Arterioscler Thromb Vasc Biol ; 37(6): 1180-1193, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28473444

RESUMEN

OBJECTIVE: A disintegrin and metalloproteinase ADAM17 (tumor necrosis factor-α [TNF]-converting enzyme) regulates soluble TNF levels. We tested the hypothesis that aging-induced activation in adipose tissue (AT)-expressed ADAM17 contributes to the development of remote coronary microvascular dysfunction in obesity. APPROACH AND RESULTS: Coronary arterioles (CAs, ≈90 µm) from right atrial appendages and mediastinal AT were examined in patients (aged: 69±11 years, BMI: 30.2±5.6 kg/m2) who underwent open heart surgery. CA and AT were also studied in 6-month and 24-month lean and obese mice fed a normal or high-fat diet. We found that obesity elicited impaired endothelium-dependent CA dilations only in older patients and in aged high-fat diet mice. Transplantation of AT from aged obese, but not from young or aged, mice increased serum cytokine levels, including TNF, and impaired CA dilation in the young recipient mice. In patients and mice, obesity was accompanied by age-related activation of ADAM17, which was attributed to vascular endothelium-expressed ADAM17. Excess, ADAM17-shed TNF from AT arteries in older obese patients was sufficient to impair CA dilation in a bioassay in which the AT artery was serially connected to a CA. Moreover, we found that the increased activity of endothelial ADAM17 is mediated by a diminished inhibitory interaction with caveolin-1, owing to age-related decline in caveolin-1 expression in obese patients and mice or to genetic deletion of caveolin-1. CONCLUSIONS: The present study indicates that aging and obesity cooperatively reduce caveolin-1 expression and increase vascular endothelial ADAM17 activity and soluble TNF release in AT, which may contribute to the development of remote coronary microvascular dysfunction in older obese patients.


Asunto(s)
Proteína ADAM17/metabolismo , Tejido Adiposo/enzimología , Envejecimiento/metabolismo , Arteriolas/enzimología , Enfermedad de la Arteria Coronaria/enzimología , Vasos Coronarios/enzimología , Vasodilatación , Proteína ADAM17/genética , Tejido Adiposo/trasplante , Adulto , Factores de Edad , Anciano , Anciano de 80 o más Años , Envejecimiento/genética , Animales , Arteriolas/fisiopatología , Caveolina 1/deficiencia , Caveolina 1/genética , Caveolina 1/metabolismo , Células Cultivadas , Enfermedad de la Arteria Coronaria/genética , Enfermedad de la Arteria Coronaria/fisiopatología , Vasos Coronarios/fisiopatología , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Células Endoteliales/enzimología , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Obesidad/enzimología , Obesidad/genética , Obesidad/fisiopatología , Interferencia de ARN , Factores de Riesgo , Transducción de Señal , Transfección , Factor de Necrosis Tumoral alfa/metabolismo
9.
J Biol Chem ; 291(45): 23440-23451, 2016 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-27645999

RESUMEN

Regulation of the epithelial sodium channel (ENaC), which regulates fluid homeostasis and blood pressure, is complex and remains incompletely understood. The TIP peptide, a mimic of the lectin-like domain of TNF, activates ENaC by binding to glycosylated residues in the extracellular loop of ENaC-α, as well as to a hitherto uncharacterized internal site. Molecular docking studies suggested three residues, Val567, Glu568, and Glu571, located at the interface between the second transmembrane and C-terminal domains of ENaC-α, as a critical site for binding of the TIP peptide. We generated Ala replacement mutants in this region of ENaC-α and examined its interaction with TIP peptide (3M, V567A/E568A/E571A; 2M, V567A/E568A; and 1M, E571A). 3M and 2M ENaC-α, but not 1M ENaC-α, displayed significantly reduced binding capacity to TIP peptide and to TNF. When overexpressed in H441 cells, 3M mutant ENaC-α formed functional channels with similar gating and density characteristics as the WT subunit and efficiently associated with the ß and γ subunits in the plasma membrane. We subsequently assayed for increased open probability time and membrane expression, both of which define ENaC activity, following addition of TIP peptide. TIP peptide increased open probability time in H441 cells overexpressing wild type and 1M ENaC-α channels, but not 3M or 2M ENaC-α channels. On the other hand, TIP peptide-mediated reduction in ENaC ubiquitination was similar in cells overexpressing either WT or 3M ENaC-α subunits. In summary, this study has identified a novel site in ENaC-α that is crucial for activation of the open probability of the channel, but not membrane expression, by the lectin-like domain of TNF.


