Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Cell Sci ; 133(3)2020 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-31964713

RESUMO

Endothelial YAP/TAZ (YAP is also known as YAP1, and TAZ as WWTR1) signaling is crucial for sprouting angiogenesis and vascular homeostasis. However, the underlying molecular mechanisms that explain how YAP/TAZ control the vasculature remain unclear. This study reveals that the focal adhesion protein deleted-in-liver-cancer 1 (DLC1) is a direct transcriptional target of the activated YAP/TAZ-TEAD complex. We find that substrate stiffening and VEGF stimuli promote expression of DLC1 in endothelial cells. In turn, DLC1 expression levels are YAP and TAZ dependent, and constitutive activation of YAP is sufficient to drive DLC1 expression. DLC1 is needed to limit F-actin fiber formation, integrin-based focal adhesion lifetime and integrin-mediated traction forces. Depletion of endothelial DLC1 strongly perturbs cell polarization in directed collective migration and inhibits the formation of angiogenic sprouts. Importantly, ectopic expression of DLC1 is sufficient to restore migration and angiogenic sprouting in YAP-depleted cells. Together, these findings point towards a crucial and prominent role for DLC1 in YAP/TAZ-driven endothelial adhesion remodeling and collective migration during angiogenesis.This article has an associated First Person interview with the first author of the paper.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Células Endoteliais , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Células Endoteliais/metabolismo , Proteínas Ativadoras de GTPase/genética , Humanos , Morfogênese , Neovascularização Patológica , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Transdução de Sinais , Proteínas Supressoras de Tumor/genética
2.
Angiogenesis ; 24(3): 677-693, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33770321

RESUMO

Endothelial barrier disruption and vascular leak importantly contribute to organ dysfunction and mortality during inflammatory conditions like sepsis and acute respiratory distress syndrome. We identified the kinase Arg/Abl2 as a mediator of endothelial barrier disruption, but the role of Arg in endothelial monolayer regulation and its relevance in vivo remain poorly understood. Here we show that depletion of Arg in endothelial cells results in the activation of both RhoA and Rac1, increased cell spreading and elongation, redistribution of integrin-dependent cell-matrix adhesions to the cell periphery, and improved adhesion to the extracellular matrix. We further show that Arg is activated in the endothelium during inflammation, both in murine lungs exposed to barrier-disruptive agents, and in pulmonary microvessels of septic patients. Importantly, Arg-depleted endothelial cells were less sensitive to barrier-disruptive agents. Despite the formation of F-actin stress fibers and myosin light chain phosphorylation, Arg depletion diminished adherens junction disruption and intercellular gap formation, by reducing the disassembly of cell-matrix adhesions and cell retraction. In vivo, genetic deletion of Arg diminished vascular leak in the skin and lungs, in the presence of a normal immune response. Together, our data indicate that Arg is a central and non-redundant regulator of endothelial barrier integrity, which contributes to cell retraction and gap formation by increasing the dynamics of adherens junctions and cell-matrix adhesions in a Rho GTPase-dependent fashion. Therapeutic inhibition of Arg may provide a suitable strategy for the treatment of a variety of clinical conditions characterized by vascular leak.


Assuntos
Matriz Extracelular/metabolismo , Junções Comunicantes/enzimologia , Células Endoteliais da Veia Umbilical Humana/enzimologia , Proteínas Tirosina Quinases/metabolismo , Alvéolos Pulmonares/enzimologia , Animais , Adesão Celular/genética , Ativação Enzimática , Matriz Extracelular/genética , Junções Comunicantes/genética , Humanos , Inflamação/enzimologia , Inflamação/genética , Camundongos , Camundongos Knockout , Proteínas Tirosina Quinases/genética
3.
J Immunol ; 196(11): 4681-91, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27183587

RESUMO

Sepsis is a systemic inflammatory response to infections associated with organ failure that is the most frequent cause of death in hospitalized patients. Exaggerated endothelial activation, altered blood flow, vascular leakage, and other disturbances synergistically contribute to sepsis-induced organ failure. The underlying signaling events associated with endothelial proinflammatory activation are not well understood, yet they likely consist of molecular pathways that act in an endothelium-specific manner. We found that LPS, a critical factor in the pathogenesis of sepsis, is internalized by endothelial cells, leading to intracellular signaling without the need for priming as found recently in immune cells. By identifying a novel role for retinoic acid-inducible gene-I (RIG-I) as a central regulator of endothelial activation functioning independent of TLR4, we provide evidence that the current paradigm of TLR4 solely being responsible for LPS-mediated endothelial responses is incomplete. RIG-I, as well as the adaptor protein mitochondrial antiviral signaling protein, regulates NF-κB-mediated induction of adhesion molecules and proinflammatory cytokine expression in response to LPS. Our findings provide essential new insights into the proinflammatory signaling pathways in endothelial cells and suggest that combined endothelial-specific inhibition of RIG-I and TLR4 will provide protection from aberrant endothelial responses associated with sepsis.


Assuntos
Proteína DEAD-box 58/metabolismo , Células Endoteliais/imunologia , Inflamação/imunologia , Lipopolissacarídeos/imunologia , Transdução de Sinais , Receptor 4 Toll-Like , Animais , Células Endoteliais/patologia , Inflamação/patologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Receptor 4 Toll-Like/imunologia
5.
Exp Cell Res ; 347(1): 161-170, 2016 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-27498166

RESUMO

In many pathological conditions the endothelium becomes activated and dysfunctional, resulting in hyperpermeability and plasma leakage. No specific therapies are available yet to control endothelial barrier function, which is regulated by inter-endothelial junctions and the generation of acto-myosin-based contractile forces in the context of cell-cell and cell-matrix interactions. However, the spatiotemporal distribution and stimulus-induced reorganization of these integral forces remain largely unknown. Traction force microscopy of human endothelial monolayers was used to visualize contractile forces in resting cells and during thrombin-induced hyperpermeability. Simultaneously, information about endothelial monolayer integrity, adherens junctions and cytoskeletal proteins (F-actin) were captured. This revealed a heterogeneous distribution of traction forces, with nuclear areas showing lower and cell-cell junctions higher traction forces than the whole-monolayer average. Moreover, junctional forces were asymmetrically distributed among neighboring cells. Force vector orientation analysis showed a good correlation with the alignment of F-actin and revealed contractile forces in newly formed filopodia and lamellipodia-like protrusions within the monolayer. Finally, unstable areas, showing high force fluctuations within the monolayer were prone to form inter-endothelial gaps upon stimulation with thrombin. To conclude, contractile traction forces are heterogeneously distributed within endothelial monolayers and force instability, rather than force magnitude, predicts the stimulus-induced formation of intercellular gaps.


Assuntos
Endotélio Vascular/fisiologia , Junções Comunicantes/metabolismo , Actinas/metabolismo , Fenômenos Biomecânicos/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/metabolismo , Endotélio Vascular/efeitos dos fármacos , Junções Comunicantes/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Trombina/farmacologia
6.
Am J Respir Crit Care Med ; 193(12): 1410-20, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-26760925

RESUMO

RATIONALE: Altered pulmonary hemodynamics and fluid flow-induced high shear stress (HSS) are characteristic hallmarks in the pathogenesis of pulmonary arterial hypertension (PAH). However, the contribution of HSS to cellular and vascular alterations in PAH is unclear. OBJECTIVES: We hypothesize that failing shear adaptation is an essential part of the endothelial dysfunction in all forms of PAH and tested whether microvascular endothelial cells (MVECs) or pulmonary arterial endothelial cells (PAECs) from lungs of patients with PAH adapt to HSS and if the shear defect partakes in vascular remodeling in vivo. METHODS: PAH MVEC (n = 7) and PAH PAEC (n = 3) morphology, function, protein, and gene expressions were compared with control MVEC (n = 8) under static culture conditions and after 24, 72, and 120 hours of HSS. MEASUREMENTS AND MAIN RESULTS: PAH MVEC showed a significantly delayed morphological shear adaptation (P = 0.03) and evidence of cell injury at sites of nonuniform shear profiles that are critical loci for vascular remodeling in PAH. In clear contrast, PAEC isolated from the same PAH lungs showed no impairments. PAH MVEC gene expression and transcriptional shear activation were not altered but showed significant decreased protein levels (P = 0.02) and disturbed interendothelial localization of the shear sensor platelet endothelial cell adhesion molecule-1 (PECAM-1). The decreased PECAM-1 levels were caused by caspase-mediated cytoplasmic cleavage but not increased cell apoptosis. Caspase blockade stabilized PECAM-1 levels, restored endothelial shear responsiveness in vitro, and attenuated occlusive vascular remodeling in chronically hypoxic Sugen5416-treated rats modeling severe PAH. CONCLUSIONS: Delayed shear adaptation, which promotes shear-induced endothelial injury, is a newly identified dysfunction specific to the microvascular endothelium in PAH. The shear response is normalized on stabilization of PECAM-1, which reverses intimal remodeling in vivo.


Assuntos
Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/fisiopatologia , Microvasos/fisiopatologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Remodelação Vascular/fisiologia , Adulto , Animais , Western Blotting , Células Cultivadas , Criança , Modelos Animais de Doenças , Feminino , Imunofluorescência , Humanos , Masculino , Microvasos/metabolismo , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , Artéria Pulmonar/metabolismo , Artéria Pulmonar/fisiopatologia , Ratos , Adulto Jovem
7.
Am J Physiol Lung Cell Mol Physiol ; 311(2): L453-66, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27343194

RESUMO

Despite considerable progress in the understanding of endothelial barrier regulation and the identification of approaches that have the potential to improve endothelial barrier function, no drug- or stem cell-based therapy is presently available to reverse the widespread vascular leak that is observed in acute respiratory distress syndrome (ARDS) and sepsis. The translational gap suggests a need to develop experimental approaches and tools that better mimic the complex environment of the microcirculation in which the vascular leak develops. Recent studies have identified several elements of this microenvironment. Among these are composition and stiffness of the extracellular matrix, fluid shear stress, interaction of endothelial cells (ECs) with pericytes, oxygen tension, and the combination of toxic and mechanic injurious stimuli. Development of novel cell culture techniques that integrate these elements would allow in-depth analysis of EC biology that closely approaches the (patho)physiological conditions in situ. In parallel, techniques to isolate organ-specific ECs, to define EC heterogeneity in its full complexity, and to culture patient-derived ECs from inducible pluripotent stem cells or endothelial progenitor cells are likely to advance the understanding of ARDS and lead to development of therapeutics. This review 1) summarizes the advantages and pitfalls of EC cultures to study vascular leak in ARDS, 2) provides an overview of elements of the microvascular environment that can directly affect endothelial barrier function, and 3) discusses alternative methods to bridge the gap between basic research and clinical application with the intent of improving the translational value of present EC culture approaches.


Assuntos
Células Endoteliais/fisiologia , Endotélio Vascular/fisiopatologia , Síndrome do Desconforto Respiratório/patologia , Animais , Permeabilidade Capilar , Comunicação Celular , Células Cultivadas , Endotélio Vascular/fisiologia , Matriz Extracelular/fisiologia , Humanos , Pulmão/irrigação sanguínea , Pulmão/patologia , Técnicas Analíticas Microfluídicas , Síndrome do Desconforto Respiratório/fisiopatologia
8.
Arterioscler Thromb Vasc Biol ; 35(5): 1071-9, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25814671

RESUMO

The vascular endothelium separates circulating fluid and inflammatory cells from the surrounding tissues. Vascular leak occurs in response to wide-spread inflammatory processes, such as sepsis and acute respiratory distress syndrome, because of the formation of gaps between endothelial cells. Although these disorders are leading causes of mortality in the intensive care unit, no medical therapies exist to restore endothelial cell barrier function. Recent evidence highlights a key role for the Abl family of nonreceptor tyrosine kinases in regulating vascular barrier integrity. These kinases have well-described roles in cancer progression and neuronal morphogenesis, but their functions in the vasculature have remained enigmatic until recently. The Abl family kinases, c-Abl (Abl1) and Abl related gene (Arg, Abl2), phosphorylate several cytoskeletal effectors that mediate vascular permeability, including nonmuscle myosin light chain kinase, cortactin, vinculin, and ß-catenin. They also regulate cell-cell and cell-matrix junction dynamics, and the formation of actin-based cellular protrusions in multiple cell types. In addition, both c-Abl and Arg are activated by hyperoxia and contribute to oxidant-induced endothelial cell injury. These numerous roles of Abl kinases in endothelial cells and the current clinical usage of imatinib and other Abl kinase inhibitors have spurred recent interest in repurposing these drugs for the treatment of vascular barrier dysfunction. This review will describe the structure and function of Abl kinases with an emphasis on their roles in mediating vascular barrier integrity. We will also provide a critical evaluation of the potential for exploiting Abl kinase inhibition as a novel therapy for inflammatory vascular leak syndromes.


Assuntos
Benzamidas/administração & dosagem , Terapia de Alvo Molecular/métodos , Piperazinas/administração & dosagem , Inibidores de Proteínas Quinases/administração & dosagem , Proteínas Proto-Oncogênicas c-abl/efeitos dos fármacos , Pirimidinas/administração & dosagem , Síndrome do Desconforto Respiratório/tratamento farmacológico , Sepse/tratamento farmacológico , Permeabilidade Capilar/efeitos dos fármacos , Feminino , Humanos , Mesilato de Imatinib , Masculino , Proteínas Proto-Oncogênicas c-abl/genética , Síndrome do Desconforto Respiratório/fisiopatologia , Sepse/fisiopatologia , Transdução de Sinais/efeitos dos fármacos , Resultado do Tratamento
9.
Ther Drug Monit ; 38(5): 621-7, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27465973

RESUMO

BACKGROUND: Azathioprine and mercaptopurine (MP) are effective in treating patients with inflammatory bowel disease (IBD). Immunosuppressive effects of thiopurines involve T-cell apoptosis after inhibition of GTPase Ras-related C3 botulinum toxin substrate 1 (Rac1). This study aimed to assess whether expression and activity of Rac1 or phosphorylated ezrin-radixin-moesin (pERM) in patients with IBD could provide a useful biomarker for the pharmacodynamic thiopurine effect and might be related to clinical effectiveness. METHODS: This was a 2-stage study: stage 1 concerned a cross-sectional cohort of patients with IBD clinically in remission and treated with (n = 10) or without stable weight-based thiopurine therapy (n = 11) and healthy controls (n = 6); stage 2 concerned a prospective study regarding IBD patients with clinically active disease who initiated MP therapy (n = 11) compared with healthy controls (n = 11). Expression and activity of Rac1 and ERM and pERM were determined. RESULTS: The median Rac1 expression was statistically significantly reduced by thiopurine maintenance therapy {0.54 [interquartile range (IQR) 0.47-0.88] versus 0.80 arbitrary units [IQR 0.64-1.46]} compared with patients without immunosuppressive therapy (P = 0.042), but not Rac1 activity and pERM. In responders to MP therapy (n = 6), both median active Rac1 [93 (IQR 81-151) to 76 ng Rac1/mg protein (IQR 62-98)] and Rac1 expression [16.2 (8.8-29.4) to 1.5 arbitrary units (0.9-5.3)] decreased (P = 0.028). In nonresponders (n = 3), Rac1 expression and activity increased. CONCLUSIONS: IBD patients treated with thiopurines had a lower expression of Rac1 compared with those not treated with thiopurine. Effective MP therapy led to decreasing concentrations of Rac1-GTP and Rac1 expression. Therefore, Rac1-GTP and expression of Rac1, but not phosphorylation of ERM, form potentially pharmacodynamic markers of therapeutic thiopurine effectiveness in patients with IBD.


Assuntos
Azatioprina/uso terapêutico , Biomarcadores Farmacológicos/sangue , Doenças Inflamatórias Intestinais/sangue , Doenças Inflamatórias Intestinais/tratamento farmacológico , Mercaptopurina/uso terapêutico , Proteínas rac1 de Ligação ao GTP/sangue , Adulto , Azatioprina/farmacocinética , Biomarcadores Farmacológicos/metabolismo , Estudos Transversais , Feminino , Humanos , Imunossupressores/farmacocinética , Imunossupressores/uso terapêutico , Masculino , Mercaptopurina/farmacocinética , Pessoa de Meia-Idade , Fosfoproteínas/biossíntese , Fosfoproteínas/sangue , Fosforilação/efeitos dos fármacos , Estudos Prospectivos , Trocadores de Sódio-Hidrogênio/biossíntese , Trocadores de Sódio-Hidrogênio/sangue , Adulto Jovem , Proteínas rac1 de Ligação ao GTP/biossíntese
10.
Crit Care Med ; 42(3): 691-700, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24158164

