Abstract
A synthetic 7-mer, HHHRHSF, was recently identified by screening a phage display library for binding to the Tie-2 receptor. A polyethylene-oxide clustered version of this peptide, termed vasculotide (VT), was reported to activate Tie-2 and promote angiogenesis in a mouse model of diabetic ulcer. We hypothesized that VT administration would defend endothelial barrier function against sepsis-associated mediators of permeability, prevent lung vascular leakage arising in endotoxemia, and improve mortality in endotoxemic mice. In confluent human microvascular endothelial cells, VT prevented endotoxin-induced (lipopolysaccharides, LPS O111:B4) gap formation, loss of monolayer resistance, and translocation of labeled albumin. In 8-wk-old male C57Bl6/J mice given a ∼70% lethal dose of endotoxin (15 mg/kg ip), VT prevented lung vascular leakage and reversed the attenuation of lung vascular endothelial cadherin induced by endotoxemia. These protective effects of VT were associated with activation of Tie-2 and its downstream mediator, Akt. Echocardiographic studies showed only a nonsignificant trend toward improved myocardial performance associated with VT. Finally, we evaluated survival in this mouse model. Pretreatment with VT improved survival by 41.4% (n = 15/group, P = 0.02) and post-LPS administration of VT improved survival by 33.3% (n = 15/group, P = 0.051). VT-mediated protection from LPS lethality was lost in Tie-2 heterozygous mice, in agreement with VT's proposed receptor specificity. We conclude that this synthetic Tie-2 agonist, completely unrelated to endogenous Tie-2 ligands, is sufficient to activate the receptor and its downstream pathways in vivo and that the Tie-2 receptor may be an important target for therapeutic evaluation in conditions of pathological vascular leakage.
Keywords: Tie-2/TEK, vasculotide, angiopoietins, vascular leakage, endothelial permeability, endotoxin, sepsis, acute lung injury
tie-2 and its agonist ligand, angiopoietin-1 (Angpt-1), were originally identified in the mid-1990s as regulators of developmental angiogenesis. Studies of knockout mice for either gene demonstrated a shared phenotype of arrested vascular development, leading to speculation that the Angpt-Tie-2 signaling axis may have a role in stabilizing nascent, and perhaps even mature, blood vessels (7, 28). Subsequent work in adult mice confirmed the latter hypothesis by demonstrating that excess Angpt-1 prevented vascular permeability induced by VEGF and other inflammatory agents (30, 31).
On the basis of this work, we and others tested what role excess Angpt-1 might play in animal models of acute lung injury, a condition notable for severe, acute pulmonary vascular leakage. Indeed, by viral overexpression (37), direct administration of recombinant protein (18), or administration of transgenic cells (19), Angpt-1 had a potent preventative effect against lung vascular leakage and tissue injury induced by local or systemic exposure to gram-negative endotoxin. Moreover, studies of human subjects with sepsis or acute lung injury have consistently shown depressed Angpt-1 in the circulation and elevated blood levels of the endogenous context-specific Tie-2 antagonist, angiopoietin-2 (Angpt-2) (9, 21, 22, 39). Finally, mice genetically deficient in Angpt-2 were found to be relatively protected from hyperoxic acute lung injury (2). These independent observations support the underlying concept that targeting a specific receptor on a specific cell type (i.e., the vascular endothelium, to which Tie-2's expression is largely limited) is sufficient to protect host organs from parenchymal injury induced by endotoxin and suggest therapeutic potential for agents that can augment Tie-2 activity.
The synthetic peptide HHHRHSF was identified in a random peptide library screened for recombinant Tie-2 binding affinity (33). Since domain-swap and point mutant experiments had already established the importance of clustering Tie-2 monomers for efficient signal transduction through the receptor (12, 23), a cysteine residue was attached to the NH2 terminus of the above 7-mer to covalently tether these monomers to a polyethylene glycol (PEG) backbone (Fig. 1). The resulting formulation, termed vasculotide (VT), was shown to induce Tie-2 phosphorylation in endothelial cell culture in a clustering-dependent fashion and to promote Angpt-1-like angiogenic actions in vivo (34).
Fig. 1.
Chemical structure of vasculotide. Four octapeptides (NH3-CHHHRHSF-COOH) are covalently attached via NH2-terminal maleimide (denoted by X) to a 10-kDa, tetrameric polyethylene oxide.
We hypothesized that VT would promote endothelial barrier function, activate Tie-2 in animals, and ameliorate lung vascular leakage and mortality in murine endotoxemia. We performed studies in human microvascular endothelial cells (HMVECs) and mice to test this hypothesis.
MATERIAL AND METHODS
In Vivo Animal Studies
All experiments were approved by the Beth Israel Deaconess Medical Center Institutional Animal Care and Use Committee. Eight-week-old male C57BL6 mice weighing 20–25 g purchased from Charles River Laboratories International (Wilmington, MA) were acclimated to the facility for at least a week before beginning an experiment. Eight-week-old male Tie-2 heterozygous mice and their wild-type littermates (CD-1 background, generated in Dumont Laboratory) were also studied (14).
Survival analysis.
Mice were injected with 500 ng VT intraperitoneally (ip) 7 h prior to the ip administration of 15 mg/kg body wt lipopolysaccharide (LPS) from Escherichia coli, serotype O111:B4 (Sigma-Aldrich, St. Louis, MO). Survival was followed over the course of 120 h. To evaluate VT's potential as a rescue treatment, a second group of mice was injected first with 15 mg/kg body wt LPS followed by 500 ng VT 2 h later and an additional 250 ng 14 h after LPS administration. To obtain a comparable level of 70–80% mortality 48 h after LPS challenge, the dose of LPS was increased to 17.5 mg/kg body wt for CD-1 mouse experiments.
Adenoviral Angpt-1 overexpression.
At 48 h prior to LPS administration, 8-wk-old male C57BL6 mice were injected with 2 × 1010 particles of recombinant adenovirus encoding human Angpt-1 (Ad-Ang1) or control backbone virus (Ad-control). Expression of Angpt-1 was confirmed by assaying blood obtained by tail nick prior to LPS administration by commercial ELISA (R&D Systems, Minneapolis, MN).
Tissue harvest.
Lung tissue was harvested from VT-pretreated (500 ng) mice 16 h after LPS administration. All animals were euthanized by exsanguination under deep anesthesia followed by rapid collection of lungs into liquid nitrogen for further immunoblot analysis or in periodate-lysine-paraformaldehyde (PLP) fixative for histology assessment.
Evans blue permeability assay.
Sixteen hours after LPS administration, animals were anesthetized with inhaled isoflurane; this procedure was performed as previously described (22). Briefly, 2% wt/vol Evans blue (100 μl) was injected into the retro-orbital sinus. At 10 min after Evans blue injection, mice were euthanized and perfused with 10 ml of PBS 2 mM EDTA for 5 min through a cannula placed in the right ventricle, after which organs were harvested and homogenized in formamide for extraction and measurement of Evans blue as previously described (22). The following formula was used to correct the optical densities for contamination with heme pigments:
Echocardiography.
Sixteen hours after LPS injection, mice were lightly anesthetized with isoflurane in medical air, and in vivo transthoracic echocardiography of the left ventricle using a 40-MHz scanhead interfaced with a Vevo 2100 (Visualsonics, ON, Canada) was used to obtain high-resolution, two-dimensional electrocardiogram-based kilohertz visualization; B-mode and M-mode images were acquired at a rate of 1,000 frames/s. The B-mode and M-mode images were used to calculate left ventricular function parameters. A single operator who was blinded to treatments (E. V. Khankin) performed all echocardiograms. Figure 2 shows an example scout image highlighting landmarks (such as papillary muscle orientation) used to obtain consistent measurements made in M-mode.
Fig. 2.
Echocardiographic landmarks. Scout image of the left ventricle short axis and resulting M-mode image showing the projection obtained for measurement of stroke volume and other parameters. In particular, note the position of the mitral valve papillary muscles at 2 and 4 o'clock used to establish consistent transducer positioning.
Synthesis of VT
Peptide NH2-CHHHRHSF-COOH (Genescript, Piscataway, NJ) was reacted with four-armed, PEG (polydisperse, average molecular mass 10 kDa) (Sunbright PTE-100MA, NOF, Tokyo, Japan) in a 12:1 molar ratio to generate the illustrated product whose molecular weight is 14 kDa (Fig. 1). Covalent attachment of the peptide to the activated maleimide groups took place in PBS, pH 6.4, at room temperature with agitation for 4 h. Products were purified by way of dialysis (Slide-A-Lyzer, 7,000 Da, MWCO, Pierce Biotechnology, Rockford, IL), at 4°C with stirring. Products were first dialyzed against PBS, pH 7.4 (2 exchanges, 300× volume, 3 h each) and then double distilled water (8 exchanges, 300× volume, 6 h each). Dialyzed products were then frozen, lyophilized, and weighed to calculate product yield. Validation of VT was assessed by MALDI-TOF to determine the potential presence of free peptide impurities and to measure the molecular size distribution of the final product (efficiency of conjugation). A control version of VT was similarly prepared by conjugating 10-kDa PEG with cysteine (PEG-Cys).
Antibodies and Reagents
Antibodies used for immunoblotting and/or immunocyto- or histochemistry were purchased from the following manufacturers: Tie-2 (C-20) and actin (C-11) (Santa Cruz Biotechnology, Santa Cruz, CA), phosphotyrosine (4G10, Upstate/Millipore, Temecula, CA), vascular endothelial (VE)-cadherin (BD Bioscience, San Jose, CA). Akt and phospho-Akt (Ser 473) antibodies were purchased from Cell Signaling Technology (Danvers, MA); fluorophore- or horseradish peroxidase (HRP)-conjugated secondary antibodies were purchased from Invitrogen (Carlsbad, CA), as well as phalloidin and 4′,6-diamidino-2-phenylindole (DAPI).
Cell Culture
Passage 5–6 dermal HMVECs (Lonza, Basel, Switzerland) were cultured in EBM-2 media (Lonza) supplemented with 5% fetal bovine serum (FBS) and growth factors according to the manufacturer's instructions at 37°C and 5% CO2. Before experimental treatments, HMVECs were starved for 2 h in EBM-2 containing 1% FBS.
Western Blot Analysis
Cells were washed with ice-cold PBS twice and lysed with ice-cold RIPA buffer [50 mM Tris·HCl (pH 7.4), 150 mM NaCl, 1% Nonidet P-40, 0.5% sodium deoxycholate, 0.1% SDS, and 1 mM EDTA] supplemented with protease inhibitors (Roche Diagnostics, Indianapolis, IN), 1 mM NaVO4, and 1 mM NaF. Lysates were sonicated and centrifuged at 10,000 rpm for 10 min at 4°C, and supernatants were collected. Protein concentrations were determined by Bradford protein assay with bovine serum albumin as a standard (Bio-Rad). A mixture of lysate, NuPAGE reducing agent, and NuPAGE sample buffer were heated at 70°C for 10 min, electrophoresed in NuPAGE 4–12% Novex Bis-Tris Gels (all from Invitrogen), transferred to nitrocellulose membrane, and immunoblotted with specific primary antibodies. Binding of primary antibodies was detected by using HRP-conjugated secondary antibodies (Amersham Pharmacia Biotech) and SuperSignal WestDura (Pierce) reagents as chemiluminescence substrates.
Immunoprecipitation
For measurement of phospho-Tie-2, murine lungs were homogenized in the above lysis buffer, and 1.5 mg of total protein was incubated with anti-Tie-2 antibody for 12 h at 4°C and then precipitated with magnetic Dynabeads conjugated with protein G (Invitrogen) for 45 min at 4°C. After the beads were washed, proteins were eluted by heating in sample buffer and detected by Western blot analysis with anti-phospho-tyrosine (4G10) as described above.
RNA Extraction and Quantitative PCR
Total RNA was extracted by using Trizol (Invitrogen, Carlsbad, CA) followed by RNeasy Mini Kit (Qiagen, Valencia, CA) with on-column DNAse-I treatment per the manufacturers' instructions. Quantification and purity of total RNA was assessed by A260/A280 absorption. Total RNA (2 μg) was then reverse transcribed by using the Omniscript Reverse Transcriptase kit (Qiagen). Quantitative PCR was performed by the Multi-Gene Transcriptional Profiling core (MGTP, http://cvbr.hms.harvard.edu) at Beth Israel Deaconess Medical Center by using the ABI 7000 Sequence Detection System (Applied Biosystems). Results were analyzed by the comparative threshold cycle method. Transcript abundances were normalized per 106 18S rRNA copies.
Immunohistochemistry
Air-dried PLP-fixed cryosections from lungs were blocked for 60 min in 10% donkey serum, incubated with primary antibodies overnight at 4°C, followed by fluorescence-conjugated affinity-purified secondary antibody labeling (60 min, 1:500, Jackson), and mounted with ProLong Gold/DAPI. All images were taken by a Zeiss LSM510 META confocal system at ×63 magnification. Of note, all images were obtained with the same laser power, gain, and offset conditions.
Immunocytochemistry
HMVECs were grown to confluence on glass coverslips coated with collagen type I. After starvation the cells were first treated with VT (100 ng/ml), equimolar PEG-Cys (70 ng/ml), or PBS 90 min prior to challenge with the following permeability mediators: LPS (100 ng/ml) plus LPS-binding protein (100 ng/ml) plus CD14 (10 ng/ml); thrombin (1 U/ml); and human septic serum (5%). A second dose of VT (or PEG-Cys) was given when permeability mediators were added. For the septic serum treatment, EBM-2 media was supplemented with 5% prefiltered human serum from a patient with septic shock collected for a prior published study (26).
Thirty minutes after mediator treatment, cells were fixed for 10 min in 2.5% paraformaldehyde and permeabilized for 5 min in 0.2% Triton X-100 in PBS. Cells were blocked overnight at 4°C with a blocking buffer (1% BSA, Triton, sodium azide), then incubated for 12 h with primary antibody, serial washes in PBS, then 60 min incubation with secondary Alexa-antibody and phalloidin. The coverslips were mounted by using ProLong Gold/DAPI. All images were taken by a Zeiss LSM510 META confocal system at ×63. Of note, all images were obtained with the same laser power, gain, and offset conditions.
TER
HMVECs were grown to confluence in polycarbonate wells containing evaporated gold microelectrodes in series with a large gold counter connected to a phase-sensitive lock-in amplifier as described previously (10, 11, 32). VT (100 ng/ml), equimolar PEG-Cys (70 ng/ml), or PBS were added to wells 90 min prior to LPS and again at the time that LPS was added. Measurements of transendothelial electrical resistance (TER) were performed by using an electrical cell-substrate impedance sensing system (ECIS; Applied BioPhysics, Troy, New York). Briefly, current was applied across the electrodes by a 4,000-Hz AC voltage source with amplitude of 1 V in series with a 1 MΩ resistance to approximate a constant current source (∼1 μA). The in-phase and out-of-phase voltages between the electrodes were monitored in real time with the lock-in amplifier and subsequently converted to scalar measurements of transendothelial impedance, of which resistance was the primary focus.
These measurements provide a highly sensitive biophysical assay that indicates the state of cell shape and focal adhesion. Values from each microelectrode were pooled at discrete time points and either plotted vs. time as means ± SD or reported as bar graphs at the time point of maximal response to a given stimulus as described elsewhere in detail (10).
Transwell Permeability Assay
Confluent HMVEC monolayers were grown on type I collagen-coated Costar Transwell membranes (polyester 0.4-μm filter, Corning, Corning, NY) and permeability was determined by measurement of fluorometric signal in the luminal and abluminal chambers at the indicated time point after luminal addition of 1 mg/ml FITC-labeled human serum albumin (Sigma) as described previously (22). Relative fluorescence units were used in the following equation to determine the permeability coefficient of albumin (Pa):
where [A] is abluminal concentration, [L] is luminal concentration, V is volume of abluminal chamber, t is time in hours, and A is area of membrane in square centimeters.
Statistical Analysis
Results are presented as means ± SE. Statistical significance was evaluated by unpaired two sided t-test unless otherwise stated. Survival data were analyzed by log-rank test and visualized by Kaplan-Meier curves. Analysis was performed in SPSS (SPSS, Chicago, IL) and graphs were made in GraphPad Prism (GraphPad Prism Software, San Diego, CA).
RESULTS
VT Prevents Endothelial Structural Rearrangements Induced by Sepsis Mediators
Given the important roles of cytoskeletal changes and VE-cadherin localization in endothelial barrier defense (5, 6), we studied these parameters on HMVECs treated with LPS in the presence of PBS, PEG-Cys, or VT. Only VT prevented the formation of F-actin stress fibers, enhanced the junctional staining of VE-cadherin, and prevented the formation of interendothelial gaps associated with LPS (Fig. 3). We also found that VT prevented morphological changes in HMVECs induced by thrombin or 5% human septic serum (Supplemental Fig. S1; the online version of this article contains supplemental data).
Fig. 3.
Vasculotide (VT) prevents endothelial cytoskeletal disruption induced by LPS. Immunofluorescence staining for vascular endothelial (VE)-cadherin (green; A, D, G), F-actin (red; B, E, H), and 4′,6-diamidino-2-phenylindole (DAPI; blue; C, F, I) was performed on 100% confluent P5 human microvascular endothelial cells (HMVECs). Cells were treated with VT (100 ng/ml; G–I), equimolar polyethylene glycol with cysteine (PEG-Cys; 70 ng/ml; D–F), or an equal volume of PBS (A–C). After incubation for 30 min with LPS (100 ng/ml), LPS-binding protein (LBP, 100 ng/ml), and CD14 (10 ng/ml), cells were fixed and stained. White arrows point to interendothelial gaps. N = 4 independent experiments per condition.
VT Ameliorates Endotoxin-Induced Endothelial Barrier Dysfunction
Having observed structural evidence of VT-mediated endothelial barrier defense, we next evaluated its performance in functional assays of the same. We first used TER to characterize the dose- and time-dependent reduction of monolayer electrical resistance induced by LPS and to identify an optimal dose of VT (Supplemental Fig. S2). From these data, we determined a time window in which to observe the maximum effect of LPS (Fig. 4A); 100 ng/ml VT prevented LPS-induced endothelial barrier dysfunction whereas an equimolar concentration of PEG-Cys had no effect (Fig. 4B). We then used a Transwell experiment to confirm our TER findings. Whereas TER reports instantaneous resistance at frequent time intervals, the Transwell assay provides a cumulative measure of monolayer leakiness to macromolecules (FITC-labeled albumin in this assay). We again found that VT prevented the barrier dysfunction induced by LPS (LPS + saline vs. LPS + VT, P = 0.001; Fig. 4C).
Fig. 4.
VT ameliorates endotoxin-induced endothelial barrier dysfunction. A: bar graph showing the normalized transendothelial electrical resistance (TER) at 4 h after LPS administered at the indicated doses (n = 3 independent experiments per condition). P values given for the comparison with control group. B: HMVECs were treated with VT (100 ng/ml), equimolar PEG-Cys (70 ng/ml), or an equal volume of PBS and then challenged with LPS (10 ng/ml, with LBP and CD14). Bar graphs indicate the % maximal TER response relative to the PBS-treated control with the indicated P values (n = 4 independent experiments per condition). C: VT (100 ng/ml) pretreated HMVECs were challenged with LPS (25 ng/ml) 2 h after VT. The luminometric reading of FITC-labeled albumin in the abluminal and luminal chambers (means ± SE) was taken 8.5 h after LPS challenge (n = 6 per condition).
VT Reduces Lung Vascular Leakage Following Endotoxin Exposure
After initial dose-ranging studies, 8-wk-old male C57BL6 mice were administered 500 ng VT (or equal volume of sterile saline) intraperitoneally 7 h prior to 15 mg/kg LPS. Mice were euthanized 16 h after LPS administration to perform several measurements. Having already observed strong evidence of endothelial barrier defense against LPS in vitro, we first surveyed vascular leakage in the viscera of these animals by the Evans blue assay (Fig. 5A). Although most organs showed a trend toward increased leakage with LPS, the lungs were affected most prominently, showing more than fourfold increase in dye extravasation (n = 4 control mice, n = 6 LPS-treated mice, P = 0.03). Notably, the lungs were also the most protected by VT (n = 6 LPS + VT treated mice, P = 0.04 vs. LPS alone).
Fig. 5.
Effects of VT on LPS-induced permeability and inflammation in vivo. A: Evans blue in vivo permeability assay was performed 16 h after the administration of LPS with or without VT pretreatment. Shown are results from lung (a), kidney (b), and spleen (c). B: coimmunofluorescence for VE-cadherin (green) and CD31 (red) from lung tissue 16 h after LPS challenge with or without VT pretreatment. In control specimens (left column), VE-cadherin is preserved in the pulmonary vasculature and colocalizes with the endothelial marker CD31 (yellow arrow). After LPS exposure (middle column), VE-cadherin staining is severely reduced as demonstrated in the inset of the merged image (red arrow). With VT pretreatment, VE-cadherin staining is enhanced, resulting in restoration of yellow signal in the merged image. Shown images are representative for n = 5 mice per group. C: measurement of local inflammation. Bar graphs showing means ± SE for quantitative PCR (qPCR) from lung homogenates normalized to 18S copies (n = 6 for LPS and LPS + VT groups; n = 4 control group). ICAM-1, intercellular adhesion molecule-1 (a); VCAM-1, vascular cell adhesion molecule-1 (b); Angpt-2, angiopoietin-2 (c); IL-6, interleukin-6 (d); TNF-α, tumor necrosis factor-α (e); E-selectin (f); P-selectin (g); PAI-1, plasminogen activator inhibitor-1 (h).
On the basis of these findings, we further evaluated the lungs of these mice for structural evidence of barrier dysfunction. As seen in vitro (Fig. 3A), VE-cadherin staining in lung endothelium (confirmed by CD31 costaining) was markedly attenuated by LPS exposure and restored to near-normal by VT pretreatment (Fig. 5B).
Finally, to discern the contribution of local inflammation to the permeability increase induced by LPS, we measured an array of inflammatory markers by quantitative PCR (Fig. 5C). As expected, LPS induced all of the markers in a highly significant fashion. However, VT pretreatment did not diminish the expression of any of these markers. These data suggest that the VT-mediated reversal of LPS-induced pulmonary vascular hyperpermeability proceeds independently of changes in the local inflammatory milieu.
VT Augments Tie-2 Signaling In Vivo
To explore the signaling changes induced by LPS and VT in the lungs, we began by performing coimmunofluorescence for Tie-2 and phosphotyrosine (pY) on lung tissue obtained 16 h after LPS challenge. Illustrative images of n = 5 mice per group are shown in Fig. 6A. Control tissue (left column) demonstrated colocalization of Tie-2 (red) and pY (green) staining, resulting in abundant yellow signal (left column, inset of merged image). LPS exposure (middle column) resulted in considerable attenuation of both Tie-2 and pY. The merged image showed distinct patches of red signal, indicative that the two stains do not colocalize and suggesting that Tie-2 is dephosphorylated. VT pretreatment (left column) enhanced pY staining to near-control intensity and reduced the patches of red signal in the merged image.
Fig. 6.
VT augments Tie-2 signaling in lungs of endotoxemic mice. A: coimmunofluorescence for Tie-2 (red) and phosphotyrosine (pY, green) from lung tissue 16 h after LPS challenge with or without VT pretreatment. Yellow signal (yellow arrow) in the inset of the merged image from the left column indicates colocalization of Tie-2 and pY in control specimens. LPS treatment (middle column) results in attenuation of Tie-2 staining and pY. Moreover, the merged image reveals distinct patches of red (red arrow), indicating a lack of pY colocalization with Tie-2, thus suggesting Tie-2 dephosphorylation. VT pretreatment (right column) does not restore Tie-2 staining to the intensity of control specimens but does augment pY staining above that observed following LPS challenge. Importantly, in the merged image, patches of red are diminished compared with LPS alone. Shown images are representative for n = 5 mice per group. B: ratio of phospho-Tie-2 (pTie-2) to total Tie-2 (tTie-2) in lung. Representative immunoprecipitation (IP) for Tie-2 and consecutive immunoblot (IB) for phosphotyrosine (4G10) from lung lysates. Control mice were injected with sterile saline only. LPS-treated mice were either pretreated with 500 ng VT (LPS + VT) or PBS (LPS only). Organs were harvested 16 h after LPS administration. IB for Tie-2 from lung homogenates from the same mice showing that total Tie-2 is downregulated after LPS but loading (i.e., IB actin) is equal. Densitometry shown for n = 5 independent experiments. C: Tie-2 protein abundance in lung. Representative immunoblot for Tie-2 from murine lung lysates (same conditions as in A) showing LPS-induced decrease in Tie-2 abundance, which is reversed after additional treatment with VT (500 ng). Densitometry shown for n = 3 independent experiments. D: Tie-2 transcript abundance in lung. Bar graph showing mean ± SE for qPCR from lung homogenates (n = 6 for LPS and LPS + VT groups; n = 4 control group) for Tie-2 mRNA normalized to 18S copies (same conditions as in A). E: induction of downstream activation by VT. Representative immunoblot for phospho-Akt (pAkt) and total Akt (tAkt) from lung lysates (same conditions as in A) showing LPS-induced decrease in Akt phosphorylation that is reversed when mice were pretreated with VT (500 ng). Densitometry shown for n = 5 independent experiments.
We next quantified these observations by immunoprecipitation studies on lung lysates obtained from these mice. The phosphorylated fraction of Tie-2 was suppressed by systemic exposure to LPS (P = 0.03 vs. control) and fully restored by VT (P = 0.007 vs. LPS alone; Fig. 6B). LPS reduced Tie-2 protein abundance by 62.6% (P < 0.0001 vs. control; Fig. 6C). This was partially prevented by VT pretreatment (P = 0.002 vs. LPS alone).
To evaluate the LPS-induced suppression of Tie-2 expression further, we performed quantitative PCR on lung homogenates from animals treated with LPS with or without VT (Fig. 6D). Endotoxemia strongly suppressed Tie-2 transcript abundance (P < 0.0001 vs. control), and VT pretreatment did not prevent this drop (P < 0.0001 vs. control and nonsignificant vs. LPS).
To resolve the impact of the changes in Tie-2 activation and protein abundance on downstream signaling, we measured the activation of Akt in lung lysates (Fig. 6E). The total level of Akt was unchanged across all three conditions. However, Akt was deactivated during endotoxemia and restored to a normal level by VT administration (LPS + saline vs. LPS + VT, P = 0.02). Together, these results suggest that the decreased Tie-2 protein abundance induced by LPS is overcome by increased activation of the remaining Tie-2 molecules when VT is given, thereby preserving net flux through this signaling pathway.
VT May Ameliorate Endotoxemic Cardiac Dysfunction
Two prior publications have suggested a cardiotropic role of exogenously administered Angpt-1 in rodents, one in the endotoxemia model (4, 37). We therefore performed serial echocardiography in mice before LPS and 16 h after LPS. Eight mice were pretreated with VT and nine with an equal volume of saline. In the group that only received LPS, and not VT, virtually all measured cardiac parameters were suppressed (Supplemental Fig. S3). Comparing the two post-LPS groups (without VT = 9, with VT = 8), we found that measures of cardiac contraction, namely ejection fraction (EF) and stroke volume (SV), were improved in the mice receiving VT compared with the LPS-only group in a statistically significant fashion (Fig. 7, P = 0.02 and 0.04, respectively).
Fig. 7.
VT may improve endotoxemic cardiac dysfunction. Bar graphs show means ± SE for following parameters: cardiac output (CO; A), ejection fraction (EF; B), stroke volume (SV; C), fractional shortening (FS; D), and heart rate (HR; E). Control values represent pooled data from all animals (n = 17) before LPS challenge. Eight mice were pretreated with VT (LPS VT), and 9 received an equal volume of PBS before LPS (LPS).
As opposed to the catheter-based technique in the prior report that evaluated Angpt-1 in endotoxemia (4, 37), the present method of assessing cardiac performance enabled repeated measurements in a single mouse. We therefore analyzed the data in a different fashion (Table 1). Within each treatment arm, we calculated a “delta” (Δ) for each parameter (e.g., ΔEF = EFpre-LPS − EFpost-LPS). When we compared ΔEF or ΔSV between VT and non-VT treated groups, we found trends that suggested a benefit of VT, but nothing that reached statistical significance. Overall, these data suggest that VT could improve cardiac contractility during endotoxemia, but analysis of the change in performance attributable to VT itself yielded encouraging trends that did not meet statistical significance.
Table 1.
Cardiac parameters in endotoxemia with and without VT pretreatment
LPS (n = 9) |
LPS + VT (n = 9) |
|||||
---|---|---|---|---|---|---|
Pre | Post (16 h) | delta | Pre | Post (16 h) | delta | |
Cardiac output, ml/min | 18.47 ± 3.6 | 6.85 ± 3.1* | −11.64 ± 4.7 | 20.86 ± 2.4 | 9.32 ± 2.8* | −11.53 ± 4.6 |
Diameter (diastolic), mm | 5.02 ± 2.4 | 3.8 ± 0.5* | −0.54 ± 2.6 | 4.12 ± 0.3 | 3.81 ± 0.4* | −0.3 ± 0.6 |
Diameter (systolic), mm | 3.15 ± 0.6 | 3.4 ± 0.4* | 0.71 ± 1.3 | 2.89 ± 0.5 | 3.19 ± 0.4* | 0.31 ± 0.74 |
Ejection fraction, % | 58.12 ± 8.6 | 22.96 ± 9.9* | −36.6 ± 17.8 | 57.5 ± 9.2 | 34.65 ± 10.3* | −22.9 ± 16.1 |
Fractional shortening, % | 27.42 ± 5.4 | 10.51 ± 5.4* | −17.59 ± 8.5 | 30.21 ± 6.2 | 16.45 ± 5.6* | −13.8 ± 9.7 |
Heart rate, beats/min | 448 ± 56 | 456 ± 31 | 11 ± 65 | 498 ± 47 | 437 ± 35 | −60 ± 48† |
Stroke volume, μl | 41.04 ± 5.5 | 14.38 ± 7.0* | −26.9 ± 8.8 | 42.57 ± 4.7 | 21.42 ± 6.5* | −21.1 ± 9.5 |
Volume (diastolic), μl | 71.79 ± 12.3 | 63.13 ± 18.5* | −6.68 ± 23.7 | 75.63 ± 14.2 | 63.4 ± 14.5* | −12.2 ± 24.3 |
Volume (systolic), μl | 30.76 ± 10.6 | 48.74 ± 15.5* | 20.18 ± 22.1 | 33.06 ± 12.6 | 41.98 ± 14.3* | 8.9 ± 22.3 |
Values are means ± SD. VT, vasculotide.
P < 0.01 vs. pre-LPS value.
P < 0.05 between deltas.
VT Improves Survival of Endotoxemic Mice
Lastly, we asked whether the specific in vivo benefits of VT could be associated with increased survival. Of mice pretreated with VT followed by 15 mg/kg LPS (n = 15), 71.4% survived indefinitely whereas only 30.0% of PBS pretreated mice lived (n = 30; P = 0.02; Fig. 8A). To confirm the specificity of VT's action in vivo, we repeated this experiment in Tie-2 heterozygous mice and found that VT-mediated protection from LPS lethality was abolished (Fig. 8B). In a second set of experiments, mice were first treated with 15 mg/kg LPS, after which they received two injections of VT (500 ng 2 h post-LPS followed by 250 ng 14 h post-LPS) or equivalent volume of sterile PBS (Fig. 8C). Of the VT-treated mice 85.0% lived whereas only 55.0% of sham-treated mice survived (n = 20 per group, P = 0.051). Finally, we used high-titer recombinant adenoviruses to confirm the results of Witzenbichler et al. (37), namely that excess human Angpt-1 could also prevent LPS-induced lethality (Supplemental Fig. S4).
Fig. 8.
VT improves survival following endotoxin exposure. A: 8-wk-old male C57BL6 mice were pretreated with 500 ng VT (n = 15) or PBS (control, n = 30) 7 h prior to injection of a ∼70% lethal LPS dose (i.e., 15 mg/kg body wt). VT improved survival by 41.4% (P = 0.02). B: 8-wk-old male Tie-2 heterozygous (Tie-2 +/−) mice were pretreated with 500 ng VT or PBS 7 h prior to injection of LPS (17.5 mg/kg body wt). Survival was indistinguishable (P = 0.66). C: 8-wk-old male C57BL6 mice were first given LPS (15 mg/kg) and 2 h later were treated with a first dose of VT (VT 500 ng, n = 20) or control (PBS, n = 20), followed by a second dose (VT 250 ng, or PBS) 12 h after the first VT dose. P = 0.051.
DISCUSSION
We found that VT counteracts the microvascular endothelial barrier dysfunction induced by LPS and other soluble sepsis mediators. In vivo, VT activated Tie-2 and its major downstream pathway in the lung, prevented endotoxemia-induced lung vascular leakage, and significantly attenuated mortality induced by endotoxemia. These effects were independent of changes in Angpt-2 expression and tissue inflammation. By the most stringent analysis, the protective actions of VT also appeared not to be mediated by changes in cardiac performance. Given that the 7-mer HHHRHSF was selected in a random peptide screen for Tie-2 affinity, the present data suggest that Tie-2 activation in the setting of strong endotoxin exposure is sufficient to confer substantial protection to the host.
These results add to the existing literature by further clarifying the role of Tie-2, and by extension the endothelium, in models of acute inflammatory lung injury. As noted in the introduction, prior studies have consistently shown a beneficial effect of Tie-2 stimulation in such models but have also relied exclusively on some form of Angpt-1 to stimulate the receptor. This approach has left unanswered the question of whether Angpt-1 has beneficial off-target effects. Since the present method of activating Tie-2 shares no homology with Angpt-1 (or any other rodent protein), and yet both approaches reduce vascular leakage and improve mortality, it is likely that Tie-2 activation per se is the critical effect of exogenous Angpt-1 in these models. Two important controls further strengthen this conclusion. First, VT failed to protect Tie-2 heterozygous mice from endotoxemic death (Fig. 8B), arguing that a reduction in Tie-2 expression (14) abrogates the benefit of VT in vivo. Second, the barrier-protective effect of VT did not appear to be dependent on the PEG backbone (Figs. 3 and 4). Prior publications showing a barrier-protective or prosurvival effect of high-molecular-weight PEG in models of sepsis have used minimum doses 50,000- to 75,000-fold higher than the highest doses in this report (3, 38). The present study may refocus therapeutic considerations of the Angpt-1/Tie-2 signaling axis in sepsis-related disorders from ligand to receptor.
Just as the present work is informative about the effects of Tie-2 activation in endotoxemia, it may also suggest which in vivo actions of Angpt-1 are dependent on Tie-2 and which are independent. Although Angpt-1 and VT share the ability to reduce pulmonary vascular leakage, they do not appear to share a cardioprotective function in endotoxemia (37). The differences could be technical in nature [e.g., cardiodepressant effect of control adenovirus used in the Witzenbichler et al. (37), study or occult cardiotoxic effect of VT] or due to differences in the degree or duration of Tie-2 activation via two disparate ligands. Notably, the prior study examining Angpt-1 action in endotoxemia was a terminal investigation using a catheter-based technique; thus no single mouse was studied both before and after LPS exposure. When we analyzed our data in the manner of Witzenbichler et al., we too found an improvement of cardiac function attributable to Tie-2 stimulation. However, when we factored in the pre- and post-LPS measurements, there was only a nonsignificant trend attributable to VT. A final possibility to account for the difference in cardiac improvement between Angpt-1 and VT is that intact Angpt-1 has an independent trophic effect on cardiomyocytes through its integrin-binding sequence QHREDGS that VT lacks (4). Thus the cardioprotection observed by Witzenbichler et al. may be a direct result of integrin ligation in the cardiac muscle rather than some Tie-2-dependent effect.
VT also failed to modulate inflammation whereas Angpt-1 has been reported to do so in experimental sepsis (18, 37) and in other models of in vivo inflammation (13, 16, 19, 27). Although additional measurements of inflammation, e.g., assaying bronchoalveolar fluid for macrophage count or cytokine concentration, may have shown an anti-inflammatory effect of VT, another possibility may relate to the recent observation that Angpt-1 binds monocytes independent of Tie-2 (1). However, Tie-2-expressing monocytes only constitute ∼5% of circulating monocytes (35) and may be more important in chronic disease processes such as tumor angiogenesis rather than acute processes such as cytokine storm (24). Moreover, reduction of inflammation may not be a desirable effect of any potential sepsis therapeutic as the human experience with immunosuppressives and anti-inflammatories has been mixed at best (25).
Despite these potential “shortcomings” of VT vs. recombinant Angpt-1, the survival benefit of the former in endotoxemia actually exceeded what Witzenbichler et al. (37), reported for the latter and was comparable to our survival experiment with overexpressed Angpt-1 (Supplemental Fig. S4). Since the only “positive” effects we were able to detect in vivo were Tie-2 activation, Akt activation, and reduction in pulmonary vascular permeability, it is possible that the main physiological consequence of Tie-2 stimulation in endotoxemia is reversal of lung endothelial barrier dysfunction, which in turn results in increased survival. If true, so-called “indirect” causes of acute lung injury, diseases such as polytrauma or pancreatitis in which the initial injury is outside the lung, but the ensuing cytokine storm promotes distant organ dysfunction affecting the lungs (36), may be particularly amenable to a Tie-2-based approach such as VT. Furthermore, if the in vivo barrier-protective effect of Tie-2 also involves the alveolar epithelium, as one recent report suggests (8), there may be a “two-compartment” barrier-defense benefit of Tie-2 stimulation in disorders such as acute lung injury or acute respiratory distress syndrome.
Does VT or some related formulation hold promise as a therapeutic in sepsis or other conditions of pathological vascular leakage such as the acute respiratory distress syndrome? Approaches that inhibit the circulating Angpt-2 excess that we and others have reported in human sepsis (15, 21, 22), e.g., by recently developed neutralizing antibodies (20, 29), may be less potent than approaches that activate Tie-2. Inhibition of an inhibitor could reasonably be expected to restore Tie-2 phosphorylation to presepsis levels whereas exogenous receptor activation offers greater titration control, even enabling one to exceed quiescent levels of Tie-2 activation. If Tie-2 protein concentration falls in human sepsis as we observed in this model, the need to maximize signal through the remaining pool of receptor molecules could become even more acute. In preliminary experiments, we have observed that a single dose of VT can sustain lung Tie-2 phosphorylation in normal mice for 72–96 h. Experiments in which the expression and activation of Tie-2 can be finely controlled will be needed to clarify this question. Finally, a VT-like drug may be preferable to a recombinant form of human Angpt-1 because the former could be more cheaply produced, be less subject to contamination during production since no bioreactor is required, and would not confer the risk to recipients of developing neutralizing antibodies against an endogenous protein, a mechanism, for example, that has led to rare cases of pure red cell aplasia in recipients of recombinant erythropoietin (17).
In summary, the present work demonstrates the beneficial in vitro and in vivo effects of a synthetic, druglike Tie-2 agonist unrelated to Angpt-1 in experimental sepsis. Future studies should further clarify the therapeutic potential of exogenous Tie-2 stimulation in sepsis, acute lung injury, and other conditions of acute vascular leakage.
GRANTS
S. David is a scholar of the German Research Foundation (DA 1209/1-1). D. Dumont is a Canada Research Chair in Angiogenesis and Lymphangiogenesis Signaling. This study was funded in part by the Heart and Stroke Foundation of Canada (Grant no. NA 7027, to P. Van Slyke and D. Dumont). This work was supported by grants to S. M. Parikh (R01HL093234, R01HL093234-01S1, and K08DK06916).
DISCLOSURES
D. Dumont and P. Van Slyke are listed as inventors on patents related to VT filed by the University of Toronto. S. M. Parikh and S. A. Karamanchi are listed as inventors on disclosures regarding angiopoietins filed by Beth Israel Deaconess Medical Center.
Supplementary Material
REFERENCES
- 1. Ahmad S, Cudmore MJ, Wang K, Hewett P, Potluri R, Fujisawa T, Ahmed A. Angiopoietin-1 induces migration of monocytes in a tie-2 and integrin-independent manner. Hypertension 56: 477–483, 2010 [DOI] [PubMed] [Google Scholar]
- 2. Bhandari V, Choo-Wing R, Lee CG, Zhu Z, Nedrelow JH, Chupp GL, Zhang X, Matthay MA, Ware LB, Homer RJ, Lee PJ, Geick A, de Fougerolles AR, Elias JA. Hyperoxia causes angiopoietin 2-mediated acute lung injury and necrotic cell death. Nat Med 12: 1286–1293, 2006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Chiang ET, Camp SM, Dudek SM, Brown ME, Usatyuk PV, Zaborina O, Alverdy JC, Garcia JG. Protective effects of high-molecular weight polyethylene glycol (PEG) in human lung endothelial cell barrier regulation: role of actin cytoskeletal rearrangement. Microvasc Res 77: 174–186, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Dallabrida SM, Ismail N, Oberle JR, Himes BE, Rupnick MA. Angiopoietin-1 promotes cardiac and skeletal myocyte survival through integrins. Circ Res 96: e8–e24, 2005 [DOI] [PubMed] [Google Scholar]
- 5. Dejana E, Tournier-Lasserve E, Weinstein BM. The control of vascular integrity by endothelial cell junctions: molecular basis and pathological implications. Dev Cell 16: 209–221, 2009 [DOI] [PubMed] [Google Scholar]
- 6. Dudek SM, Garcia JG. Cytoskeletal regulation of pulmonary vascular permeability. J Appl Physiol 91: 1487–1500, 2001 [DOI] [PubMed] [Google Scholar]
- 7. Dumont DJ, Gradwohl G, Fong GH, Puri MC, Gertsenstein M, Auerbach A, Breitman ML. Dominant-negative and targeted null mutations in the endothelial receptor tyrosine kinase, tek, reveal a critical role in vasculogenesis of the embryo. Genes Dev 8: 1897–1909, 1994 [DOI] [PubMed] [Google Scholar]
- 8. Fang X, Neyrinck AP, Matthay MA, Lee JW. Allogeneic human mesenchymal stem cells restore epithelial protein permeability in cultured human alveolar type II cells by secretion of angiopoietin-1. J Biol Chem 285: 26211–26222, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Gallagher DC, Parikh SM, Balonov K, Miller A, Gautam S, Talmor D, Sukhatme VP. Circulating angiopoietin 2 correlates with mortality in a surgical population with acute lung injury/adult respiratory distress syndrome. Shock 29: 656–661, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Garcia JG, Liu F, Verin AD, Birukova A, Dechert MA, Gerthoffer WT, Bamberg JR, English D. Sphingosine 1-phosphate promotes endothelial cell barrier integrity by Edg-dependent cytoskeletal rearrangement. J Clin Invest 108: 689–701, 2001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Giaever I, Keese CR. A morphological biosensor for mammalian cells. Nature 366: 591–592, 1993 [DOI] [PubMed] [Google Scholar]
- 12. Kim KT, Choi HH, Steinmetz MO, Maco B, Kammerer RA, Ahn SY, Kim HZ, Lee GM, Koh GY. Oligomerization and multimerization are critical for angiopoietin-1 to bind and phosphorylate Tie2. J Biol Chem 280: 20126–20131, 2005 [DOI] [PubMed] [Google Scholar]
- 13. Kim SR, Lee KS, Park SJ, Min KH, Lee KY, Choe YH, Hong SH, Koh GY, Lee YC. Angiopoietin-1 variant, COMP-Ang1 attenuates hydrogen peroxide-induced acute lung injury. Exp Mol Med 40: 320–331, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Kugathasan L, Ray JB, Deng Y, Rezaei E, Dumont DJ, Stewart DJ. The angiopoietin-1-Tie2 pathway prevents rather than promotes pulmonary arterial hypertension in transgenic mice. J Exp Med 206: 2221–2234, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Kumpers P, Lukasz A, David S, Horn R, Hafer C, Faulhaber-Walter R, Fliser D, Haller H, Kielstein JT. Excess circulating angiopoietin-2 is a strong predictor of mortality in critically ill medical patients. Crit Care 12: R147, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Lee S, Kim W, Moon SO, Sung MJ, Kim DH, Kang KP, Jang KY, Lee SY, Park BH, Koh GY, Park SK. Renoprotective effect of COMP-angiopoietin-1 in db/db mice with type 2 diabetes. Nephrol Dial Transplant 22: 396–408, 2007 [DOI] [PubMed] [Google Scholar]
- 17. Macdougall IC. Antibody-mediated pure red cell aplasia (PRCA): epidemiology, immunogenicity and risks. Nephrol Dial Transplant 20, Suppl 4: iv9–iv15, 2005 [DOI] [PubMed] [Google Scholar]
- 18. Mammoto T, Parikh SM, Mammoto A, Gallagher D, Chan B, Mostoslavsky G, Ingber DE, Sukhatme VP. Angiopoietin-1 requires p190 RhoGAP to protect against vascular leakage in vivo. J Biol Chem 282: 23910–23918, 2007 [DOI] [PubMed] [Google Scholar]
- 19. Mei SH, McCarter SD, Deng Y, Parker CH, Liles WC, Stewart DJ. Prevention of LPS-induced acute lung injury in mice by mesenchymal stem cells overexpressing angiopoietin 1. PLoS Med 4: e269, 2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Oliner J, Min H, Leal J, Yu D, Rao S, You E, Tang X, Kim H, Meyer S, Han SJ, Hawkins N, Rosenfeld R, Davy E, Graham K, Jacobsen F, Stevenson S, Ho J, Chen Q, Hartmann T, Michaels M, Kelley M, Li L, Sitney K, Martin F, Sun JR, Zhang N, Lu J, Estrada J, Kumar R, Coxon A, Kaufman S, Pretorius J, Scully S, Cattley R, Payton M, Coats S, Nguyen L, Desilva B, Ndifor A, Hayward I, Radinsky R, Boone T, Kendall R. Suppression of angiogenesis and tumor growth by selective inhibition of angiopoietin-2. Cancer Cell 6: 507–516, 2004 [DOI] [PubMed] [Google Scholar]
- 21. Orfanos SE, Kotanidou A, Glynos C, Athanasiou C, Tsigkos S, Dimopoulou I, Sotiropoulou C, Zakynthinos S, Armaganidis A, Papapetropoulos A, Roussos C. Angiopoietin-2 is increased in severe sepsis: correlation with inflammatory mediators. Crit Care Med 35: 199–206, 2007 [DOI] [PubMed] [Google Scholar]
- 22. Parikh SM, Mammoto T, Schultz A, Yuan HT, Christiani D, Karumanchi SA, Sukhatme VP. Excess circulating angiopoietin-2 may contribute to pulmonary vascular leak in sepsis in humans. PLoS Med 3: e46, 2006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Procopio WN, Pelavin PI, Lee WM, Yeilding NM. Angiopoietin-1 and -2 coiled coil domains mediate distinct homo-oligomerization patterns, but fibrinogen-like domains mediate ligand activity. J Biol Chem 274: 30196–30201, 1999 [DOI] [PubMed] [Google Scholar]
- 24. Pucci F, Venneri MA, Biziato D, Nonis A, Moi D, Sica A, Di Serio C, Naldini L, De Palma M. A distinguishing gene signature shared by tumor-infiltrating Tie2-expressing monocytes, blood “resident” monocytes, and embryonic macrophages suggests common functions and developmental relationships. Blood 114: 901–914, 2009 [DOI] [PubMed] [Google Scholar]
- 25. Russell JA. Management of sepsis. N Engl J Med 355: 1699–1713, 2006 [DOI] [PubMed] [Google Scholar]
- 26. Shapiro NI, Schuetz P, Yano K, Sorasaki M, Parikh SM, Jones AE, Trzeciak S, Ngo L, Aird WC. The association of endothelial cell signaling, severity of illness, and organ dysfunction in sepsis. Crit Care 14: R182, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Simoes DC, Vassilakopoulos T, Toumpanakis D, Petrochilou K, Roussos C, Papapetropoulos A. Angiopoietin-1 protects against airway inflammation and hyperreactivity in asthma. Am J Respir Crit Care Med 177: 1314–1321, 2008 [DOI] [PubMed] [Google Scholar]
- 28. Suri C, Jones PF, Patan S, Bartunkova S, Maisonpierre PC, Davis S, Sato TN, Yancopoulos GD. Requisite role of angiopoietin-1, a ligand for the TIE2 receptor, during embryonic angiogenesis. Cell 87: 1171–1180, 1996 [DOI] [PubMed] [Google Scholar]
- 29. Tabruyn SP, Colton K, Morisada T, Fuxe J, Wiegand SJ, Thurston G, Coyle AJ, Connor J, McDonald DM. Angiopoietin-2-driven vascular remodeling in airway inflammation. Am J Pathol 177: 3233–3243, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Thurston G, Rudge JS, Ioffe E, Zhou H, Ross L, Croll SD, Glazer N, Holash J, McDonald DM, Yancopoulos GD. Angiopoietin-1 protects the adult vasculature against plasma leakage. Nat Med 6: 460–463, 2000 [DOI] [PubMed] [Google Scholar]
- 31. Thurston G, Suri C, Smith K, McClain J, Sato TN, Yancopoulos GD, McDonald DM. Leakage-resistant blood vessels in mice transgenically overexpressing angiopoietin-1. Science 286: 2511–2514, 1999 [DOI] [PubMed] [Google Scholar]
- 32. Tiruppathi C, Malik AB, Del Vecchio PJ, Keese CR, Giaever I. Electrical method for detection of endothelial cell shape change in real time: assessment of endothelial barrier function. Proc Natl Acad Sci USA 89: 7919–7923, 1992 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Tournaire R, Simon MP, le Noble F, Eichmann A, England P, Pouyssegur J. A short synthetic peptide inhibits signal transduction, migration and angiogenesis mediated by Tie2 receptor. EMBO Rep 5: 262–267, 2004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Van Slyke P, Alami J, Martin D, Kuliszewski M, Leong-Poi H, Sefton MV, Dumont D. Acceleration of diabetic wound healing by an angiopoietin peptide mimetic. Tissue Eng Part A 15: 1269–1280, 2009 [DOI] [PubMed] [Google Scholar]
- 35. Venneri MA, De Palma M, Ponzoni M, Pucci F, Scielzo C, Zonari E, Mazzieri R, Doglioni C, Naldini L. Identification of proangiogenic TIE2-expressing monocytes (TEMs) in human peripheral blood and cancer. Blood 109: 5276–5285, 2007 [DOI] [PubMed] [Google Scholar]
- 36. Ware LB, Matthay MA. The acute respiratory distress syndrome. N Engl J Med 342: 1334–1349, 2000 [DOI] [PubMed] [Google Scholar]
- 37. Witzenbichler B, Westermann D, Knueppel S, Schultheiss HP, Tschope C. Protective role of angiopoietin-1 in endotoxic shock. Circulation 111: 97–105, 2005 [DOI] [PubMed] [Google Scholar]
- 38. Wu L, Zaborina O, Zaborin A, Chang EB, Musch M, Holbrook C, Shapiro J, Turner JR, Wu G, Lee KY, Alverdy JC. High-molecular-weight polyethylene glycol prevents lethal sepsis due to intestinal Pseudomonas aeruginosa. Gastroenterology 126: 488–498, 2004 [DOI] [PubMed] [Google Scholar]
- 39. Yuan HT, Khankin EV, Karumanchi SA, Parikh SM. Angiopoietin 2 is a partial agonist/antagonist of Tie2 signaling in the endothelium. Mol Cell Biol 29: 2011–2022, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.