Asunto(s)
Agonistas del Canal de Sodio Epitelial/farmacología , Canales Epiteliales de Sodio/metabolismo , Péptidos Cíclicos/farmacología , Línea Celular Tumoral , Canales Epiteliales de Sodio/química , Canales Epiteliales de Sodio/genética , Células HEK293 , Humanos , Simulación del Acoplamiento Molecular , Mutación Puntual , Dominios Proteicos/efectos de los fármacos , Subunidades de Proteína/química , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Ubiquitinación/efectos de los fármacos
10.
Am J Physiol Regul Integr Comp Physiol ; 313(5): R560-R571, 2017 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-28835451

RESUMEN

Obesity-induced vascular dysfunction involves pathological remodeling of the visceral adipose tissue (VAT) and increased inflammation. Our previous studies showed that arginase 1 (A1) in endothelial cells (ECs) is critically involved in obesity-induced vascular dysfunction. We tested the hypothesis that EC-A1 activity also drives obesity-related VAT remodeling and inflammation. Our studies utilized wild-type and EC-A1 knockout (KO) mice made obese by high-fat/high-sucrose (HFHS) diet. HFHS diet induced increases in body weight, fasting blood glucose, and VAT expansion. This was accompanied by increased arginase activity and A1 expression in vascular ECs and increased expression of tumor necrosis factor-α (TNF-α), monocyte chemoattractant protein-1 (MCP-1), interleukin-10 (IL-10), vascular cell adhesion molecule-1 (VCAM-1), and intercellular adhesion molecule-1 (ICAM-1) mRNA and protein in both VAT and ECs. HFHS also markedly increased circulating inflammatory monocytes and VAT infiltration by inflammatory macrophages, while reducing reparative macrophages. Additionally, adipocyte size and fibrosis increased and capillary density decreased in VAT. These effects of HFHS, except for weight gain and hyperglycemia, were prevented or reduced in mice lacking EC-A1 or treated with the arginase inhibitor 2-(S)-amino-6-boronohexanoic acid (ABH). In mouse aortic ECs, exposure to high glucose (25 mM) and Na palmitate (200 µM) reduced nitric oxide production and increased A1, TNF-α, VCAM-1, ICAM-1, and MCP-1 mRNA, and monocyte adhesion. Knockout of EC-A1 or ABH prevented these effects. HFHS diet-induced VAT inflammation is mediated by EC-A1 expression/activity. Limiting arginase activity is a possible therapeutic means of controlling obesity-induced vascular and VAT inflammation.


Asunto(s)
Arginasa/metabolismo , Grasa Intraabdominal/metabolismo , Obesidad/complicaciones , Adipocitos/metabolismo , Adipocitos/patología , Tejido Adiposo/metabolismo , Animales , Quimiocina CCL2/metabolismo , Inflamación/etiología , Inflamación/metabolismo , Interleucina-10/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/metabolismo , Aumento de Peso/fisiología
12.
Crit Care ; 21(1): 194, 2017 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-28750677