RESUMO

OBJECTIVE: Numerous studies have focused on biomarkers for acute lung injury and acute respiratory distress syndrome. Although several biomarkers have been identified, their relative performance is unclear. We aim to provide a quantitative overview of plasma-derived biomarkers associated with acute respiratory distress syndrome diagnosis or mortality. DATA SOURCES: MEDLINE (inception to January 2012) and personal databases. STUDY SELECTION: English-language studies on plasma biomarkers associated with acute respiratory distress syndrome diagnosis or mortality. DATA EXTRACTION: Demographic variables, plasma levels of biomarker, statistical data, acute respiratory distress syndrome occurrence, and mortality rates were retrieved. The methodological quality was assessed with the Quality Assessment of Diagnostic Accuracy Studies score. Clinical outcomes included 1) diagnosis of acute respiratory distress syndrome in the at-risk population and 2) mortality in acute respiratory distress syndrome patients. For each biomarker, pooled odds ratios for clinical outcome were calculated by meta-analysis, and biomarkers were ranked according to pooled odds ratio. DATA SYNTHESIS: Fifty-four studies appeared eligible for meta-analysis, together including 3,753 patients. We identified 20 biomarkers for diagnosis of acute respiratory distress syndrome in the at-risk population and 19 biomarkers for mortality of acute respiratory distress syndrome patients. The biomarkers most strongly associated with acute respiratory distress syndrome diagnosis in the at-risk population, when increased, were Krebs von den Lungen-6 (odds ratio [95% CI], 6.1 [3.0-12.1]), lactate dehydrogenase (5.7 [1.7-19.1]), soluble receptor for advanced glycation end products (3.5 [1.7-7.2]), and von Willebrand Factor (3.1 [2.0-5.2]). The biomarkers most strongly associated with acute respiratory distress syndrome mortality, when increased, were interleukin-4 (18.0 [6.0-54.2]), interleukin-2 (11.8 [4.3-32.2]), angiopoietin-2 (6.4 [1.3-30.4]), and Krebs von den Lungen-6 (5.1 [3.0-12.2]). Decreased levels of Protein C were associated with increased odds for acute respiratory distress syndrome diagnosis and mortality. CONCLUSIONS: This meta-analysis provides a unique ranking of plasma biomarkers according to their strength of association with acute respiratory distress syndrome diagnosis or acute respiratory distress syndrome mortality. The relative performance of biomarkers among studies shown in this ranking may help to improve acute respiratory distress syndrome diagnosis and outcome prediction.


Assuntos
Angiopoietina-2/sangue , Interleucinas/sangue , Proteína C/metabolismo , Síndrome do Desconforto Respiratório/sangue , Síndrome do Desconforto Respiratório/mortalidade , Lesão Pulmonar Aguda/sangue , Lesão Pulmonar Aguda/mortalidade , Lesão Pulmonar Aguda/fisiopatologia , Biomarcadores/sangue , Estado Terminal , Feminino , Mortalidade Hospitalar , Humanos , Interleucina-2/sangue , Interleucina-4/sangue , Masculino , Prognóstico , Síndrome do Desconforto Respiratório/fisiopatologia , Medição de Risco , Sensibilidade e Especificidade , Análise de Sobrevida
11.
Cell Tissue Res ; 355(3): 557-76, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24633925

RESUMO

Although the endothelium is an extremely thin single-cell layer, it performs exceedingly well in preventing blood fluids from leaking into the surrounding tissues. However, specific pathological conditions can affect this cell layer, compromising the integrity of the barrier. Vascular leakage is a hallmark of many cardiovascular diseases and despite its medical importance, no specialized therapies are available to prevent it or reduce it. Small guanosine triphosphatases (GTPases) of the Rho family are known to be key regulators of various aspects of cell behavior and studies have shown that they can exert both positive and negative effects on endothelial barrier integrity. Moreover, extracellular matrix stiffness has now been implicated in the regulation of Rho-GTPase signaling, which has a direct impact on the integrity of endothelial junctions. However, knowledge about both the precise mechanism of this regulation and the individual contribution of the specific regulatory proteins remains fragmentary. In this review, we discuss recent findings concerning the balanced activities of Rho-GTPases and, in particular, aspects of the regulation of the endothelial barrier. We highlight the role of Rho-GTPases in the intimate relationships between biomechanical forces, microenvironmental influences and endothelial intercellular junctions, which are all interwoven in a beautiful filigree-like fashion.


Assuntos
Células Endoteliais/fisiologia , Endotélio Vascular/fisiologia , Proteínas rho de Ligação ao GTP/metabolismo , Animais , Permeabilidade Capilar , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Humanos , Transdução de Sinais
12.
Circulation ; 126(23): 2728-38, 2012 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-23099479

RESUMO

BACKGROUND: Tissue edema and endothelial barrier dysfunction as observed in sepsis and acute lung injury carry high morbidity and mortality, but currently lack specific therapy. In a recent case report, we described fast resolution of pulmonary edema on treatment with the tyrosine kinase inhibitor imatinib through an unknown mechanism. Here, we explored the effect of imatinib on endothelial barrier dysfunction and edema formation. METHODS AND RESULTS: We evaluated the effect of imatinib on endothelial barrier function in vitro and in vivo. In human macro- and microvascular endothelial monolayers, imatinib attenuated endothelial barrier dysfunction induced by thrombin and histamine. Small interfering RNA knock-downs of the imatinib-sensitive kinases revealed that imatinib attenuates endothelial barrier dysfunction via inhibition of Abl-related gene kinase (Arg/Abl2), a previously unknown mediator of endothelial barrier dysfunction. Indeed, Arg was activated by endothelial stimulation with thrombin, histamine, and vascular endothelial growth factor. Imatinib limited Arg-mediated endothelial barrier dysfunction by enhancing Rac1 activity and enforcing adhesion of endothelial cells to the extracellular matrix. Using mouse models of vascular leakage as proof-of-concept, we found that pretreatment with imatinib protected against vascular endothelial growth factor-induced vascular leakage in the skin, and effectively prevented edema formation in the lungs. In a murine model of sepsis, imatinib treatment (6 hours and 18 hours after induction of sepsis) attenuated vascular leakage in the kidneys and the lungs (24 hours after induction of sepsis). CONCLUSIONS: Thus, imatinib prevents endothelial barrier dysfunction and edema formation via inhibition of Arg. These findings identify imatinib as a promising approach to permeability edema and indicate Arg as novel target for edema treatment.


Assuntos
Permeabilidade Capilar/fisiologia , Endotélio Vascular/metabolismo , Piperazinas/uso terapêutico , Edema Pulmonar/tratamento farmacológico , Edema Pulmonar/metabolismo , Pirimidinas/uso terapêutico , Animais , Benzamidas , Permeabilidade Capilar/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/fisiopatologia , Humanos , Mesilato de Imatinib , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Piperazinas/farmacologia , Edema Pulmonar/fisiopatologia , Pirimidinas/farmacologia , Pele/irrigação sanguínea , Pele/efeitos dos fármacos , Pele/metabolismo , Resultado do Tratamento
13.
Crit Care Med ; 40(3): 793-9, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22080639

RESUMO

OBJECTIVES: It is largely unknown why extravascular lung water may increase during fluid loading in the critically ill with presumed hypovolemia. In this study we evaluated the hemodynamic predictors of such an increase. DESIGN: A prospective observational study. PATIENTS: Sixty-three presumed hypovolemic mechanically ventilated patients (22 septic and 41 nonseptic patients). INTERVENTION: Fluid loading with saline or colloid fluids guided by (changes in) cardiac filling pressures. MEASUREMENTS AND MAIN RESULTS: Before and after fluid-loading, hemodynamic and respiratory variables were recorded, including variables obtained by transpulmonary dilution such as cardiac index, pulmonary blood volume index, and extravascular lung water. Baseline parameters and change in parameters were compared between patients with a change in extravascular lung water <10% and patients with a change in extravascular lung water ≥ 10%. Predictive values for change in extravascular lung water ≥ 10% were evaluated. Baseline cardiac index and pulmonary blood volume index were higher, whereas change in cardiac index, change in pulmonary blood volume index, and change in PaO2/FIO2 ratio were lower in patients with a change in extravascular lung water ≥ 10% than in patients with a change in extravascular lung water <10%. The change in extravascular lung water correlated to baseline cardiac index (r = 0.17; p = .001), baseline pulmonary blood volume index (r = 0.15; p = .001), change in pulmonary blood volume index (r = 0.16; p < .001), and change in PaO2/FIO2 ratio (r = 0.13; p = .004). In multiple logistic regression analysis baseline cardiac index, baseline pulmonary blood volume index, the change in cardiac index, change in pulmonary blood volume index, and change in PaO2/FIO2 ratio individually contributed to prediction of a change in extravascular lung water ≥ 10%, independent of the presence of sepsis, pulmonary vascular permeability, and cardiac filling pressures. A change in extravascular lung water ≥ 10% was predicted by baseline cardiac index (77% sensitivity, 98% specificity) and pulmonary blood volume index (92% sensitivity, 68% specificity), and by change in cardiac index (69% sensitivity, 59% specificity), change in pulmonary blood volume index (77% sensitivity, 82% specificity), and change in PaO2/FIO2 ratio (77% sensitivity, 66% specificity). CONCLUSION: Extravascular lung water increase during fluid loading in the critically ill is predicted by a plateau of cardiac function and pulmonary vascular filling at baseline, rather than by pulmonary vascular permeability and filling pressures. Increasing extravascular lung water is further reflected by a decrease of PaO2/FIO2 ratio. These observations may help preventing pulmonary fluid overloading.


Assuntos
Hidratação/efeitos adversos , Hipovolemia/terapia , Edema Pulmonar/etiologia , Estado Terminal , Feminino , Previsões , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos
14.
Blood ; 115(12): 2533-42, 2010 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-20032497

RESUMO

The shear stress-induced transcription factor Krüppel-like factor 2 (KLF2) confers antiinflammatory properties to endothelial cells through the inhibition of activator protein 1, presumably by interfering with mitogen-activated protein kinase (MAPK) cascades. To gain insight into the regulation of these cascades by KLF2, we used antibody arrays in combination with time-course mRNA microarray analysis. No gross changes in MAPKs were detected; rather, phosphorylation of actin cytoskeleton-associated proteins, including focal adhesion kinase, was markedly repressed by KLF2. Furthermore, we demonstrate that KLF2-mediated inhibition of Jun NH(2)-terminal kinase (JNK) and its downstream targets ATF2/c-Jun is dependent on the cytoskeleton. Specifically, KLF2 directs the formation of typical short basal actin filaments, termed shear fibers by us, which are distinct from thrombin- or tumor necrosis factor-alpha-induced stress fibers. KLF2 is shown to be essential for shear stress-induced cell alignment, concomitant shear fiber assembly, and inhibition of JNK signaling. These findings link the specific effects of shear-induced KLF2 on endothelial morphology to the suppression of JNK MAPK signaling in vascular homeostasis via novel actin shear fibers.


Assuntos
Citoesqueleto de Actina/metabolismo , Células Endoteliais/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Fator 2 Ativador da Transcrição/metabolismo , Animais , Aorta/citologia , Células Cultivadas , Células Endoteliais/citologia , Artéria Femoral/citologia , Humanos , Fatores de Transcrição Kruppel-Like/genética , Fosforilação/fisiologia , Ratos , Fluxo Sanguíneo Regional/fisiologia , Veia Safena/citologia , Estresse Mecânico , Transdução Genética , Veias Umbilicais/citologia , Quinases Associadas a rho/metabolismo
15.
Am J Physiol Cell Physiol ; 300(1): C146-54, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20861463

RESUMO

A hallmark of many, sometimes life-threatening, inflammatory diseases and disorders is vascular leakage. The extent and severity of vascular leakage is broadly mediated by the integrity of the endothelial cell (EC) monolayer, which is in turn governed by three major interactions: cell-cell and cell-substrate contacts, soluble mediators, and biomechanical forces. A potentially critical but essentially uninvestigated component mediating these interactions is the stiffness of the substrate to which the endothelial monolayer is adherent. Accordingly, we investigated the extent to which substrate stiffening influences endothelial monolayer disruption and the role of cell-cell and cell-substrate contacts, soluble mediators, and physical forces in that process. Traction force microscopy showed that forces between cell and cell and between cell and substrate were greater on stiffer substrates. On stiffer substrates, these forces were substantially enhanced by a hyperpermeability stimulus (thrombin, 1 U/ml), and gaps formed between cells. On softer substrates, by contrast, these forces were increased far less by thrombin, and gaps did not form between cells. This stiffness-dependent force enhancement was associated with increased Rho kinase activity, whereas inhibition of Rho kinase attenuated baseline forces and lessened thrombin-induced inter-EC gap formation. Our findings demonstrate a central role of physical forces in EC gap formation and highlight a novel physiological mechanism. Integrity of the endothelial monolayer is governed by its physical microenvironment, which in normal circumstances is compliant but during pathology becomes stiffer.


Assuntos
Células Endoteliais/citologia , Células Endoteliais/fisiologia , Resinas Acrílicas , Antígenos CD/metabolismo , Fenômenos Biomecânicos , Caderinas/metabolismo , Adesão Celular/efeitos dos fármacos , Adesão Celular/fisiologia , Células Cultivadas , Meios de Cultura/química , Células Endoteliais/efeitos dos fármacos , Humanos , Membranas Artificiais , Microscopia , Trombina/farmacologia , Quinases Associadas a rho/metabolismo
16.
Cell Physiol Biochem ; 28(1): 53-62, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21865848

RESUMO

AIMS: Increased levels of homocysteine (Hcy) form an independent risk factor for cardiovascular disease. In a previous study we have shown that Hcy induced phosphatidylserine (PS) exposure to the outer leaflet of the plasma membrane in cardiomyocytes, inducing a pro-inflammatory phenotype. In the present study the mechanism(s) involved in Hcy-induced PS exposure were analyzed. METHODS: H9c2 rat cardiomyoblasts were subjected to 2.5 mM D,L-Hcy and analyzed for RhoA translocation and activity, Rho Kinase (ROCK) activity and expression and flippase activity. In addition, the effect of ROCK inhibition with Y27632 on Hcy-induced PS exposure and flippase activity was analyzed. Furthermore, GTP and ATP levels were determined. RESULTS: Incubation of H9c2 cells with 2.5 mM D,L-Hcy did not inhibit RhoA translocation to the plasma membrane. Neither did it inhibit activation of RhoA, even though GTP levels were significantly decreased. Hcy did significantly inhibit ROCK activation, but not its expression, and did inhibit flippase activity, in advance of a significant decrease in ATP levels. ROCK inhibition via Y27632 did not have significant added effects on this. CONCLUSION: Hcy induced PS exposure in the outer leaflet of the plasma membrane in cardiomyocytes via inhibition of ROCK and flippase activity. As such Hcy may induce cardiomyocytes vulnerable to inflammation in vivo in hyperhomocysteinaemia patients.


Assuntos
Homocisteína/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/enzimologia , Fosfatidilserinas/metabolismo , Proteínas de Transferência de Fosfolipídeos/antagonistas & inibidores , Quinases Associadas a rho/antagonistas & inibidores , Trifosfato de Adenosina/metabolismo , Amidas/farmacologia , Animais , Células Cultivadas , Guanosina Trifosfato/metabolismo , Proteínas de Transferência de Fosfolipídeos/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Ratos , Quinases Associadas a rho/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
17.
Crit Care Med ; 39(1): 89-97, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21057316

RESUMO

OBJECTIVES: To evaluate the diagnostic value of plasma protein levels for pulmonary vascular permeability and acute respiratory distress syndrome. During acute lung injury and acute respiratory distress syndrome, increased vascular permeability induces protein-rich fluid extravasation. We hypothesized that plasma protein levels predict increased vascular permeability and acute respiratory distress syndrome. DESIGN: A prospective, observational study. PATIENTS: Eighty-three consecutive, mechanically ventilated patients with or at risk for acute lung injury/acute respiratory distress syndrome, of whom 18 had sepsis. Patients with increased pulmonary capillary wedge pressures or central venous pressures were excluded. INTERVENTIONS: Patients were subjected to pulmonary capillary wedge pressure/central venous pressure-guided fluid loading with saline or colloid fluids. MEASUREMENTS AND MAIN RESULTS: We measured plasma albumin and transferrin levels and determined the Gallium-transferrin pulmonary leak index, the American European Consensus Conference criteria, and the lung injury score. Measurements were performed before and after fluid loading to evaluate effects of fluid loading. Plasma albumin and transferrin levels were approximately 30% lower in acute respiratory distress syndrome than patients with acute lung injury (p < .01) and patients without lung injury (p < .05). Protein levels inversely related to the pulmonary leak index (standardized regression coefficient -0.28, p < .001 for albumin; standardized regression coefficient -0.30, p = .003 for transferrin) and the lung injury score (standardized regression coefficient -0.19, p = .01 for albumin), independently of presence of sepsis, severity of disease, and fluid loading. Albumin and transferrin levels had a high sensitivity (77-93%) and negative predictive value (80-98%) for elevated pulmonary vascular permeability and acute respiratory distress syndrome (American European Consensus Conference criteria and lung injury score). The addition of hypoalbuminemia (<17.5 g/L) and hypotransferrinemia (<0.98 g/L) as criteria to the American European Consensus Conference criteria or the lung injury score increased their predictive values for elevated pulmonary vascular permeability. CONCLUSIONS: In critically ill patients, decreased plasma albumin and transferrin levels parallel increased pulmonary vascular permeability irrespective of underlying disease and fluid status. While normal levels help to exclude acute respiratory distress syndrome, hypoalbuminemia and hypotransferrinemia increase the diagnostic accuracy of the American European Consensus Conference criteria and lung injury score for elevated pulmonary vascular permeability.


Assuntos
Lesão Pulmonar Aguda/sangue , Lesão Pulmonar Aguda/mortalidade , Proteínas Sanguíneas/metabolismo , Mortalidade Hospitalar , Síndrome do Desconforto Respiratório/sangue , Síndrome do Desconforto Respiratório/mortalidade , Lesão Pulmonar Aguda/terapia , Idoso , Biomarcadores/sangue , Permeabilidade Capilar , Estudos de Coortes , Cuidados Críticos/métodos , Feminino , Humanos , Unidades de Terapia Intensiva , Pulmão/irrigação sanguínea , Masculino , Pessoa de Meia-Idade , Valor Preditivo dos Testes , Prognóstico , Estudos Prospectivos , Edema Pulmonar/sangue , Edema Pulmonar/diagnóstico , Edema Pulmonar/mortalidade , Respiração Artificial , Síndrome do Desconforto Respiratório/terapia , Medição de Risco , Sepse/diagnóstico , Sepse/mortalidade , Sepse/terapia , Albumina Sérica/metabolismo , Análise de Sobrevida , Transferrina/metabolismo , Resultado do Tratamento
18.
Arterioscler Thromb Vasc Biol ; 30(6): 1137-42, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20224051

RESUMO

OBJECTIVE: To investigate the effects of activation of the AMP-activated protein kinase (AMPK) on muscle perfusion and to elucidate the mechanisms involved. METHODS AND RESULTS: In a combined approach, we studied the vasoactive actions of AMPK activator by 5-aminoimidazole-4-carboxamide-1-beta-D-ribofuranoside (AICAR) on rat cremaster muscle resistance arteries ( approximately 100 mum) ex vivo and on microvascular perfusion in the rat hindlimb in vivo. In isolated resistance arteries, AICAR increased Thr172 phosphorylation of AMPK in arteriolar endothelium, which was predominantly located in microvascular endothelium. AICAR induced vasodilation (19+/-4% at 2 mmol/L, P<0.01), which was abolished by endothelium removal, inhibition of NO synthase (with N-nitro-L-arginine), or AMPK (with compound C). Smooth muscle sensitivity to NO, determined by studying the effects of the NO donor S-nitroso-N-acetylpenicillamine (SNAP), was not affected by AICAR except at the highest dose. AICAR increased endothelial nitric oxide synthase activity, as indicated by Ser1177 phosphorylation. In vivo, infusion of AICAR markedly increased muscle microvascular blood volume (approximately 60%, P<0.05), as was evidenced by contrast-enhanced ultrasound, without effects on blood pressure, femoral blood flow, or hind leg glucose uptake. CONCLUSIONS: Activation of AMPK by AICAR activates endothelial nitric oxide synthase in arteriolar endothelium by increasing its Ser1177 phosphorylation, which leads to vasodilation of resistance arteries and recruitment of microvascular perfusion in muscle.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Endotélio Vascular/efeitos dos fármacos , Ativadores de Enzimas/farmacologia , Microcirculação/efeitos dos fármacos , Músculo Esquelético/irrigação sanguínea , Óxido Nítrico/metabolismo , Ribonucleotídeos/farmacologia , Vasodilatadores/farmacologia , Proteínas Quinases Ativadas por AMP/antagonistas & inibidores , Aminoimidazol Carboxamida/administração & dosagem , Aminoimidazol Carboxamida/farmacologia , Animais , Artérias/efeitos dos fármacos , Artérias/enzimologia , Relação Dose-Resposta a Droga , Endotélio Vascular/enzimologia , Ativação Enzimática , Ativadores de Enzimas/administração & dosagem , Inibidores Enzimáticos/farmacologia , Membro Posterior , Infusões Intravenosas , Doadores de Óxido Nítrico/farmacologia , Óxido Nítrico Sintase Tipo III/antagonistas & inibidores , Óxido Nítrico Sintase Tipo III/metabolismo , Nitroarginina/farmacologia , Fosforilação , Pirazóis/farmacologia , Pirimidinas/farmacologia , Ratos , Fluxo Sanguíneo Regional/efeitos dos fármacos , Ribonucleotídeos/administração & dosagem , S-Nitroso-N-Acetilpenicilamina/farmacologia , Serina , Treonina , Fatores de Tempo , Resistência Vascular/efeitos dos fármacos , Vasodilatação/efeitos dos fármacos , Vasodilatadores/administração & dosagem
19.
J Mol Cell Cardiol ; 49(5): 781-90, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20691698

RESUMO

Subsequent to myocardial infarction, cardiomyocytes within the infarcted areas and border zones expose phosphatidylserine (PS) in the outer plasma membrane leaflet (flip-flop). We showed earlier that in addition to apoptosis, this flip-flop can be reversible in cardiomyocytes. We now investigated a possible role for Rho and downstream effector Rho-associated kinase (ROCK) in the process of (reversible) PS exposure and apoptosis in cardiomyocytes. In rat cardiomyoblasts (H9c2 cells) and isolated adult ventricular rat cardiomyocytes Clostridium difficile Toxin B (TcdB), a Rho GTPase family inhibitor, C3 transferase (C3), a Rho(A,B,C) inhibitor and the ROCK inhibitors Y27632 and H1152 were used to inhibit Rho-ROCK signaling. PS exposure was assessed via flow cytometry and fluorescent digital imaging microscopy using annexin V. Akt expression and phosphorylation were analyzed via Western blot, and Akt activity was inhibited by wortmannin. The cellular concentration activated caspase 3 was determined as a measure of apoptosis, and flippase activity was assessed via flow cytometry using NBD-labeled PS. TcdB, C3, Y27632 and H1152 all significantly increased PS exposure. TcdB, Y27632 and H1152 all significantly inhibited phosphorylation of the anti-apoptotic protein Akt and Akt inhibition by wortmannin lead to increased PS exposure. However, only TcdB and C3, but not ROCK- or Akt inhibition led to caspase 3 activation and thus apoptosis. Notably, pancaspase inhibitor zVAD only partially inhibited TcdB-induced PS exposure indicating the existence of apoptotic and non-apoptotic PS exposure. The induced PS exposure coincided with decreased flippase activity as measured with NBD-labeled PS flip-flop. In this study, we show a regulatory role for a novel signaling route, Rho-ROCK-flippase signaling, in maintaining asymmetrical membrane phospholipid distribution in cardiomyocytes.


Assuntos
Apoptose , Miócitos Cardíacos/enzimologia , Fosfatidilserinas/metabolismo , Proteínas de Transferência de Fosfolipídeos/metabolismo , Transdução de Sinais , Proteínas rho de Ligação ao GTP/antagonistas & inibidores , Quinases Associadas a rho/antagonistas & inibidores , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/análogos & derivados , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/farmacologia , ADP Ribose Transferases/farmacologia , Trifosfato de Adenosina/metabolismo , Amidas/farmacologia , Animais , Apoptose/efeitos dos fármacos , Proteínas de Bactérias/farmacologia , Toxinas Bacterianas/farmacologia , Toxinas Botulínicas/farmacologia , Caspase 3/metabolismo , Inibidores de Caspase , Linhagem Celular , Separação Celular , Ativação Enzimática/efeitos dos fármacos , Humanos , Miócitos Cardíacos/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Piridinas/farmacologia , Ratos , Transdução de Sinais/efeitos dos fármacos , Proteínas rho de Ligação ao GTP/metabolismo , Quinases Associadas a rho/metabolismo
20.
Microvasc Res ; 80(2): 240-9, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20417648

RESUMO

We studied the potential involvement of the Ca(2+)-independent atypical protein kinase C isoform PKCzeta in mediating the thrombin-induced increase in endothelial permeability. Studies were done using human dermal microvessel endothelial cells (HMEC), which we showed constitutively expressed PKCzeta. We quantified the patency of inter-endothelial junctions (IEJs) and endothelial barrier function by measuring transendothelial electrical resistance (TER) in confluent HMEC monolayers. In control monolayers, thrombin decreased TER by approximately 50%, indicating thrombin-dependent opening of IEJs. Thrombin also elicited increases in cytosolic Ca(2+) concentration [Ca(2+)](i), actin stress fiber formation, and myosin light chain (MLC) phosphorylation. Pan-PKC inhibitors, calphostin C and chelerythrine, abrogated these responses. Thrombin also decreased TER after depletion of conventional and novel Ca(2+)-dependent PKC isoforms using phorbol 12-myristate 13-acetate (PMA). In these PMA-treated cells, thrombin induced inter-endothelial gap formation, MLC phosphorylation, and actin stress fiber formation, but failed to increase [Ca(2+)](i). Inhibition of PKCzeta activation using the PKCzeta pseudosubstrate peptide (PSI), depletion of PKCzeta protein with siRNA, and competitive inhibition of PKCzeta activity using dominant-negative (dn) PKCzeta mutant all prevented the thrombin-induced decrease in TER and MLC phosphorylation. Expression of dn-PKCzeta also inhibited thrombin-induced RhoA activation. These findings reveal a novel Ca(2+)-independent, PKCzeta-dependent mechanism of thrombin-induced increase in endothelial permeability. The results raise the possibility that inhibition of PKCzeta may be a novel drug target for thrombin-induced inflammatory hyperpermeability.


Assuntos
Derme/irrigação sanguínea , Células Endoteliais/enzimologia , Hemostáticos/farmacologia , Microvasos/enzimologia , Proteína Quinase C/metabolismo , Trombina/farmacologia , Proteína rhoA de Ligação ao GTP/biossíntese , Citoesqueleto de Actina/efeitos dos fármacos , Citoesqueleto de Actina/metabolismo , Benzofenantridinas/farmacologia , Cálcio/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Permeabilidade Capilar/efeitos dos fármacos , Permeabilidade Capilar/fisiologia , Linhagem Celular , Citosol/efeitos dos fármacos , Citosol/metabolismo , Derme/enzimologia , Impedância Elétrica , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Inativação Gênica , Humanos , Junções Intercelulares/efeitos dos fármacos , Junções Intercelulares/metabolismo , Microvasos/citologia , Microvasos/efeitos dos fármacos , Cadeias Leves de Miosina/metabolismo , Fosforilação , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/genética , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA