Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Blood ; 123(3): 442-50, 2014 Jan 16.
Article in English | MEDLINE | ID: mdl-24081657

ABSTRACT

von Willebrand factor (vWF) secretion by endothelial cells (ECs) is essential for hemostasis and thrombosis; however, the molecular mechanisms are poorly understood. Interestingly, we observed increased bleeding in EC-Gα13(-/-);Gα12(-/-) mice that could be normalized by infusion of human vWF. Blood from Gα12(-/-) mice exhibited significantly reduced vWF levels but normal vWF multimers and impaired laser-induced thrombus formation, indicating that Gα12 plays a prominent role in EC vWF secretion required for hemostasis and thrombosis. In isolated buffer-perfused mouse lungs, basal vWF levels were significantly reduced in Gα12(-/-), whereas thrombin-induced vWF secretion was defective in both EC-Gαq(-/-);Gα11(-/-) and Gα12(-/-) mice. Using siRNA in cultured human umbilical vein ECs and human pulmonary artery ECs, depletion of Gα12 and soluble N-ethylmaleimide-sensitive-fusion factor attachment protein α (α-SNAP), but not Gα13, inhibited both basal and thrombin-induced vWF secretion, whereas overexpression of activated Gα12 promoted vWF secretion. In Gαq, p115 RhoGEF, and RhoA-depleted human umbilical vein ECs, thrombin-induced vWF secretion was reduced by 40%, whereas basal secretion was unchanged. Finally, in vitro binding assays revealed that Gα12 N-terminal residues 10-15 mediated the binding of Gα12 to α-SNAP, and an engineered α-SNAP binding-domain minigene peptide blocked basal and evoked vWF secretion. Discovery of obligatory and complementary roles of Gα12 and Gαq/11 in basal vs evoked EC vWF secretion may provide promising new therapeutic strategies for treatment of thrombotic disease.


Subject(s)
Endothelial Cells/cytology , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , rhoA GTP-Binding Protein/metabolism , von Willebrand Factor/metabolism , Animals , Antibodies, Monoclonal/chemistry , Gene Expression Regulation , Hemostasis , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Knockout , Platelet Adhesiveness , Protein Binding , RNA, Small Interfering/metabolism , Signal Transduction , Soluble N-Ethylmaleimide-Sensitive Factor Attachment Proteins/chemistry , Thrombosis
2.
Microvasc Res ; 80(3): 355-64, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20888351

ABSTRACT

Monoclonal antibody (mAb) 9B9 to angiotensin-converting enzyme (ACE) demonstrates selective accumulation in lung tissue of the rat, hamster, cat, monkey and human after systemic injection. It has also been demonstrated that mAb 9B9 is the useful tool for targeting therapeutic agents or genes to lung endothelium. In this study, we describe the generation and characterization of a single-chain derivative (scFv) of mAb9B9 (scFv 9B9). In vitro, scFv9B9 retains the ability of the parental antibody to recognize human and rat ACE when expressed both on the surface of phage and as a soluble protein in prokaryotic and eukaryotic expression systems. The ability of scFv 9B9 presented by phage or the soluble protein labeled with I(125) to recognize ACE in the pulmonary circulation was also confirmed in an in vivo rat model. Sequence analysis revealed a putative glycosylation site in close proximity to the complementarity determining region 2 (CDR2) of the scFv 9B9 heavy chain. Mutation of Asn68 to Gln in the heavy chain of scFv 9B9 eliminated the glycosylation site and significantly improved the binding affinity of scFv 9B9 to human ACE as determined by cell ELISA and Western Blot. Moreover, Asn68Gln scFv 9B9 showed a greater rate of secretion at 30°C than wild type scFv 9B9, but had a decreased thermal stability at 37°C. The development of a stable and functional single-chain format of mAb 9B9 which specifically recognizes human and rat ACE represents a novel antibody-based reagent suitable for targeted delivery of drugs/genes to the pulmonary circulation.


Subject(s)
Antibodies, Monoclonal/metabolism , Cloning, Molecular , Drug Carriers , Endothelial Cells/enzymology , Lung/blood supply , Peptidyl-Dipeptidase A/immunology , Single-Chain Antibodies/metabolism , Amino Acid Sequence , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/genetics , Antibody Specificity , Base Sequence , Blotting, Western , CHO Cells , Complementarity Determining Regions , Cricetinae , Cricetulus , Endothelial Cells/immunology , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Glycosylation , Humans , Hybridomas , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Protein Conformation , Protein Processing, Post-Translational , Protein Stability , Rats , Single-Chain Antibodies/biosynthesis , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/genetics , Species Specificity , Structure-Activity Relationship , Temperature , Transfection
3.
Microvasc Res ; 80(2): 250-7, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20430040

ABSTRACT

Reduced lung capillary expression of angiotensin I-converting enzyme (ACE), a key enzyme in cardiovascular pathophysiology, and of caveolin-1, an important regulator of endothelial cell signalling, has been demonstrated in various models of pulmonary arterial hypertension (PAH). We addressed the relationship between PAH and ACE expression in caveolin-1 knockout mice (Cav1(-/-)), which have moderate PAH. Tissue ACE activity was reduced by 50% in lungs from 3-month-old Cav1(-/-) mice compared to wild type (WT). A similar reduction in lung endothelial ACE expression was observed by measuring the lung uptake of (125)I-labeled monoclonal anti-ACE antibody and by quantitative immunohistochemistry. These alterations in ACE are limited to capillary segments of the pulmonary circulation. Functionally, the increase in pulmonary artery pressure (PAP) in response to ACE conversion of angiotensin I to angiotensin II in isolated, perfused mouse lungs was reduced significantly in Cav1(-/-) mice compared to WT. Thus, these complementary approaches demonstrate the dependence of lung microvascular endothelial cell ACE protein expression on caveolin-1 expression and underscore the vital role of caveolin-1-regulated pulmonary vascular homeostasis on endothelial ACE expression and activity. In summary, we have revealed a novel role of caveolin-1 in the regulation of ACE expression in pulmonary capillary endothelial cells. Further understanding of the mechanism by which reduced caveolin-1 expression leads altered pulmonary vascular development, PAH, and reduced ACE expression may have important clinical implications in patients with these severe lung diseases.


Subject(s)
Caveolin 1/genetics , Hypertension, Pulmonary/enzymology , Lung/enzymology , Peptidyl-Dipeptidase A/metabolism , Angiotensin I/metabolism , Angiotensin I/pharmacology , Angiotensin II/metabolism , Angiotensin II/pharmacology , Animals , Blood Pressure , Capillaries/enzymology , Capillaries/pathology , Caveolin 1/metabolism , Endothelial Cells/enzymology , Endothelial Cells/pathology , Gene Expression Regulation, Enzymologic , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/physiopathology , Immunohistochemistry , Lung/blood supply , Lung/pathology , Mice , Mice, Knockout , Perfusion , Pulmonary Artery/drug effects , Pulmonary Artery/enzymology , Pulmonary Artery/physiopathology , Signal Transduction
4.
Anesth Analg ; 105(5): 1363-70, table of contents, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17959967

ABSTRACT

BACKGROUND: Acute lung injury (ALI) is a frequent complication in septic patients. Previously, we found that propofol, a highly lipid-soluble anesthetic, attenuates ischemia-reperfusion and oxidative lung injury in the isolated perfused rat lung. In the present study, we evaluated the effect of propofol on endotoxin-induced ALI and endothelial dysfunction. METHODS: The effect of propofol on endotoxin-induced lung endothelial injury was evaluated by plasma and lung tissue homogenate angiotensin I converting enzyme (ACE) activity, pulmonary vascular anti-ACE monoclonal antibody binding, and lung wet weight to body weight ratio (LW/BW). RESULTS: When injected IV into rats, endotoxin produced endothelial cell injury and lung edema, as indicated by: 1) an increase in plasma ACE activity, 2) a decrease in lung ACE activity and anti-ACE monoclonal antibody binding, and 3) an increase in LW/BW. Monoclonal antibody 1A2 was up to 1.8 times more sensitive than other anti-ACE monoclonal antibodies in detecting the decrease in ACE in lungs of endotoxin-treated rats. Pretreatment of rats with a bolus of propofol before endotoxin injection significantly inhibited the increase in ACE activity in the blood, the decrease in ACE activity in the lung, the decrease in anti-ACE monoclonal antibody binding in the lung, and the increase in LW/BW ratio. Importantly, propofol also significantly increased the survival rate of endotoxin-treated animals. The protective effect of propofol in endotoxin-treated animals in vivo was confirmed in vitro, i.e., propofol decreased endothelial cell injury and ACE shedding from endothelial cells in culture. CONCLUSIONS: These results suggest that propofol offers significant protection against endotoxin-induced pulmonary microvessel endothelial cell injury and that anti-ACE monoclonal antibody 1A2 is a sensitive probe for monitoring endothelial dysfunction and ALI during sepsis.


Subject(s)
Endothelial Cells/pathology , Endotoxins/toxicity , Peptidyl-Dipeptidase A/metabolism , Propofol/therapeutic use , Pulmonary Edema/prevention & control , Animals , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Male , Propofol/pharmacology , Pulmonary Edema/chemically induced , Pulmonary Edema/enzymology , Pulmonary Edema/pathology , Rats , Rats, Sprague-Dawley
5.
Anesthesiology ; 104(4): 777-85, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16571974

ABSTRACT

BACKGROUND: Caveolae mediated transendothelial transport of albumin has recently been shown to be the primary mechanism regulating microvascular endothelial albumin permeability. The authors investigated the effects of isoflurane and sevoflurane on pulmonary endothelial albumin permeability and assessed the potential role of the caveolae scaffold protein, caveolin-1, in these effects. METHODS: Isolated rat lungs and cultured rat lung microvessel endothelial cells (RLMVECs) were exposed to 1.0 or 2.0 minimum alveolar concentration (MAC) isoflurane or sevoflurane for 30 min. I-albumin permeability-surface area product and capillary filtration coefficient were determined in the isolated lungs. In RLMVECs, uptake and transendothelial transport of I-albumin were measured in the absence and presence of pretreatment with 2 mm methyl-beta-cyclodextrin, a caveolae-disrupting agent. Uptake of fluorescent-labeled albumin, as well as phosphorylation of Src kinase and caveolin-1, was also determined. In Y14F-caveolin-1 mutant (nonphosphorylatable) expressing RLMVECs, uptake of I-albumin and phosphorylation of caveolin-1 were evaluated. RESULTS: In the isolated lungs, 2.0 MAC isoflurane increased I-albumin permeability-surface area product by 48% without affecting capillary filtration coefficient. In RLMVECs, isoflurane more than doubled the uptake of I-albumin and caused a 54% increase in the transendothelial transport of I-albumin. These effects were blocked by pretreatment with methyl-beta-cyclodextrin. The isoflurane-induced increase in uptake of I-albumin in wild-type RLMVECs was abolished in the Y14F-caveolin-1 mutant expressing cells. Isoflurane also caused a twofold increase in Src and caveolin-1 phosphorylation. Neither 1.0 MAC isoflurane nor 1.0 or 2.0 MAC sevoflurane affected any index of albumin transport or phosphorylation of caveolin-1. CONCLUSION: Isoflurane, but not sevoflurane, increased lung transendothelial albumin permeability through enhancement of caveolae-mediated albumin uptake and transport in the isolated lung. This effect may involve an enhanced phosphorylation of caveolin-1.


Subject(s)
Albumins/metabolism , Anesthetics, Inhalation/pharmacology , Capillary Permeability/drug effects , Caveolin 1/metabolism , Endothelium, Vascular/metabolism , Isoflurane/pharmacology , Lung/metabolism , Methyl Ethers/pharmacology , Animals , Female , In Vitro Techniques , Male , Phosphorylation , Rats , Rats, Sprague-Dawley , Sevoflurane , src-Family Kinases/metabolism
6.
Anesth Analg ; 100(4): 929-936, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15781500

ABSTRACT

Lung dysfunction after cardiopulmonary bypass and lung transplantation results from oxidant-mediated cellular damage. Previously, we observed the shedding of angiotensin-converting enzyme (ACE) from the endothelial cell surface to be a more sensitive and earlier marker of oxidative lung endothelial injury than lung wet-to-dry weight ratio. The aim of this study was to evaluate the potential of the anesthetic propofol, which has antioxidant properties, to prevent oxidative lung injury by measuring ACE shedding. ACE release from isolated perfused rat lungs increased significantly after ischemia-reperfusion (I/R). Propofol significantly decreased I/R-induced ACE release by 23.4% (P < 0.05). Perfusion with 0.75 mM H(2)O(2) also caused ACE release from the lung microvasculature, which was similarly attenuated by propofol. The protective effect of propofol on H(2)O(2)-induced ACE shedding was confirmed in vitro using Chinese Hamster Ovary cells overexpressing human ACE. Thus, propofol can attenuate oxidative injury of the pulmonary endothelium as detected by ACE shedding in I/R and H(2)O(2) models of acute lung injury.


Subject(s)
Anesthetics, Intravenous/therapeutic use , Lung Diseases/prevention & control , Oxidative Stress/drug effects , Propofol/therapeutic use , Reperfusion Injury/prevention & control , Animals , CHO Cells , Cricetinae , Endothelium/pathology , Enzyme-Linked Immunosorbent Assay , Hydrogen Peroxide/toxicity , In Vitro Techniques , L-Lactate Dehydrogenase/metabolism , Lung/pathology , Lung Diseases/etiology , Lung Diseases/pathology , Male , Peptidyl-Dipeptidase A/metabolism , Perfusion , Rats , Rats, Sprague-Dawley , Reperfusion Injury/pathology
7.
Pulm Pharmacol Ther ; 18(4): 251-67, 2005.
Article in English | MEDLINE | ID: mdl-15777608

ABSTRACT

We demonstrated previously that monoclonal antibody (mAb) 9B9 to angiotensin-converting enzyme (ACE) accumulates selectively in the rat lung after systemic injection and thus is a powerful tool for immunotargeting therapeutic agents/genes to the lung microvasculature. Bearing in mind the tremendous research and therapeutic potential of lung immunotargeting via ACE, we generated a novel set of mAbs to rat ACE in order to enhance the repertoire of mAbs suitable for targeting drugs/genes to the rat lung. Five new mAbs recognizing different epitopes on rat ACE were examined for their efficacy to bind rat ACE both in vitro and in vivo. Gene delivery into cultured rat lung endothelial cells increased 30-50-fold after coating modified adenoviruses (containing Ig-binding domain) with mAbs to rat ACE. Radiolabeled mAbs specifically accumulated in the lung after systemic injection. mAb 1A2, 4H3 and 2E1 demonstrated the highest efficacy of lung uptake-around 50% of injected dose per gram of tissue; for mAb 1A2, the selectivity of lung uptake (ratio of lung to blood radioactivity) was 205. The effect of the mAbs on ACE shedding was epitope-specific: injection of mAb 1A2 and 4H3 did not change lung ACE activity, whereas injection of mAb 2E1 and 9B9 decreased rat lung ACE activity by 20%. None of the tested mAbs inhibited ACE activity in vitro. A new set of mAbs to rat ACE demonstrated highly efficient and selective lung accumulation and thus have the potential for targeting drugs/genes to the pulmonary vasculature in different rat models of lung diseases.


Subject(s)
Antibodies, Monoclonal/metabolism , Endothelium, Vascular/metabolism , Lung/metabolism , Peptidyl-Dipeptidase A/immunology , Adenoviridae/genetics , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/pharmacokinetics , Antibody Specificity/immunology , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Enzyme-Linked Immunosorbent Assay , Epitopes/immunology , Epitopes/metabolism , Genetic Vectors/genetics , Hybridomas/immunology , Immunohistochemistry , Liver/metabolism , Lung/cytology , Lung/drug effects , Peptidyl-Dipeptidase A/metabolism , Protein Binding/immunology , Rats , Spleen/metabolism , Tissue Distribution , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...