RESUMEN

BACKGROUND: High-permeability pulmonary edema is a hallmark of acute respiratory distress syndrome (ARDS) and is frequently accompanied by impaired alveolar fluid clearance (AFC). AP301 enhances AFC by activating epithelial sodium channels (ENaCs) on alveolar epithelial cells, and we investigated its effect on extravascular lung water index (EVLWI) in mechanically ventilated patients with ARDS. METHODS: Forty adult mechanically ventilated patients with ARDS were included in a randomized, double-blind, placebo-controlled trial for proof of concept. Patients were treated with inhaled AP301 (n = 20) or placebo (0.9% NaCl; n = 20) twice daily for 7 days. EVLWI was measured by thermodilution (PiCCO®), and treatment groups were compared using the nonparametric Mann-Whitney U test. RESULTS: AP301 inhalation was well tolerated. No differences in mean baseline-adjusted change in EVLWI from screening to day 7 were found between the AP301 and placebo group (p = 0.196). There was no difference in the PaO2/FiO2 ratio, ventilation pressures, Murray lung injury score, or 28-day mortality between the treatment groups. An exploratory subgroup analysis according to severity of illness showed reductions in EVLWI (p = 0.04) and ventilation pressures (p < 0.05) over 7 days in patients with initial sequential organ failure assessment (SOFA) scores ≥11 inhaling AP301 versus placebo, but not in patients with SOFA scores ≤10. CONCLUSIONS: There was no difference in mean baseline-adjusted EVLWI between the AP301 and placebo group. An exploratory post-hoc subgroup analysis indicated reduced EVLWI in patients with SOFA scores ≥11 receiving AP301. These results suggest further confirmation in future clinical trials of inhaled AP301 for treatment of pulmonary edema in patients with ARDS. TRIAL REGISTRATION: The study was prospectively registered at clinicaltrials.gov, NCT01627613 . Registered 20 June 2012.


Asunto(s)
Péptidos Cíclicos/farmacología , Edema Pulmonar/tratamiento farmacológico , Síndrome de Dificultad Respiratoria/complicaciones , Administración por Inhalación , Adulto , Anciano , Método Doble Ciego , Femenino , Humanos , Pulmón/efectos de los fármacos , Masculino , Persona de Mediana Edad , Puntuaciones en la Disfunción de Órganos , Péptidos Cíclicos/uso terapéutico , Edema Pulmonar/etiología , Respiración Artificial/métodos
13.
J Infect Dis ; 211(2): 306-16, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25183769

RESUMEN

Microbial infections can induce aberrant responses in cellular stress pathways, leading to translational attenuation, metabolic restriction, and activation of oxidative stress, with detrimental effects on cell survival. Here we show that infection of human airway epithelial cells with Streptococcus pneumoniae leads to induction of endoplasmic reticulum (ER) and oxidative stress, activation of mitogen-associated protein kinase (MAPK) signaling pathways, and regulation of their respective target genes. We identify pneumococcal H2O2 as the causative agent for these responses, as both catalase-treated and pyruvate oxidase-deficient bacteria lacked these activities. Pneumococcal H2O2 induced nuclear NF-κB translocation and transcription of proinflammatory cytokines. Inhibition of translational arrest and ER stress by salubrinal or of MAPK signaling pathways attenuate cytokine transcription. These results provide strong evidence for the notion that inhibition of translation is an important host pathway in monitoring harmful pathogen-associated activities, thereby enabling differentiation between pathogenic and nonpathogenic bacteria.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Interacciones Huésped-Patógeno , Peróxido de Hidrógeno/metabolismo , Inflamación , Transducción de Señal/efectos de los fármacos , Streptococcus pneumoniae/inmunología , Estrés Fisiológico , Línea Celular , Citocinas/metabolismo , Células Epiteliales/inmunología , Células Epiteliales/microbiología , Humanos , FN-kappa B/metabolismo
14.
J Biol Chem ; 289(8): 4710-22, 2014 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-24398689

