Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
bioRxiv ; 2023 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-37292826

RESUMO

Although lung immunity is pathogen induced, the immunity can also be induced by mechanical distortion of the lung. The causal basis of the lung's mechanosensitive immunity remains unclear. Here, through live optical imaging of mouse lungs, we show that alveolar stretch due to hyperinflation induced prolonged cytosolic Ca2+ increases in sessile alveolar macrophages (AMs). Knockout studies revealed that the Ca2+ increases resulted from Ca2+ diffusion from the alveolar epithelium to sessile AMs through connexin 43 (Cx43)-containing gap junctions. Lung inflammation and injury in mice exposed to injurious mechanical ventilation were inhibited by AM-specific Cx43 knockout, or AM-specific delivery of a calcium inhibitor. We conclude, Cx43 gap junctions and calcium mobilization in sessile AMs determine the lung's mechanosensitive immunity, providing a therapeutic strategy against hyperinflation-induced lung injury.

2.
Nat Commun ; 13(1): 5837, 2022 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-36192486

RESUMO

Acute Lung Injury (ALI) due to inhaled pathogens causes high mortality. Underlying mechanisms are inadequately understood. Here, by optical imaging of live mouse lungs we show that a key mechanism is the viability of cytosolic Ca2+ buffering by the mitochondrial Ca2+ uniporter (MCU) in the lung's surfactant-secreting, alveolar type 2 cells (AT2). The buffering increased mitochondrial Ca2+ and induced surfactant secretion in wild-type mice, but not in mice with AT2-specific MCU knockout. In the knockout mice, ALI due to intranasal LPS instillation caused severe pulmonary edema and mortality, which were mitigated by surfactant replenishment prior to LPS instillation, indicating surfactant's protective effect against alveolar edema. In wild-type mice, intranasal LPS, or Pseudomonas aeruginosa decreased AT2 MCU. Loss of MCU abrogated buffering. The resulting mortality was reduced by spontaneous recovery of MCU expression, or by MCU replenishment. Enhancement of AT2 mitochondrial buffering, hence endogenous surfactant secretion, through MCU replenishment might be a therapy against ALI.


Assuntos
Lesão Pulmonar Aguda , Lipopolissacarídeos , Lesão Pulmonar Aguda/induzido quimicamente , Animais , Cálcio/metabolismo , Canais de Cálcio , Lipopolissacarídeos/toxicidade , Pulmão/metabolismo , Camundongos , Camundongos Knockout , Tensoativos
3.
Nat Commun ; 12(1): 6309, 2021 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-34728633

RESUMO

Lung fibrosis is increasingly detected with aging and has been associated with poor outcomes in acute lung injury or infection. However, the molecular programs driving this pro-fibrotic evolution are unclear. Here we profile distal lung samples from healthy human donors across the lifespan. Gene expression profiling by bulk RNAseq reveals both increasing cellular senescence and pro-fibrotic pathway activation with age. Quantitation of telomere length shows progressive shortening with age, which is associated with DNA damage foci and cellular senescence. Cell type deconvolution analysis of the RNAseq data indicates a progressive loss of lung epithelial cells and an increasing proportion of fibroblasts with age. Consistent with this pro-fibrotic profile, second harmonic imaging of aged lungs demonstrates increased density of interstitial collagen as well as decreased alveolar expansion and surfactant secretion. In this work, we reveal the transcriptional and structural features of fibrosis and associated functional impairment in normal lung aging.


Assuntos
Colágeno/metabolismo , Regulação da Expressão Gênica , Fibrose Pulmonar Idiopática/patologia , Pulmão/patologia , Encurtamento do Telômero , Adolescente , Adulto , Fatores Etários , Idoso , Senescência Celular/fisiologia , Estudos de Coortes , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Fibroblastos/metabolismo , Fibroblastos/patologia , Humanos , Fibrose Pulmonar Idiopática/metabolismo , Pulmão/metabolismo , Masculino , Pessoa de Meia-Idade , Análise de Sequência de RNA , Proteína Supressora de Tumor p53/metabolismo , Adulto Jovem
4.
JCI Insight ; 6(6)2021 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-33749665

RESUMO

High mortality in acute lung injury (ALI) results from sustained proinflammatory signaling by alveolar receptors, such as TNF-α receptor type 1 (TNFR1). Factors that determine the sustained signaling are not known. Unexpectedly, optical imaging of live alveoli revealed a major TNF-α-induced surge of alveolar TNFR1 due to a Ca2+-dependent mechanism that decreased the cortical actin fence. Mouse mortality due to inhaled LPS was associated with cofilin activation, actin loss, and the TNFR1 surge. The constitutively active form of the GTPase, Rac1 (V12Rac1), given intranasally (i.n.) as a noncovalent construct with a cell-permeable peptide, enhanced alveolar filamentous actin (F-actin) and blocked the TNFR1 surge. V12Rac1 also protected against ALI-induced mortality resulting from i.n. instillation of LPS or of Pseudomonas aeruginosa. We propose a potentially new therapeutic paradigm in which actin enhancement by exogenous Rac1 strengthens the alveolar actin fence, protecting against proinflammatory receptor hyperexpression, and therefore blocking ALI.


Assuntos
Actinas/uso terapêutico , Lesão Pulmonar Aguda/prevenção & controle , Neuropeptídeos/uso terapêutico , Proteínas rac1 de Ligação ao GTP/uso terapêutico , Lesão Pulmonar Aguda/metabolismo , Animais , Humanos , Masculino , Camundongos , Microscopia Confocal , Alvéolos Pulmonares/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo
5.
bioRxiv ; 2021 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-33469582

RESUMO

Acute lung immunity to inhaled pathogens elicits defensive pneumonitis that may convert to the Acute Respiratory Distress Syndrome (ARDS), causing high mortality. Mechanisms underlying the conversion are not understood, but are of intense interest because of the ARDS-induced mortality in the ongoing Covid-19 pandemic. Here, by optical imaging of live lungs we show that key to the lethality is the functional status of mitochondrial Ca2+ buffering across the mitochondrial Ca2+ uniporter (MCU) in the alveolar type 2 cells (AT2), which protect alveolar stability. In mice subjected to ARDS by airway exposure to lipopolysaccharide (LPS), or to Pseudomonas aeruginosa, there was marked loss of MCU expression in AT2. The ability of mice to survive ARDS depended on the extent to which the MCU expression recovered, indicating that the viability of Ca2+ buffering by AT2 mitochondria critically determines ARDS severity. Mitochondrial transfer to enhance AT2 MCU expression might protect against ARDS.

6.
J Control Release ; 329: 205-222, 2021 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-33245954

RESUMO

This work explores the potential for strategizing pulmonary surfactant (PS) for drug delivery over the respiratory air-liquid interface: the interfacial delivery. The efficacy of PS- and interface-assisted drug vehiculization was determined both in vitro and in vivo using a native purified porcine PS combined with the hydrophobic anti-inflammatory drug Tacrolimus (TAC), a calcineurin inhibitor. In vitro assays were conducted in a novel double surface balance setup designed to emulate compression-expansion dynamics applied to interfacially connected drug donor and recipient compartments. In this setup, PS transported TAC efficiently over air-liquid interfaces, with compression/expansion breathing-like dynamics enhancing rapid interface-assisted diffusion and drug release. The efficacy of PS-assisted TAC vehiculization was also evaluated in vivo in a mouse model of lipopolysaccharide (LPS)-induced acute lung injury (ALI). In anesthetized mice, TAC combined with PS was intra-nasally (i.n) instilled prior administering i.n. LPS. PS/TAC pre-treatment caused greater TAC internalization into a higher number of lung cells obtained from bronchoalveolar lavages (BAL) than TAC pre-treatment alone. Additionally, the PS/TAC combination but not TAC or PS alone attenuated the LPS-induced pro-inflammatory effects reducing cells and proteins in BAL fluid. These findings indicated that PS-mediated increase in TAC uptake blunted the pro-injurious effects of LPS, suggesting a synergistic anti-inflammatory effect of PS/drug formulations. These in vitro and in vivo results establish the potential utility of PS to open novel effective delivery strategies for inhaled drugs.


Assuntos
Preparações Farmacêuticas , Surfactantes Pulmonares , Animais , Sistemas de Liberação de Medicamentos , Camundongos , Tensoativos , Suínos , Tacrolimo
7.
Pulm Circ ; 8(3): 2045894018783735, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29869931

RESUMO

Crosstalk signaling between the closely juxtaposed epithelial and endothelial membranes of pulmonary alveoli establishes the lung's immune defense against inhaled and blood-borne pathogens. The crosstalk can occur in a forward direction, as from alveolus to capillary, or in a reverse direction, as from capillary to alveolus. The crosstalk direction likely depends on the site at which pathogens first initiate signaling. Thus, forward crosstalk may occur when inhaled pathogens encounter the alveolar epithelium, while reverse crosstalk may result from interactions of blood-borne pathogens with the endothelium. Here, we review the factors that regulate these two directions of signaling.

8.
Eur Respir J ; 51(4)2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29545281

RESUMO

The European Respiratory Society (ERS) Research Seminar entitled "Pulmonary vascular endothelium: orchestra conductor in respiratory diseases - highlights from basic research to therapy" brought together international experts in dysfunctional pulmonary endothelium, from basic science to translational medicine, to discuss several important aspects in acute and chronic lung diseases. This review will briefly sum up the different topics of discussion from this meeting which was held in Paris, France on October 27-28, 2016. It is important to consider that this paper does not address all aspects of endothelial dysfunction but focuses on specific themes such as: 1) the complex role of the pulmonary endothelium in orchestrating the host response in both health and disease (acute lung injury, chronic obstructive pulmonary disease, high-altitude pulmonary oedema and pulmonary hypertension); and 2) the potential value of dysfunctional pulmonary endothelium as a target for innovative therapies.


Assuntos
Endotélio Vascular/patologia , Endotélio Vascular/fisiopatologia , Doenças Respiratórias/fisiopatologia , Congressos como Assunto , Desenho de Fármacos , Humanos , Paris , Artéria Pulmonar/patologia , Remodelação Vascular
9.
J Clin Invest ; 128(3): 1074-1086, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29431734

RESUMO

Infection by Staphylococcus aureus strain USA300 causes tissue injury, multiorgan failure, and high mortality. However, the mechanisms by which the bacteria adhere to, then stabilize on, mucosal surfaces before causing injury remain unclear. We addressed these issues through the first real-time determinations of USA300-alveolar interactions in live lungs. We found that within minutes, inhaled USA300 established stable, self-associated microaggregates in niches at curved, but not at flat, regions of the alveolar wall. The microaggregates released α-hemolysin toxin, causing localized alveolar injury, as indicated by epithelial dye loss, mitochondrial depolarization, and cytosolic Ca2+ increase. Spread of cytosolic Ca2+ through intercellular gap junctions to adjoining, uninfected alveoli caused pulmonary edema. Systemic pretreatment with vancomycin, a USA300-cidal antibiotic, failed to protect mice infected with inhaled WT USA300. However, vancomycin pretreatment markedly abrogated mortality in mice infected with mutant USA300 that lacked the aggregation-promoting factor PhnD. We interpret USA300-induced mortality as having resulted from rapid bacterial aggregation in alveolar niches. These findings indicate, for the first time to our knowledge, that alveolar microanatomy is critical in promoting the aggregation and, hence, in causing USA300-induced alveolar injury. We propose that in addition to antibiotics, strategies for bacterial disaggregation may constitute novel therapy against USA300-induced lung injury.


Assuntos
Lesão Pulmonar/microbiologia , Lesão Pulmonar/prevenção & controle , Pulmão/microbiologia , Infecções Estafilocócicas/patologia , Staphylococcus aureus/efeitos dos fármacos , Vancomicina/farmacologia , Animais , Antibacterianos/farmacologia , Cálcio/metabolismo , Citosol/metabolismo , Junções Comunicantes/metabolismo , Proteínas Hemolisinas/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Perfusão , Alvéolos Pulmonares/microbiologia
10.
Curr Opin Genet Dev ; 38: 97-101, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27235808

RESUMO

Mitochondrial transfer from donor cells to cells of injured tissues is a promising cell-based therapy for effectively bringing about recovery of tissue bioenergetics. Here, we review recent studies on intercellular mitochondrial transfer in organs and cells. We also review studies that shed light on our current understanding of the known mechanisms and conditions that lead to intercellular mitochondrial transfer.


Assuntos
Comunicação Celular/genética , Metabolismo Energético , Mitocôndrias/genética , Humanos , Mitocôndrias/metabolismo
11.
Am J Respir Cell Mol Biol ; 48(1): 78-86, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23043086

RESUMO

Although exposure to ambient hypoxia is known to cause proinflammatory vascular responses, the mechanisms initiating these responses are not understood. We tested the hypothesis that in systemic hypoxia, erythrocyte-derived H(2)O(2) induces proinflammatory gene transcription in vascular endothelium. We exposed mice or isolated, perfused murine lungs to 4 hours of hypoxia (8% O(2)). Leukocyte counts increased in the bronchoalveolar lavage. The expression of leukocyte adhesion receptors, reactive oxygen species, and protein tyrosine phosphorylation increased in freshly recovered lung endothelial cells (FLECs). These effects were inhibited by extracellular catalase and by the removal of erythrocytes, indicating that the responses were attributable to erythrocyte-derived H(2)O(2). Concomitant nuclear translocation of the p65 subunit of NF-κB and hypoxia-inducible factor-1α stabilization in FLECs occurred only in the presence of erythrocytes. Hemoglobin binding to the erythrocyte membrane protein, band 3, induced the release of H(2)O(2) from erythrocytes and the p65 translocation in FLECs. These data indicate for the first time, to our knowledge, that erythrocytes are responsible for endothelial transcriptional responses in hypoxia.


Assuntos
Eritrócitos/fisiologia , Hipóxia/sangue , Hipóxia/fisiopatologia , Animais , Proteína 1 de Troca de Ânion do Eritrócito/metabolismo , Líquido da Lavagem Broncoalveolar/citologia , Selectina E/sangue , Endotélio Vascular/fisiopatologia , Peróxido de Hidrogênio/sangue , Hipóxia/genética , Mediadores da Inflamação/sangue , Pulmão/irrigação sanguínea , Pulmão/fisiopatologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Fatores de Transcrição/metabolismo
12.
Am J Physiol Lung Cell Mol Physiol ; 302(11): L1209-20, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22505671

RESUMO

Although the lung expresses procoagulant proteins under inflammatory conditions, underlying mechanisms remain unclear. Here, we addressed lung endothelial expression of tissue factor (TF), which initiates the coagulation cascade and expression of which signifies development of a procoagulant phenotype in the vasculature. To establish the model of acid-induced acute lung injury (ALI), we intranasally instilled anesthetized mice with saline or acid. Then 2 h later, we isolated pulmonary vascular cells for flow cytometry and confocal microscopy to detect the leukocyte antigen, CD45 and the endothelial markers VE-cadherin and von Willebrand factor (vWf). Acid increased both the number of vWf-expressing cells as well as TF and P-selectin expressions on these cells. All of these effects were markedly inhibited by treating mice with antiplatelet serum, suggesting the involvement of platelets. The increased expressions of TF, vWf, and P-selectin in response to acid also occurred in platelets. Moreover, the effects were replicated in endothelial cells derived from isolated, blood-perfused lungs. However, the effect was inhibited completely in lungs perfused with platelet-depleted and, to a lesser extent, with leukocyte-depleted blood. Acid injury increased endothelial expressions of the platelet proteins, CD41 and CD42b, providing evidence that platelet proteins were transferred to the vascular surface. Reactive oxygen species (ROS) were implicated in these responses, in that the endothelial and platelet protein expressions were inhibited. We conclude that acid-induced ALI causes NOX2-mediated ROS generation that activates platelets, which then generate a procoagulant endothelial surface.


Assuntos
Lesão Pulmonar Aguda/sangue , Lesão Pulmonar Aguda/induzido quimicamente , Plaquetas/metabolismo , Endotélio Vascular/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Tromboplastina/biossíntese , Animais , Antígenos CD/biossíntese , Coagulação Sanguínea , Plaquetas/imunologia , Caderinas/biossíntese , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Ácido Clorídrico/efeitos adversos , Ácido Clorídrico/toxicidade , Antígenos Comuns de Leucócito/biossíntese , Pulmão/imunologia , Pulmão/patologia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , NADPH Oxidase 2 , NADPH Oxidases/genética , NADPH Oxidases/metabolismo , Selectina-P/biossíntese , Selectina-P/metabolismo , Ativação Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/imunologia , Complexo Glicoproteico GPIb-IX de Plaquetas/biossíntese , Glicoproteína IIb da Membrana de Plaquetas/biossíntese , Tromboplastina/metabolismo , Fator de von Willebrand/biossíntese , Fator de von Willebrand/metabolismo
13.
Nat Med ; 18(5): 759-65, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-22504485

RESUMO

Bone marrow-derived stromal cells (BMSCs) protect against acute lung injury (ALI). To determine the role of BMSC mitochondria in this protection, we airway-instilled mice first with lipopolysaccharide (LPS) and then with either mouse BMSCs (mBMSCs) or human BMSCs (hBMSCs). Live optical studies revealed that the mBMSCs formed connexin 43 (Cx43)-containing gap junctional channels (GJCs) with the alveolar epithelia in these mice, releasing mitochondria-containing microvesicles that the epithelia engulfed. The presence of BMSC-derived mitochondria in the epithelia was evident optically, as well as by the presence of human mitochondrial DNA in mouse lungs instilled with hBMSCs. The mitochondrial transfer resulted in increased alveolar ATP concentrations. LPS-induced ALI, as indicated by alveolar leukocytosis and protein leak, inhibition of surfactant secretion and high mortality, was markedly abrogated by the instillation of wild-type mBMSCs but not of mutant, GJC-incompetent mBMSCs or mBMSCs with dysfunctional mitochondria. This is the first evidence, to our knowledge, that BMSCs protect against ALI by restituting alveolar bioenergetics through Cx43-dependent alveolar attachment and mitochondrial transfer.


Assuntos
Lesão Pulmonar Aguda/prevenção & controle , Células da Medula Óssea/fisiologia , Mitocôndrias/fisiologia , Alvéolos Pulmonares/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Conexina 43/fisiologia , Metabolismo Energético , Junções Comunicantes/fisiologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células Estromais/fisiologia
14.
J Clin Invest ; 121(5): 1986-99, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21519143

RESUMO

Shedding of the extracellular domain of cytokine receptors allows the diffusion of soluble receptors into the extracellular space; these then bind and neutralize their cytokine ligands, thus dampening inflammatory responses. The molecular mechanisms that control this process, and the extent to which shedding regulates cytokine-induced microvascular inflammation, are not well defined. Here, we used real-time confocal microscopy of mouse lung microvascular endothelium to demonstrate that mitochondria are key regulators of this process. The proinflammatory cytokine soluble TNF-α (sTNF-α) increased mitochondrial Ca2+, and the purinergic receptor P2Y2 prolonged the response. Concomitantly, the proinflammatory receptor TNF-α receptor-1 (TNFR1) was shed from the endothelial surface. Inhibiting the mitochondrial Ca2+ increase blocked the shedding and augmented inflammation, as denoted by increases in endothelial expression of the leukocyte adhesion receptor E-selectin and in microvascular leukocyte recruitment. The shedding was also blocked in microvessels after knockdown of a complex III component and after mitochondria-targeted catalase overexpression. Endothelial deletion of the TNF-α converting enzyme (TACE) prevented the TNF-α receptor shedding response, which suggests that exposure of microvascular endothelium to sTNF-α induced a Ca2+-dependent increase of mitochondrial H2O2 that caused TNFR1 shedding through TACE activation. These findings provide what we believe to be the first evidence that endothelial mitochondria regulate TNFR1 shedding and thereby determine the severity of sTNF-α-induced microvascular inflammation.


Assuntos
Cálcio/química , Pulmão/irrigação sanguínea , Mitocôndrias/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Proteínas ADAM/metabolismo , Proteína ADAM17 , Animais , Cálcio/metabolismo , Selectina E/metabolismo , Humanos , Peróxido de Hidrogênio/química , Inflamação , Leucócitos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Microcirculação , Modelos Biológicos , Estrutura Terciária de Proteína , Espécies Reativas de Oxigênio , Fator de Necrose Tumoral alfa/metabolismo
15.
Blood ; 113(18): 4197-205, 2009 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-19036701

RESUMO

Mesenchymal stem cells (MSCs), which potentially transdifferentiate into multiple cell types, are increasingly reported to be beneficial in models of organ system injury. However, the molecular mechanisms underlying interactions between MSCs and host cells, in particular endothelial cells (ECs), remain unclear. We show here in a matrigel angiogenesis assay that MSCs are capable of inhibiting capillary growth. After addition of MSCs to EC-derived capillaries in matrigel at EC:MSC ratio of 1:1, MSCs migrated toward the capillaries, intercalated between ECs, established Cx43-based intercellular gap junctional communication (GJC) with ECs, and increased production of reactive oxygen species (ROS). These events led to EC apoptosis and capillary degeneration. In an in vivo tumor model, direct MSC inoculation into subcutaneous melanomas induced apoptosis and abrogated tumor growth. Thus, our findings show for the first time that at high numbers, MSCs are potentially cytotoxic and that when injected locally in tumor tissue they might be effective antiangiogenesis agents suitable for cancer therapy.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Endotélio Vascular/citologia , Melanoma Experimental/irrigação sanguínea , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Neovascularização Patológica/terapia , Animais , Apoptose/fisiologia , Comunicação Celular , Células Cultivadas , Colágeno/metabolismo , Combinação de Medicamentos , Fibroblastos/citologia , Fibroblastos/metabolismo , Citometria de Fluxo , Junções Comunicantes/fisiologia , Immunoblotting , Técnicas Imunoenzimáticas , Imunoprecipitação , Laminina/metabolismo , Pulmão/citologia , Pulmão/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteoglicanas/metabolismo , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Engenharia Tecidual
16.
Am J Respir Cell Mol Biol ; 39(5): 569-75, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18483418

RESUMO

Although platelets induce lung inflammation, leading to acute lung injury (ALI), the extent of platelet-endothelial cell (EC) interactions remains poorly understood. Here, in a ventilation-stress model of lung inflammation, we show that platelet-EC interactions are important. We obtained freshly isolated lung endothelial cells (FLECs) from isolated, blood-perfused rat lungs exposed to ventilation at low tidal volume (LV) or stress-inducing high tidal volume (HV). Immunofluorescence and immunoprecipitation studies revealed HV-induced increases in cell-surface von Willebrand factor (vWf) expression on FLEC. This increased expression was inhibited by platelet removal from the lung perfusion and by including a P-selectin-blocking antibody in the lung perfusion. The expression was also blocked in lungs from P-selectin knockout (P sel(-/-)) mice perfused with autologous blood, but not with heterologous wild-type blood containing P-selectin-expressing platelets. These findings indicate that in ventilation stress, platelets transfer vWf to the EC surface and that platelet P-selectin plays a critical role in this transfer. Further evidence for such intercellular transfers was the HV-induced FLEC expressions of platelet glycoprotein 1b and of platelet P-selectin. We conclude that in ventilation stress, platelets deposit leukocyte- and platelet-binding proteins on the EC surface, thereby establishing the proinflammatory phenotype of the vascular lining.


Assuntos
Plaquetas/citologia , Plaquetas/metabolismo , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Ventilação de Alta Frequência , Animais , Adesão Celular , Camundongos , Camundongos Knockout , Selectina-P/genética , Selectina-P/metabolismo , Ligação Proteica , Ratos , Fator de von Willebrand/metabolismo
17.
Am J Respir Cell Mol Biol ; 33(6): 549-54, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16123392

RESUMO

Although high tidal volume ventilation exacerbates lung injury, the mechanisms underlying the inflammatory response are not clear. Here, we exposed isolated lungs to high or low tidal volume ventilation, while perfusing lungs with whole blood, or blood depleted of leukocytes and platelets. Then, we determined signaling responses in freshly isolated lung endothelial cells by means of immunoblotting and immunofluorescence approaches. In depleted blood perfusion, high tidal volume induced modest increases in both P-selectin expression on the endothelial surface, and in endothelial protein tyrosine phosphorylation. Both high tidal volume-induced responses were markedly enhanced in the presence of whole blood perfusion. However, a P-selectin-blocking antibody given together with whole blood perfusion inhibited the responses down to levels corresponding to those for depleted blood perfusion. These findings indicate that the full proinflammatory response occurs in two stages. First, lung distension causes modest endothelial activation. Second, subsequent endothelial-inflammatory cell interactions augment P-selectin expression and tyrosine phosphorylation. We conclude that interactions of circulating inflammatory cells with P-selectin critically determine proinflammatory endothelial activation during high tidal volume ventilation.


Assuntos
Endotélio Vascular/metabolismo , Pulmão/fisiologia , Selectina-P/metabolismo , Transdução de Sinais , Volume de Ventilação Pulmonar/fisiologia , Animais , Endotélio Vascular/citologia , Endotélio Vascular/imunologia , Imunofluorescência , Immunoblotting , Inflamação/etiologia , Inflamação/patologia , Perfusão , Fosforilação , Proteínas Tirosina Quinases/metabolismo , Ratos , Respiração Artificial , Tirosina/metabolismo
18.
Am J Respir Cell Mol Biol ; 28(2): 218-24, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12540489

RESUMO

Alveolar overdistension during mechanical ventilation causes leukocyte sequestration, leading to lung injury. However, underlying endothelial cell (EC) mechanisms are undefined. In a new approach, we exposed isolated blood-perfused rat lungs to high tidal volume ventilation (HV) for 2 h, then obtained fresh lung endothelial cells (FLEC) by immunosorting at 4 degrees C. Immunoblotting experiments indicated that as compared with FLEC derived from lungs ventilated at low volume (LV), HV markedly enhanced tyrosine phosphorylation (TyrP). The tyrosine kinase blocker, genistein, inhibited this response. HV also induced focal adhesion (FA) formation in FLEC, as detected by immunofluorescent aggregates of the alpha(v)beta(3) integrin that co-localized with aggregations of focal adhesion kinase (FAK). Immunoprecipitation and blotting experiments revealed that HV increased TyrP of the FA protein, paxillin. In addition, HV induced a paxillin-associated P-selectin expression on FLEC that was also inhibited by genistein. However, HV did not increase lung water. These results indicate that in HV, EC signaling in situ causes FA formation and induces TyrP-dependent P-selectin expression. These signaling mechanisms may promote leukocyte-mediated responses in HV.


Assuntos
Pulmão/irrigação sanguínea , Pulmão/fisiologia , Volume de Ventilação Pulmonar/fisiologia , Animais , Proteínas do Citoesqueleto/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/fisiologia , Adesões Focais , Técnicas In Vitro , Inflamação/etiologia , Inflamação/patologia , Inflamação/fisiopatologia , Mediadores da Inflamação/metabolismo , Selectina-P/metabolismo , Paxilina , Perfusão , Fosfoproteínas/metabolismo , Fosforilação , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Tirosina/metabolismo
19.
Am J Physiol Lung Cell Mol Physiol ; 283(4): L678-82, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12225942

RESUMO

A lung-protective ventilator strategy significantly reduces mortality in patients with acute lung injury. Substantial progress has been made in understanding how mechanical stress can injure the lung, both in terms of alterations in barrier properties of the pulmonary endothelium and epithelium as well as in stimulating proinflammatory responses of macrophages and neutrophils.


Assuntos
Lesão Pulmonar , Pulmão/fisiopatologia , Respiração Artificial/efeitos adversos , Animais , Humanos , Técnicas In Vitro , Pulmão/imunologia , Macrófagos/imunologia , Neutrófilos/imunologia , Estresse Mecânico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA