The receptor tyrosine kinase Tie2 is highly expressed in endothelial cells and is crucial for angiogenesis and vascular maintenance. The ligands for Tie2 are the angiopoietins, of which angiopoietin-1 and angiopoietin-2 have been the most studied. Angiopoietin-1 has been characterized as the primary activating ligand for Tie2 whereas the role of angiopoietin-2 remains controversial; activating Tie2 in some studies and inhibiting Tie2 in others. Our studies were aimed at understanding the regulation of Tie2 in endothelial cells by angiopoietin-1 and angiopoietin-2 and revealed that both ligands activated Tie2 in a concentration-dependent manner. Angiopoietin-2 was considerably weaker at activating Tie2 compared with angiopoietin-1 suggesting that angiopoietin-2 may be a partial agonist. Activation of Tie2 by these ligands resulted in differential turnover of the receptor where binding of angiopoietin-1, and to a lesser extent angiopoietin-2, induced rapid internalization and degradation of Tie2. Furthermore, our binding studies demonstrate that both ligands are differentially released from the endothelial cell surface after receptor activation and accumulate in the surrounding medium. Altogether, these data begin our understanding of the regulation of Tie2 and the activity of the angiopoietins after engaging the endothelial cell surface.

The receptor tyrosine kinase Tie2 is highly expressed in endothelial cells and is crucial for angiogenesis and for vascular maintenance in the embryo and in the adult (Dumont et al., 1992; Dumont et al., 1993; Dumont et al., 1994; Sato et al., 1995; Wong et al., 1997; Jones et al., 2001). The ligands for Tie2 are the angiopoietins, which are comprised of three members: angiopoietin-1 (Ang1) (Davis et al., 1996), Ang2 (Maisonpierre et al., 1997), and the mouse/human interspecies orthologs Ang3/Ang4 respectively (Valenzuela et al., 1999). Ang1 has been identified as the primary activating ligand for Tie2 and it is believed that most of the angiogenic functions of Tie2 can be attributed to the actions of Ang1 (Suri et al., 1996). Binding of Ang1 to the extracellular domain of Tie2 in endothelial cells results in receptor auto-phosphorylation and the activation of several intracellular signaling pathways leading to endothelial cell migration (Witzenbichler et al., 1998; Jones et al., 1999), tube formation (Hayes et al., 1999; Teichert-Kuliszewska et al., 2001), sprouting (Koblizek et al., 1998) and survival (Kwak et al., 1999; Jones et al., 1999). The role of Ang2 has been controversial. Ang2 possesses similar receptor affinity to Ang1 but was initially characterized as a Tie2 antagonist (Maisonpierre et al., 1997). In some studies, the binding of Ang2 did not activate Tie2 and competitively inhibited the binding of Ang1, establishing an agonist-antagonist ligand pair regulating vascular development (Maisonpierre et al., 1997). In other studies, the binding of Ang2 to Tie2 did result in receptor activation (Kim et al., 2000b; Teichert-Kuliszewska et al., 2001) with similar biological outcomes to Ang1, namely, endothelial cell migration (Mochizuki et al., 2002), tube formation (Teichert-Kuliszewska et al., 2001; Mochizuki et al., 2002) and survival (Kim et al., 2000b), thus it was thought that Ang2 may act as a context-dependent antagonist.

Tyrosine kinase receptors are regulated by their cognate ligands (Sorkin and Waters, 1993; Gaborik and Hunyady, 2004). For example, binding of the epidermal growth factor (EGF) to the epidermal growth factor receptor (EGFR) induces rapid internalization of the receptor-ligand complex into the cell followed by their delivery to lysosomes for degradation (Carpenter and Cohen, 1976; Beguinot et al., 1984). Rapid internalization and degradation of activated receptors following ligand binding, a phenomenon referred to as receptor downregulation, has been observed for numerous tyrosine kinase receptors, some examples include the platelet-derived growth factor receptor and the insulin receptor (Kasuga et al., 1981; Keating and Williams, 1987). Receptor downregulation is considered to be a primary mechanism for the attenuation of downstream signal transduction and results in an overall loss of receptors from the cell surface thereby diminishing the ability of the cell to respond to further stimulation (Sorkin and Waters, 1993). It has also been suggested that internalization of activated receptors may be necessary to facilitate downstream signaling and that a proper signaling response may require receptor endocytosis (Sorkin and von Zastrow, 2002; Wiley, 2003).

In our study, we demonstrate that the binding of Ang1 and Ang2 to Tie2 impart different regulatory effects on Tie2 which may ultimately have an impact on the biological output of these two related ligands. These observed differences may provide an important link in further understanding the complex nature of the angiogenic and lymphangiogenic ligands - the angiopoietins.

Ang1 and Ang2 activate Tie2 in a concentration-dependent manner

The regulation of many tyrosine kinase receptors is dependent on ligand concentration (Kasuga et al., 1981) and consequently, on receptor activation where higher levels of receptor activation stimulate faster rates of receptor internalization and degradation. To understand the relationship between the concentration of Ang1 and Ang2, and Tie2 activation, we examined Tie2 tyrosine phosphorylation in response to increasing concentrations of each ligand (Fig. 1). Pre-incubating recombinant Ang1 where a polyhistidine tag has been added at the C-terminus together with an anti-polyhistidine monoclonal antibody enhanced Tie2 tyrosine phosphorylation compared with Ang1 alone presumably by cross-linking Ang1 multimers (Fig. 1A,C). Under these conditions, Ang1 activated Tie2 in a concentration-dependent manner with maximal levels of Tie2 activation observed when the cells were stimulated with 800 ng/ml Ang1 (Fig. 1A). The anti-polyhistidine antibody alone did not induce Tie2 activation (Fig. 1C) and was included in Ang1 stimulations to achieve higher levels of Tie2 activation. Ang2 also activated Tie2 in a concentration-dependent manner (Fig. 1B). Similar to Ang1, maximal activation of Tie2 was observed when the cells were stimulated with 800 ng/ml Ang2 (Fig. 1B). In contrast to Ang1, pre-incubating Ang2 with the anti-polyhistidine antibody did not enhance Tie2 phosphorylation (data not shown). To study the regulation of Tie2 by Ang1 and Ang2, 800 ng/ml of each ligand was used in subsequent experiments because these concentrations produced maximal activation of Tie2.

When tyrosine phosphorylation of Tie2 induced by Ang1 and Ang2 was compared, Tie2 phosphorylation induced by Ang1 was considerably higher compared with Ang2 (Fig. 1C). We examined the phosphorylation of tyrosine residues 1102 and 1108 (mouse 1100 and 1106) on Tie2 in response to Ang1 and Ang2. Phosphorylation on tyrosine residues 1102 and 1108 is important for the initiation of downstream signalling pathways (Jones et al., 1999; Jones et al., 2003). Both Ang1 and Ang2 stimulated phosphorylation of Tyr1102 and 1108 on Tie2 although phosphorylation induced by Ang2 was weaker compared with Ang1 (Fig. 1D).

Fig. 1.

Ang1 and Ang2 activate Tie2 in a concentration-dependent manner. (A,B) HUVECs were stimulated with the indicated concentrations of Ang1 pre-clustered with 10 μg/ml anti-polyhistidine antibody (A) or Ang2 (B) for 15 minutes at 37°C. Top panels are representative immunoblots. Intensities of Tie2 tyrosine phosphorylation were quantified by densitometry and plotted versus Ang1 or Ang2 concentration (lower panels). The value at 800 ng/ml was designated 100%. (C) Comparison of Tie2 tyrosine phosphorylation when HUVECs are stimulated with Ang1 (800 ng/ml), Ang1 (800 ng/ml) pre-clustered with the anti-polyhistidine (anti-his) antibody (10 μg/ml), Ang2 (800 ng/ml), or the anti-polyhistidine antibody (10 μg/ml) alone. (D) EA.hy926 cells were stimulated with Ang1 (800 ng/ml, pre-clustered) or Ang2 (800 ng/ml) for 15 minutes at 37°C. Tie2 was immunoprecipitated and immunoblotted with an anti-phospho Tyr1102 and Tyr1108 antibody (top panel) or an anti-Tie2 antibody (lower panel).

Fig. 1.

Ang1 and Ang2 activate Tie2 in a concentration-dependent manner. (A,B) HUVECs were stimulated with the indicated concentrations of Ang1 pre-clustered with 10 μg/ml anti-polyhistidine antibody (A) or Ang2 (B) for 15 minutes at 37°C. Top panels are representative immunoblots. Intensities of Tie2 tyrosine phosphorylation were quantified by densitometry and plotted versus Ang1 or Ang2 concentration (lower panels). The value at 800 ng/ml was designated 100%. (C) Comparison of Tie2 tyrosine phosphorylation when HUVECs are stimulated with Ang1 (800 ng/ml), Ang1 (800 ng/ml) pre-clustered with the anti-polyhistidine (anti-his) antibody (10 μg/ml), Ang2 (800 ng/ml), or the anti-polyhistidine antibody (10 μg/ml) alone. (D) EA.hy926 cells were stimulated with Ang1 (800 ng/ml, pre-clustered) or Ang2 (800 ng/ml) for 15 minutes at 37°C. Tie2 was immunoprecipitated and immunoblotted with an anti-phospho Tyr1102 and Tyr1108 antibody (top panel) or an anti-Tie2 antibody (lower panel).

Tie2 turnover in HUVECs

To examine the effect of Ang1 and Ang2 on the turnover of Tie2 in HUVECs, we determined the half-life of Tie2 by pulse-chase in unstimulated cells, and in cells stimulated with either Ang1 or Ang2. HUVECs were first incubated in media containing [35S]methionine and [35S]cysteine to label newly synthesized Tie2. The cells were then washed and incubated in normal growth medium (with or without ligands) for various times. Upon transfer of the cells to normal growth medium the initial radioactively labelled Tie2 population disappeared over time. This approach allowed us to measure the rate of Tie2 degradation and how this may be modulated by angiopoietin stimulation. Under these conditions, Tie2 degraded exponentially over time (Fig. 2A). Semi-logarithmic plots of Tie2 levels versus time were generated and the slopes of the lines were used to calculate the time required for half of the initial receptor mass to decrease by 50%. The half-life of Tie2 in unstimulated cells was ∼9 hours (range 7.5 to 12 hours), in the presence of Ang1 the half-life was determined to be ∼3 hours (range 1.8 to 4.0 hours), and in the presence of Ang2 the half-life was ∼7 hours (range 4.9 to 8.2 hours) (Fig. 2B). A one-way analysis of variance (ANOVA) followed by the Tukey's HSD Test revealed that the half-life values of Tie2 differ significantly between unstimulated cells and in cells treated with either Ang1 and Ang2 (P<0.05). The rate of Tie2 synthesis was unchanged in the presence of either ligand compared with unstimulated cells (Fig. 3).

Fig. 2.

Turnover of Tie2 in HUVECs. [35S]methionine and [35S]cysteine labelled HUVECs were washed and incubated at 37°C for the indicated times in either HUVEC medium alone, or containing pre-clustered Ang1 (800 ng/ml), or Ang2 (800 ng/ml). The cells were lysed, Tie2 immunoprecipitated with an anti-Tie2 antibody, and immunoprecipitates resolved by SDS-PAGE. (A) Representative autoradiograph. (B) Semi-logarithmic plot of the relative Tie2 intensities versus time in unstimulated cells (•), in cells stimulated with Ang1 (⋄) or Ang2 (▪). Each point on the graph is an average value from three to nine independent experiments.

Fig. 2.

Turnover of Tie2 in HUVECs. [35S]methionine and [35S]cysteine labelled HUVECs were washed and incubated at 37°C for the indicated times in either HUVEC medium alone, or containing pre-clustered Ang1 (800 ng/ml), or Ang2 (800 ng/ml). The cells were lysed, Tie2 immunoprecipitated with an anti-Tie2 antibody, and immunoprecipitates resolved by SDS-PAGE. (A) Representative autoradiograph. (B) Semi-logarithmic plot of the relative Tie2 intensities versus time in unstimulated cells (•), in cells stimulated with Ang1 (⋄) or Ang2 (▪). Each point on the graph is an average value from three to nine independent experiments.

The increased rate of degradation of Tie2 in the presence of Ang1 and Ang2 could also be observed in cell lysates by western blot analysis (Fig. 4) further suggesting that the rate of Tie2 synthesis was unable to compensate for the ligand-induced degradation of the receptor and that activation of the Tie2 pathway does not up-regulate its own synthesis.

Tie2 internalization

Upon ligand binding, many tyrosine kinase receptors are rapidly internalized into intracellular compartments (Sorkin and Waters, 1993; Gaborik and Hunyady, 2004). The internalization of Tie2 upon binding Ang1 or Ang2 was indirectly assessed by tagging Tie2 at the cell surface with biotin. Biotinylated Tie2 was isolated with streptavidin conjugated to agarose beads and detected by immunoblotting (Fig. 5). To ensure that only Tie2 at the plasma membrane was being isolated, biotin was stripped from the cell surface and proteins pulled down with streptavidin agarose. After stripping the cell surface, most of the Tie2-specific signal in the immunoblot was lost confirming that Tie2 receptors isolated with streptavidin agarose represented the receptors at the plasma membrane (Fig. 5A).

Internalization of Tie2 in the presence of Ang1 and Ang2 was determined by examining the amount of receptor remaining at the cell surface at various times after the addition of either ligand. HUVECs were incubated with either Ang1 or Ang2 for various times at 37°C followed by biotinylation of cell surface proteins. Cell lysates containing equal amounts of protein were incubated with streptavidin agarose and the amount of Tie2 remaining at the cell surface was determined by immunoblotting for Tie2. In the presence of Ang1, Tie2 was rapidly internalized into the cell evidenced by the reduced amount of Tie2 isolated with streptavidin agarose (Fig. 5B). By contrast, in the presence of Ang2, Tie2 internalization was considerably slower compared with Ang1, with almost all of the receptor still remaining at the cell surface after 90 minutes of stimulation (Fig. 5B). When cell stimulations with Ang1 were performed at 0°C, a temperature where receptor endocytosis is suppressed, the disappearance of Tie2 from the cell surface was inhibited indicating that the reduced amount of Tie2 isolated with streptavidin agarose was a consequence of receptor internalization (data not shown).

Fig. 3.

Ang1 and Ang2 do not influence Tie2 synthesis. [35S]methionine and [35S]cysteine labelled HUVECs were incubated for various times together with pre-clustered Ang1 (800 ng/ml), or Ang2 (800 ng/ml). Tie2 immunoprecipitates were resolved by SDS-PAGE and the dried gel exposed to a phosphorimager screen. (A) Representative autoradiograph. (B) The intensities of the bands were quantified and plotted against time. Time=12 hours was designated 100%. All other values were expressed as a percentage of the 12-hour time point. Unstimulated cells (•), Ang1 (⋄), Ang2 (▪). Results are mean ± s.e.m. of three independent experiments.

Fig. 3.

Ang1 and Ang2 do not influence Tie2 synthesis. [35S]methionine and [35S]cysteine labelled HUVECs were incubated for various times together with pre-clustered Ang1 (800 ng/ml), or Ang2 (800 ng/ml). Tie2 immunoprecipitates were resolved by SDS-PAGE and the dried gel exposed to a phosphorimager screen. (A) Representative autoradiograph. (B) The intensities of the bands were quantified and plotted against time. Time=12 hours was designated 100%. All other values were expressed as a percentage of the 12-hour time point. Unstimulated cells (•), Ang1 (⋄), Ang2 (▪). Results are mean ± s.e.m. of three independent experiments.

Fig. 4.

Tie2 protein levels decrease in Ang1 and Ang2 stimulated HUVECs. (A) HUVECs were stimulated with pre-clustered Ang1 (800 ng/ml) or Ang2 (800 ng/ml) for the indicated times. Total cell lysates were analyzed by western immunoblotting using a monoclonal Tie2 antibody to detect total receptor levels and an anti-β-actin antibody to verify that equal amounts of protein were analyzed. Time 0 represents cells stimulated with medium alone. (B) The intensities of the bands in A were quantified and expressed as a percentage of the t=0 value and plotted against time, Ang1 (⋄) Ang2 (▪). Results are mean ± s.e.m. of three independent experiments.

Fig. 4.

Tie2 protein levels decrease in Ang1 and Ang2 stimulated HUVECs. (A) HUVECs were stimulated with pre-clustered Ang1 (800 ng/ml) or Ang2 (800 ng/ml) for the indicated times. Total cell lysates were analyzed by western immunoblotting using a monoclonal Tie2 antibody to detect total receptor levels and an anti-β-actin antibody to verify that equal amounts of protein were analyzed. Time 0 represents cells stimulated with medium alone. (B) The intensities of the bands in A were quantified and expressed as a percentage of the t=0 value and plotted against time, Ang1 (⋄) Ang2 (▪). Results are mean ± s.e.m. of three independent experiments.

Release of bound Ang1 and Ang2 upon Tie2 activation

Most peptide growth factors that bind to their cognate receptors are co-internalized upon activation and internalization of the receptor. In order to determine whether Ang1 or Ang2 are also internalized together with Tie2, HUVECs were stimulated with iodinated Ang1 or Ang2. Iodination of Ang1 and Ang2 did not disrupt the multimerization state of either ligand as shown by gel electrophoresis and western blot analysis. Under reducing conditions, both native Ang1 and 125I-labeled Ang1 (125I-Ang1) migrated with a molecular mass of ∼70 kDa, and under non-reducing conditions migrated with a minimum molecular mass of ∼200 kDa suggesting that the minimal multimeric state of Ang1 and 125I-Ang1 is a trimer as reported previously for Ang1 (Procopio et al., 1999) (Fig. 6A). Ang2 and 125I-Ang2 migrated, under reducing conditions, with a molecular mass of ∼70 kDa, and under non-reducing conditions migrated with a minimum molecular mass of ∼150 kDa, suggesting that the minimal multimeric state of Ang2 and 125I-Ang2 is a dimer as reported previously for Ang2 (Procopio et al., 1999) (Fig. 6B).

Fig. 5.

Ang1 and Ang2 induce differential internalization of Tie2 in HUVECs. Surface biotinylation of HUVECs was used to assess the amount of Tie2 remaining at the cell surface upon Ang1 and Ang2 stimulation. (A) Surface biotinylated HUVECs were lysed and incubated with streptavidin conjugated to agarose beads. Non-biotinylated cells (Non), biotinylated (Biotin), biotinylated followed by stripping (Biotin + Stripping) and total cell lysate (Cell Lysate) proteins were resolved by SDS-PAGE and immunoblotted using a monoclonal anti-Tie2 antibody. (B) HUVECs were incubated with pre-clustered Ang1 (800 ng/ml), or Ang2 (800 ng/ml) for the indicated times at 37°C. Cell surface proteins were biotinylated and isolated with streptavidin agarose followed by immunoblotting as described in (A), t=0 represents cells stimulated with media alone. (C) The intensities of the bands in B were quantified and plotted against time, Ang1 (⋄) Ang2 (▪). Results are mean ± s.e.m. of four independent experiments.

Fig. 5.

Ang1 and Ang2 induce differential internalization of Tie2 in HUVECs. Surface biotinylation of HUVECs was used to assess the amount of Tie2 remaining at the cell surface upon Ang1 and Ang2 stimulation. (A) Surface biotinylated HUVECs were lysed and incubated with streptavidin conjugated to agarose beads. Non-biotinylated cells (Non), biotinylated (Biotin), biotinylated followed by stripping (Biotin + Stripping) and total cell lysate (Cell Lysate) proteins were resolved by SDS-PAGE and immunoblotted using a monoclonal anti-Tie2 antibody. (B) HUVECs were incubated with pre-clustered Ang1 (800 ng/ml), or Ang2 (800 ng/ml) for the indicated times at 37°C. Cell surface proteins were biotinylated and isolated with streptavidin agarose followed by immunoblotting as described in (A), t=0 represents cells stimulated with media alone. (C) The intensities of the bands in B were quantified and plotted against time, Ang1 (⋄) Ang2 (▪). Results are mean ± s.e.m. of four independent experiments.

The binding of 125I-Ang1 and 125I-Ang2 to HUVECs was performed at 0°C. At this temperature, receptor endocytosis is inhibited thereby allowing ligand binding with minimal receptor internalization. Following ligand binding at 0°C, the cells were washed to remove unbound ligands and then transferred to 37°C to initiate receptor internalization. Immediately upon transfer of the cells to 37°C there was a progressive loss of radioactivity from the cells evident within 5 minutes of incubation (Fig. 6C). This was observed for both 125I-Ang1 and 125I-Ang2 with the loss of radioactivity from the cells faster when 125I-Ang2 was bound (Fig. 6C). When 125I-Ang2 was bound, more than half of the initial radioactivity was lost from the cells after 30 minutes of incubation at 37°C, whereas when 125I-Ang1 was bound, half of the initial radioactivity was lost after 90 minutes of incubation. Cross-linking 125I-Ang1 with the anti-polyhistidine antibody did not significantly influence the rate of release of the ligand from the cell surface and was therefore omitted from the binding studies.

Fig. 6.

Binding and release of 125I-Ang1 and 125I-Ang2 to HUVECs. Electrophoretic mobility of 125I-Ang1 and Ang1 (A) and 125I-Ang2 and Ang2 (B). Aliquots of 125I-Ang1 and 125I-Ang2 and 50 ng Ang1 and Ang2 were resolved by SDS-PAGE under reduced (R) and non-reduced (NR) conditions. To visualize 125I-Ang1 and 125I-Ang2, the gel was dried and exposed to X-ray film. To visualize Ang1 and Ang2, proteins were transferred to PVDF membranes and immunoblotted using an anti-polyhistidine antibody. Numbers indicate relative molecular masses. (C) 125I-Ang1 and 125I-Ang2 were allowed to bind to HUVECs at 0°C. The cells were washed to remove unbound ligands and then transferred to 37°C. At the indicated times, the cells were solubilized and cell-associated radioactivity counted and plotted against time. Radioactivity at t=0 was designated 100%, 125I-Ang1 (⋄) 125I-Ang2 (▪). (D) Time-dependent appearance of radioactivity in the medium collected from cells bound with 125I-Ang1 (⋄) and 125I-Ang2 (▪). Radioactivity was expressed as a percentage of the total radioactivity contained within the sample. (E) Medium was collected at the indicated times from HUVECs bound by 125I-Ang1 and immunoprecipitated with an anti-polyhistidine antibody. Immunoprecipitates were resolved by SDS-PAGE and exposed to X-ray film. Cold refers to cells incubated at 0°C for 120 minutes. Numbers on the left represent relative molecular masses. (F) Media was collected at the indicated times from cells bound with 125I-Ang2 and processed as in E. White line indicates that intervening lanes have been spliced out. (G) Ang1 and Ang2 were allowed to bind to HUVECs. Cells were washed and incubated at 37°C in fresh binding medium. At the indicated times, the medium was immunoprecipitated and immunoblotted using an anti-polyhistidine antibody. As controls for the position of Ang1 and Ang2, 50 ng of each ligand were resolved alongside the media samples. Cold refers to media collected from cells incubated at 0°C for 3 hours.

Fig. 6.

Binding and release of 125I-Ang1 and 125I-Ang2 to HUVECs. Electrophoretic mobility of 125I-Ang1 and Ang1 (A) and 125I-Ang2 and Ang2 (B). Aliquots of 125I-Ang1 and 125I-Ang2 and 50 ng Ang1 and Ang2 were resolved by SDS-PAGE under reduced (R) and non-reduced (NR) conditions. To visualize 125I-Ang1 and 125I-Ang2, the gel was dried and exposed to X-ray film. To visualize Ang1 and Ang2, proteins were transferred to PVDF membranes and immunoblotted using an anti-polyhistidine antibody. Numbers indicate relative molecular masses. (C) 125I-Ang1 and 125I-Ang2 were allowed to bind to HUVECs at 0°C. The cells were washed to remove unbound ligands and then transferred to 37°C. At the indicated times, the cells were solubilized and cell-associated radioactivity counted and plotted against time. Radioactivity at t=0 was designated 100%, 125I-Ang1 (⋄) 125I-Ang2 (▪). (D) Time-dependent appearance of radioactivity in the medium collected from cells bound with 125I-Ang1 (⋄) and 125I-Ang2 (▪). Radioactivity was expressed as a percentage of the total radioactivity contained within the sample. (E) Medium was collected at the indicated times from HUVECs bound by 125I-Ang1 and immunoprecipitated with an anti-polyhistidine antibody. Immunoprecipitates were resolved by SDS-PAGE and exposed to X-ray film. Cold refers to cells incubated at 0°C for 120 minutes. Numbers on the left represent relative molecular masses. (F) Media was collected at the indicated times from cells bound with 125I-Ang2 and processed as in E. White line indicates that intervening lanes have been spliced out. (G) Ang1 and Ang2 were allowed to bind to HUVECs. Cells were washed and incubated at 37°C in fresh binding medium. At the indicated times, the medium was immunoprecipitated and immunoblotted using an anti-polyhistidine antibody. As controls for the position of Ang1 and Ang2, 50 ng of each ligand were resolved alongside the media samples. Cold refers to media collected from cells incubated at 0°C for 3 hours.

When the culture medium was analyzed for the presence of Ang1 and Ang2 there was a progressive accumulation of radioactivity in the media. This release into the medium occurred more rapidly with 125I-Ang2 compared with 125I-Ang1. Radioactivity was detected in the medium within 5 minutes at 37°C and continued to increase up to 2 hours of incubation for both ligands (Fig. 6D). When the cells were maintained at 0°C the release of 125I-Ang1 was significantly reduced. By contrast, 125I-Ang2 had a tendency to be released at 0°C (Table 1).

Table 1.

Cell and media distribution of 125I-Ang1 and 125I-Ang2 at 0°C and 37°C

0°C
37°C
% Cell associated % Media % Cell associated % Media
125I-Ang1   82±5.4   24±3.8   28±3.0   76±2.8  
125I-Ang2   53±7.7   56±5.9   14±2.2   86±2.8  
0°C
37°C
% Cell associated % Media % Cell associated % Media
125I-Ang1   82±5.4   24±3.8   28±3.0   76±2.8  
125I-Ang2   53±7.7   56±5.9   14±2.2   86±2.8  

HUVECs were incubated with 125I-Ang1 and 125I-Ang2 at 0°C to allow ligand binding as described. The cells were washed to remove unbound ligand and incubated at 0°C or 37°C in fresh binding medium for 2 hours. The medium was collected and radioactivity quantified in a gamma counter. The cells were washed, solubilized, and counted in a gamma counter. Values are expressed as a percentage of the total radioactivity contained within the sample (cell-associated radioactivity + media radioactivity = 100%). Results are mean ± s.e.m. of four independent experiments

To determine whether the released radioactivity represented intact Ang1 or Ang2, or possibly a proteolytic fragment of the ligands, the media was collected and incubated with anti-polyhistidine antibodies pre-coupled to protein-G-Sepharose. Following SDS-PAGE under reducing conditions and autoradiography, a band with a molecular mass of ∼70 kDa was immunoprecipitated from the medium collected from cells previously incubated with 125I-Ang1 or 125I-Ang2 (Fig. 6E,F). The ∼70 kDa proteins isolated from the medium collected from 125I-Ang1- and 125I-Ang2-bound cells migrated at the same molecular mass as full-length 125I-Ang1 and 125I-Ang2 respectively, suggesting that the 125I-Ang1 and 125I-Ang2 released from the cells upon Tie2 internalization were not proteolytically modified upon release from the receptor. As predicted from our earlier studies, there was significantly less 125I-Ang1 in the medium from cells incubated at 0°C (Fig. 6E), whereas 125I-Ang2 was released into the medium even when the cells were maintained at 0°C (Fig. 6F).

To further investigate the observation that 125I-Ang1 and 125I-Ang2 remain at the cell surface and may not be internalized into endothelial cells, surface bound ligands were differentiated from internalized ligands by cleaving surface proteins with trypsin. Surface-bound 125I-Ang1 and 125I-Ang2 were not readily removed by washing the cells with high salt and acid buffers which are commonly used to remove surface ligands from their receptors. By treating HUVECs with trypsin at 0°C, ∼90% of surface-bound 125I-Ang1 and 125I-Ang2 were removed by this procedure.

To determine whether Ang1 and Ang2 remained at the cell surface, HUVECs were incubated with 125I-Ang1 or 125I-Ang2 for 30 minutes at 37°C. The cells were washed to remove unbound ligands and the proteins at the cell surface were then cleaved with trypsin. Following digestion of surface proteins by trypsin, 90% of 125I-Ang1 and 86% of 125I-Ang2 were removed from the cells as evidenced by the loss of radioactivity associated with the cells and the concomitant increase of radioactivity in the supernatant fraction (Table 2). These findings suggest that 125I-Ang1 and 125I-Ang2 were not internalized into endothelial cells but remained at the cell surface where they were accessible to trypsin. Similar results were obtained when the cells were incubated with 125I-Ang1 or 125I-Ang2 for 15 minutes, 1 hour, or 2 hours (data not shown).

Table 2.

Trypsin digestion of cell surface proteins

Trypsin treatment
Total cell radioactivity Pellet Supernatant
125I-Ang1   44,570   4681   33,599  
125I-Ang2   29,400   4780   23,664  
Trypsin treatment
Total cell radioactivity Pellet Supernatant
125I-Ang1   44,570   4681   33,599  
125I-Ang2   29,400   4780   23,664  

HUVECs were incubated in binding medium containing 125I-Ang1 or 125I-Ang2 for 30 minutes at 37°C. The cells were washed and incubated at 0°C with 1 mg/ml trypsin for 55 minutes followed by 5 minutes at 37°C. The cells were collected and centrifuged. The supernatant containing digested extracellular proteins was removed and the radioactivity quantified in a gamma counter. The cell pellet was solubilized and counted in a gamma counter. Numbers refer to radioactive counts per minute (cpm)

To ensure that the surface release of the ligands was not a consequence of the iodination, we incubated HUVECs with non-iodinated Ang1 or Ang2 and repeated our release experiment. Western blot analysis using the anti-polyhistidine antibody revealed that Ang1 and Ang2 were released into the media with similar kinetics as that observed with 125I-Ang1 and 125I-Ang2 (Fig. 6G).

Potential mechanisms regulating ligand release

To determine whether Ang1 and Ang2 release was due to shedding of the extracellular domain of Tie2, we measured the amount of Tie2 present in the culture medium of cells after stimulation with either Ang1 or Ang2 (Fig. 7A). Our results reveal that although soluble Tie2 is present in the culture medium of HUVECs as described previously (Reusch et al., 2001), the amount of soluble Tie2 released to the media was not increased by Ang1 or Ang2 stimulation indicating that the release of Ang1 and Ang2 does not involve cleavage of the extracellular domain of Tie2. This observation is consistent with our preliminary experiments suggesting that released Ang1 and Ang2 are biologically active and are capable of rebinding to fresh cells (data not shown).

Blocking Tie2 internalization using chemical inhibitors such as phenylarsine oxide (PAO) or hypertonic sucrose (0.5 M), agents shown to block internalization of a variety of cell surface receptors (Hertel et al., 1985; Faussner et al., 2004), had no effect on the release of Ang1 although treatment of cells with 0.5 M sucrose only partially blocked Tie2 internalization in response to Ang1 (data not shown). Receptor internalization is probably not sufficient for ligand release because Ang2 did not significantly promote Tie2 internalization but was released faster compared with Ang1.

To stimulate intracellular signalling within endothelial cells, we exposed endothelial cells to pervanadate, a powerful protein tyrosine phosphatase inhibitor known to considerably enhance phosphorylation on tyrosine, serine and threonine residues of proteins. Exposure of HUVECs to pervanadate led to marked increases in tyrosine phosphorylation of cellular proteins (data not shown). When endothelial cells were exposed to pervanadate Ang1 was released faster compared with endothelial cells not treated with pervanadate (Fig. 7B). Examination of Tie2 internalization in the presence of Ang1 and pervanadate revealed that although Ang1 induced Tie2 internalization, the addition of pervanadate did not further stimulate Tie2 internalization (Fig. 7C) indicating that receptor internalization alone is not sufficient for Ang1 release. Pervanadate also enhanced Ang2 release but more consistent results were obtained with Ang1 (data not shown). In addition, stimulation of endothelial cells with pervanadate led to increased levels of Tie2 tyrosine phosphorylation (data not shown).

Fig. 7.

Potential mechanisms of angiopoietin release. (A) HUVECs were treated with either Ang1 (800 ng/ml cross-linked, white bars), Ang2 (800 ng/ml) (black bars), Mock (cross-linking antibody alone, hatched bars) or left untreated (grey bars). At the indicated times, the media was collected and the amount of soluble Tie2 (sTie2) quantified using a Human Tie2 Immunoassay Kit. (B) HUVECs were incubated with Ang1 (800 ng/ml) for 90 minutes at 0°C in the presence or absence of 100 μM pervanadate. The cells were washed to remove unbound Ang1 and incubated for 30 minutes at 37°C in fresh medium alone or containing 100 μM pervanadate. Ang1 was immunoprecipitated from the media and immunoblotted using an anti-polyhistidine antibody. (C) The cells from B were washed, surface proteins were biotinylated and isolated with streptavidin agarose, followed by immunoblotting for Tie2. As a control for Tie2 internalization, cells incubated in media alone in the absence of Ang1 or pervanadate were included (Non). White line indicates that intervening lanes have been spliced out.

Fig. 7.

Potential mechanisms of angiopoietin release. (A) HUVECs were treated with either Ang1 (800 ng/ml cross-linked, white bars), Ang2 (800 ng/ml) (black bars), Mock (cross-linking antibody alone, hatched bars) or left untreated (grey bars). At the indicated times, the media was collected and the amount of soluble Tie2 (sTie2) quantified using a Human Tie2 Immunoassay Kit. (B) HUVECs were incubated with Ang1 (800 ng/ml) for 90 minutes at 0°C in the presence or absence of 100 μM pervanadate. The cells were washed to remove unbound Ang1 and incubated for 30 minutes at 37°C in fresh medium alone or containing 100 μM pervanadate. Ang1 was immunoprecipitated from the media and immunoblotted using an anti-polyhistidine antibody. (C) The cells from B were washed, surface proteins were biotinylated and isolated with streptavidin agarose, followed by immunoblotting for Tie2. As a control for Tie2 internalization, cells incubated in media alone in the absence of Ang1 or pervanadate were included (Non). White line indicates that intervening lanes have been spliced out.

In this study, we demonstrate that in response to Ang1, Tie2 is rapidly internalized and targeted for degradation. By contrast, Ang2 only weakly activated Tie2 and did not significantly stimulate receptor internalization compared with Ang1, while mildly inducing Tie2 degradation. More importantly, we show that the angiopoietins are released from the endothelial cell surface after binding and accumulate in the surrounding medium.

The observation that Ang1 induced rapid internalization and degradation of Tie2 is consistent with studies of other tyrosine kinase receptors, such as the EGFR (Stoscheck and Carpenter, 1984), where degradation of the receptor may serve to turn off signal transduction, a process crucial for maintaining cellular homeostasis. For example, a mutant EGFR that cannot undergo ligand-induced downregulation transforms cells in culture (Wells et al., 1990). Evidence that Tie2 signalling may be regulated in the vasculature comes from studies that have identified constitutively active mutants of Tie2 causing vascular abnormalities known as venous malformations in humans (Vikkula et al., 1996; Calvert et al., 1999). The degradation of Tie2 upon binding Ang1 may serve to regulate the magnitude and duration of Ang1 signal transduction. Consistent with this possibility, Ang2 only weakly activated Tie2 and resulted in a correspondingly reduced rate of Tie2 degradation.

Similar to our study, Hashimoto et al. (Hashimoto et al., 2004) have shown that Tie2 is downregulated in cell lysates in the presence of Ang1 but not Ang2. In our study, the range of half-life values obtained for Tie2 in the presence of Ang2 overlapped with the range of half-life values in unstimulated cells indicating that in some experiments the downregulation of Tie2 by Ang2 may not be as pronounced.

The differences in the rate of Tie2 internalization stimulated by Ang1 and Ang2 might be related to the level of Tie2 activation induced by these ligands, where higher levels of Tie2 activation promoted a more rapid rate of Tie2 internalization. It is possible that owing to the higher multimerization state of Ang1, a larger population of Tie2 receptors were activated and subsequently targeted for degradation. In line with this possibility, rapid downregulation of the EGFR is dependent on the tyrosine kinase activity of the receptor (Wiley et al., 1991). Recently, receptor dimerization has also been shown to be sufficient to induce rapid internalization of the EGFR (Wang et al., 2005). This could also be applied to Tie2 where Ang1, owing to its higher multimerization state would induce corresponding higher multimerization states of Tie2 compared with Ang2. To date, the mechanism(s) regulating Tie2 internalization are not known.

Our study has characterized Ang2 as a partial agonist for Tie2 rather than a complete antagonist because both Ang1 and Ang2 exerted similar effects on Tie2 but with varying potency and kinetics. In addition to inducing phosphorylation of Tyr1102 and Tyr1108 on Tie2, Ang1 and Ang2 activate similar signalling pathways such as the activation of Akt (Kim et al., 2000a; Papapetropoulos et al., 2000; Kim et al., 2000b), although a higher concentration of Ang2 is required (Kim et al., 2000b). Nevertheless, our characterization of Ang2 as a partial agonist does not rule out the possibility that Ang2 could still antagonize the Ang1 signal.

The role of Ang2 in Tie2 signalling has been controversial in the literature with some studies describing Ang2 as a Tie2 antagonist (Maisonpierre et al., 1997) whereas other studies reported the agonist properties of Ang2 (Kim et al., 2000b; Teichert-Kuliszewska et al., 2001). The reason for the controversy among the various studies is not clear. In our study, the activation of Tie2 by Ang2 is weak and concentration dependent. Perhaps the weak induction of Tie2 by Ang2 may be interpreted as background phosphorylation. Another angiopoietin family member, Ang4, elicits similar levels of Tie2 activation as Ang2 (our unpublished observations) but is described in the literature as a Tie2 agonist (Valenzuela et al., 1999).

The observation that Ang1 and Ang2 were not internalized into endothelial cells but instead were released from the cell surface into the surrounding medium was surprising because many ligands have been shown to internalize together with their receptors. Similar to our results, Wang et al. (Wang et al., 2002) have shown that VEGF is released into the surrounding medium after binding to human colonic vascular endothelial cells and that released VEGF can be reused. Ang1 and Ang2, which are released into the medium after binding to endothelial cells, are capable of rebinding to fresh cells (data not shown), suggesting that these ligands may also be recycled or reused by endothelial cells.

Our data have indicated that shedding of the extracellular domain of Tie2, and receptor internalization are not responsible for the release of the angiopoietins but suggests that the mechanism(s) may be dependent in part or can be modulated by intracellular signalling. The observation that pervanadate appears to enhance angiopoietin release suggests that activation of Tie2 together with the activation or inhibition of particular signalling pathways or molecules is responsible for the release of Ang1 and Ang2 from the endothelial cell surface. Although the data obtained with pervanadate may suggest the involvement of a signalling pathway(s), the mechanism(s) involved might be more complex. The mechanism(s) regulating Ang1 and Ang2 release may be different. For example, Ang2 only weakly induces tyrosine phosphorylation of Tie2 but was released faster from endothelial cells compared with Ang1. One possibility is that angiopoietin release may be related more to changes in receptor affinity after binding to these ligands. Further work will be required to completely elucidate the mechanism(s) regulating ligand release.

The affinities of Ang1 and Ang2 for Tie2 were reported to be the same when binding of either ligand to the extracellular domain of Tie2 was examined in vitro (Maisonpierre et al., 1997). The interaction of Ang1 and Ang2 with the endothelial cell surface may be more complex, because several reports have shown that Ang1 can bind to other surface receptors such as Tie1 and integrins (Carlson et al., 2001; Saharinen et al., 2005). Ang2 can also bind to integrins and activate intracellular signalling pathways (Hu et al., 2006). The fact that Ang2 was released faster after binding compared with Ang1 suggests that the interaction between these ligands and the endothelial cell surface may be different. One possibility is that Ang1 may bind with higher affinity to other receptors such as Tie1 and/or integrins and therefore remain on the cell surface longer compared with Ang2. The release of the angiopoietins after binding and activation of Tie2 might make them available to bind to other receptors on endothelial cells and on non-endothelial cells.

The angiopoietins together with VEGF are an emerging class of ligands, which, instead of being co-internalized together with their cognate receptors as described for numerous other receptor-ligand systems, are released. We believe that the ability of ligands to be released after receptor activation will be a consideration when studying endothelial cell biology.

Cell culture and stimulation

Human umbilical vein endothelial cells (HUVECs) (GlycoTech, Rockville, MD) were grown on gelatinized (2% gelatin, Sigma) tissue culture plates in F12K medium (ATCC) supplemented with 10% fetal bovine serum, penicillin-streptomycin, 0.1 mg/ml heparin sodium, 10 ng/ml EGF, 10 ng/ml VEGF, 5 ng/ml bFGF, and 2 mM L-glutamine, and were used up to passage 10. EA.hy926 endothelial cells were cultured as described previously (Jones et al., 1999). Human recombinant Ang1 and Ang2 containing a polyhistidine tag were obtained from R&D Systems. All cell stimulations were performed in supplemented F12K medium containing varying concentrations of Ang1 or Ang2. For some stimulations with Ang1, recombinant Ang1 was pre-incubated together with an anti-polyhistidine monoclonal antibody (10 μg/ml, R&D Systems) to facilitate cross-linking of Ang1 multimers.

Cell lysis and tie2 immunoprecipitation

HUVECs were washed twice with ice-cold phosphate buffered saline (PBS) and lysed with RIPA lysis buffer (50 mM Tris-HCl pH 7.5, 150 mM NaCl, 1% Igepal, 0.5% sodium deoxycholate, 0.1% SDS, 2 mM sodium orthovanadate and protease inhibitors). The cells were incubated on ice for 20 minutes followed by centrifugation at 14,000 g for 5 minutes at 4°C. Tie2 was immunoprecipitated from the supernatant using 2-4 μg of anti-Tie2 antibody C20 (Santa Cruz Biotechnology) that had been pre-coupled to 25 μl protein-A-Sepharose (Amersham Biosciences).

Tie2 immunoblotting

Proteins were resolved on a 7.5% polyacrylamide gel and transferred to PVDF (Perkin Elmer) membranes. The membranes were blocked for 1 hour at room temperature in 5% bovine serum albumin (BSA) prior to detecting phosphotyrosine, or in 5% non-fat dry milk for detecting total Tie2. The membranes were incubated with 1 μg/ml anti-phosphotyrosine antibody 4G10 (Upstate), anti-phospho-Tie2 Tyr1102/1108 antibody (1:5000 dilution, Calbiochem) or 0.5 μg/ml anti-Tie2 monoclonal antibody (BD Biosciences Pharmingen). Proteins were visualized using secondary antibodies conjugated to HRP (Bio-Rad) followed by enhanced chemiluminescence.

Tie2 half-life determination and synthesis

The half-life of Tie2 in unstimulated cells and in cells treated with either Ang1 or Ang2 was determined by 35S labelling of proteins as described (Burke and Wiley, 1999). HUVECs were incubated for 16-18 hours at 37°C in DMEM lacking methionine and cysteine (Gibco) supplemented with 10% dialyzed fetal bovine serum (Gibco), penicillin-streptomycin, 2 mM L-glutamine, and 100 μCi/ml [35S]methionine and [35S]cysteine (Tran35S-Label, MP Biomedicals). Cells were washed six times with HUVEC growth medium and incubated in the same medium either alone, or containing 800 ng/ml Ang1 (cross-linked with the anti-polyhistidine monoclonal antibody) or 800 ng/ml Ang2 for various times at 37°C. The cells were lysed and Tie2 immunoprecipitated and resolved on a 7.5% polyacrylamide gel. The gel was dried and exposed to a Molecular Dynamics phosphorimager screen. The intensities of the Tie2 bands were quantified using the Bio-Rad Molecular Imager FX. The intensity of the band at time=0 was designated 100%. The intensities of all other bands were expressed as a percentage of the initial (t=0) value. The natural logarithm of the percentage values was taken and plotted against time. The slope of the line was used to determine the time required for the initial value (100%) to decrease to 50%.

To examine Tie2 synthesis in the presence of Ang1 or Ang2, cells were incubated in DMEM lacking methionine and cysteine as described above, containing 100 μCi/ml [35S]methionine and [35S]cysteine and either 800 ng/ml Ang1 (cross-linked) or 800 ng/ml Ang2 for various times at 37°C. The cells were lysed and Tie2 immunoprecipitated and resolved on a 7.5% polyacrylamide gel and the bands were quantified as above.

Tie2 biotinylation

HUVECs were incubated for various times with either Ang1 (800 ng/ml, cross-linked) or Ang2 (800 ng/ml) at 37°C. The cells were placed on ice, washed twice with ice-cold PBS and incubated for 15 minutes at 4°C with 0.5 mg/ml biotin (Sulfo-NHS-SS-Biotin, Pierce) in PBS containing Ca2+ and Mg2+. The biotin solution was removed and the cells were washed once with 25 mM Tris-HCl pH 8.0 to quench non-reacted biotin followed by three washes with cold PBS. The cells were lysed with RIPA lysis buffer and cell lysates containing equal amounts of protein were incubated with 25 μl streptavidin conjugated to agarose (Pierce). The beads were washed three times with 1 ml RIPA lysis buffer, resuspended in 85 μl sample buffer, and boiled for 5 minutes. Supernatants (60 μl) were immunoblotted for Tie2 as described. For control purposes, surface biotin was removed by incubating cells three times in cold stripping solution (50 mM reduced glutathione, 75 mM NaCl, 75 mM NaOH, 1 mM EDTA and 1% BSA) for 10 minutes each at 4°C followed by two washes with cold PBS as described previously (Burke et al., 2001).

Iodination of Ang1 and Ang2

Carrier-free human recombinant Ang1 and Ang2 (25 μg each, R&D Systems) were iodinated using IODO-GEN pre-coated iodination tubes (Pierce) and 0.2-0.5 mCi Na[125I] (MP Biomedicals). All steps were performed according to the manufacturer's instructions. Free iodine was separated using 10 ml Dextran desalting columns (Pierce).

Internalization of Ang1 and Ang2

HUVECs grown to confluency were washed once with PBS and incubated in ice-cold binding medium (F12K medium containing 0.1% BSA) containing either 125I-Ang1 or 125I-Ang2 for 90 minutes on ice at 4°C to allow ligand binding. Cell monolayers were washed six times (4 ml per wash) with cold binding medium, fresh binding medium was added, and the cells transferred to 37°C. At various times, the cells were removed from the incubator and the medium collected and counted. The cells were lysed with solubilization buffer (0.1 M NaOH, 0.1% SDS) as described previously (Waterman et al., 1998). Radioactivity contained within the media and the cells was counted in a Compugamma CS gamma counter. To differentiate between surface bound or internalized ligands, surface ligands were removed by trypsin. Cell monolayers were placed on ice, washed three times with cold PBS, and incubated with trypsin (1 mg/ml) for 55 minutes on ice at 4°C followed by 5 minute incubation at 37°C. The cells were collected and centrifuged at 1000 g for 5 minutes. The supernatant, which contained digested extracellular proteins including cleaved surface ligands, was removed and counted. The cell pellet consisting of internalized ligand, was resuspended in solubilization buffer and counted.

Angiopoietin immunoprecipitation and immunoblotting

Ang1 and Ang2 were immunoprecipitated from cell culture media using 2 μg anti-polyhistidine monoclonal antibody (R&D Systems) that was pre-coupled to 25 μl protein-G-Sepharose (Amersham Biosciences). The immunoprecipitates were immunoblotted using 1 μg/ml anti-polyhistidine monoclonal antibody (R&D Systems). Proteins were visualized using goat anti-mouse secondary antibodies conjugated to HRP (Bio-Rad) followed by enhanced chemiluminescence.

Soluble Tie2 determination

HUVECs were grown to 25% confluency in 24-well dishes for 24 hours before treatment with Ang1 (cross-linked) or Ang2. Each treatment was done in triplicate. Following cell stimulations, the culture media was collected and cleared by brief centrifugation. Soluble Tie2 present in the culture media was quantified by the Quantikine Human Tie2 Immunoassay (R&D Systems) according to the manufacturer's instructions.

The authors would like thank Dr Mariana Capurro, Dr Vladimir Lhotak and John Ebos, for helpful suggestions. This work was supported by operating grants from the Canadian Institutes of Health Research and the Heart and Stroke Foundation of Ontario. D.J.D. holds a Canada Research Chair in Angiogenic and Lymphangiogenic Signalling. E.B. is supported by the Ontario Graduate Scholarship in Science and Technology. V.P.K.H.N. is a recipient of the Heart and Stroke Master's Studentship.

Beguinot, L., Lyall, R. M., Willingham, M. C. and Pastan, I. (
1984
). Down-regulation of the epidermal growth factor receptor in KB cells is due to receptor internalization and subsequent degradation in lysosomes.
Proc. Natl. Acad. Sci. USA
81
,
2384
-2388.
Burke, P. M. and Wiley, H. S. (
1999
). Human mammary epithelial cells rapidly exchange empty EGFR between surface and intracellular pools.
J. Cell Physiol.
180
,
448
-460.
Burke, P., Schooler, K. and Wiley, H. S. (
2001
). Regulation of epidermal growth factor receptor signalling by endocytosis and intracellular trafficking.
Mol. Biol. Cell
12
,
1897
-1910.
Calvert, J. T., Riney, T. J., Kontos, C. D., Cha, E. H., Prieto, V. G., Shea, C. R., Berg, J. N., Nevin, N. C., Simpson, S. A., Pasyk, K. A. et al. (
1999
). Allelic and locus heterogeneity in inherited venous malformations.
Hum. Mol. Genet.
8
,
1279
-1289.
Carlson, T. R., Feng, Y., Maisonpierre, P. C., Mrksich, M. and Morla, A. O. (
2001
). Direct cell adhesion to the angiopoietins mediated by integrins.
J. Biol. Chem.
276
,
26516
-26525.
Carpenter, G. and Cohen, S. (
1976
). 125I-labeled human epidermal growth factor. Binding, internalization, and degradation in human fibroblasts.
J. Cell Biol.
71
,
159
-171.
Davis, S., Aldrich, T. H., Jones, P. F., Acheson, A., Compton, D. L., Jain, V., Ryan, T. E., Bruno, J., Radziejewski, C., Maisonpierre, P. C. et al. (
1996
). Isolation of angiopoietin-1, a ligand for the Tie2 receptor, by secretion-trap expression cloning.
Cell
87
,
1161
-1169.
Dumont, D. J., Yamaguchi, T. P., Conlon, R. A., Rossant, J. and Breitman, M. L. (
1992
). Tek, a novel tyrosine kinase gene located on mouse chromosome 4, is expressed in endothelial cells and their presumptive precursors.
Oncogene
7
,
1471
-1480.
Dumont, D. J., Gradwohl, G. J., Fong, G. H., Auerbach, R. and Breitman, M. L. (
1993
). The endothelial-specific receptor tyrosine kinase, tek, is a member of a new subfamily of receptors.
Oncogene
8
,
1293
-1301.
Dumont, D. J., Gradwohl, G., Fong, G. H., Puri, M. C., Gertsenstein, M., Auerbach, A. and Breitman, M. L. (
1994
). 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.
Faussner, A., Schuessler, S., Seidl, C. and Jochum, M. (
2004
). Inhibition of sequestration of human B2 bradykinin receptor by phenylarsine oxide or sucrose allows determination of receptor affinity shift and ligand dissociation in intact cells.
Biol. Chem.
385
,
835
-843.
Gaborik, Z. and Hunyady, L. (
2004
). Intracellular trafficking of hormone receptors.
Trends Endocrinol. Metab.
15
,
286
-293.
Hashimoto, T., Wu, Y., Boudreau, N. J., Li, J. F., Matsumoto, M. M. and Young, W. L. (
2004
). Regulation of Tie2 expression by angiopoietin - potential feedback system.
Endothelium
11
,
207
-210.
Hayes, A. J., Huang, W. Q., Mallah, J., Yang, D., Lippman, M. E. and Li, L. Y. (
1999
). Angiopoietin-1 and its receptor Tie-2 participate in the regulation of capillary-like tube formation and survival of endothelial cell.
Microvasc. Res.
58
,
224
-237.
Hertel, C., Coulter, S. J. and Perkins, J. P. (
1985
). A comparison of catecholamine-induced internalization of beta-adrenergic receptors and receptor-mediated endocytosis of epidermal growth factor in human astrocytoma cells. Inhibition by phenylarsine oxide.
J. Biol. Chem.
260
,
12547
-12553.
Hu, B., Jarzynka, M. J., Guo, P., Imanishi, Y., Schlaepfer, D. D. and Cheng, S. Y. (
2006
). Angiopoietin 2 induces glioma cell invasion by stimulating matrix metalloprotease 2 expression through αvβ1 integrin and focal adhesion kinase signalling pathway.
Cancer Res.
66
,
775
-783.
Jones, N., Master, Z., Jones, J., Bouchard, D., Gunji, Y., Sasaki, H., Daly, R., Alitalo, K. and Dumont, D. J. (
1999
). Identification of Tek/Tie2 binding partners.
J. Biol. Chem.
274
,
30896
-30905.
Jones, N., Voskas, D., Master, Z., Sarao, R., Jones, J. and Dumont, D. J. (
2001
). Rescue of the early vascular defects in Tek/Tie2 null mice reveals an essential survival function.
EMBO Rep.
2
,
438
-445.
Jones, N., Chen, S. H., Sturk, C., Master, Z., Tran, J., Kerbel, R. S. and Dumont, D. J. (
2003
). A unique autophosphorylation site on Tie2/Tek mediates Dok-R phosphotyrosine binding domain binding and function.
Mol. Cell. Biol.
23
,
2658
-2668.
Kasuga, M., Kahn, C. R., Hedo, J. A., van Obberghen, E. and Yamada, K. M. (
1981
). Insulin-induced receptor loss in cultured human lymphocytes is due to accelerated receptor degradation.
Proc. Natl. Acad. Sci. USA
78
,
6917
-6921.
Keating, M. T. and Williams, L. T. (
1987
). Processing of the platelet-derived growth factor receptor. Biosynthetic and degradation studies using anti-receptor antibodies.
J. Biol. Chem.
262
,
7932
-7937.
Kim, I., Kim, H. G., So, J. N., Kim, J. H., Kwak, H. J. and Koh, G. Y. (
2000a
). Angiopoietin-1 regulates endothelial cell survival through the phosphatidylinositol 3′-kinase/Akt signal transduction pathway.
Circ. Res.
86
,
24
-29.
Kim, I., Kim, J. H., Moon, S. O., Kwak, H. J., Kim, N. G. and Koh, G. Y. (
2000b
). Angiopoietin-2 at high concentration can enhance endothelial cell survival through the phosphatidylinositol 3′-kinase/Akt signal transduction pathway.
Oncogene
19
,
4549
-4552.
Koblizek, T. I., Weiss, C., Yancopoulos, G. D., Deutsch, U. and Risau, W. (
1998
). Angiopoietin-1 induces sprouting angiogenesis in vitro.
Curr. Biol.
8
,
529
-532.
Kwak, H. J., So, J. N., Lee, S. J., Kim, I. and Koh, G. Y. (
1999
). Angiopoietin-1 is an apoptosis survival factor for endothelial cells.
FEBS Lett.
448
,
249
-253.
Maisonpierre, P. C., Suri, C., Jones, P. F., Bartunkova, S., Wiegand, S. J., Radziejewski, C., Compton, D., McClain, J., Aldrich, T. H., Papadopoulos, N. et al. (
1997
). Angiopoietin-2, a natural antagonist for Tie2 that disrupts in vivo angiogenesis.
Science
277
,
55
-60.
Mochizuki, Y., Nakamura, T., Kanetake, H. and Kanda, S. (
2002
). Angiopoietin 2 stimulates migration and tube-like structure formation of murine brain capillary endothelial cells through c-Fes and c-Fyn.
J. Cell Sci.
115
,
175
-183.
Papapetropoulos, A., Fulton, D., Mahboubi, K., Kalb, R. G., O'Connor, D. S., Li, F., Altieri, D. C. and Sessa, W. C. (
2000
). Angiopoietin-1 inhibits endothelial cell apoptosis via the Akt/Survivin pathway.
J. Biol. Chem.
275
,
9102
-9105.
Procopio, W. N., Pelavin, P. I., Lee, W. M. F. and Yeilding, N. M. (
1999
). 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.
Reusch, P., Barleon, B., Weindel, K., Martiny-Baron, G., Godde, A., Siemeister, G. and Marme, D. (
2001
). Identification of a soluble form of the angiopoietin receptor TIE-2 released from endothelial cells and present in human blood.
Angiogenesis
4
,
123
-131.
Saharinen, P., Kerkela, K., Ekman, N., Marron, M., Brindle, N., Lee, G. M., Augustin, H., Koh, G. Y. and Alitalo, K. (
2005
). Multiple angiopoietin recombinant proteins activate the Tie1 receptor tyrosine kinase and promote its interaction with Tie2.
J. Cell Biol.
169
,
239
-243.
Sato, T. N., Tozawa, Y., Deutsch, U., Wolburg-Buchholz, K., Fujiwara, Y., Gendron-Maguire, M., Gridley, T., Wolburg, H., Risau, W. and Qin, Y. (
1995
). Distinct roles of the receptor tyrosine kinases Tie1 and Tie-2 in blood vessel formation.
Nature
376
,
70
-74.
Sorkin, A. and Waters, C. M. (
1993
). Endocytosis of growth factor receptors.
BioEssays
15
,
375
-382.
Sorkin, A. and von Zastrow, M. (
2002
). Signal transduction and endocytosis: close encounters of many kinds.
Nat. Rev. Mol. Cell Biol.
3
,
600
-614.
Stoscheck, C. M. and Carpenter, G. (
1984
). Characterization of the metabolic turnover of epidermal growth factor receptor protein in A-431 cells.
J. Cell Physiol.
120
,
296
-302.
Suri, C., Jones, P. F., Patan, S., Bartunkova, S., Maisonpierre, P. C., Davis, S., Sato, T. N. and Yancopoulos, G. D. (
1996
). Requisite role of angiopoietin-1, a ligand for the TIE2 receptor, during embryonic angiogenesis.
Cell
87
,
1171
-1180.
Teichert-Kuliszewska, K., Maisonpierre, P. C., Jones, N., Campbell, A. I. M., Master, Z., Bendeck, M. P., Alitalo, K., Dumont, D. J., Yancopoulos, G. D. and Stewart, D. J. (
2001
). Biological action of angiopoietin-2 in a fibrin matrix model of angiogenesis is associated with activation of Tie2.
Circ. Res.
49
,
659
-670.
Valenzuela, D. M., Griffiths, J. A., Rojas, J., Aldrich, T. H., Jones, P. F., Zhou, H., McClain, J., Copeland, N. G., Gilbert, D. J., Jenkins, N. A. et al. (
1999
). Angiopoietins 3 and 4, diverging gene counterparts in mice and humans.
Proc. Natl. Acad. Sci. USA
96
,
1904
-1909.
Vikkula, M., Boon, L. M., Carraway, K. L., III, Calvert, J. T., Diamonti, A. J., Goumnerov, B., Pasyk, K. A., Marchuk, D. A., Warman, M. L., Cantley, L. C. et al. (
1996
). Vascular dysmorphogenesis caused by an activating mutation in the receptor tyrosine kinase TIE2.
Cell
87
,
1181
-1190.
Wang, D., Lehman, R. E., Donner, D. B., Matli, M. R., Warren, R. S. and Welton, M. L. (
2002
). Expression and endocytosis of VEGF and its receptors in human colonic vascular endothelial cells.
Am. J. Physiol. Gastrointest. Liver Physiol.
282
,
G1088
-G1096.
Wang, Q., Villenueve, G. and Wang, Z. (
2005
). Control of epidermal growth factor receptor endocytosis by receptor dimerization rather than kinase activation.
EMBO Rep.
6
,
942
-948.
Waterman, H., Sabanai, I., Geiger, B. and Yarden, Y. (
1998
). Alternative intracellular routing of ErbB receptors may determine signalling potency.
J. Biol. Chem.
273
,
13819
-13827.
Wells, A., Welsh, J. B., Lazar, C. S., Wiley, H. S., Gill, G. N. and Rosenfeld, M. G. (
1990
). Ligand-induced transformation by a noninternalizing epidermal growth factor receptor.
Science
247
,
962
-964.
Wiley, H. S. (
2003
). Trafficking of the ErbB receptors and its influence on signalling.
Exp. Cell Res.
284
,
78
-88.
Wiley, H. S., Herbst, J. J., Walsh, B. J., Lauffenburger, D. A., Rosenfeld, M. G. and Gill, G. N. (
1991
). The role of tyrosine kinase activity in endocytosis, compartmentation, and down-regulation of the epidermal growth factor receptor.
J. Biol. Chem.
266
,
11083
-11094.
Witzenbichler, B., Maisonpierre, P. C., Jones, P., Yancopoulos, G. D. and Isner, J. M. (
1998
). Chemotactic properties of angiopoietin-1 and -2, ligands for the endothelial-specific tyrosine kinase Tie2.
J. Biol. Chem.
273
,
18514
-18521.
Wong, A. L., Haroon, Z. A., Werner, S., Dewhirst, M. W., Greenberg, C. S. and Peters, K. G. (
1997
). Tie2 expression and phosphorylation in angiogenic and quiescent adult tissues.
Circ. Res.
81
,
567
-574.