RESUMEN

Acute lung injury (ALI) is characterized by increased endothelial hyperpermeability. Protein nitration is involved in the endothelial barrier dysfunction in LPS-exposed mice. However, the nitrated proteins involved in this process have not been identified. The activation of the small GTPase RhoA is a critical event in the barrier disruption associated with LPS. Thus, in this study we evaluated the possible role of RhoA nitration in this process. Mass spectroscopy identified a single nitration site, located at Tyr(34) in RhoA. Tyr(34) is located within the switch I region adjacent to the nucleotide-binding site. Utilizing this structure, we developed a peptide designated NipR1 (nitration inhibitory peptide for RhoA 1) to shield Tyr(34) against nitration. TAT-fused NipR1 attenuated RhoA nitration and barrier disruption in LPS-challenged human lung microvascular endothelial cells. Further, treatment of mice with NipR1 attenuated vessel leakage and inflammatory cell infiltration and preserved lung function in a mouse model of ALI. Molecular dynamics simulations suggested that the mechanism by which Tyr(34) nitration stimulates RhoA activity was through a decrease in GDP binding to the protein caused by a conformational change within a region of Switch I, mimicking the conformational shift observed when RhoA is bound to a guanine nucleotide exchange factor. Stopped flow kinetic analysis was used to confirm this prediction. Thus, we have identified a new mechanism of nitration-mediated RhoA activation involved in LPS-mediated endothelial barrier dysfunction and show the potential utility of "shielding" peptides to prevent RhoA nitration in the management of ALI.


Asunto(s)
Lesión Pulmonar/enzimología , Lesión Pulmonar/patología , Proteína de Unión al GTP rhoA/metabolismo , Lesión Pulmonar Aguda/enzimología , Lesión Pulmonar Aguda/patología , Lesión Pulmonar Aguda/fisiopatología , Secuencia de Aminoácidos , Animales , Líquido del Lavado Bronquioalveolar , Citocinas/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/enzimología , Células Endoteliales/patología , Activación Enzimática , Humanos , Lipopolisacáridos , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Microvasos/patología , Datos de Secuencia Molecular , Óxido Nítrico Sintasa de Tipo III/metabolismo , Nitrosación , Péptidos/metabolismo , Sustancias Protectoras/metabolismo , Tirosina/metabolismo , Proteína de Unión al GTP rhoA/química
15.
Am J Physiol Heart Circ Physiol ; 308(5): H376-85, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25527780

RESUMEN

The type 1 angiotensin II (ANG II) receptor (AT1R) undergoes internalization following stimulation by ANG II. Internalization reduces cell surface AT1Rs, and it is required for AT1R resensitization. In this process AT1R may interact with caveolin-1 (Cav1), the main scaffolding protein of caveolae. We hypothesized that the interaction between Cav1 and AT1R delays AT1R resensitization and thereby prevents sustained ANG II-induced resistance artery (RA) constriction under normal conditions and in experimental obesity. In rat and mouse skeletal muscle RA (diameter: ∼90-120 µm) ANG II-induced constrictions were reduced upon repeated (30-min apart) administrations. Upon disruption of caveolae with methyl-ß-cyclodextrin or in RA of Cav1 knockout mice, repeated ANG II applications resulted in essentially maintained constrictions. In vascular smooth muscle cells, AT1R interacted with Cav1, and the degree of cell surface interactions was reduced by long-term (15-min), but not short-term (2-min), exposure to ANG II. When Cav1 was silenced, the amount of membrane-associated AT1R was significantly reduced by a short-term ANG II exposure. Moreover, Cav1 knockout mice fed a high-fat diet exhibited augmented and sustained RA constriction to ANG II and had elevated systemic blood pressure, when compared with normal or high-fat fed wild-type mice. Thus, Cav1, through a direct interaction, delays internalization and subsequent resensitization of AT1R. We suggest that this mechanism prevents sustained ANG II-induced RA constriction and elevated systemic blood pressure in diet-induced obesity.


Asunto(s)
Arterias/fisiopatología , Caveolina 2/metabolismo , Hipertensión/metabolismo , Obesidad/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Resistencia Vascular , Vasoconstricción , Angiotensina II/farmacología , Animales , Arterias/efectos de los fármacos , Arterias/metabolismo , Caveolas/metabolismo , Caveolina 2/genética , Membrana Celular/metabolismo , Hipertensión/etiología , Hipertensión/fisiopatología , Masculino , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiología , Obesidad/complicaciones , Unión Proteica , Ratas , Ratas Wistar , Receptor de Angiotensina Tipo 1/agonistas , Vasoconstrictores/farmacología
16.
Am J Respir Crit Care Med ; 190(5): 522-32, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-25029038

