Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 8(1): 1002, 2018 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-29343719

RESUMO

Exploration of human pulmonary artery endothelial cell (EC) as a prototypical biomechanical system has important pathophysiologic relevance because this cell type plays a key role in the development of a wide variety of clinical conditions. The complex hierarchical organization ranging from the molecular scale up to the cellular level has an intimate and intricate relationship to the barrier function between lung tissue and blood. To understand the innate molecule-cell-tissue relationship across varied length-scales, the functional role of c-Abl kinase in the cytoskeletal nano-biomechanics of ECs in response to barrier-altering agonists was investigated using atomic force microscopy. Concurrently, the spatially specific arrangement of cytoskeleton structure and dynamic distribution of critical proteins were examined using scanning electron microscopy and immunofluorescence. Reduction in c-Abl expression by siRNA attenuates both thrombin- and sphingosine 1-phosphate (S1P)-mediated structural changes in ECs, specifically spatially-defined changes in elastic modulus and distribution of critical proteins. These results indicate that c-Abl kinase is an important determinant of cortical actin-based cytoskeletal rearrangement. Our findings directly bridge the gap between kinase activity, structural complexity, and functional connectivity across varied length-scales, and suggest that manipulation of c-Abl kinase activity may be a potential target for the treatment of pulmonary barrier disorders.


Assuntos
Citoesqueleto de Actina/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Mecanotransdução Celular , Proteínas Proto-Oncogênicas c-abl/genética , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/ultraestrutura , Antígenos CD/genética , Antígenos CD/metabolismo , Fenômenos Biomecânicos , Caderinas/genética , Caderinas/metabolismo , Linhagem Celular , Cortactina/genética , Cortactina/metabolismo , Módulo de Elasticidade , Células Endoteliais/metabolismo , Células Endoteliais/ultraestrutura , Regulação da Expressão Gênica , Humanos , Lisofosfolipídeos/farmacologia , Microscopia de Força Atômica , Paxilina/genética , Paxilina/metabolismo , Proteínas Proto-Oncogênicas c-abl/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-abl/metabolismo , Artéria Pulmonar/citologia , Artéria Pulmonar/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Esfingosina/análogos & derivados , Esfingosina/farmacologia , Trombina/farmacologia
2.
Sci Rep ; 7(1): 14152, 2017 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-29075042

RESUMO

The endothelium serves as a size-selective barrier and tightly controls the fluid exchange from the circulation to the surrounding tissues. In this study, a multiplexed microscopy characterization is developed to study the spatio-temporal effects of Abl kinases on endothelial cytoskeletal structure using AFM, SEM, and immunofluorescence. Sphingosine 1-phosphate (S1P) produces significant endothelial barrier enhancement by means of peripheral actin rearrangement. However, Abl kinase inhibition by imatinib reduces rapid redistribution of the important cytoskeletal proteins to the periphery and their association with the cortical actin ring. Herein, it moderates the thickness of the cortical actin ring, and diminishes the increase in elastic modulus at the periphery and cytoplasm. These findings demonstrate that imatinib attenuates multiple cytoskeletal changes associated with S1P-mediated endothelial barrier enhancement and suggest a novel role for Abl kinases in mediating these S1P effects. These observations bridge the gap between molecule dynamics, structure complexity and function connectivity across varied length-scales to improve our understanding on human pulmonary endothelial barrier regulation. Moreover, our study suggests a framework for understanding form-function relationships in other biomechanical subsystems, wherein complex hierarchical organization programmed from the molecular scale to the cellular and tissue levels has an intimate relationship to the overall physiological function.


Assuntos
Citoesqueleto/efeitos dos fármacos , Mesilato de Imatinib/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Artéria Pulmonar/citologia , Antígenos CD/metabolismo , Caderinas/metabolismo , Células Cultivadas , Cortactina/metabolismo , Citoesqueleto/ultraestrutura , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Imunofluorescência , Humanos , Lisofosfolipídeos/farmacologia , Microscopia de Força Atômica , Microscopia Eletrônica de Varredura , Paxilina/metabolismo , Proteínas Proto-Oncogênicas c-abl/agonistas , Proteínas Proto-Oncogênicas c-abl/antagonistas & inibidores , Artéria Pulmonar/efeitos dos fármacos , Esfingosina/análogos & derivados , Esfingosina/farmacologia
3.
Biochem Biophys Res Commun ; 478(2): 599-605, 2016 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-27473658

RESUMO

Vascular integrity is primarily determined by endothelial cell (EC) cytoskeletal structure that is differentially regulated by various stimuli. In this study, atomic force microscopy (AFM) was used to characterize structural and mechanical properties in the cytoskeleton of cultured human pulmonary artery EC (HPAEC) and human lung microvascular EC (HLMVEC) by determining elastic properties (Young's modulus) in response to endogenous barrier protective agents sphingosine 1-phosphate (S1P) and hepatocyte growth factor (HGF), or the barrier disruptive molecule thrombin. Initial studies in unstimulated cells indicate higher baseline peripheral elastic modulus values in HPAEC (mean 2.9 KPa) than in HLMVEC (1.8 KPa). After 30 min of stimulation, S1P induced the highest Young's modulus increase (6.1 KPa) compared to the other barrier enhancing stimuli, HGF (5.8 KPa) and the pharmaceutical agent and S1P analog FTY720 (4.1 KPa). In contrast, the barrier disruptive agent thrombin decreased values from 2.5 KPa to 0.7 KPa depending on the cell type and treatment time. AFM topographical imaging supports these quantitative biophysical data regarding differential peripheral elastic properties in EC. Overall, these AFM studies provide novel insights into the biomechanical properties of human lung EC that regulate vascular barrier function and have potential applicability to pathophysiologic vascular leak syndromes such as acute lung injury.


Assuntos
Módulo de Elasticidade , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Pulmão/irrigação sanguínea , Fenômenos Biomecânicos , Linhagem Celular , Citoesqueleto/metabolismo , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Lisofosfolipídeos/metabolismo , Microscopia de Força Atômica , Artéria Pulmonar/citologia , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Trombina/metabolismo
4.
Am J Physiol Lung Cell Mol Physiol ; 308(3): L259-69, 2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-25480336

RESUMO

Endothelial dysfunction underlies the pathophysiology of vascular disorders such as acute lung injury (ALI) syndromes. Recent work has identified the Abl family kinases (c-Abl and Arg) as important regulators of endothelial cell (EC) barrier function and suggests that their inhibition by currently available pharmaceutical agents such as imatinib may be EC protective. Here we describe novel and differential effects of imatinib in regulating lung pathophysiology in two clinically relevant experimental models of ALI. Imatinib attenuates endotoxin (LPS)-induced vascular leak and lung inflammation in mice but exacerbates these features in a mouse model of ventilator-induced lung injury (VILI). We next explored these discrepant observations in vitro through investigation of the roles for Abl kinases in cultured lung EC. Imatinib attenuates LPS-induced lung EC permeability, restores VE-cadherin junctions, and reduces inflammation by suppressing VCAM-1 expression and inflammatory cytokine (IL-8 and IL-6) secretion. Conversely, in EC exposed to pathological 18% cyclic stretch (CS) (in vitro model of VILI), imatinib decreases VE-cadherin expression, disrupts cell-cell junctions, and increases IL-8 levels. Downregulation of c-Abl expression with siRNA attenuates LPS-induced VCAM-1 expression, whereas specific reduction of Arg reduces VE-cadherin expression in 18% CS-challenged ECs to mimic the imatinib effects. In summary, imatinib exhibits pulmonary barrier-protective and anti-inflammatory effects in LPS-injured mice and lung EC; however, imatinib exacerbates VILI as well as dysfunction in 18% CS-EC. These findings identify the Abl family kinases as important modulators of EC function and potential therapeutic targets in lung injury syndromes.


Assuntos
Anti-Inflamatórios/farmacologia , Benzamidas/farmacologia , Piperazinas/farmacologia , Pirimidinas/farmacologia , Lesão Pulmonar Induzida por Ventilação Mecânica/tratamento farmacológico , Animais , Anti-Inflamatórios/uso terapêutico , Benzamidas/uso terapêutico , Fenômenos Biomecânicos , Permeabilidade Capilar/efeitos dos fármacos , Células Cultivadas , Citocinas/biossíntese , Avaliação Pré-Clínica de Medicamentos , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/imunologia , Endotélio Vascular/fisiopatologia , Humanos , Mesilato de Imatinib , Lipopolissacarídeos/farmacologia , Pulmão/irrigação sanguínea , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Masculino , Camundongos Endogâmicos C57BL , Piperazinas/uso terapêutico , Proteínas Proto-Oncogênicas c-abl/metabolismo , Pirimidinas/uso terapêutico , Estresse Fisiológico , Lesão Pulmonar Induzida por Ventilação Mecânica/imunologia , alfa-Fetoproteínas/metabolismo
5.
Pharmacogenomics J ; 13(3): 218-26, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22310353

RESUMO

Management of severe sepsis, an acute illness with high morbidity and mortality, suffers from the lack of effective biomarkers and largely empirical predictions of disease progression and therapeutic responses. We conducted a genome-wide association study using a large randomized clinical trial cohort to discover genetic biomarkers of response to therapy and prognosis utilizing novel approaches, including combination markers, to overcome limitations of single-marker analyses. Sepsis prognostic models were dominated by clinical variables with genetic markers less informative. In contrast, evidence for gene-gene interactions were identified for sepsis treatment responses with genetic biomarkers dominating models for predicting therapeutic responses, yielding candidates for replication in other cohorts.


Assuntos
Biomarcadores Farmacológicos , Marcadores Genéticos , Proteína C/genética , Sepse/tratamento farmacológico , Sepse/genética , Progressão da Doença , Epistasia Genética , Estudo de Associação Genômica Ampla , Humanos , Polimorfismo de Nucleotídeo Único , Prognóstico , Ensaios Clínicos Controlados Aleatórios como Assunto , Proteínas Recombinantes/genética , Sepse/patologia
6.
Eur Respir J ; 38(1): 78-88, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21071472

RESUMO

Strategies to improve pulmonary endothelial barrier function are needed to reverse the devastating effects of vascular leak in acute respiratory distress syndrome. FTY720 is a pharmaceutical analogue of the potent barrier-enhancing phospholipid sphingosine 1-phosphate (S1P). FTY720 decreases vascular permeability by an incompletely characterised mechanism that differs from S1P. Here, we describe its barrier-promoting effects on intracellular signalling and junctional assembly formation in human pulmonary endothelium. Permeability of cultured human pulmonary endothelial cells was assessed using transendothelial electrical resistance and dextran transwell assays. Junctional complex formation was assessed using membrane fractionation and immunofluorescence. Pharmacological inhibitors and small interfering (si)RNA were utilised to determine the effects of individual components on permeability. Unlike S1P, FTY720 failed to induce membrane translocation of adherens junction or tight junction proteins. ß-catenin, occludin, claudin-5 or zona occludens protein (ZO)-1/ZO-2 siRNAs did not alter FTY720-induced barrier enhancement. FTY720 induced focal adhesion kinase (FAK) phosphorylation and focal adhesion formation, with FAK siRNA partially attenuating the prolonged phase of barrier enhancement. Inhibition of Src, protein kinase (PK)A, PKG, PKC or protein phosphatase 2A failed to alter FTY720-induced barrier enhancement. FTY720 increased c-Abl tyrosine kinase activity and c-Abl siRNA attenuated peak barrier enhancement after FTY720. FTY720 enhances endothelial barrier function by a novel pathway involving c-Abl signalling.


Assuntos
Células Endoteliais/citologia , Regulação da Expressão Gênica , Pulmão/efeitos dos fármacos , Propilenoglicóis/farmacologia , Proteínas Proto-Oncogênicas c-abl/metabolismo , Esfingosina/análogos & derivados , Junções Aderentes/patologia , Células Cultivadas , Cloridrato de Fingolimode , Humanos , Inflamação , Lisofosfolipídeos/metabolismo , Permeabilidade , Fosforilação , Artéria Pulmonar/citologia , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Esfingosina/metabolismo , Esfingosina/farmacologia , Frações Subcelulares/metabolismo , Junções Íntimas/patologia
7.
J Pharmacol Exp Ther ; 331(1): 54-64, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19592667

RESUMO

Novel therapies are needed to address the vascular endothelial cell (EC) barrier disruption that occurs in inflammatory diseases such as acute lung injury (ALI). We previously demonstrated the potent barrier-enhancing effects of both sphingosine 1-phosphate (S1P) and the structurally similar compound FTY720 [2-amino-2-(2-[4-octylphenyl]ethyl)-1,3-propanediol] in inflammatory lung injury. In this study, we examined the therapeutic potential of several novel FTY720 analogs to reduce vascular leak. Similar to S1P and FTY720, the (R)- and (S)-enantiomers of FTY720 phosphonate and enephosphonate analogs produce sustained EC barrier enhancement in vitro, as seen by increases in transendothelial electrical resistance (TER). In contrast, the (R)- and (S)-enantiomers of FTY720-regioisomeric analogs disrupt EC barrier integrity in a dose-dependent manner. Barrier-enhancing FTY720 analogs demonstrate a wider protective concentration range in vitro (1-50 microM) and greater potency than either S1P or FTY720. In contrast to FTY720-induced EC barrier enhancement, S1P and the FTY720 analogs dramatically increase TER within minutes in association with cortical actin ring formation. Unlike S1P, these FTY720 analogs exhibit differential phosphorylation effects without altering the intracellular calcium level. Inhibitor studies indicate that barrier enhancement by these analogs involves signaling via G(i)-coupled receptors, tyrosine kinases, and lipid rafts. Consistent with these in vitro responses, the (S)-phosphonate analog of FTY720 significantly reduces multiple indices of alveolar and vascular permeability in a lipopolysaccharide-mediated murine model of ALI (without significant alterations in leukocyte counts). These results demonstrate the capacity for FTY720 analogs to significantly decrease pulmonary vascular leakage and inflammation in vitro and in vivo.


Assuntos
Permeabilidade Capilar/efeitos dos fármacos , Permeabilidade Capilar/fisiologia , Cloridrato de Fingolimode/análogos & derivados , Mediadores da Inflamação/síntese química , Mediadores da Inflamação/farmacologia , Organofosfonatos/síntese química , Organofosfonatos/farmacologia , Propilenoglicóis/síntese química , Propilenoglicóis/farmacologia , Artéria Pulmonar/efeitos dos fármacos , Esfingosina/análogos & derivados , Animais , Linhagem Celular , Cloridrato de Fingolimode/síntese química , Cloridrato de Fingolimode/farmacologia , Humanos , Pulmão/irrigação sanguínea , Pulmão/efeitos dos fármacos , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Artéria Pulmonar/patologia , Esfingosina/síntese química , Esfingosina/farmacologia
8.
Eur Respir J ; 33(3): 612-24, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19010997

RESUMO

Despite an encouraging outcome of antioxidant therapy in animal models of acute lung injury, effective antioxidant agents for clinical application remain to be developed. The present study investigated the effect of pre-treatment with amifostine, a thiol antioxidant compound, on lung endothelial barrier dysfunction induced by Gram-negative bacteria wall-lipopolysaccharide (LPS). Endothelial permeability was monitored by changes in transendothelial electrical resistance. Cytoskeletal remodelling and reactive oxygen species (ROS) production was examined by immunofluorescence. Cell signalling was assessed by Western blot. Measurements of Evans blue extravasation, cell count and protein content in bronchoalveolar lavage fluid were used as in vivo parameters of lung vascular permeability. Hydrogen peroxide, LPS and interleukin-6 caused cytoskeletal reorganisation and increased permeability in the pulmonary endothelial cells, reflecting endothelial barrier dysfunction. These disruptive effects were inhibited by pre-treatment with amifostine and linked to the amifostine-mediated abrogation of ROS production and redox-sensitive signalling cascades, including p38, extracellular signal regulated kinase 1/2, mitogen-activated protein kinases and the nuclear factor-kappaB pathway. In vivo, concurrent amifostine administration inhibited LPS-induced oxidative stress and p38 mitogen-activated protein kinase activation, which was associated with reduced vascular leak and neutrophil recruitment to the lungs. The present study demonstrates, for the first time, protective effects of amifostine against lipopolysaccharide-induced lung vascular leak in vitro and in animal models of lipopolysaccharide-induced acute lung injury.


Assuntos
Amifostina/farmacologia , Permeabilidade Capilar/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Protetores contra Radiação/farmacologia , Animais , Antioxidantes/metabolismo , Líquido da Lavagem Broncoalveolar , Citoesqueleto/metabolismo , Inflamação , Interleucina-6/metabolismo , Lipopolissacarídeos/metabolismo , Pulmão/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Espécies Reativas de Oxigênio , Transdução de Sinais
9.
Eur Respir J ; 30(3): 429-35, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17537765

RESUMO

The pathogenesis of acute lung injury includes transendothelial diapedesis of leukocytes into lung tissues and disruption of endothelial/epithelial barriers leading to protein-rich oedema. In vitro studies show that the microtubule network plays a role in the regulation of endothelial permeability as well as in neutrophil locomotion. It was hypothesised that the microtubule-stabilising agent, taxol, might attenuate inflammation and vascular leak associated with acute lung injury in vivo. The effect of intravenously delivered taxol was assessed using a model of murine lung injury induced by intratracheal lipopolysaccharide (LPS) administration. Parameters of lung injury and inflammation were assessed 18 h after treatment. Intravenously delivered taxol significantly reduced inflammatory histological changes in lung parenchyma and parameters of LPS-induced inflammation: infiltration of proteins and inflammatory cells into bronchoalveolar lavage fluid, lung myeloperoxidase activity, and extravasation of Evans blue-labelled albumin into lung tissue. Taxol alone (in the absence of LPS) had no appreciable effect on these parameters. In addition to lung proteins, intravenous taxol reduced accumulation of leukocytes in ascitic fluid in a model of LPS-induced peritonitis. Taken together, the present data demonstrate that microtubule stabilisation with taxol systemically attenuates lipopolysaccharide-induced inflammation and vascular leak.


Assuntos
Endotoxemia/imunologia , Lipopolissacarídeos/imunologia , Paclitaxel/farmacologia , Síndrome do Desconforto Respiratório/imunologia , Síndrome de Resposta Inflamatória Sistêmica/imunologia , Moduladores de Tubulina/farmacologia , Animais , Líquido da Lavagem Broncoalveolar/imunologia , Síndrome de Vazamento Capilar/imunologia , Síndrome de Vazamento Capilar/patologia , Endotoxemia/patologia , Extravasamento de Materiais Terapêuticos e Diagnósticos/imunologia , Extravasamento de Materiais Terapêuticos e Diagnósticos/patologia , Contagem de Leucócitos , Pulmão/imunologia , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Peritonite/imunologia , Peritonite/patologia , Peroxidase/metabolismo , Edema Pulmonar/imunologia , Edema Pulmonar/patologia , Síndrome do Desconforto Respiratório/patologia , Síndrome de Resposta Inflamatória Sistêmica/patologia
10.
Cell Signal ; 19(8): 1754-64, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17475445

RESUMO

Novel therapeutic strategies are needed to reverse the loss of endothelial cell (EC) barrier integrity that occurs during inflammatory disease states such as acute lung injury. We previously demonstrated potent EC barrier augmentation in vivo and in vitro by the platelet-derived phospholipid, sphingosine 1-phosphate (S1P) via ligation of the S1P1 receptor. The S1P analogue, FTY720, similarly exerts barrier-protective vascular effects via presumed S1P1 receptor ligation. We examined the role of the S1P1 receptor in sphingolipid-mediated human lung EC barrier enhancement. Both S1P and FTY-induced sustained, dose-dependent barrier enhancement, reflected by increases in transendothelial electrical resistance (TER), which was abolished by pertussis toxin indicating Gi-coupled receptor activation. FTY-mediated increases in TER exhibited significantly delayed onset and intensity relative to the S1P response. Reduction of S1P1R expression (via siRNA) attenuated S1P-induced TER elevations whereas the TER response to FTY was unaffected. Both S1P and FTY rapidly (within 5 min) induced S1P1R accumulation in membrane lipid rafts, but only S1P stimulated S1P1R phosphorylation on threonine residues. Inhibition of PI3 kinase activity attenuated S1P-mediated TER increases but failed to alter FTY-induced TER elevation. Finally, S1P, but not FTY, induced significant myosin light chain phosphorylation and dramatic actin cytoskeletal rearrangement whereas reduced expression of the cytoskeletal effectors, Rac1 and cortactin (via siRNA), attenuated S1P-, but not FTY-induced TER elevations. These results mechanistically characterize pulmonary vascular barrier regulation by FTY720, suggesting a novel barrier-enhancing pathway for modulating vascular permeability.


Assuntos
Células Endoteliais/efeitos dos fármacos , Imunossupressores/farmacologia , Propilenoglicóis/farmacologia , Receptores de Lisoesfingolipídeo/metabolismo , Esfingosina/análogos & derivados , Adenoviridae/genética , Permeabilidade Capilar , Células Cultivadas , Citoesqueleto/metabolismo , Impedância Elétrica , Endotélio Vascular/citologia , Cloridrato de Fingolimode , Humanos , Pulmão/citologia , Modelos Biológicos , Fosforilação , Artéria Pulmonar/citologia , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Esfingosina/farmacologia , Treonina/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo
11.
Curr Drug Targets ; 8(4): 509-14, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17430121

RESUMO

Sepsis is characterized physiologically by an aberrant systemic inflammatory response and microvascular dysfunction. While appropriate antibiotics and supportive care are essential in the management of the septic patient, therapies targeting specific aspects of the pathophysiology could have a significant impact on the morbidity and mortality associated with both sepsis and its sequlea, including acute lung injury (ALI). We have characterized several mediators of endothelial cell (EC) barrier function that may serve as novel therapies for sepsis-induced microvascular dysfunction including simvastatin, adenosine triphosphate (ATP), sphingosine 1-phosphate (S1P), and activated protein C (APC). Notably, APC is already available for the treatment of severe sepsis, however, to date its mechanism of action has been unclear. While distinct in many ways, we have found that these agonists have in common the ability to induce dynamic rearrangement of the EC actin cytoskeleton that corresponds to barrier protection. In addition, we have extended our in vitro findings to relevant animal models of endotoxin-induced acute lung injury and have confirmed beneficial effects of both simvastatin and S1P which are associated with evidence of decreased vascular permeability in this setting. Moreover, our data also indicate that APC effects in sepsis may be largely due to augmentation of EC barrier function affecting decreased microvascular permeability. We speculate that the administration of direct modulators of EC barrier function and microvascular permeability, such as those described here, may ultimately become the standard of care for the septic patient.


Assuntos
Capilares/fisiopatologia , Permeabilidade Capilar/efeitos dos fármacos , Sepse/fisiopatologia , Vênulas/fisiopatologia , Trifosfato de Adenosina/farmacologia , Trifosfato de Adenosina/uso terapêutico , Animais , Capilares/efeitos dos fármacos , Cães , Humanos , Lisofosfolipídeos/farmacologia , Lisofosfolipídeos/uso terapêutico , Proteína C/farmacologia , Proteína C/uso terapêutico , Sepse/tratamento farmacológico , Sinvastatina/farmacologia , Sinvastatina/uso terapêutico , Esfingosina/análogos & derivados , Esfingosina/farmacologia , Esfingosina/uso terapêutico , Vênulas/efeitos dos fármacos
12.
Microvasc Res ; 72(1-2): 3-11, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16820176

RESUMO

Angiogenesis or the formation of new blood vessels is important in the growth and metastatic potential of various cancers. Therefore, agents that inhibit angiogenesis have important therapeutic implications in numerous malignancies. We examined the effects of methylnaltrexone (MNTX), a peripheral mu opioid receptor antagonist, on agonist-induced human EC proliferation and migration, two key components in angiogenesis. Using human dermal microvascular EC, we observed that morphine sulfate (MS), the active metabolite, morphine-6-glucuronide (M6G), DAMGO ([d-Ala(2), N-Me-Phe(4), Gly(5)-ol]enkaphalin) and VEGF induced migration which were inhibited by pretreatment with MNTX at therapeutically relevant concentration (0.1 microM). The biologically inactive metabolite morphine-3-glucuronide (M3G) did not affect EC migration. We next examined the mechanism(s) by which MNTX inhibits opioid and VEGF-induced angiogenesis using human pulmonary microvascular EC. MS and DAMGO induced Src activation which was required for VEGF receptor transactivation and opioid-induced EC proliferation and migration. MNTX inhibited MS, DAMGO and VEGF induced tyrosine phosphorylation (transactivation) of VEGF receptors 1 and 2. Furthermore, MS, DAMGO and VEGF induced RhoA activation which was inhibited by MNTX or VEGF receptor tyrosine kinase inhibition. Finally, MNTX or silencing RhoA expression (siRNA) blocked MS, DAMGO and VEGF-induced EC proliferation and migration. Taken together, these results indicate that MNTX inhibits opioid-induced EC proliferation and migration via inhibition of VEGF receptor phosphorylation/transactivation with subsequent inhibition of RhoA activation. These results suggest that MNTX inhibition of angiogenesis can be a useful therapeutic intervention for cancer treatment.


Assuntos
Naltrexona/análogos & derivados , Neoplasias/tratamento farmacológico , Neovascularização Patológica , Peptídeos Opioides/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Inibidores da Angiogênese/farmacologia , Proliferação de Células , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Humanos , Pulmão/irrigação sanguínea , Pulmão/patologia , Morfina/farmacologia , Derivados da Morfina/farmacologia , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Compostos de Amônio Quaternário/farmacologia , Ativação Transcricional
13.
Genes Immun ; 7(1): 27-35, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16355111

RESUMO

Proinflammatory and immunoregulatory products from C3 play a major role in phagocytosis, respiratory burst, and airways inflammation. C3 is critical in adaptive immunity; studies in mice deficient in C3 demonstrate that features of asthma are significantly attenuated in the absence of C3. To test the hypothesis that the C3 gene on chromosome 19p13.3-p13.2 contains variants associated with asthma and related phenotypes, we genotyped 25 single nucleotide polymorphism (SNP) markers distributed at intervals of approximately 1.9 kb within the C3 gene in 852 African Caribbean subjects from 125 nuclear and extended pedigrees. We used the multiallelic test in the family-based association test program to examine sliding windows comprised of 2-6 SNPs. A five-SNP window between markers rs10402876 and rs366510 provided strongest evidence for linkage in the presence of linkage disequilibrium for asthma, high log[total IgE], and high log[IL-13]/[log[IFN-gamma] in terms of global P-values (P = 0.00027, 0.00013, and 0.003, respectively). A three-SNP haplotype GGC for the first three of these markers showed best overall significance for the three phenotypes (P = 0.003, 0.007, 0.018, respectively) considering haplotype-specific tests. Taken together, these results implicate the C3 gene as a priority candidate controlling risk for asthma and allergic disease in this population of African descent.


Assuntos
Asma/genética , População Negra , Complemento C3/genética , Predisposição Genética para Doença , Barbados/etnologia , População Negra/etnologia , Região do Caribe/etnologia , Variação Genética , Genótipo , Humanos , Fenótipo , Polimorfismo de Nucleotídeo Único
14.
Artigo em Inglês | MEDLINE | ID: mdl-15853752

RESUMO

L-Mevalonic acid is the distant precursor of cholesterol, in contrast to cholesterol, L-mevalonic acid, its distant precursor gives rise to farnesyl and geranylgeranyl pyrophosphates in relatively few metabolic steps. These isoprenyl pyrophophates covalently conjugate with specific G-proteins and serve as membrane anchors enabling them to carry out their function. Although farnesyl-proteins may participate in signal transduction, geranylgeranyl-proteins (e.g., Rho GTP binding proteins) are well known to downregulate signaling pathways by inhibiting L-mevalonic acid synthesis. Such inhibitors include 3-hydroxy-3-methylglutaryl CoA reductase inhibitors, drugs (statins) and isoprenoids of dietary origins, where Rho protein activation appears to be necessary for cellular-mediated thrombin generation. Thrombin and other proteases (e.g., coagulation factor Xa, tryptase) upregulate protease-activated receptor (PAR) synthesis and PAR activation promotes synthesis and expression of other proteins [e.g., tissue factor (TF) and plasminogen activator inhibitor-1 (PAI-1)]. With the PAR-1 activating peptide SSFLRNP, we found that either cerivastatin or atorvastatin mitigated platelet stimulation in a time- and dose-dependent manner, as predicted if a statin-mediated Rho pathway is required. We also found that simvastatin decreased prothrombin fragments F1+2 in plasma from type 2 diabetics, demonstrating that statins downregulate thrombin generation. Thus, independent of cholesterol, statins and dietary isoprenoids behave as inhibitors of TF-dependent thrombin generation. Because thrombin has multiple physiological functions, the 20 pleiotropic effects reported for statins may reflect a common mechanism for downregulation of thrombin-mediated events, in particular at the cellular level.


Assuntos
Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Trombina/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Trombina/metabolismo , Trombina/uso terapêutico
15.
Stem Cells Dev ; 13(5): 496-508, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15588507

RESUMO

Angiogenesis is a multistep process involving the endothelial cell (EC) cytoskeleton in migration, proliferation, and barrier stabilization. Although precise intracellular pathways by which angiogenic tube formation occurs remain poorly understood, we speculated that interactions between the cytoskeleton and apoptosis are involved and explored cytoskeletal rearrangement and caspase activation in human lung microvascular EC capillary-like tube formation induced by sphingosine 1-phosphate (Sph 1-P) and vascular endothelial growth factor (VEGF). Sph 1-P and VEGF enhance tube formation quantified by a Tube Immaturity Index (TII) generated from the ratio of cell number to tube length, with concomitant morphologic and actomyosin network changes. Angiogenesis was temporally grouped into three stages with early changes characterized by cortical actin localization, whereas midstage tube development demonstrated elongated EC with peripheral actin labeling with transcellular stress fibers. Late tube formation was characterized by broad actin distribution and presence of caspase-positive EC. Phosphorylated MLC immunoreactivity was present at all stages, suggesting that coordinate Rho kinase and MLCK involvement is important to Sph 1-P-induced cell motility; however, chemical inhibition of either MLCK or Rho kinase failed to alter early tube formation. To address whether gaps created by apoptosis expand the lumen, Sph 1-P-induced tubes were differentiated in the presence of caspase inhibitor z-Val-Ala-Asp-fluoromethylketone (zVAD-FMK). Capillary-like tube maturation, but not length, was decreased by zVAD-FMK treatment. These studies suggest that Sph 1-P may induce EC tube formation by regulating early cytoskeletal rearrangement, whereas EC apoptosis within capillary-like tubes is necessary for late stage Sph 1-P-induced tube maturation and lumen formation.


Assuntos
Capilares/patologia , Caspases/metabolismo , Citoesqueleto/química , Pulmão/irrigação sanguínea , Lisofosfolipídeos/química , Esfingosina/análogos & derivados , Esfingosina/química , Actinas/metabolismo , Actomiosina/química , Clorometilcetonas de Aminoácidos/farmacologia , Apoptose , Membrana Basal/patologia , Capilares/metabolismo , Diferenciação Celular , Movimento Celular , Proliferação de Células , Colágeno/farmacologia , Citoesqueleto/metabolismo , Combinação de Medicamentos , Células Endoteliais/citologia , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Humanos , Laminina/farmacologia , Pulmão/metabolismo , Microcirculação , Microscopia de Fluorescência , Neovascularização Patológica , Fosforilação , Proteoglicanas/farmacologia , Fatores de Tempo , Fator A de Crescimento do Endotélio Vascular/metabolismo
16.
Biochemistry (Mosc) ; 67(1): 75-84, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11841342

RESUMO

Confluent endothelium serves as a selective barrier between the vascular space of blood vessels and underlying tissues. Compromised barrier function of the endothelium in response to inflammation mediators, such as thrombin, is accompanied by reversible cell rounding and interendothelial gap formation. Endothelial barrier integrity substantially depends on the cytoskeleton, which ensures actin stress fiber formation and via actomyosin-driven contraction regulates cell shape and adhesion. Recent studies have shown the sequence of events that mediate signal transduction in endothelial cells. Binding of thrombin with its receptor initiates activation of heterotrimeric G-proteins, which, in turn, entails a decrease in cAMP level in the cell, increase in intracellular Ca2+ and diacylglycerol concentration, and activation of the small G-protein Rho. Phosphorylation of myosin light chains as a result of activation of myosin light chain kinase and inactivation of myosin phosphatases stimulates stress fiber formation and triggers actomyosin contraction. In addition, thrombin-induced rearrangement in the endothelial cytoskeleton is regulated by Ca2+/calmodulin-dependent protein kinase, protein kinase C, and tyrosine protein kinases. This review focuses on presently known biochemical mechanisms of cell response to thrombin and their role in endothelial barrier dysfunction.


Assuntos
Endotélio Vascular/metabolismo , Trombina/metabolismo , Animais , Cálcio/metabolismo , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , AMP Cíclico/metabolismo , Diglicerídeos/metabolismo , Endotélio/metabolismo , Humanos , Inositol 1,4,5-Trifosfato/metabolismo , Cadeias Leves de Miosina/metabolismo , Miosinas/metabolismo , Permeabilidade , Fosforilação , Proteína Quinase C/metabolismo , Proteínas Tirosina Quinases/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...