Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Adv Anat Embryol Cell Biol ; 228: 41-61, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29288385

RESUMO

Pulmonary blood vessels act as a well-regulated barrier to the flux of fluid and solutes between the lumen and the air space. Perturbation of the barrier function results in excessive fluid leak into the interstitium and alveoli, and impairs gas exchange. Recent studies provide deeper insight into the precise control mechanisms involved in the regulation of the barrier. This chapter will highlight these mechanisms and discuss the current understanding on the fluid and solute transport pathways across the vascular endothelial layer. In addition, the chapter summarizes the contributions of extra-endothelial structures such as pericytes and glycocalyx in regulating fluid flux across pulmonary vessels. The chapter concludes with an analysis on the impact of pulmonary endothelial heterogeneity and experimental models on current interpretations of barrier function and regulatory mechanisms.


Assuntos
Barreira Alveolocapilar/fisiologia , Endotélio Vascular/fisiologia , Pulmão/fisiologia , Mucosa Respiratória/fisiologia , Animais , Transporte Biológico/fisiologia , Barreira Alveolocapilar/citologia , Capilares/citologia , Capilares/fisiologia , Células Endoteliais/fisiologia , Endotélio Vascular/citologia , Glicocálix/fisiologia , Humanos , Pulmão/irrigação sanguínea , Pulmão/citologia , Modelos Animais , Pericitos/fisiologia , Artéria Pulmonar/citologia , Artéria Pulmonar/fisiologia , Veias Pulmonares/citologia , Veias Pulmonares/fisiologia , Mucosa Respiratória/citologia , Equilíbrio Hidroeletrolítico/fisiologia
2.
JCI Insight ; 2(11)2017 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-28570277

RESUMO

Pulmonary function is dependent upon the precise regulation of alveolar surfactant. Alterations in pulmonary surfactant concentrations or function impair ventilation and cause tissue injury. Identification of the molecular pathways that sense and regulate endogenous alveolar surfactant concentrations, coupled with the ability to pharmacologically modulate them both positively and negatively, would be a major therapeutic advance for patients with acute and chronic lung diseases caused by disruption of surfactant homeostasis. The orphan adhesion GPCR GPR116 (also known as Adgrf5) is a critical regulator of alveolar surfactant concentrations. Here, we show that human and mouse GPR116 control surfactant secretion and reuptake in alveolar type II (AT2) cells by regulating guanine nucleotide-binding domain α q and 11 (Gq/11) signaling. Synthetic peptides derived from the ectodomain of GPR116 activated Gq/11-dependent inositol phosphate conversion, calcium mobilization, and cortical F-actin stabilization to inhibit surfactant secretion. AT2 cell-specific deletion of Gnaq and Gna11 phenocopied the accumulation of surfactant observed in Gpr116-/- mice. These data provide proof of concept that GPR116 is a plausible therapeutic target to modulate endogenous alveolar surfactant pools to treat pulmonary diseases associated with surfactant dysfunction.

3.
Am J Pathol ; 187(4): 921-935, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28188112

RESUMO

Activation of plasma membrane receptors initiates compartmentalized second messenger signaling. Whether this compartmentalization facilitates the preferential intercellular diffusion of specific second messengers is unclear. Toward this, the receptor-mediated agonist, thrombin, was instilled into microvessels in a restricted region of isolated blood-perfused mouse lungs. Subsequently, the thrombin-induced increase in endothelial F-actin was determined using confocal fluorescence microscopy. Increased F-actin was evident in microvessels directly treated with thrombin and in those located in adjoining thrombin-free regions. This increase was abrogated by inhibiting inositol trisphosphate-mediated calcium release with Xestospongin C (XeC). XeC also inhibited the thrombin-induced increase in the amplitude of endothelial cytosolic Ca2+ oscillations. Instillation of thrombin and XeC into adjacent restricted regions increased F-actin in microvessels in the thrombin-treated and adjacent regions but not in those in the XeC-treated region. Thus, inositol trisphosphate, and not calcium, diffused interendothelially to the spatially remote thrombin-free microvessels. Thus, activation of plasma membrane receptors increased the ambit of inflammatory responses via a second messenger different from that used by stimuli that induce cell-wide increases in second messengers. Thrombin however failed to induce the spatially extensive response in microvessels of mice lacking endothelial connexin43, suggesting a role for connexin43 gap junctions. Compartmental second messenger signaling and interendothelial communication define the specific second messenger involved in exacerbating proinflammatory responses to receptor-mediated agonists.


Assuntos
Inositol 1,4,5-Trifosfato/metabolismo , Pulmão/irrigação sanguínea , Microvasos/metabolismo , Trombina/farmacologia , Actinas/metabolismo , Animais , Cálcio/metabolismo , Comunicação Celular/efeitos dos fármacos , Conexina 43/metabolismo , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Camundongos Endogâmicos C57BL , Microvasos/efeitos dos fármacos , Permeabilidade/efeitos dos fármacos , Fibras de Estresse/efeitos dos fármacos , Fibras de Estresse/metabolismo
4.
Am J Physiol Lung Cell Mol Physiol ; 309(6): L584-92, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26163513

RESUMO

Endothelial barrier restoration reverses microvessel hyperpermeability and facilitates recovery from lung injury. Because inhibiting connexin 43 (Cx43)-dependent interendothelial communication blunts hyperpermeability in single microvessels, we determined whether endothelial Cx43 levels correlate with changes in microvessel permeability during recovery from lung injury. Toward this, bacterial endotoxin was instilled intratracheally into rat lungs, and at different durations postinstillation the lungs were isolated and blood perfused. Microvessel Cx43 expression was quantified by in situ immunofluorescence and microvessel permeability via a fluorescence method. To supplement the immunofluorescence data, protein levels were determined by immunoblots of lung tissue from endotoxin-instilled rats. Immunofluorescence and immunoblot together revealed that both Cx43 expression and microvessel permeability increased above baseline within a few hours after endotoxin instillation but declined progressively over the next few days. On day 5 postendotoxin, microvessel Cx43 declined to negligible levels, resulting in complete absence of intermicrovessel communication determined by photolytic uncaging of Ca(2+). However, by day 14, both Cx43 expression and microvessel permeability returned to baseline levels. In contrast to Cx43, expression of microvessel vascular endothelial (VE)-cadherin, a critical determinant of vascular barrier integrity, exhibited an inverse trend by initially declining below baseline and then returning to baseline at a longer duration. Knockdown of vascular Cx43 by tail vein injection of Cx43 shRNA increased VE-cadherin expression, suggesting that reduction in Cx43 levels may modulate VE-cadherin levels in lung microvessels. Together, the data suggest that endotoxin challenge initiates interrelated changes in microvessel Cx43, VE-cadherin, and microvessel permeability, with changes in Cx43 temporally leading the other responses.


Assuntos
Antígenos CD/metabolismo , Caderinas/metabolismo , Conexina 43/metabolismo , Pulmão/imunologia , Microvasos/metabolismo , Animais , Permeabilidade Capilar , Movimento Celular , Proliferação de Células , Células Cultivadas , Quimiocina CXCL2/antagonistas & inibidores , Quimiocina CXCL2/metabolismo , Conexina 43/genética , Lipopolissacarídeos/farmacologia , Pulmão/irrigação sanguínea , Pulmão/crescimento & desenvolvimento , Pulmão/metabolismo , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Neovascularização Fisiológica/imunologia , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais
5.
J Biol Chem ; 290(18): 11246-57, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-25762723

RESUMO

Diarrhea is one of the most common adverse side effects observed in ∼7% of individuals consuming Food and Drug Administration (FDA)-approved drugs. The mechanism of how these drugs alter fluid secretion in the gut and induce diarrhea is not clearly understood. Several drugs are either substrates or inhibitors of multidrug resistance protein 4 (MRP4), such as the anti-colon cancer drug irinotecan and an anti-retroviral used to treat HIV infection, 3'-azido-3'-deoxythymidine (AZT). These drugs activate cystic fibrosis transmembrane conductance regulator (CFTR)-mediated fluid secretion by inhibiting MRP4-mediated cAMP efflux. Binding of drugs to MRP4 augments the formation of MRP4-CFTR-containing macromolecular complexes that is mediated via scaffolding protein PDZK1. Importantly, HIV patients on AZT treatment demonstrate augmented MRP4-CFTR complex formation in the colon, which defines a novel paradigm of drug-induced diarrhea.


Assuntos
AMP Cíclico/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Diarreia/induzido quimicamente , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Animais , Camptotecina/efeitos adversos , Camptotecina/análogos & derivados , Aprovação de Drogas , Células HT29 , Humanos , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Irinotecano , Camundongos , Modelos Moleculares , Proteínas Associadas à Resistência a Múltiplos Medicamentos/química , Proteínas Associadas à Resistência a Múltiplos Medicamentos/deficiência , Conformação Proteica , Estados Unidos , United States Food and Drug Administration
6.
J Biol Chem ; 289(52): 35757-69, 2014 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-25542932

RESUMO

Chemotactic migration of fibroblasts toward growth factors relies on their capacity to sense minute extracellular gradients and respond to spatially confined receptor-mediated signals. Currently, mechanisms underlying the gradient sensing of fibroblasts remain poorly understood. Using single-particle tracking methodology, we determined that a lysophosphatidic acid (LPA) gradient induces a spatiotemporally restricted decrease in the mobility of LPA receptor 2 (LPA2) on chemotactic fibroblasts. The onset of decreased LPA2 mobility correlates to the spatial recruitment and coupling to LPA2-interacting proteins that anchor the complex to the cytoskeleton. These localized PDZ motif-mediated macromolecular complexes of LPA2 trigger a Ca(2+) puff gradient that governs gradient sensing and directional migration in response to LPA. Disruption of the PDZ motif-mediated assembly of the macromolecular complex of LPA2 disorganizes the gradient of Ca(2+) puffs, disrupts gradient sensing, and reduces the directional migration of fibroblasts toward LPA. Our findings illustrate that the asymmetric macromolecular complex formation of chemoattractant receptors mediates gradient sensing and provides a new mechanistic basis for models to describe gradient sensing of fibroblasts.


Assuntos
Fibroblastos/fisiologia , Microdomínios da Membrana/metabolismo , Receptores de Ácidos Lisofosfatídicos/metabolismo , Animais , Sinalização do Cálcio , Quimiotaxia , Lisofosfolipídeos/fisiologia , Camundongos , Células NIH 3T3 , Fosfolipase C beta/metabolismo , Multimerização Proteica , Transporte Proteico
7.
J Vis Exp ; (88): e51552, 2014 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-25045895

RESUMO

The isolated blood-perfused lung preparation is widely used to visualize and define signaling in single microvessels. By coupling this preparation with real time imaging, it becomes feasible to determine permeability changes in individual pulmonary microvessels. Herein we describe steps to isolate rat lungs and perfuse them with autologous blood. Then, we outline steps to infuse fluorophores or agents via a microcatheter into a small lung region. Using these procedures described, we determined permeability increases in rat lung microvessels in response to infusions of bacterial lipopolysaccharide. The data revealed that lipopolysaccharide increased fluid leak across both venular and capillary microvessel segments. Thus, this method makes it possible to compare permeability responses among vascular segments and thus, define any heterogeneity in the response. While commonly used methods to define lung permeability require postprocessing of lung tissue samples, the use of real time imaging obviates this requirement as evident from the present method. Thus, the isolated lung preparation combined with real time imaging offers several advantages over traditional methods to determine lung microvascular permeability, yet is a straightforward method to develop and implement.


Assuntos
Pulmão/irrigação sanguínea , Microscopia de Fluorescência/métodos , Modelos Animais , Animais , Permeabilidade Capilar , Dextranos/química , Dextranos/farmacocinética , Fluoresceína-5-Isotiocianato/análogos & derivados , Fluoresceína-5-Isotiocianato/química , Fluoresceína-5-Isotiocianato/farmacocinética , Corantes Fluorescentes/química , Corantes Fluorescentes/farmacocinética , Pulmão/cirurgia , Masculino , Microscopia de Fluorescência/instrumentação , Perfusão/métodos , Ratos , Ratos Sprague-Dawley
8.
FEBS J ; 281(3): 957-69, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24289040

RESUMO

Patients with acute lung injury are administered high concentrations of oxygen during mechanical ventilation, and while both hyperoxia and mechanical ventilation are necessary, each can independently cause additional injury. However, the precise mechanisms that lead to injury are not well understood. We hypothesized that alveolar epithelial cells may be more susceptible to injury caused by mechanical ventilation because hyperoxia causes cells to be stiffer due to increased filamentous actin (f-actin) formation via the GTPase RhoA and its effecter Rho kinase (ROCK). We examined cytoskeletal structures in cultured murine lung alveolar epithelial cells (MLE-12) under normoxic and hyperoxic (48 h) conditions. We also measured cell elasticity (E) using an atomic force microscope in the indenter mode. Hyperoxia caused increased f-actin stress fibers and bundle formation, an increase in g- and f-actin, an increase in nuclear area and a decrease in nuclear height, and cells became stiffer (higher E). Treatment with an inhibitor (Y-27632) of ROCK significantly decreased E and prevented the cytoskeletal changes, while it did not influence the nuclear height and area. Pre-exposure of cells to hyperoxia promoted detachment when cells were subsequently stretched cyclically, but the ROCK inhibitor prevented this effect. Hyperoxia caused thickening of vinculin focal adhesion plaques, and inhibition of ROCK reduced the formation of distinct focal adhesion plaques. Phosphorylation of focal adhesion kinase was significantly reduced by both hyperoxia and treatment with Y-27632. Hyperoxia caused increased cell stiffness and promoted cell detachment during stretch. These effects were ameliorated by inhibition of ROCK.


Assuntos
Citoesqueleto/química , Estresse Oxidativo , Oxigênio/efeitos adversos , Alvéolos Pulmonares/metabolismo , Mucosa Respiratória/metabolismo , Quinases Associadas a rho/metabolismo , Animais , Adesão Celular/efeitos dos fármacos , Linhagem Celular , Núcleo Celular/química , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Forma do Núcleo Celular/efeitos dos fármacos , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/metabolismo , Regulação para Baixo/efeitos dos fármacos , Módulo de Elasticidade/efeitos dos fármacos , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Camundongos , Microscopia de Força Atômica , Estresse Oxidativo/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Alvéolos Pulmonares/química , Alvéolos Pulmonares/citologia , Alvéolos Pulmonares/efeitos dos fármacos , Mucosa Respiratória/química , Mucosa Respiratória/citologia , Mucosa Respiratória/efeitos dos fármacos , Vinculina/metabolismo , Quinases Associadas a rho/antagonistas & inibidores
9.
PLoS One ; 8(5): e63465, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23675486

RESUMO

The pulmonary microvasculature plays a critical role in endotoxin-induced acute lung injury. However, the relevant signaling remain unclear. Specifically the role of endothelial Ca(2+) in the induction of endotoxin-mediated responses in lung microvessels remains undefined. Toward elucidating this, we used the isolated blood-perfused rat lung preparation. We loaded microvessels with the Ca(2+) indicator, Fura 2 AM and then determined Ca(2+) responses to infusions of lipopolysaccharide (LPS) into the microvessels. LPS induced a more than two-fold increase in the amplitude of cytosolic Ca(2+) oscillations. Inhibiting inositol 1,4,5 trisphosphate receptors on endoplasmic reticulum (ER) Ca(2+) stores with Xestospongin C (XeC), blocked the LPS-induced increase in the Ca(2+) oscillation amplitude. However, XeC did not affect entry of external Ca(2+) via plasma membrane Ca(2+) channels in lung microvascular endothelial cells. This suggested that LPS augmented the oscillations via release of Ca(2+) from ER stores. In addition, XeC also blocked LPS-mediated activation and nuclear translocation of nuclear factor-kappa B in lung microvessels. Further, inhibiting ER Ca(2+) release blunted increases in intercellular adhesion molecule-1 expression and retention of naïve leukocytes in LPS-treated microvessels. Taken together, the data suggest that LPS-mediated Ca(2+) release from ER stores underlies nuclear factor-kappa B activation and downstream inflammatory signaling in lung microvessels. Thus, we show for the first time a role for inositol 1,4,5 trisphosphate-mediated ER Ca(2+) release in the induction of LPS responses in pulmonary microvascular endothelium. Mechanisms that blunt this signaling may mitigate endotoxin-induced morbidity.


Assuntos
Cálcio/metabolismo , Citosol/metabolismo , Inflamação/metabolismo , Lipopolissacarídeos/farmacologia , Pulmão/metabolismo , Microvasos/efeitos dos fármacos , Microvasos/metabolismo , Animais , Sinalização do Cálcio/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Ativação Enzimática/efeitos dos fármacos , Inflamação/genética , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/metabolismo , Leucócitos/efeitos dos fármacos , Leucócitos/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/patologia , Masculino , NF-kappa B/metabolismo , Ratos
10.
Am J Physiol Lung Cell Mol Physiol ; 303(1): L33-42, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22561459

RESUMO

Acid aspiration, a common cause of acute lung injury, leads to alveolar edema. Increase in lung vascular permeability underlies this pathology. To define mechanisms, isolated rat lungs were perfused with autologous blood. Hydrochloric acid and rhodamine-dextran 70 kDa (RDx70) were coinstilled into an alveolus by micropuncture. RDx70 fluorescence was used to establish the spatial distribution of acid. Subsequently, FITC-dextran 20 kDa (FDx20) was infused into microvessels for 60 min followed by a 10-min HEPES-buffered saline wash. During the infusion, FITC fluorescence changes were recorded to quantify the ratio of peak to postwash fluorescence. The ratio, termed normalized fluorescence, was low for acid compared with buffer instillation both in microvessels abutting acid-treated alveoli and those located more than 700 µm away. In contrast, the normalized fluorescence was similar to buffer controls when a higher molecular weight tracer (FITC-dextran 70 kDa) was infused instead of FDx20, suggesting that normalized FDx20 fluorescence faithfully represented microvascular permeability. Inhibiting endothelial connexin43 (Cx43) gap junction communication with Gap27 blunted the acid-induced reduction in normalized fluorescence, although scrambled Gap27 did not have any effect. The blunting was evident not only in microvessels away from the site of injury, but also in those abutting directly injured alveoli. Thus the new fluorescence-based method reveals that acid increases microvascular permeability both at acid-instilled and away sites. Inhibiting endothelial Cx43 blocked the permeability increase even at the direct injury sites. These data indicate for the first time that Cx43-dependent mechanisms mediate acid-induced increases in microvascular permeability. Cx43 may be a therapeutic target in acid injury.


Assuntos
Ácidos/metabolismo , Permeabilidade Capilar/fisiologia , Conexina 43/metabolismo , Endotélio Vascular/fisiologia , Ácido Clorídrico/metabolismo , Pulmão/irrigação sanguínea , Alvéolos Pulmonares/fisiologia , Animais , Dextranos/metabolismo , Endotélio Vascular/metabolismo , Fluoresceína-5-Isotiocianato/análogos & derivados , Fluoresceína-5-Isotiocianato/metabolismo , Junções Comunicantes/metabolismo , Junções Comunicantes/fisiologia , Masculino , Alvéolos Pulmonares/metabolismo , Ratos , Ratos Sprague-Dawley
11.
Microvasc Res ; 83(3): 323-31, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22342350

RESUMO

Endotoxemia, a major feature of sepsis, is a common cause of acute lung injury and initiates rapid accumulation of leukocytes in the lung vasculature. Endothelial mechanisms that underlie this accumulation remain unclear, as current experimental models of endotoxemia are less suitable for targeted activation of the endothelium. Toward elucidating this, we used the isolated blood-perfused rat lung preparation. With a microcatheter inserted through a left atrial cannula, we cleared blood cells from a small lung region and then infused lipopolysaccharide (LPS) into microvessels. After a Ringer's wash to remove residual LPS, we infused fluorescently-labeled autologous leukocytes and imaged their transit through the treated microvessels. Image analysis revealed that leukocytes infused 90 min after LPS treatment were retained more in treated venules and capillaries than untreated vessels. Further, pretreatment with either the intercellular adhesion molecule-1 (ICAM-1) mAb or polymyxin-B blunted LPS-induced leukocyte retention in both microvessel segments. In addition, retention of leukocytes treated ex vivo with LPS in LPS-treated microvessels was higher compared to retention of untreated leukocytes. In situ immunofluorescence experiments revealed that LPS significantly increased microvessel ICAM-1 expression at 90 min post treatment. Polymyxin pretreatment inhibited this increase. Taken together, the data suggest that LPS increased leukocyte retention in both venules and capillaries and this response was mediated by the increased expression of endothelial ICAM-1. Thus, endothelial mechanisms may themselves play a major role in LPS-induced leukocyte retention in lung microvessels. Blunting the endothelial responses may mitigate endotoxin-induced morbidity.


Assuntos
Endotélio Vascular/patologia , Leucócitos/citologia , Lipopolissacarídeos/metabolismo , Pulmão/irrigação sanguínea , Microcirculação , Animais , Capilares/patologia , Diagnóstico por Imagem/métodos , Corantes Fluorescentes/farmacologia , Humanos , Molécula 1 de Adesão Intercelular/biossíntese , Selectina L/biossíntese , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência/métodos , Perfusão , Polimixina B/biossíntese , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Fatores de Tempo
12.
Am J Respir Cell Mol Biol ; 46(4): 461-9, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22052879

RESUMO

Both hyperoxia and mechanical ventilation can independently cause lung injury. In combination, these insults produce accelerated and severe lung injury. We recently reported that pre-exposure to hyperoxia for 12 hours, followed by ventilation with large tidal volumes, induced significant lung injury and epithelial cell apoptosis compared with either stimulus alone. We also reported that such injury and apoptosis are inhibited by antioxidant treatment. In this study, we hypothesized that apoptosis signal-regulating kinase-1 (ASK-1), a redox-sensitive, mitogen-activated protein kinase kinase kinase, plays a role in lung injury and apoptosis in this model. To determine the role of ASK-1 in lung injury, the release of inflammatory mediators and apoptosis, attributable to 12 hours of hyperoxia, were followed by large tidal volume mechanical ventilation with hyperoxia. Wild-type and ASK-1 knockout mice were subjected to hyperoxia (Fi(O(2)) = 0.9) for 12 hours before 4 hours of large tidal mechanical ventilation (tidal volume = 25 µl/g) with hyperoxia, and were compared with nonventilated control mice. Lung injury, apoptosis, and cytokine release were measured. The deletion of ASK-1 significantly inhibited lung injury and apoptosis, but did not affect the release of inflammatory mediators, compared with the wild-type mice. ASK-1 is an important regulator of lung injury and apoptosis in this model. Further study is needed to determine the mechanism of lung injury and apoptosis by ASK-1 and its downstream mediators in the lung.


Assuntos
MAP Quinase Quinase Quinase 5/genética , MAP Quinase Quinase Quinase 5/metabolismo , Lesão Pulmonar Induzida por Ventilação Mecânica/enzimologia , Lesão Pulmonar Induzida por Ventilação Mecânica/prevenção & controle , Animais , Apoptose/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Ativação Enzimática , Células Epiteliais/patologia , Feminino , Hiperóxia/enzimologia , Mediadores da Inflamação/metabolismo , Masculino , Camundongos , Camundongos Knockout , Alvéolos Pulmonares/patologia , Lesão Pulmonar Induzida por Ventilação Mecânica/patologia
13.
Anat Rec (Hoboken) ; 294(9): 1585-91, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21809471

RESUMO

Acid aspiration is a major cause of acute lung injury. However, the mechanisms that underlie this spatial expansion of the injury remain undefined. In current animal models of acid injury, intratracheal acid instillation replicates the lung injury. However intratracheal instillation causes a global effect, precluding studies of how the injury spreads. Here, we report an airway catheter-based method for localized acid delivery in the isolated blood-perfused rat lung. We co-instilled hydrochloric acid with evans blue through the catheter into one lung and determined blood-free extravascular lung water in tissue samples from regions that either received, or did not receive the instilled acid. Tissue samples from the noncatheterized contralateral lung were used as controls. Lung water increased both in the regions that received acid, as well as in adjacent regions that did not. Pretreating the lung with vascular infusions of the gap junctional blocker, glycerrhetinic acid, blunted the acid-induced lung water increase at the adjacent regions. These findings indicate that endothelial gap junction communication causes spread of lung injury from regions that were directly acid injured, to adjacent sites that did not directly receive acid. Our new method for establishing localized acid injury provides evidence for a novel role for vascular gap junctions in the spatial expansion of acid injury.


Assuntos
Lesão Pulmonar Aguda/metabolismo , Endotélio Vascular/metabolismo , Junções Comunicantes/metabolismo , Ácido Clorídrico/toxicidade , Pulmão/irrigação sanguínea , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/patologia , Animais , Permeabilidade Capilar/efeitos dos fármacos , Cateterismo , Endotélio Vascular/efeitos dos fármacos , Junções Comunicantes/efeitos dos fármacos , Ácido Clorídrico/administração & dosagem , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley
14.
Am J Physiol Lung Cell Mol Physiol ; 299(5): L711-9, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20833778

RESUMO

Both high tidal volume mechanical ventilation (HV) and hyperoxia (HO) have been implicated in ventilator-induced lung injury. However, patients with acute lung injury are often exposed to HO before the application of mechanical ventilation. The potential priming of the lungs for subsequent injury by exposure to HO has not been extensively studied. We provide evidence that HO (90%) for 12 h followed by HV (25 µl/g) combined with HO for 2 or 4 h (HO-12h+HVHO-2h or -4h) induced severe lung injury in mice. Analysis of lung homogenates showed that lung injury was associated with cleavage of executioner caspases, caspases-3 and -7, and their downstream substrate poly(ADP-ribose) polymerase-1 (PARP-1). No significant lung injury or caspase cleavage was seen with either HO for 16 h or HV for up to 4 h. Ventilation for 4 h with HO (HVHO) did not cause significant lung injury without preexposure to HO. Twelve-hour HO followed by lower tidal volume (6 µl/g) mechanical ventilation failed to produce significant injury or caspase cleavage. We also evaluated the initiator caspases, caspases-8 and -9, to determine whether the death receptor or mitochondrial-mediated pathways were involved. Caspase-9 cleavage was observed in HO-12h+HVHO-2h and -4h as well as HO for 16 h. Caspase-8 activation was observed only in HO-12h+HVHO-4h, indicating the involvement of both pathways. Immunohistochemistry and in vitro stretch studies showed caspase cleavage in alveolar epithelial cells. In conclusion, preexposure to HO followed by HV produced severe lung injury associated with alveolar epithelial cell apoptosis.


Assuntos
Apoptose/fisiologia , Células Epiteliais/patologia , Células Epiteliais/fisiologia , Hiperóxia/complicações , Volume de Ventilação Pulmonar , Lesão Pulmonar Induzida por Ventilação Mecânica/etiologia , Lesão Pulmonar Induzida por Ventilação Mecânica/patologia , Animais , Caspases/metabolismo , Linhagem Celular , Ativação Enzimática , Células Epiteliais/citologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Poli Adenosina Difosfato Ribose/metabolismo , Alvéolos Pulmonares/citologia , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/patologia , Respiração Artificial/efeitos adversos , Estresse Mecânico
15.
Am J Physiol Lung Cell Mol Physiol ; 296(6): L901-10, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19304909

RESUMO

Although the vascular bed is a major source of nitric oxide (NO) production, factors regulating the production remain unclear. We considered the role played by paracrine signaling. Determinations by fluorescence microscopy in isolated, blood-perfused rat and mouse lungs revealed that a brief lung expansion enhanced cytosolic Ca(2+) (Ca(2+)cyt) oscillations in alveolar epithelial (AEC) and endothelial (EC) cells, and NO production in EC. Furthermore, as assessed by a novel microlavage assay, alveolar ATP production increased. Intra-alveolar microinfusion of the purinergic receptor antagonist, PPADS, and the nucleotide hydrolyzing enzyme, apyrase, each completely blocked the Ca(2+)cyt and NO responses in EC. Lung expansion induced Ca(2+)cyt oscillations in mice lacking the P2Y1, but not the P2Y2, purinergic receptors, which were located in the perivascular interstitium basolateral to AEC. Prolonged lung expansion instituted by mechanical ventilation at high tidal volume increased EC expression of nitrotyrosine, indicating development of nitrosative stress in lung microvessels. These findings reveal a novel mechanism in which mechanically induced purinergic signaling couples cross-compartmental Ca(2+)cyt oscillations to microvascular NO production.


Assuntos
Sinalização do Cálcio/fisiologia , Óxido Nítrico Sintase Tipo III/metabolismo , Comunicação Parácrina/fisiologia , Alvéolos Pulmonares/enzimologia , Animais , Citosol/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Microcirculação/fisiologia , Óxido Nítrico/metabolismo , Alvéolos Pulmonares/irrigação sanguínea , Circulação Pulmonar/fisiologia , Ratos , Ratos Sprague-Dawley , Receptores Purinérgicos P2/genética , Receptores Purinérgicos P2/metabolismo , Receptores Purinérgicos P2Y1 , Receptores Purinérgicos P2Y2 , Tirosina/análogos & derivados , Tirosina/metabolismo
16.
Mol Med ; 13(11-12): 615-24, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17948065

RESUMO

A number of Lys-Pro-containing short peptides have been described as possessing a variety of biological activities in vitro. Because of limited metabolic stability, however, their efficacy in vivo is uncertain. To exploit the pharmacological potential of Lys-Pro-containing short peptides, we synthesized a series of chemically modified forms of these peptides. One of them, ITF1697 (Gly-(Nalpha-Et)Lys-Pro-Arg) was stable in vivo and particularly efficacious in experimental models of disseminated endotoxemia and of cardiovascular disorders. Using intravital fluorescence microscopy, we studied the peptide cellular and molecular basis of protection in the Syrian hamster cheek pouch microcirculation subjected to ischemia/reperfusion (I/R) and in pressure elevation-induced proinflammatory responses in isolated Sprague-Dawley rat lungs. Continuous intravenous infusion of ITF1697 at 0.1 to 100 mug/kg/min nearly completely protected the cheek pouch microcirculation from I/R injury as measured by decreased vascular permeability and increased capillary perfusion. Adhesion of leukocytes and platelets to blood vessels was strongly inhibited by the peptide. ITF1697 exerted its activity at the early stages of endothelial activation and inhibited P-selectin and von Willebrand factor secretion. Further mechanistic studies in the rat lung preparation revealed that the peptide inhibited the intracellular Ca(2+)-dependent fusion of Weibel-Palade bodies with the plasma membrane. The ability of ITF1697 to inhibit the early functions of activated endothelial cells, such as the exocytosis of Weibel-Palade bodies, represents a novel and promising pharmacological tool in model of pathologies of a variety of microvascular disorders.


Assuntos
Exocitose/efeitos dos fármacos , Oligopeptídeos/farmacologia , Traumatismo por Reperfusão/metabolismo , Corpos de Weibel-Palade/efeitos dos fármacos , Animais , Cricetinae , Masculino , Pressão , Ratos , Ratos Sprague-Dawley , Corpos de Weibel-Palade/metabolismo
17.
Am J Respir Cell Mol Biol ; 36(6): 688-96, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17290033

RESUMO

The liquid layer lining the pulmonary alveolar wall critically determines the lung's immune defense against inhaled pathogens, because it provides a liquid milieu in the air-filled alveolus for dispersal of immune cells and defensive surfactant proteins. However, mechanisms underlying formation of the liquid are unknown. We achieved visualization of the alveolar wall liquid (AWL) in situ in mouse lungs by means of optical-sectioning microscopy. Continuous liquid secretion was present in alveoli of wild-type (WT) mice under baseline conditions. This secretion was blocked by inhibitors of the cystic fibrosis transmembrane regulator (CFTR). The secretion was absent in Cftr(-/-) mice, and it was blocked when chloride was depleted from the perfusate of WT mice, providing the first evidence that CFTR-dependent chloride secretion causes AWL formation. Injected microparticles demonstrated flow of the AWL. The flow was blocked by CFTR inhibition and was absent in Cftr(-/-) mice. We conclude that CFTR-dependent liquid secretion is present in alveoli of the adult mouse. Defective alveolar secretion might impair alveolar immune defense and promote alveolar disease.


Assuntos
Líquidos Corporais/química , Cloretos/metabolismo , Microscopia de Fluorescência/métodos , Alvéolos Pulmonares/metabolismo , Animais , Líquidos Corporais/imunologia , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Dextranos/metabolismo , Fluoresceína-5-Isotiocianato/análogos & derivados , Fluoresceína-5-Isotiocianato/metabolismo , Corantes Fluorescentes/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Alvéolos Pulmonares/imunologia
18.
J Appl Physiol (1985) ; 102(3): 1255-64, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17095639

RESUMO

Although proinflammatory cell signaling in the alveolo-capillary region predisposes to acute lung injury, key cell-signaling mechanisms remain inadequately understood. Alveolo-capillary inflammation is likely to involve coordinated signaling among cells of different phenotypes. For example, migration of inflammatory cells into the alveolus might entail coordinated signaling between adjoining alveolar epithelial and microvascular endothelial cells. The popular cultured cell experimental strategy fails to replicate this multicellular environment. Cultured lung cells, both alveolar and endothelial, undergo phenotypic transformations; hence they might inadequately reflect innate responses of native cells. Consequently, new approaches are required for the investigation of cell signaling in the native setting. Here we summarize new developments in classical intravital microscopy and discuss real-time fluorescence imaging as a novel technique for studying second-messenger mechanisms in the alveolo-capillary region.


Assuntos
Microscopia/métodos , Alvéolos Pulmonares/citologia , Animais , Corantes Fluorescentes
19.
J Clin Invest ; 116(8): 2193-200, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16878174

RESUMO

Acute lung injury (ALI), which is associated with a mortality of 30-40%, is attributable to inflammation that develops rapidly across the lung's vast vascular surface, involving an entire lung or even both lungs. No specific mechanism explains this extensive inflammatory spread, probably because of the lack of approaches for detecting signal conduction in lung capillaries. Here, we addressed this question by applying the photolytic uncaging approach to induce focal increases in Ca2+ levels in targeted endothelial cells of alveolar capillaries. Uncaging caused Ca2+ levels to increase not only in the targeted cell, but also in vascular locations up to 150 microm from the target site, indicating that Ca2+ was conducted from the capillary to adjacent vessels. No such conduction was evident in mouse lungs lacking endothelial connexin 43 (Cx43), or in rat lungs in which we pretreated vessels with peptide inhibitors of Cx43. These findings provide the first direct evidence to our knowledge that interendothelial Ca2+ conduction occurs in the lung capillary bed and that Cx43-containing gap junctions mediate the conduction. A proinflammatory effect was evident in that induction of increases in Ca2+ levels in the capillary activated expression of the leukocyte adherence receptor P-selectin in venules. Further, peptide inhibitors of Cx43 completely blocked thrombin-induced microvascular permeability increases. Together, our findings reveal a novel role for Cx43-mediated gap junctions, namely as conduits for the spread of proinflammatory signals in the lung capillary bed. Gap junctional mechanisms require further consideration in the understanding of ALI.


Assuntos
Cálcio/fisiologia , Capilares/fisiopatologia , Conexina 43/fisiologia , Junções Comunicantes/fisiologia , Inflamação/fisiopatologia , Pulmão/fisiopatologia , Animais , Conexina 43/antagonistas & inibidores , Conexina 43/deficiência , Camundongos , Peptídeos/farmacologia , Fotólise , Circulação Pulmonar/fisiologia , Ratos , Ratos Sprague-Dawley
20.
Am J Physiol Lung Cell Mol Physiol ; 291(4): L596-601, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16698857

RESUMO

Although clusters of alveoli form the acinus, which is the most distal respiratory unit, it is not known whether interalveolar communication coordinates acinar surfactant secretion. To address this, we applied real-time digital imaging in conjunction with photo-excited Ca2+ uncaging in intact alveoli of the isolated, blood-perfused rat lung. We loaded alveolar cells with the Ca2+ cage o-nitrophenyl EGTA-AM (NP-EGTA-AM) together with the fluorophores, fluo 4, or LysoTracker green (LTG) to determine, respectively, the cytosolic Ca2+ concentration ([Ca2+]cyt) or type 2 cell secretion. To uncage Ca2+ from NP-EGTA, we exposed a region in a selected alveolus to high-intensity UV illumination. As a result, fluo 4 fluorescence increased, whereas LTG fluorescence decreased, in the photo-targeted region, indicating that uncaging both increased [Ca2+]cyt and induced secretion. Concomitantly, [Ca2+]cyt increases conducted from the uncaging site induced type 2 cell secretion in both the selected alveolus as well as in neighboring alveoli, indicating the presence of interalveolar communication. These conducted responses were inhibited by pretreating alveoli with the connexin43 (Cx43)-inhibiting peptides gap 26 and gap 27. However, although the conducted [Ca2+]cyt increase diminished with distance from the uncaging site, type 2 cell secretion rates were similar at all locations. We conclude that Cx43-dependent, interalveolar Ca2+ signals regulate type 2 cell secretion in adjacent alveoli. Such interalveolar communication might facilitate acinar coordination of alveolar function.


Assuntos
Sinalização do Cálcio/fisiologia , Pulmão/metabolismo , Alvéolos Pulmonares/metabolismo , Surfactantes Pulmonares/metabolismo , Compostos de Anilina , Animais , Transporte Biológico , Cálcio/metabolismo , Conexinas/farmacologia , Citosol/metabolismo , Ácido Egtázico/análogos & derivados , Ácido Egtázico/metabolismo , Corantes Fluorescentes , Junções Comunicantes/metabolismo , Pulmão/citologia , Oligopeptídeos , Peptídeos/farmacologia , Alvéolos Pulmonares/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Xantenos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...