RESUMEN

RATIONALE: Alveolar liquid clearance is regulated by Na(+) uptake through the apically expressed epithelial sodium channel (ENaC) and basolaterally localized Na(+)-K(+)-ATPase in type II alveolar epithelial cells. Dysfunction of these Na(+) transporters during pulmonary inflammation can contribute to pulmonary edema. OBJECTIVES: In this study, we sought to determine the precise mechanism by which the TIP peptide, mimicking the lectin-like domain of tumor necrosis factor (TNF), stimulates Na(+) uptake in a homologous cell system in the presence or absence of the bacterial toxin pneumolysin (PLY). METHODS: We used a combined biochemical, electrophysiological, and molecular biological in vitro approach and assessed the physiological relevance of the lectin-like domain of TNF in alveolar liquid clearance in vivo by generating triple-mutant TNF knock-in mice that express a mutant TNF with deficient Na(+) uptake stimulatory activity. MEASUREMENTS AND MAIN RESULTS: TIP peptide directly activates ENaC, but not the Na(+)-K(+)-ATPase, upon binding to the carboxy-terminal domain of the α subunit of the channel. In the presence of PLY, a mediator of pneumococcal-induced pulmonary edema, this binding stabilizes the ENaC-PIP2-MARCKS complex, which is necessary for the open probability conformation of the channel and preserves ENaC-α protein expression, by means of blunting the protein kinase C-α pathway. Triple-mutant TNF knock-in mice are more prone than wild-type mice to develop edema with low-dose intratracheal PLY, correlating with reduced pulmonary ENaC-α subunit expression. CONCLUSIONS: These results demonstrate a novel TNF-mediated mechanism of direct ENaC activation and indicate a physiological role for the lectin-like domain of TNF in the resolution of alveolar edema during inflammation.


Asunto(s)
Agonistas del Canal de Sodio Epitelial/metabolismo , Canales Epiteliales de Sodio/metabolismo , Péptidos Cíclicos/metabolismo , Alveolos Pulmonares/metabolismo , Edema Pulmonar/metabolismo , Estreptolisinas , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Proteínas Bacterianas , Agonistas del Canal de Sodio Epitelial/química , Canales Epiteliales de Sodio/química , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Péptidos Cíclicos/química , Alveolos Pulmonares/microbiología , Edema Pulmonar/microbiología , Factor de Necrosis Tumoral alfa/química
17.
Proc Natl Acad Sci U S A ; 109(6): 2084-9, 2012 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-22308467

RESUMEN

Aggressive treatment with antibiotics in patients infected with Streptococcus pneumoniae induces release of the bacterial virulence factor pneumolysin (PLY). Days after lungs are sterile, this pore-forming toxin can still induce pulmonary permeability edema in patients, characterized by alveolar/capillary barrier dysfunction and impaired alveolar liquid clearance (ALC). ALC is mainly regulated through Na(+) transport by the apically expressed epithelial sodium channel (ENaC) and the basolaterally expressed Na(+)/K(+)-ATPase in type II alveolar epithelial cells. Because no standard treatment is currently available to treat permeability edema, the search for novel therapeutic candidates is of high priority. We detected mRNA expression for the active receptor splice variant SV1 of the hypothalamic polypeptide growth hormone-releasing hormone (GHRH), as well as for GHRH itself, in human lung microvascular endothelial cells (HL-MVEC). Therefore, we have evaluated the effect of the GHRH agonist JI-34 on PLY-induced barrier and ALC dysfunction. JI-34 blunts PLY-mediated endothelial hyperpermeability in monolayers of HL-MVEC, in a cAMP-dependent manner, by means of reducing the phosphorylation of myosin light chain and vascular endothelial (VE)-cadherin. In human airway epithelial H441 cells, PLY significantly impairs Na(+) uptake, but JI-34 restores it to basal levels by means of increasing cAMP levels. Intratracheal instillation of PLY into C57BL6 mice causes pulmonary alveolar epithelial and endothelial hyperpermeability as well as edema formation, all of which are blunted by JI-34. These findings point toward a protective role of the GHRH signaling pathway in PLY-induced permeability edema.


Asunto(s)
Hormona Liberadora de Hormona del Crecimiento/agonistas , Edema Pulmonar/patología , Estreptolisinas/toxicidad , Animales , Antígenos CD/metabolismo , Proteínas Bacterianas/toxicidad , Cadherinas/metabolismo , Células Endoteliales/metabolismo , Células Endoteliales/patología , Regulación de la Expresión Génica , Hormona Liberadora de Hormona del Crecimiento/genética , Hormona Liberadora de Hormona del Crecimiento/metabolismo , Humanos , Activación del Canal Iónico , Pulmón/metabolismo , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Microvasos/patología , Cadenas Ligeras de Miosina/metabolismo , Permeabilidad , Fosforilación , Alveolos Pulmonares/efectos de los fármacos , Alveolos Pulmonares/patología , Edema Pulmonar/genética , Edema Pulmonar/fisiopatología , Empalme del ARN/genética , Receptores de Neuropéptido/genética , Receptores de Neuropéptido/metabolismo , Receptores de Hormona Reguladora de Hormona Hipofisaria/genética , Receptores de Hormona Reguladora de Hormona Hipofisaria/metabolismo , Canales de Sodio/metabolismo
18.
Am J Respir Cell Mol Biol ; 50(3): 614-25, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24134589

RESUMEN

Acute lung injury (ALI) is a severe hypoxemic respiratory insufficiency associated with lung leak, diffuse alveolar damage, inflammation, and loss of lung function. Decreased dimethylaminohydrolase (DDAH) activity and increases in asymmetric dimethylarginine (ADMA), together with exaggerated oxidative/nitrative stress, contributes to the development of ALI in mice exposed to LPS. Whether restoring DDAH function and suppressing ADMA levels can effectively ameliorate vascular hyperpermeability and lung injury in ALI is unknown, and was the focus of this study. In human lung microvascular endothelial cells, DDAH II overexpression prevented the LPS-dependent increase in ADMA, superoxide, peroxynitrite, and protein nitration. DDAH II also attenuated the endothelial barrier disruption associated with LPS exposure. Similarly, in vivo, we demonstrated that the targeted overexpression of DDAH II in the pulmonary vasculature significantly inhibited the accumulation of ADMA and the subsequent increase in oxidative/nitrative stress in the lungs of mice exposed to LPS. In addition, augmenting pulmonary DDAH II activity before LPS exposure reduced lung vascular leak and lung injury and restored lung function when DDAH activity was increased after injury. Together, these data suggest that enhancing DDAH II activity may prove a useful adjuvant therapy to treat patients with ALI.


Asunto(s)
Lesión Pulmonar Aguda/prevención & control , Amidohidrolasas/metabolismo , Células Endoteliales/enzimología , Terapia Genética , Lipopolisacáridos , Pulmón/irrigación sanguínea , Microvasos/enzimología , Edema Pulmonar/prevención & control , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/enzimología , Lesión Pulmonar Aguda/genética , Amidohidrolasas/genética , Animales , Arginina/análogos & derivados , Arginina/metabolismo , Líquido del Lavado Bronquioalveolar/química , Permeabilidad Capilar , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales/patología , Humanos , Pulmón/enzimología , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Microvasos/patología , Estrés Oxidativo , Ácido Peroxinitroso/metabolismo , Edema Pulmonar/inducido químicamente , Edema Pulmonar/enzimología , Edema Pulmonar/genética , Superóxidos/metabolismo , Factores de Tiempo , Transfección , Regulación hacia Arriba
19.
Am J Physiol Lung Cell Mol Physiol ; 306(6): L497-507, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24414256

RESUMEN

Acute lung injury and acute respiratory distress syndrome (ALI/ARDS) affect 200,000 people a year in the USA. Pulmonary vascular and specifically endothelial cell (EC) barrier compromise is a hallmark of these diseases. We have recently shown that extracellular adenosine enhances human pulmonary (EC) barrier via activation of adenosine receptors (ARs) in cell cultures. On the basis of these data, we hypothesized that activation of ARs might exert barrier-protective effects in a model of ALI/ARDS in mice. To test this hypothesis, we examined the effects of pre- and posttreatment of adenosine and 5'-N-ethylcarboxamidoadenosine (NECA), a nonselective stable AR agonist, on LPS-induced lung injury. Mice were given vehicle or LPS intratracheally followed by adenosine, NECA, or vehicle instilled via the internal jugular vein. Postexperiment cell counts, Evans Blue Dye albumin (EBDA) extravasation, levels of proteins, and inflammatory cytokines were analyzed. Harvested lungs were used for histology and myeloperoxidase studies. Mice challenged with LPS alone demonstrated an inflammatory response typical of ALI. Cell counts, EBDA extravasation, as well as levels of proteins and inflammatory cytokines were decreased in adenosine-treated mice. Histology displayed reduced infiltration of neutrophils. NECA had a similar effect on LPS-induced vascular barrier compromise. Importantly, posttreatment with adenosine or NECA recovers lung vascular barrier and reduces inflammation induced by LPS challenge. Furthermore, adenosine significantly attenuated protein degradation of A2A and A3 receptors induced by LPS. Collectively, our results demonstrate that activation of ARs protects and restores vascular barrier functions and reduces inflammation in LPS-induced ALI.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Adenosina/metabolismo , Endotelio/metabolismo , Receptores Purinérgicos P1/metabolismo , Lesión Pulmonar Aguda/inducido químicamente , Adenosina-5'-(N-etilcarboxamida)/metabolismo , Animales , Líquido del Lavado Bronquioalveolar/citología , Permeabilidad Capilar/efectos de los fármacos , Recuento de Células , Citocinas/metabolismo , Células Endoteliales/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Interleucina-6/metabolismo , Lipopolisacáridos , Pulmón/metabolismo , Pulmón/fisiología , Ratones , Ratones Endogámicos C57BL , Agonistas del Receptor Purinérgico P1/metabolismo , Síndrome de Dificultad Respiratoria/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
20.
Chin Med J Pulm Crit Care Med ; 2(2): 80-87, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-39006829

RESUMEN

Endothelial cells (ECs) form a semi-permeable barrier between the interior space of blood vessels and the underlying tissues. Pulmonary endothelial barrier integrity is maintained through coordinated cellular processes involving receptors, signaling molecules, junctional complexes, and protein-regulated cytoskeletal reorganization. In acute lung injury (ALI) or its more severe form acute respiratory distress syndrome (ARDS), the loss of endothelial barrier integrity secondary to endothelial dysfunction caused by severe pulmonary inflammation and/or infection leads to pulmonary edema and hypoxemia. Pro-inflammatory agonists such as histamine, thrombin, bradykinin, interleukin 1ß, tumor necrosis factor α, vascular endothelial growth factor, angiopoietin-2, and platelet-activating factor, as well as bacterial toxins and reactive oxygen species, cause dynamic changes in cytoskeletal structure, adherens junction disorganization, and detachment of vascular endothelial cadherin (VE-cadherin) from the actin cytoskeleton, leading to an increase in endothelial permeability. Endothelial interactions with leukocytes, platelets, and coagulation enhance the inflammatory response. Moreover, inflammatory infiltration and the associated generation of pro-inflammatory cytokines during infection cause EC death, resulting in further compromise of the structural integrity of lung endothelial barrier. Despite the use of potent antibiotics and aggressive intensive care support, the mortality of ALI is still high, because the mechanisms of pulmonary EC barrier disruption are not fully understood. In this review, we summarized recent advances in the studies of endothelial cytoskeletal reorganization, inter-endothelial junctions, endothelial inflammation, EC death, and endothelial repair in ALI and ARDS, intending to shed some light on the potential diagnostic and therapeutic targets in the clinical management of the disease.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA