Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(8)2024 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-38673806

RESUMEN

We have recently reported that transcription factor Runx3 is required for pulmonary generation of CD8+ cytotoxic T lymphocytes (CTLs) that play a crucial role in the clearance of influenza A virus (IAV). To understand the underlying mechanisms, we determined the effects of Runx3 knockout (KO) on CD8+ T cell local expansion and phenotypes using an inducible general Runx3 KO mouse model. We found that in contrast to the lungs, Runx3 general KO promoted enlargement of lung-draining mediastinal lymph node (mLN) and enhanced CD8+ and CD4+ T cell expansion during H1N1 IAV infection. We further found that Runx3 deficiency greatly inhibited core 2 O-glycosylation of selectin ligand CD43 on activated CD8+ T cells but minimally affected the cell surface expression of CD43, activation markers (CD44 and CD69) and cell adhesion molecules (CD11a and CD54). Runx3 KO had a minor effect on lung effector CD8+ T cell death by IAV infection. Our findings indicate that Runx3 differently regulates CD8+ T cell expansion in mLNs and lungs by H1N1 IAV infection. Runx3 is required for CD43 core 2 O-glycosylation on activated CD8+ T cells, and the involved Runx3 signal pathway may mediate CD8+ T cell phenotype for pulmonary generation of CTLs.


Asunto(s)
Linfocitos T CD8-positivos , Subunidad alfa 3 del Factor de Unión al Sitio Principal , Subtipo H1N1 del Virus de la Influenza A , Infecciones por Orthomyxoviridae , Animales , Ratones , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Proliferación Celular , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Glicosilación , Subtipo H1N1 del Virus de la Influenza A/inmunología , Leucosialina/metabolismo , Pulmón/virología , Pulmón/metabolismo , Pulmón/inmunología , Pulmón/patología , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/inmunología , Activación de Linfocitos/inmunología , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/virología
2.
Am J Physiol Lung Cell Mol Physiol ; 325(5): L689-L708, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37642665

RESUMEN

Chronic obstructive pulmonary disease (COPD) is a debilitating lung disease with no effective treatment that can reduce mortality or slow the disease progression. COPD is the third leading cause of global death and is characterized by airflow limitations due to chronic bronchitis and alveolar damage/emphysema. Chronic cigarette smoke (CS) exposure damages airway and alveolar epithelium and remains a major risk factor for the pathogenesis of COPD. We found that the expression of caveolin-1, a tumor suppressor protein; p53; and plasminogen activator inhibitor-1 (PAI-1), one of the downstream targets of p53, was markedly increased in airway epithelial cells (AECs) as well as in type II alveolar epithelial (AT2) cells from the lungs of patients with COPD or wild-type mice with CS-induced lung injury (CS-LI). Moreover, p53- and PAI-1-deficient mice resisted CS-LI. Furthermore, treatment of AECs, AT2 cells, or lung tissue slices from patients with COPD or mice with CS-LI with a seven amino acid caveolin-1 scaffolding domain peptide (CSP7) reduced mucus hypersecretion in AECs and improved AT2 cell viability. Notably, induction of PAI-1 expression via increased caveolin-1 and p53 contributed to mucous cell metaplasia and mucus hypersecretion in AECs, and reduced AT2 viability, due to increased senescence and apoptosis, which was abrogated by CSP7. In addition, treatment of wild-type mice having CS-LI with CSP7 by intraperitoneal injection or nebulization via airways attenuated mucus hypersecretion, alveolar injury, and significantly improved lung function. This study validates the potential therapeutic role of CSP7 for treating CS-LI and COPD. NEW & NOTEWORTHY Chronic cigarette smoke (CS) exposure remains a major risk factor for the pathogenesis of COPD, a debilitating disease with no effective treatment. Increased caveolin-1 mediated induction of p53 and downstream plasminogen activator inhibitor-1 (PAI-1) expression contributes to CS-induced airway mucus hypersecretion and alveolar wall damage. This is reversed by caveolin-1 scaffolding domain peptide (CSP7) in preclinical models, suggesting the therapeutic potential of CSP7 for treating CS-induced lung injury (CS-LI) and COPD.


Asunto(s)
Caveolina 1 , Fumar Cigarrillos , Lesión Pulmonar , Enfermedad Pulmonar Obstructiva Crónica , Enfisema Pulmonar , Animales , Humanos , Ratones , Caveolina 1/farmacología , Fumar Cigarrillos/efectos adversos , Pulmón/metabolismo , Lesión Pulmonar/patología , Péptidos/farmacología , Inhibidor 1 de Activador Plasminogénico/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/patología , Enfisema Pulmonar/patología , Proteína p53 Supresora de Tumor/metabolismo
3.
Int J Mol Sci ; 23(6)2022 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-35328736

