当前位置: 首页 > 医学版 > 期刊论文 > 内科学 > 循环研究杂志 > 2005年 > 第12期 > 正文
编号:11256034
Gab1, SHP2, and Protein Kinase A Are Crucial for the Activation of the Endothelial NO Synthase by Fluid Shear Stress
     the Vascular Signalling Group, Institut fe Kardiovaskulre Physiologie (M.D., A.E.L., A.M., B.F., R.B., I.F.), Johann Wolfgang Goethe-Universitt, Frankfurt am Main, Germany

    Institut National de la Santee et de la Recherche Meedicale (INSERM) (C.M.B.), Unit 541, Hpital Lariboisieere, Paris, France

    Department of Physiology (B.C., A.H.), University of Tennessee Health Sciences Center, Memphis, Tenn.

    Abstract

    Fluid shear stress enhances NO production in endothelial cells by a mechanism involving the activation of the phosphatidylinositol 3-kinase and the phosphorylation of the endothelial NO synthase (eNOS). We investigated the role of the scaffolding protein Gab1 and the tyrosine phosphatase SHP2 in this signal transduction cascade in cultured and native endothelial cells. Fluid shear stress elicited the phosphorylation and activation of Akt and eNOS as well as the tyrosine phosphorylation of Gab1 and its association with the p85 subunit of phosphatidylinositol 3-kinase and SHP2. Overexpression of a Gab1 mutant lacking the pleckstrin homology domain abrogated the shear stresseCinduced phosphorylation of Akt but failed to affect the phosphorylation or activity of eNOS. The latter response, however, was sensitive to a protein kinase A (PKA) inhibitor. Mutation of Gab1 Tyr627 to phenylalanine (YF-Gab1) to prevent the binding of SHP2 completely prevented the shear stresseCinduced phosphorylation of eNOS, leaving the Akt response intact. A dominant-negative SHP2 mutant prevented the activation of PKA and phosphorylation of eNOS without affecting that of Akt. Moreover, shear stress elicited the formation of a signalosome complex including eNOS, Gab1, SHP2 and the catalytic subunit of PKA. In isolated murine carotid arteries, flow-induced vasodilatation was prevented by a PKA inhibitor as well as by overexpression of either the YF-Gab1 or the dominant-negative SHP2 mutant. Thus, the shear stresseCinduced activation of eNOS depends on Gab1 and SHP2, which, in turn, regulate the phosphorylation and activity of eNOS by a PKA-dependent but Akt-independent mechanism.

    Key Words: Akt blood flow endothelial nitric oxide synthase mechanotransduction protein kinase A

    Introduction

    Although the endothelial NO synthase (eNOS) was initially described as a Ca2+-dependent enzyme, it is now clear that the changes in the phosphorylation of several serine and threonine (and possibly also tyrosine) residues regulate NO production in response to Ca2+-elevating agonists as well as to hemodynamic stimuli, such as cyclic stretch and fluid shear stress (for reviews, see Fleming and Busse1 and Boo and Jo2). Most is known about the role played by Ser1177, which is situated in the reductase domain of the enzyme, and Thr495, which is situated in the calmodulin-binding domain, in the regulation of NO production. The phosphorylation of these sites appears to play a reciprocal role in the regulation of eNOS activity as Ser1177 becomes phosphorylated in response to endothelial cell activation, whereas Thr495 is constitutively phosphorylated but dephosphorylated on stimulation as a consequence of the activation of phosphatases.3eC5 The dephosphorylation of Thr495 facilitates the Ca2+-dependent association of calmodulin with eNOS,5 whereas the phosphorylation of eNOS on Ser1177 increases NO output in an apparently Ca2+-independent manner.6,7 As the maintained production of endothelium-derived NO in response to fluid shear stress is a Ca2+-independent process,8,9 it is generally assumed that the phosphorylation of eNOS on Ser1177 plays the predominant role in regulating eNOS activity in response to hemodynamic stimuli.

    The kinases responsible for the phosphorylation of eNOS on Ser1177 vary with the stimuli applied and can be attributed not only to the activation of the Ca2+/calmodulin-dependent kinase II in bradykinin-stimulated endothelial cells5 but to Akt in cells stimulated with vascular endothelial growth factor7 or hepatocyte growth factor (HGF)10 and to Akt6,7 and, probably more importantly, to protein kinase A (PKA)11 in cells exposed to fluid shear stress. Relatively little is known about the molecular events that determine eNOS activity in response to fluid shear stress. It is generally assumed that fluid shear stress simultaneously initiates signaling within caveolae, focal adhesion sites and intercellular junctions (for review, see Davies et al12). We and others have recently reported that platelet-endothelial cell adhesion molecule-1 modulates endothelial cell activation in response to shear stress,13,14 most probably by virtue of its ability to interact with signaling molecules such as the tyrosine phosphatase SHP2 (Src Homology 2-containing Protein tyrosine phosphatase 2) and the scaffolding protein Gab1 (Grb2-associated binder 1) (for recent reviews, see Newman and Newman15 and Ilan and Madri16). Indeed, Gab-1 translocates from the cytoplasm to endothelial cell junctions in response to flow17 and can associate with the p85 subunit of the phosphatidylinositol 3-kinase (PI3-K),18,19 which is essential for the shear stresseCinduced phosphorylation of eNOS.6

    In the present study, we determined the role of Gab1 and SHP2 in the shear stresseCinduced phosphorylation and activation of Akt and eNOS in cultured and native endothelial cells. We assessed the effect of a Gab1 mutant lacking the pleckstrin homology domain (PHGab1) and compared its effects with those of a mutant in which Tyr627 was substituted with phenylalanine (YF-Gab1) to interfere with its interaction with SHP2,19,20 as well as with a dominant-negative SHP2 mutant.

    Materials and Methods

    Materials

    The phospho-specific antibodies recognizing eNOS Ser1177, phospho-Akt (Ser473), and Gab1 were from Cell Signaling (Munich, Germany). The antibody against SHP2 was from BD Biosciences (Heidelberg, Germany), the anti-p85 PI3-K antibody was from Upstate (Lake Placid, NY), the anti-eNOS and antieCphospho-tyrosine antibodies were from Transduction Laboratories (Heidelberg, Germany), and the antibody against the catalytic subunit of PKA was from Santa Cruz Biotechnology (Heidelberg, Germany). HGF was purchased from R&D Biosystems (Abingdon, UK), the PKA inhibitor Rp-3',5'-cyclic monophosphorothioate sodium salt (RpAMPS) was from Alexis Biochemicals (Greberg, Germany), and H89 was from Calbiochem (Bad Soden, Germany). All the other substances were from Sigma (Munich, Germany).

    Cell Culture and Shear Stress

    Porcine aortic endothelial cells were isolated and cultured as described previously.21 Because of the loss of several signaling components with time in culture, endothelial cells were used only after 1 passage. Confluent endothelial cells were washed twice with M-199 medium containing 0.1% BSA and after serum starvation for 4 hours were subjected to shear stress of 12 dynes/cm2 in a cone-plate viscosimeter as described22 or to HGF (50 ng/mL). In some experiments, the intracellular concentration of cGMP was measured in presence of the phosphodiesterase inhibitor (isobutyl methylxanthine, 0.1 mmol/L) using a specific radioimmunoassay (Amersham, Freiburg, Germany) as described22 or PKA activity was assessed using a commercially available kit (Upstate) according to the instructions of the manufacturer.

    Immunoprecipitation and Immunoblotting

    Cells were lysed in a Triton X-100 lysis buffer, the immunoprecipitation of Gab1 was performed as previously described,23 and proteins in the cell supernatants or immunoprecipitates were subjected to SDS-PAGE.5 Proteins were detected using their respective antibodies and visualized by enhanced chemiluminescence using a commercially available kit (Amersham, Germany).

    Preparation and Expression of Adenoviral Vectors

    Details regarding the generation of the recombinant adenoviruses expressing WT-Gab1, a Gab1 mutant lacking the N-terminal pleckstrin homology domain (PH-Gab1), and Y627F-Gab1 (YF-Gab1) as well as the dominant-negative SHP2 mutant (DSH2) are provided in the online data supplement available at http://circres.ahajournals.org.

    For efficient expression of the recombinant proteins, confluent primary cultures of porcine aortic endothelial cells were infected with the respective adenovirus (3x106 pfu/mL). Four hours after infection in serum-free medium, cells were washed extensively in PBS and fresh medium containing 4% FCS was added and cells were incubated for a further 44 hours.

    Adenoviral Infection of Mouse Carotid Arteries

    Male C57BL/6 mice, 6 to 9 weeks of age (Charles River, Sulzfeld, Germany) were anesthetized with isoflurane. The investigation conforms to the Guide for the Care and Use of Laboratory Animals published by the US National Institutes of Health (NIH Publication No. 85-23, revised 1996). The carotid arteries were perfused with saline solution and partially freed of connective tissue but maintained in situ. Virus solution (3x106 pfu in 20 e蘈) was introduced into the lumen before the arteries were ligated, excised, and placed in culture medium (MCDB 131 containing penicillin, 50 IU/mL streptomycin, 50 e/mL, and 2% FCS) in a standard CO2 incubator (37°C) for 4 hours. Thereafter, the ligatures were removed and the vessels washed extensively to remove the virus and then maintained overnight in culture medium. See the online data supplement for additional experimental details.

    Flow-Induced Vasodilatation

    Freshly isolated or adenovirus-treated mouse carotid arteries were cannulated and perfused in a video-monitored perfusion system (Living Systems Instruments, Burlington, Vt) as described.24 Transluminal pressure was set at 80 mm Hg and vessels were constricted with phenylephrine (10 to 100 nmol/L) to approximately 80% of the baseline diameter. Once a stable contraction was achieved, flow was increased stepwise from 10 to 800 e蘈/min. Endothelium-dependent relaxation to acetylcholine (1 eol/L) was assessed at the end of each experiment, and only vessels that responded with a dilatation of 60% or more of the phenylephrine-induced contraction were included in the study. Segments were then recovered, incubated with lysis buffer, and subjected to Western blotting to determine the effectiveness of the adenoviral infection.

    Statistics

    Data are expressed as the mean±SEM, and statistical evaluation was performed using Student’s t test for unpaired data and 1-way ANOVA or ANOVA for repeated measures followed by Dunnett’s post hoc test where appropriate. Values of P<0.05 were considered statistically significant.

    Results

    Effect of Shear Stress on the Tyrosine Phosphorylation of Gab1

    A weak tyrosine phosphorylation of Gab1 was detected in confluent cultures of porcine aortic endothelial cells maintained under static conditions. However, exposure to fluid shear stress (12 dynes/cm2) induced the rapid and marked tyrosine phosphorylation of Gab1 (Figure 1A). The tyrosine phosphorylation of Gab1 was evident as early as 2 minutes after stimulation and peaked after 30 minutes. Thereafter, tyrosine phosphorylation of Gab1 decreased slightly but remained elevated above control levels as long as shear stress was applied, ie, up to 24 hours. Phosphorylation of Gab1 was accompanied by its association with the protein tyrosine phosphatase, SHP2, and the p85 subunit of the PI3-K (Figure 1B).

    Effect of a Gab1 Mutant Lacking the PHGab1 on the Phosphorylation of Akt and eNOS in Endothelial Cells

    Because exposure to fluid shear stress stimulated the association of Gab1 with the PI3-K, and a Gab1 PH domain decoy is reported to suppress the activity of a constitutively active Akt,25 we assessed the consequences of the overexpression of the PHGab1 mutant on the phosphorylation of Akt and eNOS in endothelial cells stimulated by either fluid shear stress or HGF, another stimulus for PI3-K and Akt activation.

    Under static conditions the phosphorylation of Akt on Ser473 was low. Both fluid shear stress (12 dynes/cm2) and HGF (50 ng/mL) elicited a pronounced increase in the phosphorylation of Akt in cells infected with a control virus (Figure 2A and 2B). However, only HGF was able to elicit the phosphorylation of Akt in cells expressing the PHGab1 mutant (Figure 2B).

    Because PHGab1 blunted the shear stresseCinduced activation of Akt, we next determined its effect on the phosphorylation of eNOS. The phosphorylation of eNOS on Ser1177 was low in cells maintained under static conditions. In cells treated with a control virus, the phosphorylation of eNOS Ser1177 rapidly increased following the application of shear stress (Figure 2C) or the addition of HGF (Figure 2D). Overexpression of the PHGab1 mutant failed to affect either the shear stresseC or the HGF-induced phosphorylation of eNOS (Figure 2C and 2D).

    Effect of the PHGab1 Mutant on the Activation of eNOS

    The PHGab1 mutant attenuated basal NO production in unstimulated endothelial cells, whereas the overexpression of wild-type Gab1 increased basal NO production (supplementary Figure II). In cells treated with the control virus, shear stress elicited a 3.1±0.6-fold increase in cGMP levels. Despite the complete inhibition of the shear stresseCinduced activation of Akt, a similar increase (3.0±0.5-fold) in cGMP was detected in cells infected with the PHGab1 mutant (P<0.05, n=4). Consistent with its lack of effect on the HGF-induced phosphorylation of Akt and eNOS, PHGab1 did not affect the ability of HGF to increase endothelial NO production (supplementary Figure II).

    As PKA can be activated by fluid shear stress and can also phosphorylate eNOS on Ser1177, we determined the effects of a PKA inhibitor on the shear stresseCinduced phosphorylation of eNOS on Ser1177. As reported above, the overexpression of PHGab1 failed to affect the shear stresseCinduced phosphorylation of eNOS; however, the pretreatment of these cells with RpAMPS (10 eol/L) prevented eNOS Ser1177 phosphorylation in cells expressing PHGab1 (Figure 3A). These data indicate that although the PHGab1 mutant completely prevented the shear stresseCinduced activation of Akt, the phosphorylation of eNOS on Ser1177 remained intact because of the activation of PKA. The shear stresseCinduced increase in the phosphorylation of eNOS Ser1177 was not observed in uninfected cells pretreated with the PKA inhibitors RpAMPS or H89 (10 eol/L), whereas the ability of shear stress to induce the phosphorylation of Akt remained intact (Figure 3B and supplementary Figure III). The PKA inhibitors also prevented the shear stresseCinduced increase in endothelial GMP levels (Figure 3C).

    In contrast to cells infected with the PHGab1 mutant, the downregulation of Gab1 using an siRNA approach abrogated the shear stresseCinduced phosphorylation of both Akt and eNOS (supplementary Figure IV).

    Effect of YF-Gab1 on the Shear StresseCInduced Activation of Akt and eNOS

    Because we detected an increased association of Gab1 with SHP2 in response to shear stress (see Figure 1B), we next determined whether the association of these 2 proteins affects the shear stresseCinduced phosphorylation of eNOS. We reasoned that if the interaction between Gab1 and SHP2 is a prerequisite for the shear stresseCinduced activation of eNOS activation, the expression of a Gab1 mutant that is incapable of interacting with SHP2, ie, YF-Gab1,20 may prevent the shear stresseCinduced activation of eNOS.

    The YF-Gab1 mutant failed to affect the HGF-induced phosphorylation of either Akt or eNOS (data not shown). However, exposure of endothelial cells overexpressing YF-Gab1 to shear stress prevented the phosphorylation of eNOS (Figure 4A and 4B), without affecting that of Akt (Figure 4A and 4C). Thus, although Gab1 appears to be necessary for the shear stresseCinduced activation of Akt through its N-terminal pleckstrin homology domain, its role in the shear stresseCinduced phosphorylation of eNOS is independent of Akt and dependent on its interaction with a protein, such as SHP2, that interacts with Gab1 at Tyr627.

    Effect of a DSH2 on Responses to Shear Stress

    The SHP2 mutant used expresses the tandem SH2 domains of SHP2 but lacks the catalytic domain and thus competes with endogenous SHP2 to act as a dominant-negative mutant. Infection of endothelial cells with the DSH2 viruses resulted in a significant overexpression of the protein without affecting endogenous levels of SHP2 (Figure 5A). Subsequent exposure of these cells to shear stress failed to induce the phosphorylation of eNOS, whereas the phosphorylation of Akt was unaffected (Figure 5). The shear stresseCinduced increase in cGMP was also attenuated in cells expressing the DSH2 protein. Levels of cGMP increased in response to shear stress by 2.8±0.2-fold (P<0.005, n=8) in cells infected with a control virus, whereas cGMP levels were only 91±14% (n=5, P=0.55) of basal levels in DHS2-expressing cells exposed to fluid shear stress.

    Effect of Shear Stress on the Activity of PKA and Its Association With the eNOS Signalosome

    As the PKA inhibitor RpAMPS, like the dominant-negative SHP2 mutant, prevented the shear stresseCinduced phosphorylation of eNOS and the subsequent increase in endothelial cell cGMP levels, we determined whether shear stress affected PKA activity and whether or not SHP2 was implicated in this process. The stimulation of cultured endothelial cells with fluid shear stress elicited a significant increase in PKA activity, an effect that was attenuated in cells expressing the DSH2 protein (Figure 6A). In parallel experiments, we assessed the association between the catalytic subunit of PKA and SHP2. In cells maintained under static conditions, we failed to detect the association of the 2 proteins, whereas an association was regularly observed in cells exposed to fluid shear stress for 30 minutes (Figure 6B). Although there was no apparent association in cells maintained under static conditions, endothelial cell stimulation with shear stress elicited the time-dependent formation of a PKA protein complex that also included Gab1 and eNOS (Figure 7A). Moreover, in parallel experiments SHP2 was coimmunoprecipitated with eNOS and the association of the 2 proteins increased in response to cell stimulation (Figure 7B).

    Flow-Mediated Responses in Carotid Arteries

    In isolated phenylephrine-constricted murine carotid arteries, acetylcholine (1 eol/L) resulted in vasodilatation of approximately 80% (82±11%, n=4), whereas increasing flow resulted in dilatation of up to 60% (Figure 8A). The PKA inhibitor RpAMPS (10 eol/L) failed to affect the response to acetylcholine (80±4%, n=4, P=0.87 versus the solvent-treated group) but markedly blunted flow-induced vasodilatation (Figure 8A).

    To determine the role of Gab1 and SHP2 on endothelium-dependent vasodilatation in response to acetylcholine and increased flow, we infected mouse coronary arteries with the corresponding recombinant adenoviruses and monitored flow-induced vasodilatation. Acetylcholine-induced vasodilatation was not significantly different in vessels treated with the green fluorescent protein (GFP), PHGab1, YF-Gab1, or DSH2 viruses; vasodilatation to acetylcholine was 87±5%, 74±4%, 78±4%, and 83±5%, respectively (n=11 to 15, P=0.36), indicating that the infection procedure did not compromise endothelial cell function. The efficiency of infection was monitored using GFP and confirmed by assessing the expression of individual mutant proteins by Western blot analysis at the end of each experiment (Figure 8B).

    Baseline diameter was not significantly different in arteries expressing GFP, PHGab1, YF-Gab1, or DSH2 (501±7, 481±15, 486±8 and 502±9 e, respectively, n=11 to 15, P=0.39). Increasing flow elicited a stepwise vasodilatation that was not significantly different in control and GFP-expressing carotid arteries (compare Figure 8A and 8B), and although flow-induced vasodilatation was slightly attenuated in arteries expressing the PH Gab1 mutant, this effect was not significant (P=0.16, n=11). However, flow-induced vasodilatation was significantly attenuated in arteries expressing either DSH2 or the YF-Gab1 mutant (Figure 8B).

    Discussion

    The results of the present study demonstrate that the shear stresseC and the HGF-induced activation of Akt are mediated by signaling pathways differentially dependent on Gab1. Indeed, whereas the pleckstrin homology domain of Gab1 seems to be crucial for the shear stresseCinduced phosphorylation of Akt, it is completely dispensable for the activation of the kinase by HGF. More importantly, inhibition of the shear stresseCinduced activation of Akt was without consequence on the phosphorylation and activation of eNOS, which was unchanged because of the activation of PKA. The shear stresseCinduced phosphorylation and activation of eNOS was, however, causally linked with the Gab1-dependent activation of the tyrosine phosphatase SHP2 inasmuch as a Gab1 mutant (YF-Gab1) to which SHP2 cannot bind, as well as a dominant-negative SHP2 mutant abrogated the shear stresseCinduced activation of eNOS in cultured as well as in native endothelial cells.

    Gab1 is an adapter protein that belongs to the insulin receptor substrate-1 family and is a mammalian homologue of the Drosophila protein DOS (Daughter Of Sevenless), which is involved in multiple signaling events mediated by cytokine and tyrosine kinase receptors, including that of the HGF receptor c-Met.26 HGF has previously been reported to enhance the phosphorylation of eNOS on Ser1177 and increase endothelial NO production.10 However, although interfering with the function of Gab1 is reported to attenuate the activation of the PI3-K and Akt in several different cell types in response to growth factor stimulation,27 we observed no effect of either PHGab1 or the YF-Gab1 mutant on the activation of Akt or eNOS in HGF-stimulated endothelial cells. The latter observations cannot rule out a role for Gab1 in the HGF-induced activation of PI3-K and Akt in endothelial cells as the binding of Gab1 to the c-Met receptor is thought to be dependent on its Met-binding sequence,28 which remained intact in both of the Gab1 mutants assessed in the present study.

    There does, however, appear to be a role for Gab1 in regulating the shear stresseCinduced activation of Akt as the PHGab1 mutant abrogated the shear stresseCinduced phosphorylation of Akt. This indicates that the translocation of Gab1 to the plasma membrane plays an important role in the regulation of Akt by fluid shear stress. Indeed, a second Gab1 mutant (YF-Gab1), which should be able to translocate normally within endothelial cells but is unable to bind SHP2, did not prevent the shear stresseCinduced phosphorylation of Akt.

    Fluid shear stress elicits the phosphorylation of eNOS on Ser1177 and although we and others initially implicated Akt in this process,6 the results of the present investigation revealed that a kinase other than Akt must be responsible for the shear stresseCinduced phosphorylation and activation of eNOS. This conclusion is based on the observations that PH Gab1 failed to affect the shear stresseCinduced phosphorylation of eNOS, although the activation of Akt was abrogated, and that the YF-Gab1 mutant abolished the phosphorylation of eNOS while leaving the shear stresseCinduced activation of Akt intact. During the preparation of this manuscript, Jin et al29 reported that Gab1 regulates the phosphorylation and activation of eNOS via Akt as the downregulation of Gab1 (using a siRNA approach) and a Gab1 mutant that was unable to bind PI3-K attenuated both processes. Although the downregulation of Gab1 also prevented the shear stresseCinduced phosphorylation of Akt and eNOS in our experiments, studies aimed at determining the consequences of the specific mutation of Gab1 indicate that Akt is not responsible for the phosphorylation of eNOS. Despite the apparent contradiction, the report by Jin et al29 is not in direct conflict with the results shown here. Indeed, preventing the shear stresseCinduced activation of PI3-K would be expected to affect the activation of both Akt and PKA,2 and the global downregulation of Gab1 would be expected to affect both the activation of Akt, which we propose requires the translocation of Gab1 to the plasma membrane, as well as the association of SHP2 with Gab1, which is dependent on the phosphorylation of Tyr627 and which does affect the shear stresseCinduced phosphorylation of eNOS.

    Several events in the signaling cascade activated by hemodynamic stimuli in endothelial cells are associated with tyrosine phosphorylation and/or dephosphorylation (for review, see Shyy and Chien30). The fact that both the YF-Gab1 mutant, which cannot bind SHP2,20 and the dominant-negative SHP2 mutant prevented the shear stresseCinduced phosphorylation and activation of eNOS, indicate that SHP2 is also an integral part of this signal transduction process. Fluid shear stress is known to attenuate the tumor necrosis factor-eCinduced activation of SHP2 in endothelial cells,31 but, to the best of our knowledge, this is the first report demonstrating the involvement of SHP2 in regulating the shear stresseCinduced activation of eNOS. Little is known about substrates for SHP2 in endothelial cells, but eNOS itself is reported to be tyrosine phosphorylated,22 and SHP2 has been implicated in the regulation of neuronal NOS activity.32 As we were able to detect SHP2 as part of the eNOS signalosome in shear stresseCstimulated cells, it is likely that either eNOS or an eNOS-associated protein is a SHP2 substrate.

    Given that our findings indicate that the phosphorylation and activation of eNOS in response to shear stress is largely independent of Akt, we assessed the role of PKA in response to shear stress. We observed that a PKA inhibitor abrogated the phosphorylation of eNOS on Ser1177, prevented the shear stresseCinduced accumulation of cGMP, and significantly attenuated the flow-induced vasodilatation of mouse carotid arteries. These data confirm and extend previously published reports on the importance of PKA in mediating eNOS activation in response to fluid shear stress,11 although it is unclear how the activity of PKA is stimulated under the conditions studied. Our data suggest a link between SHP2 and the activation of PKA inasmuch as shear stress stimulated the association of SHP2 with the catalytic subunit of PKA and the shear stresseCinduced activation of PKA was attenuated in endothelial cells expressing the DSH2 mutant. Because shear stress increases PKA activity without increasing endothelial cAMP levels,33 and PKA, SHP2, Gab1, and eNOS coexist as a signalosome complex in shear stresseCstimulated endothelial cells, it is tempting to suggest that SHP2 can directly regulate PKA activity.

    In conclusion, our data clearly indicate that the adaptor protein Gab1 and the tyrosine phosphatase SHP2 play crucial roles in the activation of eNOS by fluid shear stress in cultured and native endothelial cells. The shear stresseCinduced phosphorylation of Gab1 and the association with SHP2 are essential for the activation of PKA, which is ultimately responsible for the increase in endothelial NO production.

    Acknowledgments

    This study was supported by the Deutsche Forschungsgemeinschaft (SFB 553, B1) (to R.B. and I.F.), US Public Health Service grants HL63886 and HL72902 (to A.H.), and by the European Vascular Genomic Network, a Network of Excellence supported by the European Community’s Sixth Framework Program (contract no. LSHM-CT-2003-503254). We are indebted to Isabel Winter, Mechtild Piepenbrock-Gyamfi, and Tanja-Maria Mareczek for expert technical assistance and to Dr Re筪iger Popp for measuring NO production with the electrode.

    This manuscript was sent to Peter Libby, Consulting Editor, for review by expert referees, editorial decision, and final disposition.

    References

    Fleming I, Busse R. Molecular mechanisms involved in the regulation of the endothelial nitric oxide synthase. Am J Physiol Regul Integr Comp Physiol. 2003; 284: R1eCR12.

    Boo YC, Jo H. Flow-dependent regulation of endothelial nitric oxide synthase: role of protein kinases. Am J Physiol Cell Physiol. 2003; 285: C499eCC508.

    Harris MB, Ju H, Venema VJ, Liang H, Zou R, Michell BJ, Chen Z-P, Kemp BE, Venema RC. Reciprocal phosphorylation and regulation of the endothelial nitric oxide synthase in response to bradykinin stimulation. J Biol Chem. 2001; 19: 16587eC16591.

    Michell BJ, Chen Z, Tiganis T, Stapleton D, Katsis F, Power DA, Sim AT, Kemp BE. Coordinated control of endothelial nitric-oxide synthase phosphorylation by protein kinase C and the cAMP-dependent protein kinase. J Biol Chem. 2001; 276: 17625eC17628.

    Fleming I, Fisslthaler B, Dimmeler S, Kemp BE, Busse R. Phosphorylation of Thr495 regulates Ca2+/calmodulin-dependent endothelial nitric oxide synthase activity. Circ Res. 2001; 88: e68eCe75.

    Dimmeler S, Fleming I, Fisslthaler B, Hermann C, Busse R, Zeiher AM. Activation of nitric oxide synthase in endothelial cells by Akt-dependent phosphorylation. Nature. 1999; 399: 601eC605.

    Fulton D, Gratton J-P, Mccabe TJ, Fontana J, Fujio Y, Walsh K, Franke TF, Papapetropoulos A, Sessa WC. Regulation of endothelium-derived nitric oxide production by the protein kinase Akt. Nature. 1999; 399: 597eC601.

    Kuchan MJ, Frangos JA. Role of calcium and calmodulin in flow-induced nitric oxide production in endothelial cells. Am J Physiol. 1994; 266: C628eCC636.

    Ayajiki K, Kindermann M, Hecker M, Fleming I, Busse R. Intracellular pH and tyrosine phosphorylation but not calcium determine shear stress-induced nitric oxide production in native endothelial cells. Circ Res. 1996; 78: 750eC758.

    Makondo K, Kimura K, Kitamura N, Kitamura T, Yamaji D, Jung BD, Saito M. Hepatocyte growth factor activates endothelial nitric oxide synthase by Ca2+- and phosphoinositide 3-kinase/Akt-dependent phosphorylation in aortic endothelial cells. Biochem J. 2003; 374: 63eC69.

    Boo YC, Sorescu G, Boyd N, Shiojima I, Walsh K, Du J, Jo H. Shear stress stimulates phosphorylation of endothelial nitric-oxide synthase at Ser1179 by Akt-independent mechanisms: role of protein kinase A. J Biol Chem. 2002; 277: 3388eC3396.

    Davies PF, Zilberberg J, Helmke BP. Spatial microstimuli in endothelial mechanosignaling. Circ Res. 2003; 92: 359eC370.

    Fleming I, Fisslthaler B, Dixit M, Busse R. Role of PECAM-1 in the shear-stress-induced activation of Akt and the endothelial nitric oxide synthase (eNOS) in endothelial cells. J Cell Sci. 2005; 118: 4103eC4111.

    Bagi Z, Frangos JA, Yeh JC, White CR, Kaley G, Koller A. PECAM-1 mediates NO-dependent dilation of arterioles to high temporal gradients of shear stress. Arterioscler Thromb Vasc Biol. 2005; 25: 1590eC1595.

    Newman PJ, Newman DK. Signal transduction pathways mediated by PECAM-1: new roles for an old molecule in platelet and vascular cell biology. Arterioscler Thromb Vasc Biol. 2003; 23: 953eC964.

    Ilan N, Madri JA. PECAM-1: old friend, new partners. Curr Opin Cell Biol. 2003; 15: 515eC524.

    Osawa M, Masuda M, Kusano K, Fujiwara K. Evidence for a role of platelet endothelial cell adhesion molecule-1 in endothelial cell mechanosignal transduction:is it a mechanoresponsive molecule J Cell Biol. 2002; 158: 773eC785.

    Nishida K, Hirano T. The role of Gab family scaffolding adapter proteins in the signal transduction of cytokine and growth factor receptors. Cancer Sci. 2003; 94: 1029eC1033.

    Gu H, Neel BG. The "Gab" in signal transduction. Trends Cell Biol. 2003; 13: 122eC130.

    Cunnick JM, Mei L, Doupnik CA, Wu J. Phosphotyrosines 627 and 659 of Gab1 constitute a bisphosphoryl tyrosine-based activation motif (BTAM) conferring binding and activation of SHP2. J Biol Chem. 2001; 276: 24380eC24387.

    Busse R, Lamontagne D. Endothelium-derived bradykinin is responsible for the increase in calcium produced by angiotensin-converting enzyme inhibitors in human endothelial cells. Naunyn Schmiedebergs Arch Pharmacol. 1991; 344: 126eC129.

    Fleming I, Bauersachs J, Fisslthaler B, Busse R. Ca2+-independent activation of the endothelial nitric oxide synthase in response to tyrosine phosphatase inhibitors and fluid shear stress. Circ Res. 1998; 82: 686eC695.

    Dixit M, Zhuang D, Ceacareanu B, Hassid A. Treatment with insulin uncovers the motogenic capacity of nitric oxide in aortic smooth muscle cells. Dependence on Gab1 and Gab1-SHP2 association. Circ Res. 2003; 93: e113eCe123.

    Henrion D, Terzi F, Matrougui K, Duriez M, Boulanger CM, Colucciguyon E, Babinet C, Briand P, Friedlander G, Poitevin P, Levy BI. Impaired flow-induced dilation in mesenteric resistance arteries from mice lacking vimentin. J Clin Invest. 1997; 100: 2909eC2914.

    Ren Y, Wu J. Simultaneous suppression of Erk and Akt/PKB activation by a Gab1 pleckstrin homology (PH) domain decoy. Anticancer Res. 2003; 23: 3231eC3236.

    Weidner KM, Di Cesare S, Sachs M, Brinkmann V, Behrens J, Birchmeier W. Interaction between Gab1 and the c-Met receptor tyrosine kinase is responsible for epithelial morphogenesis. Nature. 1996; 384: 173eC176.

    Lamothe B, Yamada M, Schaeper U, Birchmeier W, Lax I, Schlessinger J. The docking protein Gab1 is an essential component of an indirect mechanism for fibroblast growth factor stimulation of the phosphatidylinositol 3-kinase/Akt antiapoptotic pathway. Mol Cell Biol. 2004; 24: 5657eC5666.

    Lock LS, Frigault MM, Saucier C, Park M. Grb2-independent recruitment of Gab1 requires the C-terminal lobe and structural integrity of the Met receptor kinase domain. J Biol Chem. 2003; 278: 30083eC30090.

    Jin ZG, Wong C, Wu J, Berk BC. Flow shear stress stimulates Gab1 tyrosine phosphorylation to mediate protein kinase B and endothelial nitric oxide synthase activation in endothelial cells. J Biol Chem. 2005; 280: 12305eC12309.

    Shyy JYJ, Chien S. Role of integrins in endothelial mechanosensing of shear stress. Circ Res. 2002; 91: 769eC775.

    Lerner-Marmarosh N, Yoshizumi M, Che W, Surapisitchat J, Kawakatsu H, Akaike M, Ding B, Huang Q, Yan C, Berk BC, Abe J. Inhibition of tumor necrosis factor-a-induced SHP-2 phosphatase activity by shear stress: a mechanism to reduce endothelial inflammation. Arterioscler Thromb Vasc Biol. 2003; 23: 1775eC1781.

    Cordelier P, Esteve JP, Rivard N, Marletta M, Vaysse N, Susini C, Buscail L. The activation of neuronal NO synthase is mediated by G-protein bg subunit and the tyrosine phosphatase SHP-2. FASEB J. 1999; 13: 2037eC2050.

    Malek AM, Greene AL, Izumo S. Regulation of endothelin 1 gene by fluid shear stress is transcriptionally mediated and independent of protein kinase C and cAMP. Proc Natl Acad Sci U S A. 1993; 90: 5999eC6003.(Madhulika Dixit, Annemari)