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a fatal disease characterized by an excess deposition of extracellular matrix in the pulmonary interstitium. Caveolin-1 scaffolding domain peptide (CSP) has been found to mitigate pulmonary fibrosis in several animal models. However, its pathophysiological role in IPF is obscure, and it remains critical to understand the mechanism by which CSP protects against pulmonary fibrosis. We first studied the delivery of CSP into cells and found that it is internalized and accumulated in the Endoplasmic Reticulum (ER). Furthermore, CSP reduced ER stress via suppression of inositol requiring enzyme1α (IRE1α) in transforming growth factor ß (TGFß)-treated human IPF lung fibroblasts (hIPF-Lfs). Moreover, we found that CSP enhanced the gelatinolytic activity of TGFß-treated hIPF-Lfs. The IRE1α inhibitor; 4µ8C also augmented the gelatinolytic activity of TGFß-treated hIPF-Lfs, supporting the concept that CSP induced inhibition of the IRE1α pathway. Furthermore, CSP significantly elevated expression of MMPs in TGFß-treated hIPF-Lfs, but conversely decreased the secretion of collagen 1. Similar results were observed in two preclinical murine models of PF, bleomycin (BLM)- and adenovirus expressing constitutively active TGFß (Ad-TGFß)-induced PF. Our findings provide new insights into the mechanism by which lung fibroblasts contribute to CSP dependent protection against lung fibrosis.


Asunto(s)
Fibrosis Pulmonar Idiopática , Animales , Bleomicina , Caveolina 1/genética , Caveolina 1/metabolismo , Endorribonucleasas/metabolismo , Fibroblastos/metabolismo , Fibrosis Pulmonar Idiopática/metabolismo , Pulmón/metabolismo , Ratones , Péptidos/metabolismo , Proteínas Serina-Treonina Quinasas , Factor de Crecimiento Transformador beta/metabolismo
4.
Am J Pathol ; 188(10): 2207-2222, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30253845

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a debilitating, incurable, and life-threatening disease. A cardinal feature of the pathogenesis of IPF is excessive extracellular matrix deposition attributable to proliferation of activated fibrotic lung fibroblasts (fLfs). To assess the underlying mechanism, we analyzed the status of the tumor suppressor protein p53 in fLfs from the lungs of IPF patients or mice with bleomycin-induced established PF. We report that basal expression of p53 is markedly reduced in fLfs. Forced expression of caveolin-1 in fLfs increased basal p53 and reduced profibrogenic proteins, including collagen-1. Transduction of fLfs with adenovirus expressing p53 reduced expression of these proteins. Conversely, inhibition of baseline p53 in control lung fibroblasts from lung tissues increased profibrogenic protein expression. Lung transduction of adenovirus expressing p53 reduced bleomycin-induced PF in wild-type or caveolin-1-deficient mice. Furthermore, treatment of fLfs or fibrotic lung tissues with caveolin-1 scaffolding domain peptide (CSP) or its fragment, CSP7, restored p53 and reduced profibrogenic proteins. Treatment of wild-type mice with i.p. CSP or CSP7 resolved bleomycin-induced PF. These peptides failed to resolve PF in inducible conditional knockout mice lacking p53 in fLfs, indicating the induction of baseline fLf p53 as the basis of the antifibrotic effects.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias)/fisiología , Fibroblastos/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Antibióticos Antineoplásicos/toxicidad , Bleomicina/toxicidad , Caveolina 1/deficiencia , Caveolina 1/metabolismo , Caveolina 1/farmacología , Humanos , Fibrosis Pulmonar Idiopática/fisiopatología , Ratones Endogámicos C57BL , Fragmentos de Péptidos/farmacología , Transducción Genética , Proteína p53 Supresora de Tumor/antagonistas & inhibidores
5.
Toxicol Appl Pharmacol ; 370: 178-183, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30910594

RESUMEN

HDAC6 is a member of the class II histone deacetylase. HDAC6 inhibition possesses anti-inflammatory effects. However, the effects of HDAC6 inhibition in acute lung inflammation have not been studied. Here, we investigated the effects of a highly selective and potent HDAC6 inhibitor CAY10603 in LPS-induced acute inflammatory lung injury. We also conducted a series of experiments including immunoblotting, ELISA, and histological assays to explore the inflammatory signaling pathways modulated by the selective HDAC6 inhibition. We observed that HDAC6 activity was increased in the lung tissues after LPS challenge, which was associated with a decreased level of ɑ-tubulin acetylation in the lung tissues. HDAC6 inhibition by CAY10603 prevented LPS-induced ɑ-tubulin deacetylation in the lung tissues. HDAC6 inhibition also exhibited protective effects against LPS-induced acute lung inflammation, which was demonstrated by the reduced production of pro-inflammatory cytokines TNF-α, IL-1ß, and IL-6 and decreased leukocyte infiltration. Furthermore, HDAC6 inhibition blocked the decrease of E-cadherin level and inhibited the increase of MMP9 expression in the lung tissues, which could prevent the destruction of the lung architecture in LPS-induced inflammatory injury. Given the important roles of NFĸB and inflammasome activation in inflammatory responses, we investigated their regulation by HDAC6 inhibition in LPS-induced lung injury. Our results showed that HDAC6 inhibition blocked the activation of NFĸB by inhibiting IĸB phosphorylation in LPS-induced acute lung injury, and LPS-induced-inflammasome activity was reduced by HDAC6 inhibition as demonstrated by the decreased IL-1ß and caspase-1 cleavage and activation. Collectively, our data suggest that selective HDAC6 inhibition suppresses inflammatory signaling pathways and alleviates LPS-induced acute lung inflammation.


Asunto(s)
Lesión Pulmonar Aguda/tratamiento farmacológico , Antiinflamatorios/administración & dosificación , Carbamatos/administración & dosificación , Caspasa 1/metabolismo , Histona Desacetilasa 6/antagonistas & inhibidores , Inhibidores de Histona Desacetilasas/administración & dosificación , Oxazoles/administración & dosificación , Neumonía/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Acetilación , Enfermedad Aguda , Lesión Pulmonar Aguda/inducido químicamente , Animales , Caspasa 1/efectos de los fármacos , Citocinas/metabolismo , Activación Enzimática/efectos de los fármacos , Inflamasomas/antagonistas & inhibidores , Lipopolisacáridos , Pulmón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B , Neumonía/inducido químicamente , Transducción de Señal/fisiología , Tubulina (Proteína)/metabolismo
6.
Am J Physiol Lung Cell Mol Physiol ; 314(5): L757-L768, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29345198

RESUMEN

Recent studies have shed new light on the role of the fibrinolytic system in the pathogenesis of pleural organization, including the mechanisms by which the system regulates mesenchymal transition of mesothelial cells and how that process affects outcomes of pleural injury. The key contribution of plasminogen activator inhibitor-1 to the outcomes of pleural injury is now better understood as is its role in the regulation of intrapleural fibrinolytic therapy. In addition, the mechanisms by which fibrinolysins are processed after intrapleural administration have now been elucidated, informing new candidate diagnostics and therapeutics for pleural loculation and failed drainage. The emergence of new potential interventional targets offers the potential for the development of new and more effective therapeutic candidates.


Asunto(s)
Fibrina/metabolismo , Enfermedades Pleurales/fisiopatología , Animales , Humanos , Enfermedades Pleurales/metabolismo
7.
Am J Pathol ; 187(5): 1016-1034, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28273432

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a progressive and fatal interstitial lung disease. The pathogenesis of interstitial lung diseases, including its most common form, IPF, remains poorly understood. Alveolar epithelial cell (AEC) apoptosis, proliferation, and accumulation of myofibroblasts and extracellular matrix deposition results in progressive loss of lung function in IPF. We found induction of tumor suppressor protein, p53, and apoptosis with suppression of urokinase-type plasminogen activator (uPA) and the uPA receptor in AECs from the lungs of IPF patients, and in mice with bleomycin, cigarette smoke, silica, or sepsis-induced lung injury. Treatment with the caveolin-1 scaffolding domain peptide (CSP) reversed these effects. Consistent with induction of p53, AECs from IPF lungs or mice with diverse types of lung injuries showed increased p53 acetylation and miR-34a expression with reduction in Sirt1. This was significantly reduced after treatment of wild-type mice with CSP, and uPA-deficient mice were unresponsive. Bleomycin failed to induce miR-34a in p53- or plasminogen activator inhibitor-1 (PAI-1)-deficient mice. CSP-mediated inhibition of miR-34a restored Sirt1, suppressed p53 acetylation and apoptosis in injured AECs, and prevented pulmonary fibrosis (PF). AEC-specific suppression of miR-34a inhibited bleomycin-induced p53, PAI-1, and apoptosis and prevented PF, whereas overexpression of precursor-miR-34a increased p53, PAI-1, and apoptosis in AECs of mice unexposed to bleomycin. Our study validates p53-miR-34a feedback as a potential therapeutic target in PF.


Asunto(s)
Fibrosis Pulmonar Idiopática/etiología , Lesión Pulmonar/etiología , MicroARNs/fisiología , Proteína p53 Supresora de Tumor/fisiología , Células Epiteliales Alveolares/fisiología , Animales , Apoptosis/fisiología , Caveolina 1/farmacología , Células Cultivadas , Retroalimentación , Humanos , Ratones , Ratones Endogámicos C57BL , Mucosa Respiratoria/fisiología
8.
Drug Dev Ind Pharm ; 44(2): 184-198, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28835128

RESUMEN

A caveolin-1 scaffolding domain, CSP7, is a newly developed peptide for the treatment of idiopathic pulmonary fibrosis. To develop a CSP7 formulation for further use we have obtained, characterized and compared a number of lyophilized formulations of CSP7 trifluoroacetate with DPBS and in combination with excipients (mannitol and lactose at molar ratios 1:5, 70 and 140). CSP7 trifluoroacetate was stable (>95%) in solution at 5 and 25 °C for up to 48 h and tolerated at least 5 freeze/thaw cycles. Lyophilized cakes of CSP7 trifluoroacetate with excipients were stable (>96%) for up to 4 weeks at room temperature (RT), and retained more than 98% of the CSP7 trifluoroacetate in the solution at 8 h after reconstitution at RT. The lyophilized CSP7 formulations were stable for up to 10 months at 5 °C protected from moisture. Exposure of the lyophilized cakes of CSP7 to 75% relative humidity (RH) resulted in an increase in the absorbed moisture, promoted crystallization of the excipients and induced reversible formation of CSP7 aggregates. Increased molar ratio of mannitol slightly affected formation of the aggregates. In contrast, lactose significantly decreased (up to 20 times) aggregate formation with apparent saturation at the molar ratio of 1:70. The possible mechanisms of stabilization of CSP7 trifluoroacetate in solid state by lactose include physical state of the bulking agent and the interactions between lactose and CSP7 trifluoroacetate (e.g. formation of a Schiff base with the N-terminal amino group of CSP7). Finally, CSP7 trifluoroacetate exhibited excellent stability during nebulization of formulations containing mannitol or lactose.


Asunto(s)
Química Farmacéutica/métodos , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Administración por Inhalación , Rastreo Diferencial de Calorimetría , Liberación de Fármacos , Estabilidad de Medicamentos , Liofilización , Humedad , Lactosa/química , Manitol/química , Ácido Trifluoroacético/química
9.
Am J Physiol Lung Cell Mol Physiol ; 312(6): L783-L796, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28385810

RESUMEN

Pulmonary surfactant protein C (SP-C) expression by type II alveolar epithelial cells (AECs) is markedly reduced in diverse types of lung injuries and is often associated with AEC apoptosis. It is unclear whether loss of SP-C contributes to the increased p53 and urokinase-type plasminogen activator (uPA) system cross-talk and apoptosis of AECs. Therefore, we inhibited SP-C expression in human and murine AECs using lentivirus vector expressing shRNA and tested p53 and downstream changes in the uPA-fibrinolytic system. Inhibition of SP-C expression in AECs induced p53 and activated caspase-3, indicating AEC apoptosis. We also found that bleomycin or cigarette smoke exposure failed to inhibit SP-C expression or apoptosis in AECs in p53- and plasminogen activator inhibitor-1 (PAI-1)-deficient mice. Depletion of SP-C expression by lentiviral SP-C shRNA in PAI-1-deficient mice failed to induce p53 or apoptosis in AECs, whereas it increased both AEC p53 and apoptosis in wild-type and uPA-deficient mice. SP-C inhibition in AECs also increased in CXCL1 and CXCL2 and their receptor CXCR2 as well as ICAM-1 expression, which is indicative of a proinflammatory response. Overexpression of p53-binding 3'-UTR sequences in AECs inhibited PAI-1 induction while maintaining uPA and uPAR protein and mRNA expression. Furthermore, caveolin-1 expression and phosphorylation were increased in AECs, indicating an intricate link between caveolin-1 and Src kinase-mediated cell signaling and AEC apoptosis due to loss of SP-C expression through p53 and uPA system-mediated cross-talk. The role of uPA, PAI-1, and p53 in the regulation of AEC apoptosis after injury was also determined in knockout mice.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Fibrinólisis , Lesión Pulmonar/metabolismo , Proteína C Asociada a Surfactante Pulmonar/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Regiones no Traducidas 3'/genética , Acetilación , Células Epiteliales Alveolares/patología , Animales , Apoptosis , Caspasa 3/metabolismo , Caveolina 1/metabolismo , Activación Enzimática , Humanos , Lesión Pulmonar/complicaciones , Lesión Pulmonar/patología , Ratones , Modelos Biológicos , Fosforilación , Fosfoserina/metabolismo , Inhibidor 1 de Activador Plasminogénico/metabolismo , Neumonía/complicaciones , Neumonía/patología , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Familia-src Quinasas/metabolismo
10.
Clin Pulm Med ; 24(4): 163-169, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29081644

RESUMEN

Complicated pleural effusions and empyema with loculation and failed drainage are common clinical problems. In adults, intrapleural fibrinolytic therapy is commonly used with variable results and therapy remains empiric. Despite the intrapleural use of various plasminogen activators; fibrinolysins, for about sixty years, there is no clear consensus about which agent is most effective. Emerging evidence demonstrates that intrapleural administration of plasminogen activators is subject to rapid inhibition by plasminogen activator inhibitor-1 and that processing of fibrinolysins is importantly influenced by other factors including the levels and quality of pleural fluid DNA. Current therapy for loculation that accompanies pleural infections also includes surgery, which is invasive and for which patient selection can be problematic. Most of the clinical literature published to date has used flat dosing of intrapleural fibrinolytic therapy in all subjects but little is known about how that strategy influences the processing of the administered fibrinolysin or how this influences outcomes. We developed a new test of pleural fluids ex vivo, which is called the Fibrinolytic Potential or FP, in which a dose of a fibrinolysin is added to pleural fluids ex vivo after which the fibrinolytic activity is measured and normalized to baseline levels. Testing in preclinical and clinical empyema fluids reveals a wide range of responses, indicating that individual patients will likely respond differently to flat dosing of fibrinolysins. The test remains under development but is envisioned as a guide for dosing of these agents, representing a novel candidate approach to personalization of intrapleural fibrinolytic therapy.

11.
J Biol Chem ; 290(15): 9428-41, 2015 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-25648892

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a fatal lung disease characterized by progressive interstitial scarification. A hallmark morphological lesion is the accumulation of myofibroblasts or fibrotic lung fibroblasts (FL-fibroblasts) in areas called fibroblastic foci. We previously demonstrated that the expression of both urokinase-type plasminogen activator (uPA) and the uPA receptor are elevated in FL-fibroblasts from the lungs of patients with IPF. FL-fibroblasts isolated from human IPF lungs and from mice with bleomycin-induced pulmonary fibrosis showed an increased rate of proliferation compared with normal lung fibroblasts (NL-fibroblasts) derived from histologically "normal" lung. Basal expression of plasminogen activator inhibitor-1 (PAI-1) in human and murine FL-fibroblasts was reduced, whereas collagen-I and α-smooth muscle actin were markedly elevated. Conversely, alveolar type II epithelial cells surrounding the fibrotic foci in situ, as well as those isolated from IPF lungs, showed increased activation of caspase-3 and PAI-1 with a parallel reduction in uPA expression. Transduction of an adenovirus PAI-1 cDNA construct (Ad-PAI-1) suppressed expression of uPA and collagen-I and attenuated proliferation in FL-fibroblasts. On the contrary, inhibition of basal PAI-1 in NL-fibroblasts increased collagen-I and α-smooth muscle actin. Fibroblasts isolated from PAI-1-deficient mice without lung injury also showed increased collagen-I and uPA. These changes were associated with increased Akt/phosphatase and tensin homolog proliferation/survival signals in FL-fibroblasts, which were reversed by transduction with Ad-PAI-1. This study defines a new role of PAI-1 in the control of fibroblast activation and expansion and its role in the pathogenesis of fibrosing lung disease and, in particular, IPF.


Asunto(s)
Células Epiteliales/metabolismo , Fibroblastos/metabolismo , Fibrosis Pulmonar Idiopática/metabolismo , Inhibidor 1 de Activador Plasminogénico/metabolismo , Alveolos Pulmonares/metabolismo , Actinas/genética , Actinas/metabolismo , Animales , Apoptosis/genética , Bleomicina , Western Blotting , Proliferación Celular/genética , Células Cultivadas , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Células Epiteliales/patología , Fibroblastos/patología , Expresión Génica , Fibrosis Pulmonar Idiopática/genética , Fibrosis Pulmonar Idiopática/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Músculo Liso/metabolismo , Inhibidor 1 de Activador Plasminogénico/genética , Alveolos Pulmonares/patología , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Activador de Plasminógeno de Tipo Uroquinasa/genética , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo
12.
Am J Physiol Lung Cell Mol Physiol ; 311(1): L39-47, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27190059

RESUMEN

Lung endothelial damage contributes to the pathogenesis of acute lung injury. New strategies against lung endothelial barrier dysfunction may provide therapeutic benefits against lung vascular injury. Cell-cell junctions and microtubule cytoskeleton are basic components in maintaining endothelial barrier integrity. HDAC6, a deacetylase primarily localized in the cytoplasm, has been reported to modulate nonnuclear protein function through deacetylation. Both α-tubulin and ß-catenin are substrates for HDAC6. Here, we examined the effects of tubastatin A, a highly selective HDAC6 inhibitor, on TNF-α induced lung endothelial cell barrier disruption and endotoxin-induced pulmonary edema. Selective HDAC6 inhibition by tubastatin A blocked TNF-α-induced lung endothelial cell hyperpermeability, which was associated with increased α-tubulin acetylation and microtubule stability. Tubastatin A pretreatment inhibited TNF-α-induced endothelial cell contraction and actin stress fiber formation with reduced myosin light chain phosphorylation. Selective HDAC6 inhibition by tubastatin A also induced ß-catenin acetylation in human lung endothelial cells, which was associated with increased membrane localization of ß-catenin and stabilization of adherens junctions. HDAC6 knockdown by small interfering RNA also prevented TNF-α-induced barrier dysfunction and increased α-tubulin and ß-catenin acetylation in endothelial cells. Furthermore, in a mouse model of endotoxemia, tubastatin A was able to prevent endotoxin-induced deacetylation of α-tubulin and ß-catenin in lung tissues, which was associated with reduced pulmonary edema. Collectively, our data indicate that selective HDAC6 inhibition by tubastatin A is a potent approach against lung endothelial barrier dysfunction.


Asunto(s)
Células Endoteliales/inmunología , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Ácidos Hidroxámicos/farmacología , Indoles/farmacología , Edema Pulmonar/prevención & control , Factor de Necrosis Tumoral alfa/fisiología , Acetilación , Animales , Permeabilidad Capilar/efectos de los fármacos , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/inmunología , Endotelio Vascular/metabolismo , Histona Desacetilasa 6 , Humanos , Lipopolisacáridos/farmacología , Masculino , Ratones Endogámicos C57BL , Microtúbulos/metabolismo , Multimerización de Proteína , Procesamiento Proteico-Postraduccional , Edema Pulmonar/inmunología , Edema Pulmonar/metabolismo , Tubulina (Proteína)/metabolismo
13.
Am J Physiol Lung Cell Mol Physiol ; 310(6): L496-506, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26747783

RESUMEN

We previously demonstrated that tumor suppressor protein p53 augments plasminogen activator inhibitor-1 (PAI-1) expression in alveolar epithelial cells (AECs) during chronic cigarette smoke (CS) exposure-induced lung injury. Chronic lung inflammation with elevated p53 and PAI-1 expression in AECs and increased susceptibility to and exacerbation of respiratory infections are all associated with chronic obstructive pulmonary disease (COPD). We recently demonstrated that preventing p53 from binding to the endogenous PAI-1 mRNA in AECs by either suppressing p53 expression or blockading p53 interactions with the PAI-1 mRNA mitigates apoptosis and lung injury. Within this context, we now show increased expression of the C-X-C chemokines (CXCL1 and CXCL2) and their receptor CXCR2, and the intercellular cellular adhesion molecule-1 (ICAM-1), in the lung tissues of patients with COPD. We also found a similar increase in lung tissues and AECs from wild-type (WT) mice exposed to passive CS for 20 wk and in primary AECs treated with CS extract in vitro. Interestingly, passive CS exposure of mice lacking either p53 or PAI-1 expression resisted an increase in CXCL1, CXCL2, CXCR2, and ICAM-1. Furthermore, inhibition of p53-mediated induction of PAI-1 expression by treatment of WT mice exposed to passive CS with caveolin-1 scaffolding domain peptide reduced CXCL1, CXCL2, and CXCR2 levels and lung inflammation. Our study reveals that p53-mediated induction of PAI-1 expression due to chronic CS exposure exacerbates lung inflammation through elaboration of CXCL1, CXCL2, and CXCR2. We further provide evidence that targeting this pathway mitigates lung injury associated with chronic CS exposure.


Asunto(s)
Quimiocina CXCL1/metabolismo , Quimiocina CXCL2/metabolismo , Pulmón/metabolismo , Receptores de Interleucina-8B/metabolismo , Serpina E2/fisiología , Humo/efectos adversos , Proteína p53 Supresora de Tumor/fisiología , Animales , Humanos , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Pulmón/inmunología , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Enfermedad Pulmonar Obstructiva Crónica/etiología , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Receptores de Interleucina-8B/genética , Fumar/efectos adversos , Nicotiana/efectos adversos , Activación Transcripcional
14.
Am J Pathol ; 185(1): 55-68, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25447049

RESUMEN

Alveolar type II epithelial (ATII) cell injury precedes development of pulmonary fibrosis. Mice lacking urokinase-type plasminogen activator (uPA) are highly susceptible, whereas those deficient in plasminogen activator inhibitor (PAI-1) are resistant to lung injury and pulmonary fibrosis. Epithelial-mesenchymal transition (EMT) has been considered, at least in part, as a source of myofibroblast formation during fibrogenesis. However, the contribution of altered expression of major components of the uPA system on ATII cell EMT during lung injury is not well understood. To investigate whether changes in uPA and PAI-1 by ATII cells contribute to EMT, ATII cells from patients with idiopathic pulmonary fibrosis and chronic obstructive pulmonary disease, and mice with bleomycin-, transforming growth factor ß-, or passive cigarette smoke-induced lung injury were analyzed for uPA, PAI-1, and EMT markers. We found reduced expression of E-cadherin and zona occludens-1, whereas collagen-I and α-smooth muscle actin were increased in ATII cells isolated from injured lungs. These changes were associated with a parallel increase in PAI-1 and reduced uPA expression. Further, inhibition of Src kinase activity using caveolin-1 scaffolding domain peptide suppressed bleomycin-, transforming growth factor ß-, or passive cigarette smoke-induced EMT and restored uPA expression while suppressing PAI-1. These studies show that induction of PAI-1 and inhibition of uPA during fibrosing lung injury lead to EMT in ATII cells.


Asunto(s)
Transición Epitelial-Mesenquimal/fisiología , Fibrinólisis , Lesión Pulmonar/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/fisiología , Actinas/metabolismo , Animales , Bleomicina/efectos adversos , Cadherinas/metabolismo , Colágeno Tipo I/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/fisiopatología , Pulmón/metabolismo , Lesión Pulmonar/fisiopatología , Ratones , Ratones Endogámicos C57BL , Fosforilación , Inhibidor 1 de Activador Plasminogénico/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/fisiopatología , Factores de Riesgo , Serpina E2/metabolismo , Fumar , Factor de Crecimiento Transformador beta/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteína de la Zonula Occludens-1/metabolismo
15.
Am J Physiol Lung Cell Mol Physiol ; 308(8): L847-53, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25659903

RESUMEN

Excessive inflammation is a major cause of organ damage during sepsis. The elderly are highly susceptible to sepsis-induced organ injury. Sirt1 expression is reduced during aging. In the present study, we investigated the role of Sirt1, a histone deacetylase, in controlling inflammatory responses in a murine sepsis model induced by cecal ligation and puncture (CLP). We examined lung inflammatory signaling in inducible Sirt1 knockout (Sirt1(-/-)) mice and wild-type littermates (Sirt1(+/+)) after CLP. Our results demonstrated that Sirt1 deficiency led to severe lung inflammatory injury. To further investigate molecular mechanisms of Sirt1 regulation of lung inflammatory responses in sepsis, we conducted a series of experiments to assess lung inflammasome activation after CLP. We detected increased lung inflammatory signaling including NF-κB, signal transducer and activator of transcription 3, and ERK1/2 activation in Sirt1(-/-) mice after CLP. Furthermore, inflammasome activity was increased in Sirt1(-/-) mice after CLP, as demonstrated by increased IL-1ß and caspase-7 cleavage and activation. Aggravated inflammasome activation in Sirt1(-/-) mice was associated with the increased production of lung proinflammatory mediators, including ICAM-1 and high-mobility group box 1, and further disruption of tight junctions and adherens junctions, as demonstrated by dramatic reduction of lung claudin-1 and vascular endothelial-cadherin expression, which was associated with the upregulation of matrix metallopeptidase 9 expression. In summary, our results suggest that Sirt1 suppresses acute lung inflammation during sepsis by controlling inflammasome activation pathway.


Asunto(s)
Inflamasomas/metabolismo , Pulmón/enzimología , Sepsis/inmunología , Sirtuina 1/fisiología , Animales , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Modelos Animales de Enfermedad , Femenino , Proteína HMGB1/metabolismo , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Pulmón/inmunología , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones Noqueados , Sepsis/enzimología
16.
Am J Physiol Lung Cell Mol Physiol ; 309(11): L1376-81, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26453518

RESUMEN

Overwhelming acute inflammation often leads to tissue damage during endotoxemia. In the present study, we investigated the role of Lyn, a member of the Src family tyrosine kinases, in modulating inflammatory responses in a murine model of endotoxemia. We examined lung inflammatory signaling in Lyn knockout (Lyn(-/-)) mice and wild-type littermates (Lyn(+/+)) during endotoxemia. Our data indicate that Lyn deletion aggravates endotoxin-induced pulmonary inflammation and proinflammatory signaling. We found increased activation of proinflammatory transcription factor NF-κB in the lung tissues of Lyn(-/-) mice after endotoxin challenge. Furthermore, during endotoxemia, the lung tissues of Lyn(-/-) mice showed increased inflammasome activation indicated by augmented caspase-1 and IL-1ß cleavage and activation. The aggravated lung inflammatory signaling in Lyn(-/-) mice was associated with increased production of proinflammatory mediators and elevated matrix metallopeptidase 9 and reduced VE-cadherin levels. Our results suggest that Lyn kinase modulates inhibitory signaling to suppress endotoxin-induced lung inflammation.


Asunto(s)
Eliminación de Gen , Neumonía/enzimología , Neumonía/patología , Familia-src Quinasas/metabolismo , Animales , Antígenos CD/metabolismo , Cadherinas/metabolismo , Caspasa 1/metabolismo , Inflamasomas/metabolismo , Mediadores de Inflamación/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Interleucina-1beta/metabolismo , Lipopolisacáridos , Pulmón/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones Noqueados , FN-kappa B/metabolismo , Familia-src Quinasas/deficiencia
17.
Toxicol Appl Pharmacol ; 283(2): 92-8, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25596429

RESUMEN

Silica is the major component of airborne dust generated by wind, manufacturing and/or demolition. Chronic occupational inhalation of silica dust containing crystalline quartz is by far the predominant form of silicosis in humans. Silicosis is a progressive lung disease that typically arises after a very long latency and is a major occupational concern with no known effective treatment. The mechanism of silicosis is not clearly understood. However, silicosis is associated with increased cell death, expression of redox enzymes and pro-fibrotic cytokines and chemokines. Since alveolar epithelial cell (AEC) death and disruption of alveolar fibrinolysis is often associated with both acute and chronic lung injuries, we explored whether p53-mediated changes in the urokinase-type plasminogen activator (uPA) system contributes to silica-induced lung injury. We further sought to determine whether caveolin-1 scaffolding domain peptide (CSP), which inhibits p53 expression, mitigates lung injury associated with exposure to silica. Lung tissues and AECs isolated from wild-type (WT) mice exposed to silica exhibit increased apoptosis, p53 and PAI-1, and suppression of uPA expression. Treatment of WT mice with CSP inhibits PAI-1, restores uPA expression and prevents AEC apoptosis by suppressing p53, which is otherwise induced in mice exposed to silica. The process involves CSP-mediated inhibition of serine-15 phosphorylation of p53 by inhibition of protein phosphatase 2A-C (PP2A-C) interaction with silica-induced caveolin-1 in AECs. These observations suggest that changes in the p53-uPA fibrinolytic system cross-talk contribute to lung injury caused by inhalation of silica dust containing crystalline quartz and is protected by CSP by targeting this pathway.


Asunto(s)
Fibrinólisis/efectos de los fármacos , Fibrinólisis/fisiología , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/metabolismo , Cuarzo/toxicidad , Proteína p53 Supresora de Tumor/fisiología , Animales , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo
18.
Am J Physiol Lung Cell Mol Physiol ; 307(8): L609-17, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25172911

RESUMEN

Epithelial sodium channels (ENaC) govern transepithelial salt and fluid homeostasis. ENaC contributes to polarization, apoptosis, epithelial-mesenchymal transformation, etc. Fibrinolytic proteases play a crucial role in virtually all of these processes and are elaborated by the airway epithelium. We hypothesized that urokinase-like plasminogen activator (uPA) regulates ENaC function in airway epithelial cells and tested that possibility in primary murine tracheal epithelial cells (MTE). Both basal and cAMP-activated Na(+) flow through ENaC were significantly reduced in monolayers of uPA-deficient cells. The reduction in ENaC activity was further confirmed in basolateral membrane-permeabilized cells. A decrease in the Na(+)-K(+)-ATPase activity in the basolateral membrane could contribute to the attenuation of ENaC function in intact monolayer cells. Dysfunctional fluid resolution was seen in uPA-disrupted cells. Administration of uPA and plasmin partially restores ENaC activity and fluid reabsorption by MTEs. ERK1/2, but not Akt, phosphorylation was observed in the cells and lungs of uPA-deficient mice. On the other hand, cleavage of γ ENaC is significantly depressed in the lungs of uPA knockout mice vs. those of wild-type controls. Expression of caspase 8, however, did not differ between wild-type and uPA(-/-) mice. In addition, uPA deficiency did not alter transepithelial resistance. Taken together, the mechanisms for the regulation of ENaC by uPA in MTEs include augmentation of Na(+)-K(+)-ATPase, proteolysis, and restriction of ERK1/2 phosphorylation. We demonstrate for the first time that ENaC may serve as a downstream signaling target by which uPA controls the biophysical profiles of airway fluid and epithelial function.


Asunto(s)
Células Epiteliales/metabolismo , Canales Epiteliales de Sodio/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Tráquea/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/fisiología , Animales , Apoptosis , Permeabilidad de la Membrana Celular , Células Cultivadas , Células Epiteliales/citología , Canales Epiteliales de Sodio/química , Canales Epiteliales de Sodio/genética , Immunoblotting , Transporte Iónico , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oocitos/citología , Oocitos/metabolismo , Fosforilación , Proteolisis , Alveolos Pulmonares/citología , Alveolos Pulmonares/metabolismo , ARN Interferente Pequeño/genética , Tráquea/citología , Xenopus laevis/metabolismo
19.
Am J Pathol ; 183(1): 131-43, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23665346

RESUMEN

Alveolar type II epithelial cell (ATII) apoptosis and proliferation of mesenchymal cells are the hallmarks of idiopathic pulmonary fibrosis, a devastating disease of unknown cause characterized by alveolar epithelial injury and progressive fibrosis. We used a mouse model of bleomycin (BLM)-induced lung injury to understand the involvement of p53-mediated changes in urokinase-type plasminogen activator (uPA) and plasminogen activator inhibitor-1 (PAI-1) levels in the regulation of alveolar epithelial injury. We found marked induction of p53 in ATII cells from mice exposed to BLM. Transgenic mice expressing transcriptionally inactive dominant negative p53 in ATII cells showed augmented apoptosis, whereas those deficient in p53 resisted BLM-induced ATII cell apoptosis. Inhibition of p53 transcription failed to suppress PAI-1 or induce uPA mRNA in BLM-treated ATII cells. ATII cells from mice with BLM injury showed augmented binding of p53 to uPA, uPA receptor (uPAR), and PAI-1 mRNA. p53-binding sequences from uPA, uPAR, and PAI-1 mRNA 3' untranslated regions neither interfered with p53 DNA binding activity nor p53-mediated promoter transactivation. However, increased expression of p53-binding sequences from uPA, uPAR, and PAI-1 mRNA 3' untranslated regions in ATII cells suppressed PAI-1 and induced uPA after BLM treatment, leading to inhibition of ATII cell apoptosis and pulmonary fibrosis. Our findings indicate that disruption of p53-fibrinolytic system cross talk may serve as a novel intervention strategy to prevent lung injury and pulmonary fibrosis.


Asunto(s)
Lesión Pulmonar/metabolismo , Inhibidor 1 de Activador Plasminogénico/metabolismo , Alveolos Pulmonares/metabolismo , Fibrosis Pulmonar/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Animales , Biomarcadores/metabolismo , Bleomicina , Western Blotting , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Inmunohistoquímica , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Alveolos Pulmonares/patología , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/patología
20.
Toxicol Appl Pharmacol ; 277(2): 118-23, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24709673

RESUMEN

The presence of endotoxin in blood can lead to acute kidney injury (AKI) and septic shock. Resolvins, the endogenous lipid mediators derived from docosahexaenoic acid, have been reported to exhibit potent anti-inflammatory action. Using a mouse model of lipopolysaccharide (LPS)-induced AKI, we investigated the effects of aspirin-triggered resolvin D1 (AT-RvD1) on inflammatory kidney injury. Administration of AT-RvD1 1h after LPS challenge protected the mice from kidney injury as indicated by the measurements of blood urea nitrogen, serum creatinine, and morphological alterations associated with tubular damage. The protective effects were evidenced by decreased neutrophil infiltration in the kidney indicating reduction in inflammation. AT-RvD1 treatment restored kidney cell junction protein claudin-4 expression, which was otherwise reduced after LPS challenge. AT-RvD1 treatment inhibited endotoxin-induced NF-κB activation and suppressed LPS-induced ICAM-1 and VCAM-1 expression in the kidney. Moreover, AT-RvD1 treatment markedly decreased LPS-induced IL-6 level in the kidney and blocked IL-6-mediated signaling including STAT3 and ERK phosphorylation. Our findings demonstrate that AT-RvD1 is a potent anti-inflammatory mediator in LPS-induced kidney injury, and AT-RvD1 has therapeutic potential against AKI during endotoxemia.


Asunto(s)
Lesión Renal Aguda/prevención & control , Antiinflamatorios no Esteroideos/farmacología , Aspirina/farmacología , Ácidos Docosahexaenoicos/metabolismo , Endotoxinas , Mediadores de Inflamación/metabolismo , Riñón/efectos de los fármacos , Lesión Renal Aguda/sangre , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/inmunología , Lesión Renal Aguda/patología , Animales , Biomarcadores/sangre , Nitrógeno de la Urea Sanguínea , Claudina-4/metabolismo , Creatinina/sangre , Citoprotección , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Interleucina-6/metabolismo , Riñón/inmunología , Riñón/metabolismo , Riñón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Infiltración Neutrófila/efectos de los fármacos , Fosforilación , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Molécula 1 de Adhesión Celular Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA