Europe PMC

This website requires cookies, and the limited processing of your personal data in order to function. By using the site you are agreeing to this as outlined in our privacy notice and cookie policy.

Abstract 


Atherogenesis involves activation of NF-kappaB in endothelial cells by fluid shear stress. Because this pathway involves integrins, we investigated the involvement of focal adhesion kinase (FAK). We found that FAK was not required for flow-stimulated translocation of the p65 NF-kappaB subunit to the nucleus but was essential for phosphorylation of p65 on serine 536 and induction of ICAM-1, an NF-kappaB-dependent gene. NF-kappaB activation by TNF-alpha or hydrogen peroxide was FAK independent. Events upstream of NF-kappaB, including integrin activation, Rac activation, reactive oxygen production, and degradation of IkappaB, were FAK independent. FAK therefore regulates NF-kappaB phosphorylation and transcriptional activity in response to flow by a novel mechanism.

Free full text 


Logo of ajpcellLink to Publisher's site
Am J Physiol Cell Physiol. 2009 Oct; 297(4): C814–C822.
Published online 2009 Jul 8. https://doi.org/10.1152/ajpcell.00226.2009
PMCID: PMC2770750
PMID: 19587216

Focal adhesion kinase modulates activation of NF-κB by flow in endothelial cells

Associated Data

Supplementary Materials

Abstract

Atherogenesis involves activation of NF-κB in endothelial cells by fluid shear stress. Because this pathway involves integrins, we investigated the involvement of focal adhesion kinase (FAK). We found that FAK was not required for flow-stimulated translocation of the p65 NF-κB subunit to the nucleus but was essential for phosphorylation of p65 on serine 536 and induction of ICAM-1, an NF-κB-dependent gene. NF-κB activation by TNF-α or hydrogen peroxide was FAK independent. Events upstream of NF-κB, including integrin activation, Rac activation, reactive oxygen production, and degradation of IκB, were FAK independent. FAK therefore regulates NF-κB phosphorylation and transcriptional activity in response to flow by a novel mechanism.

Keywords: atherosclerosis, fluid shear stress, integrin signaling, mechanotransduction

current models for atherosclerosis suggest that local endothelial dysfunction results in monocyte recruitment and lipid deposition in the vessel intima (29). In addition to systemic risk factors, fluid shear stress from flowing blood plays a crucial role. Atherosclerotic lesions develop preferentially in regions of oscillatory or disturbed flow that have lower mean shear stress, multidirectionality, and flow separation (33).

The proinflammatory transcription factor NF-κB is thought to be a key determinant of atherogenesis (8). NF-κB is activated in atheroprone regions in vivo concomitant with expression of its target genes including ICAM-1, VCAM-1 (20), monocyte chemoattractant protein 1, tissue factor, and PDGF (8). NF-κB is a dimer, usually consisting of a p65 and a p50 subunit, though other combinations also exist. Inactive NF-κB is sequestered in the cytoplasm by IκB proteins (8, 20). The IκB kinase (IKK) complex phosphorylates IκB, leading to its ubiquitination and proteosomal degradation. Free NF-κB dimer then translocates into the nucleus and binds to κB enhancer sites. Phosphorylation, acetylation, and sumoylation regulates the subsequent recruitment of cofactors and other members of the transcriptional machinery required to drive target gene expression.

Integrins have been implicated in activation of NF-κB and several other proinflammatory pathways in response to flow (19). Shear stress induces conversion of integrins to a high-affinity state (i.e., integrin activation), which is followed by binding of these high-affinity integrins to the extracellular matrix beneath the cells to form new integrin-matrix contacts (53). The newly ligated integrins trigger downstream signals, including RhoA, Rac, and Cdc42 activation (5355). These GTPases mediate cell alignment, sterol regulatory element-binding protein (SREBP) cleavage (30), and NF-κB activation (54). Consistent with this model, flow-induced activation of NF-κB is integrin and matrix dependent, occurring in cells on fibronectin (FN) or fibrinogen but not on collagen or laminin (38).

Focal adhesion kinase (FAK), a nonreceptor tyrosine kinase involved in integrin signaling (43, 46), is present in endothelial cells and has been implicated in responses of endothelial cells to fluid shear stress. Complete deletion of FAK in mice is lethal at embryonic (E) day 8.5, and endothelium-specific deletion is lethal after day E11.5. In endothelial cells, flow stimulates FAK recruitment into focal adhesions and increased tyrosine phosphorylation (27). Furthermore, dominant-negative FAK inhibited activation of the sterol regulatory element-binding protein 1 (SREBP-1) by flow (31), and introduction of an antibody against FAK phosphorylated on Y397 decreased endothelial nitric oxide synthetase (eNOS) phosphorylation and vasodilation in vivo (25). Thus, several reports show that FAK is involved in endothelial responses to flow, most likely through the integrin activation pathway described above.

These considerations led us to examine the contribution of FAK to flow-induced inflammatory signaling. Many studies have shown that onset of laminar shear induces transient activation of the same pathways via the same mechanisms as disturbed flow does in a sustained fashion (14, 34, 38, 40, 41). We therefore used onset of shear as a simple model system for understanding the basic mechanisms of cell response to flow. Using endothelial cells isolated from the conditional FAK-knockout mouse, our results reveal a novel and unexpected role for FAK in NF-κB activation.

MATERIAL AND METHODS

Antibodies.

Antibodies used were rabbit anti-phospho-p65 (Ser536), rabbit anti-ERK, rabbit anti-IkBα, rabbit anti-phospho eNOS (Ser1179) and rabbit anti-phospho-ERK (Thr202/Tyr204), and rabbit phospho-JNK (Thr183/Tyr185) (Cell Signaling Technology, Beverly, MA); mouse-anti-Pyk2 (BD Bioscience, San Diego, CA); mouse anti-FAK (NH2-terminal; Upstate Biotechnology, Lake Placid, NY) and mouse anti-Rac (Upstate Biotechnology); rabbit anti-p65, rabbit anti-SREBP, rabbit anti-eNOS, goat anti-ICAM-1, mouse anti-β-catenin, mouse anti-glutathione S-transferase (GST), and secondary horseradish peroxidase (HRP) donkey anti-goat (Santa Cruz Biotechnology, Santa Cruz, CA); mouse anti-paxillin (Zymed); Alexa568-goat anti-mouse, Alexa488-goat anti-mouse- and Alexa488-conjugated anti-rabbit (Molecular Probes); HRP-goat anti-rabbit and goat anti-mouse (Jackson ImmunoResearch Laboratories, West Grove, PA); and mouse anti-tubulin (Developmental Studies Hybridoma Bank, Iowa city, IA).

Mouse aortic endothelial cell isolation.

All animal procedures were approved by the Institutional Animal Care and Use Committee of the University of Virginia. Mouse aortic endothelial cells (MAECs) were isolated from 8-wk-old FAK conditional knockout mice as described previously (49). Cells were initially grown on 0.2% gelatin-coated dishes with l-valine DMEM growth medium (17) containing 15% fetal bovine serum (FBS), 30 μg/ml endothelial cell growth supplement, and 50 μg/ml heparin. Cells expressed VE-cadherin, and >95% of cells showed uptake of 1,1'-dioctadecyl-3,3,3',3'-tetramethyl-indocarbocyanine perchlorate-labeled acetylated LDL (Dil-Ac-LDL).

Following passage 2, cells were maintained in Cambrex EBM-2 endothelial cell medium containing 10% FBS, 10 U/ml penicillin and 10 μg/ml streptomycin (Invitrogen), and 2 mM l- glutamine (Invitrogen). Cells were immortalized with polyoma middle T retrovirus (produced in the BOSC23 packaging cell line) (4). At passage 7, cells were infected with adenovirus carrying Cre-recombinase-green fluorescent protein (GFP) to delete the FAK gene (2). Transient transfections used Amaxa nucleofection (Amaxa Biotechnology, according to manufacturer's instructions) with 4 μg of DNA encoding wild-type FAK (FAK-WT) or green fluorescent protein pmaxGFP (Amaxa) 48 h before use.

Small interfering RNA experiments.

Bovine aortic endothelial cells (BAECs) were cultured in DMEM supplemented with 10% fetal bovine serum, 10 U/ml penicillin and 10 μg/ml streptomycin, and 2 mM l-glutamine (Invitrogen). Cells were transfected with small interfering RNA (siRNA) oligonucleotides against FAK using Lipofectamine 2000 (Invitrogen) according to the manufacturer's instructions. The sequence was 5′-GCUAGUGACGUAUGGAUGU-3′ as described previously (52).

Flow experiments.

MAECs were starved for 2 h in EBM-2 medium containing 0.2% FBS before use. BAECS were starved in DMEM with 0.2% serum before use. The parallel plate laminar flow chamber system (15) was maintained at 37°C and perfused with 7% CO2-93% air. Glass slides (38 × 75 mm, Corning) were coated with 10 μg/ml FN in PBS (for 2 h) and blocked with 0.2% BSA in PBS for 30 min. Cells were plated overnight to reach confluence the next day. Glass slides were kept under static conditions or exposed to 24 dyn/cm2 in the same starvation medium.

Immunocytochemistry.

Cells on coverslips were washed with cold PBS, fixed for 30 min in 2% formaldehyde in PBS, permeabilized with 0.1% Triton-PBS, and blocked with Tris-buffered saline (TBS) containing 0.05% Tween 20 (TBST) and 10% goat serum. Primary antibodies were incubated overnight at 4°C in TBST. Antibodies were anti-FAK (1:500), anti-p65 (1:200), anti-paxillin (1:500), anti-β-catenin (1:500), or anti-phospho-S536 p65 (1:100). Cells were then washed, incubated with secondary Alexa-conjugated antibody (1:500) for 1 h, washed again, and mounted in Fluorochrome G (Southern Biotech). Confocal images were acquired using a Zeiss LSM 510 with ×63 oil or ×40 lenses. Images were processed with Adobe Photoshop 7 and Image J software.

Immunoblotting.

Cells were lysed in 120 μl 5× Laemmli's buffer, sonicated, and boiled for 5 min. Proteins were separated by SDS-PAGE gel electrophoresis and transferred to polyvinylidene difluoride membranes. Membranes were blocked with 5% nonfat milk powder in TBST for 1 h and incubated with primary antibody at 4°C overnight. Blots were washed with TBST and incubated with HRP-conjugated secondary antibodies (Jackson ImmunoResearch) for 1 h at room temperature. Membranes were then treated with ECL reagents (Amersham Biosciences) and exposed to film. Resultant protein bands were scanned and quantified using Image J software. Membranes were reprobed after stripping with ReBlot Strong solution (Chemicon).

Integrin activation assay.

To measure the level of high-affinity, unoccupied integrins on the cell surface, cells were sheared for 5 min and incubated with 10 μg/ml GST-FNIII9–11 in PBS containing 1 mM Mg2+ for 30 min at 37°C as described previously (39). Cells were washed three times with PBS + 1 mM Mg2+ to remove unbound protein, lysed, and analyzed by immunoblotting for GST. Values were normalized for total protein loading (determined by tubulin Western blot).

RNA extraction and quantitative RT-PCR.

Total RNA was isolated using TRIzol reagent (Invitrogen) according to the manufacturer's instructions. cDNA was synthesized by reverse transcription (RT-PCR) using the iScript cDNA Synthesis Kit (Bio-Rad). To quantify mRNA expression, cDNA was analyzed by real-time RT-PCR (iCycler, Bio-Rad) using SYBR green. The following primers were used: ICAM-1 primer 1, 5′-CTGGCTGTCACAGAACAGGA-3′; ICAM-1 primer 2, 5′-AAAGTAGGTGGGGAGGTGCT-3′; 18S primer 1, 5′-CGGCTACCACATCCAAGGAA-3′; and 18S primer 2, 5′-AGCTGGAATTACCGCGGC-3′. ICAM-1 mRNA levels were quantified using a standard curve and were normalized to 18S.

Rac activity assays.

Cells were sheared for the indicated times, then extracted in lysis buffer [50 mM Tris, 500 mM NaCl, 10 mM MgCl2, 0.1% SDS, 1% Triton, 0.5% deoxycholate, and protease inhibitor mix (Sigma)]. Lysates from four slides were combined for each point. GST-p21 binding domain (20 μg) (10) and glutathione-Sepharose beads (20 μl, Amersham Biosciences) were added and incubated for 30 min at 4°C under rotation. Beads were washed with 50 mM Tris, 150 mM NaCl, 10 mM MgCl2, 1% Triton, and protease inhibitor mix. Bound Rac was eluted by adding 5× Laemmli's buffer and boiling for 5 min. Samples were analyzed by immunoblotting, and bound Rac was normalized for total Rac in the whole cell lysates.

Reactive oxygen species assay using dichlorofluorescein-diacetate.

Cells were incubated in media containing 10 μg/ml dichlorofluorescein-diacetate (DCFH-DA) (57) (Molecular Probes) for 30 min, then exposed to flow at 24 dyn/cm2 for 30 min in the continuous presence of DCFH-DA. Cells were rinsed with PBS and scraped into 1% Triton in PBS containing 3 μl/ml butylated hydroxyl toluene to prevent further oxidation. Samples were analyzed in a 96-well plate reader with excitation at 488 nm and emission at 528 nm. Values were normalized to total protein levels measured by Lowry protein assay (32).

Statistical analysis.

Statistical comparisons between groups were performed using Student's t-test with the indicated significance level (P = 0.05 or P = 0.01). For data in Figs. 2 and and55 and supplemental data, analysis by ANOVA with a post hoc test yielded similar results.

An external file that holds a picture, illustration, etc.
Object name is zh00090960340002.jpg

FAK regulation of NF-κB. Control FAKfl/fl (fl/fl) cells and FAK−/− cells were exposed to 24 dyn/cm2 laminar shear stress (LSS) for 45 min. A: cells were fixed and stained for p65, and >100 cells per condition in each experiment were scored for nuclear translocation. Scale bars = 50 μm. Values are means ± SE (n = 3). B: cells were exposed to flow for the indicated times and p65 Ser536 phosphorylation was analyzed. Results were quantified and normalized to total p65 levels. Values are means ± SE (n = 6). *P < 0.05, **P < 0.01. C: cells with or without 45 min of flow were fixed and stained for phospho-Ser536 p65. Images are representative of 3 independent experiments.

An external file that holds a picture, illustration, etc.
Object name is zh00090960340005.jpg

NF-κB activation by integrins. A: MAECs were treated with TS2/16 for 30 min and p65 nuclear localization was scored (>100 cells/condition in each experiment). Values are means ± SE (n = 4). **P < 0.01. B: FAKfl/fl and FAK−/− MAECs were treated with TS2/16 (20 μg/ml) and p65 phosphorylation was then determined as in Fig 2B. Values are means ± SE (n = 3). *P < 0.05, **P < 0.01. C: WT cells plated on fibronectin were pretreated with blocking antifibronectin antibody 16G3 or nonblocking antibody 11E5. Cells were then exposed to shear and extracted and p65 Ser536 phosphorylation was analyzed (n = 3).

RESULTS

FAK−/− endothelial cells.

Aortic endothelial cells were derived from 8-wk-old mice in which the second exon of the kinase domain of the FAK gene was flanked by LoxP sites (FAKfl/fl mice), immortalized with Polyoma middle t-antigen, and infected with Cre-recombinase adenovirus to yield FAK−/− mouse aortic endothelial cells. These FAK−/− endothelial cells contained no detectable FAK by Western blotting (Fig. 1A). The related kinase Pyk2 and the p65 NF-κB subunit were unchanged. Both focal adhesions and cell-cell junctions, structures implicated in shear stress sensing, were similar in FAK−/− cells (Fig. 1B). We conclude that infection with Cre deletes the FAK gene without perturbing Pyk2, NF-κB, or adhesive structures.

An external file that holds a picture, illustration, etc.
Object name is zh00090960340001.jpg

Characterization of focal adhesion kinase-knockout (FAK/) mouse aortic endothelial cells (MAECs). A: aortic endothelial cells from homozygous conditional FAK floxed/floxed mice (FAKfl/fl or fl/fl cells). Lysates were analyzed by Western blotting for FAK, NF-κB p65 subunit, Pyk2, or tubulin as a loading control. B: cells were fixed and stained for paxillin and β-catenin. Scale bars represent 20 μm (×63) or 50 μm (×40).

SREBP cleavage, ERK, JNK, and eNOS activation by flow.

eNOS (12, 51) and the MAP kinases ERK and JNK (42, 51) are activated by fluid shear stress. We found that phosphorylation of these enzymes in FAKfl/fl (control) and FAK−/− (knockout) cells was similar (supplemental Fig. 1, AC; supplemental data for this article can be found online at the American Journal of Physiology-Cell Physiology website). Thus, deletion of FAK does not affect a number of previously characterized cellular responses to shear.

Onset of flow also triggers activation of SREBP, which occurs through cleavage of its precursor protein into a 68-kDa mature form that drives target gene expression (30, 31). Activation of this pathway by flow is suppressed by dominant-negative Y397F FAK (31). Accumulation of mature SREBP protein after shear stimulation was abolished in FAK−/− cells (supplemental Fig. 1D), confirming that this process is FAK dependent.

NF-κB activation in FAK−/− endothelial cells.

NF-κB is activated by flow (8). In addition to phosphorylation and degradation of the inhibitory IkBα subunit, which allows NF-κB to translocate into the nucleus, both subunits of NF-κB undergo posttranslational modification (6, 44), including phosphorylation of p65 Ser536 in the carboxyl-terminal transactivation domain, which is important for expression of target genes (6). FAK expression did not affect nuclear translocation of p65 after flow, although FAK−/− cells showed somewhat less nuclear p65 in the absence of flow (Fig. 2A). By contrast, loss of FAK slightly increased the baseline Ser536 phosphorylation but abolished the response to flow (Fig. 2B).

To test the dependence on FAK, FAK−/− cells were transfected with a vector for WT FAK or for GFP. Cells were then sheared for 30 min. Although expression of the exogenous FAK was substantially lower than endogenous protein in control FAKfl/fl cells (Fig. 3A), there was a significant rescue of Ser536 phosphorylation after flow (Fig. 3B). Although the rescue is incomplete, this result is very likely due to the low expression or transfection efficiency. These data indicate that FAK is dispensable for NF-κB nuclear translocation but is required for Ser536 phosphorylation.

An external file that holds a picture, illustration, etc.
Object name is zh00090960340003.jpg

Confirming the FAK requirement for p65 phosphorylation. A: FAK−/− cells were transiently transfected with wild-type (WT) FAK or green fluorescent protein (GFP). FAK was analyzed by Western blotting using the FAKfl/fl cells for comparison. B: cells expressing FAK or GFP were exposed to shear for 30 min and p65 Ser536 phosphorylation was assayed. Results were quantified and normalized to total p65 levels. Values are means ± SE (n = 4). Statistical analysis compared GFP-transfected cells to FAK-transfected cells after 30 min of shear. *P < 0.05. C: bovine aortic endothelial cells (BAECs) were transfected with small interfering RNA (siRNA) oligonucleotides against FAK or with control oligonucleotides against luciferase (GL2). At 72 h, cells were exposed to flow for 30 min and harvested and p65 phosphorylation was analyzed. Inset: Western blots from a representative experiment. Graph values are means ± SE (n = 6). **P < 0.02. Results show that shear induces p65 phosphorylation in control but not FAK siRNA cells.

As a second way to confirm these results, we asked whether loss of FAK affects NF-κB in BAECs, a commonly used model for shear studies. Transfection of siRNA into BAECs resulted in depletion of FAK by >90% when compared with the luciferase (GL2) controls (Fig. 3C). Flow was found to stimulate p65 phosphorylation on Ser536 FAK in control knockdown cells as in control murine cells. By contrast, FAK siRNA-treated cells again showed an increase in the baseline Ser536 phosphorylation, but the response to flow was completely inhibited. Thus, effects of FAK deletion in MAECs and knockdown in BAECs are similar.

ICAM-1 expression.

We next examined expression of ICAM-1, a gene that is induced by flow through activation of NF-κB (11, 23, 38). ICAM-1 is a transmembrane receptor that mediates leukocyte adhesion to the endothelium (20) and is upregulated in areas of atheroprone flow in vivo (36). Induction of both ICAM-1 mRNA (Fig. 4A) and protein (Fig. 4B) were strongly dependent on FAK. Thus, flow-induced expression of a well-characterized NF-κB target gene also requires FAK.

An external file that holds a picture, illustration, etc.
Object name is zh00090960340004.jpg

ICAM-1 mRNA levels and protein expression. A: mRNA was isolated from cells with or without flow for 1.5 h. ICAM-1 expression was quantified by RT-PCR and normalized to 18S. Values are means ± SE (n = 3). **P < 0.01. B: ICAM-1 protein levels were assayed in cells exposed to flow for 1.5 or 3 h by Western blotting (n = 4). Bands were quantified and normalized to ERK. Values are means ± SE (n = 4). *P < 0.05.

The role of integrins.

Previous studies showed that shear stimulates NF-κB by triggering integrin activation followed by binding to matrix proteins (54). To test whether integrin activation was sufficient to trigger the NF-κB pathway, we treated endothelial cells with the activating β1-integrin antibody TS2/16, which induces conversion of integrins to the high-affinity conformation (5, 53). TS2/16 triggered nuclear translocation of p65 equally well in FAKfl/fl and FAK−/− endothelial cells (Fig. 5A); however, p65 Ser536 phosphorylation strictly required FAK (Fig. 5B). FAK is therefore required for NF-κB activation downstream of integrin activation and ligation.

To confirm the requirement for integrins in the flow system, we used a blocking antibody to FN that inhibits binding of integrins α5β1 and αvβ3 to the central cell-binding domain (35). Previous studies showed that short-term treatment of endothelial cells on FN with anti-FN antibody blocks free FN molecules and prevents new integrin binding, however, on this time scale does not disrupt existing adhesions (53, 54). Pretreatment of cells plated on FN with the blocking antibody 16G3 abolished NF-κB activation, whereas the nonblocking antibody 11E5 that binds a different region of FN had no effect (Fig. 5C). Taken together, these data demonstrate that activation of NF-κB by flow occurs though integrins, which requires FAK for induction of NF-κB p536 phosphorylation, whereas nuclear translocation is FAK independent.

Activation of NF-κB by TNF-α.

To determine whether FAK is required for NF-κB activation in response to all stimuli, cells were treated with TNF-α. Binding of TNF-α to its receptor 1 (TNFR-1) recruits a signaling complex consisting of TRADD, TRAF2, and RIP to the cytoplasmic domain of TNFR-1, which triggers the downstream activation of the IKK complex. A recent study using FAK−/− fibroblasts suggested a role for FAK in NF-κB activation by TNF-α (16). However, FAKfl/fl and FAK−/− MAECs treated with 10 ng/ml TNF-α showed no difference in phosphorylation of Ser536 (Fig. 6A) or ICAM-1 expression (Fig. 6B). These results argue that the requirement for FAK in NF-κB function in endothelial cells is not a general requirement.

An external file that holds a picture, illustration, etc.
Object name is zh00090960340006.jpg

NF-κB activation by TNF-α. A: cells were treated with 10 ng/ml TNF-α and p65 phosphorylation was measured. Bands were quantified and normalized to total p65 levels. Values are means ± SE (n = 3). B: cells were treated with 10 ng/ml TNF-α and ICAM-1 protein was determined. Bands were quantified and normalized to ERK. Values are means ± SE (n = 4). *P < 0.05.

Analysis of the upstream pathway.

Onset of flow activates NF-κB via a pathway that involves integrin activation, followed by their binding to matrix proteins, followed by activation of Rac (54), which leads to production of reactive oxygen species (ROS), activation of IKK, and degradation of IκB (3, 26, 47, 50). To determine at what point FAK is required, we first measured integrin activation using a FN fragment that binds α5β1- and αvβ3-integrins in an activation-dependent manner (13, 39). When cells were sheared for 5 min (the time at which integrin activation is maximal), the increase was independent of FAK expression (Fig. 7A). Increased Rac activity after flow was also independent of FAK expression (Fig. 7B). When cellular ROS was assayed, shear stimulated ROS in FAK−/− cells slightly better than controls (Fig. 8A). Though the reason for the increase is unclear, FAK is clearly not required for ROS production. To test whether FAK determines the sensitivity of NF-κB phosphorylation to ROS, cells were treated with 100 μM H2O2 for 30 min. Both cell types phosphorylated p65 to a similar extent (Fig. 8B). Thus, the ability of ROS to activate NF-κB is also independent of FAK. Degradation of IκBα in response to flow occurred similarly in both cell lines (Fig. 8C). This result suggests that IKK activation is independent of FAK, consistent with the movement of p65 to the nucleus (Fig. 2).

An external file that holds a picture, illustration, etc.
Object name is zh00090960340007.jpg

Integrin and Rac activation by shear. A: sheared cells (24 dyn/cm2, LSS) were incubated with 10 μg/ml glutathione S-transferase-fibronectin III9–11 (GST-FN) for 30 min, and bound GST-FN was analyzed by Western blotting for GST. Tubulin was used as a loading control. Values are means ± SE (n = 4). B: cells exposed to shear were analyzed for active Rac using pull-down assays. Values are means ± SE (n = 3) normalized to total Rac in whole cell lysates.

An external file that holds a picture, illustration, etc.
Object name is zh00090960340008.jpg

Reactive oxygen species (ROS) and IκB. A: ROS generation was measured by dichlorofluorescein-diacetate (DCFH-DA) fluorescence as described in materials and methods. DCFH-DA fluorescence after shear (24 dyn/cm2) was normalized to total protein. Values are means ± SE (n = 3). B: cells were treated with 100 μM H2O2 and p65 Ser536 phosphorylation was assayed. Bands were quantified and normalized to total p65. Values are means ± SE (n = 3). C: cells were exposed to shear for the indicated times and IκB was analyzed by Western blotting. Bands were quantified and normalized to total p65. Values are means ± SE (n = 3).

DISCUSSION

This study used onset of fluid shear stress as a simple model system to explore the role of FAK in NF-κB activation by flow. In vivo, various types of disturbed shear are associated with NF-κB activation and atherogenesis (9, 18). In vitro, NF-κB as well as other events associated with atherosclerosis are stimulated transiently by the onset of shear but are downregulated at later times (34, 40). By contrast, the same events are stimulated by oscillatory or other disturbed shear profiles in a sustained manner. We and others have proposed that disturbed shear induces continual stimulation of the integrin pathway, whereas cells in laminar flow adapt and downregulate these signals (7, 19). Thus, onset of shear provides a convenient means to investigate biologically relevant mechanisms.

Our studies used endothelial cells from mice with a floxed FAK gene that, on treatment with Cre recombinase, resulted in loss of detectable FAK. No change in the FAK homolog Pyk2 was observed, which removes a potential complicating factor. Two previous studies where FAK was deleted in endothelial cells obtained opposite results on this point, with one study reporting no change in Pyk2 levels (48) and a second reporting increased Pyk2 expression after Cre-mediated recombination (58). The reasons for these differences are unknown, but our results clearly demonstrate no change in Pyk2 under our conditions.

Previous studies showed that FAK phosphorylation increased in response to flow and that dominant-negative FAK inhibited activation of MAP kinases and SREBP (28, 31). Our data confirmed the role of FAK in SREBP cleavage, although we did not observe attenuation of ERK activation. However, Fujiwara and coworkers (42) showed that ERK activation by shear is quite rapid, similar to our data, and is a direct consequence of platelet endothelial cell adhesion molecule signaling. The rapidity of ERK activation is not consistent with the integrin pathway, which requires integrin activation and binding such that signals peak at 15–30 min (53, 56). However, it would not be surprising if dominant-negative FAK constructs have effects on MAP kinases that are not entirely specific.

Our major finding is that although FAK is not required for IκB degradation and nuclear translocation of NF-κB, it is essential for p65 phosphorylation and expression of ICAM-1. FAK is not required for any component of the known upstream events including integrin activation, Rac activation, or production of ROS. Thus, flow-induced NF-κB liberation and transactivation are at least partially regulated by distinct signaling pathways. By contrast, responses to TNF-α or hydrogen peroxide were FAK-independent.

IKK kinases are believed to phosphorylate p65 Ser536 (45), although IKK family members other than the classic IKKα or β may carry out this phosphorylation (1). Ser536 is within the transactivation domain, and its phosphorylation has been linked to transcriptional activity in several (22, 24, 59) but not all systems (37). How FAK controls p65 phosphorylation is currently unknown, and at present it is difficult to even propose viable hypotheses. However, it seems noteworthy that activated IKK localizes to focal adhesions in endothelial cells exposed to shear (38). This result supports the integrin dependence of this pathway and further suggests that localized FAK-dependent signaling events within the adhesion sites may explain the distinct effects of FAK on activation of NF-κB by shear or integrin vs. TNF-α or H2O2. Taken together, these data indicate an unexpected and potentially important role for FAK in the activation of endothelium in response to flow. This pathway may therefore contribute to the initiation of atherosclerosis in regions of disturbed flow.

GRANTS

This work was supported by United States Public Health Service Grant RO1 HL75092 to M. A. Schwartz. A. W. Orr and C. Hahn were supported by National Institutes of Health Training Grant 5T32 HL-7284. T. Petzold was supported by a Fulbright Foundation scholarship and American Heart student fellowship AHA 0525532U.

Supplementary Material

[Supplemental Figures]

ACKNOWLEDGMENTS

We thank David Bolick for help with endothelial cell isolation and Konstandinos Moissoglu for helpful discussions.

REFERENCES

1. Adli M, Baldwin AS. IKK-i/IKKepsilon controls constitutive, cancer cell-associated NF-kappaB activity via regulation of Ser-536 p65/RelA phosphorylation. J Biol Chem 281: 26976–26984, 2006 [Abstract] [Google Scholar]
2. Beggs HE, Schahin-Reed D, Zang K, Goebbels S, Nave KA, Gorski J, Jones KR, Sretavan D, Reichardt LF. FAK deficiency in cells contributing to the basal lamina results in cortical abnormalities resembling congenital muscular dystrophies. Neuron 40: 501–514, 2003 [Europe PMC free article] [Abstract] [Google Scholar]
3. Bhullar IS, Li YS, Miao H, Zandi E, Kim M, Shyy JY, Chien S. Fluid shear stress activation of IkappaB kinase is integrin-dependent. J Biol Chem 273: 30544–30549, 1998 [Abstract] [Google Scholar]
4. Carmeliet P, Lampugnani MG, Moons L, Breviario F, Compernolle V, Bono F, Balconi G, Spagnuolo R, Oostuyse B, Dewerchin M, Zanetti A, Angellilo A, Mattot V, Nuyens D, Lutgens E, Clotman F, de Ruiter MC, Gittenberger-de Groot A, Poelmann R, Lupu F, Herbert JM, Collen D, Dejana E. Targeted deficiency or cytosolic truncation of the VE-cadherin gene in mice impairs VEGF-mediated endothelial survival and angiogenesis. Cell 98: 147–157, 1999 [Abstract] [Google Scholar]
5. Chan BMC, Hemler ME. Multiple functional forms of the integrin VLA-2 can be derived from a single α2 cDNA clone: interconversion of forms induced by an anti-β1 antibody. J Cell Biol 120: 537–543, 1993 [Europe PMC free article] [Abstract] [Google Scholar]
6. Chen LF, Greene WC. Shaping the nuclear action of NF-kappaB. Nat Rev Mol Cell Biol 5: 392–401, 2004 [Abstract] [Google Scholar]
7. Chien S. Effects of disturbed flow on endothelial cells. Ann Biomed Eng 36: 554–562, 2008 [Abstract] [Google Scholar]
8. Collins T, Cybulsky MI. NF-kappaB: pivotal mediator or innocent bystander in atherogenesis? J Clin Invest 107: 255–264, 2001 [Europe PMC free article] [Abstract] [Google Scholar]
9. Cunningham KS, Gotlieb AI. The role of shear stress in the pathogenesis of atherosclerosis. Lab Invest 85: 9–23, 2005 [Abstract] [Google Scholar]
10. Del Pozo MA, Price LS, Alderson NB, Ren XD, Schwartz MA. Adhesion to the extracellular matrix regulates the coupling of the small GTPase Rac to its effector PAK. EMBO J 19: 2008–2014, 2000 [Europe PMC free article] [Abstract] [Google Scholar]
11. Denk A, Goebeler M, Schmid S, Berberich I, Ritz O, Lindemann D, Ludwig S, Wirth T. Activation of NF-kappa B via the Ikappa B kinase complex is both essential and sufficient for proinflammatory gene expression in primary endothelial cells. J Biol Chem 276: 28451–28458, 2001 [Abstract] [Google Scholar]
12. Dusserre N, L'Heureux N, Bell KS, Stevens HY, Yeh J, Otte LA, Loufrani L, Frangos JA. PECAM-1 interacts with nitric oxide synthase in human endothelial cells: implication for flow-induced nitric oxide synthase activation. Arterioscler Thromb Vasc Biol 24: 1796–1802, 2004 [Abstract] [Google Scholar]
13. Faull RJ, Kovach NL, Harlan JM, Ginsberg MH. Affinity modulation of integrin α5β1: regulation of the functional response by soluble fibronectin. J Cell Biol 121: 155–162, 1993 [Europe PMC free article] [Abstract] [Google Scholar]
14. Feaver RE, Hastings NE, Pryor A, Blackman BR. GRP78 upregulation by atheroprone shear stress via p38-, alpha2beta1-dependent mechanism in endothelial cells. Arterioscler Thromb Vasc Biol 28: 1534–1541, 2008 [Europe PMC free article] [Abstract] [Google Scholar]
15. Frangos JA, Eskin SG, McIntire LV, Ives CL. Flow effects on prostacyclin production by cultured human endothelial cells. Science 227: 1477–1479, 1985 [Abstract] [Google Scholar]
16. Funakoshi-Tago M, Sonoda Y, Tanaka S, Hashimoto K, Tago K, Tominaga S, Kasahara T. Tumor necrosis factor-induced nuclear factor kappaB activation is impaired in focal adhesion kinase-deficient fibroblasts. J Biol Chem 278: 29359–29365, 2003 [Abstract] [Google Scholar]
17. Gilbert SF, Migeon BR. D-valine as a selective agent for normal human and rodent epithelial cells in culture. Cell 5: 11–17, 1975 [Abstract] [Google Scholar]
18. Gimbrone MA, Jr, Topper JN, Nagel T, Anderson KR, Garcia-Cardena G. Endothelial dysfunction, hemodynamic forces, and atherogenesis. Ann NY Acad Sci 902: 230–239; discussion 239–240, 2000 [Abstract] [Google Scholar]
19. Hahn C, Schwartz MA. The role of cellular adaptation to mechanical forces in atherosclerosis. Arterioscler Thromb Vasc Biol 28: 2101–2107, 2008 [Europe PMC free article] [Abstract] [Google Scholar]
20. Hajra L, Evans AI, Chen M, Hyduk SJ, Collins T, Cybulsky MI. The NF-kappa B signal transduction pathway in aortic endothelial cells is primed for activation in regions predisposed to atherosclerotic lesion formation. Proc Natl Acad Sci USA 97: 9052–9057, 2000 [Europe PMC free article] [Abstract] [Google Scholar]
22. Hu J, Nakano H, Sakurai H, Colburn NH. Insufficient p65 phosphorylation at S536 specifically contributes to the lack of NF-kappaB activation and transformation in resistant JB6 cells. Carcinogenesis 25: 1991–2003, 2004 [Abstract] [Google Scholar]
23. Imberti B, Morigi M, Zoja C, Angioletti S, Abbate M, Remuzzi A, Remuzzi G. Shear stress-induced cytoskeleton rearrangement mediates NF-kappaB-dependent endothelial expression of ICAM-1. Microvasc Res 60: 182–188, 2000 [Abstract] [Google Scholar]
24. Jiang X, Takahashi N, Matsui N, Tetsuka T, Okamoto T. The NF-kappa B activation in lymphotoxin beta receptor signaling depends on the phosphorylation of p65 at serine 536. J Biol Chem 278: 919–926, 2003 [Abstract] [Google Scholar]
25. Koshida R, Rocic P, Saito S, Kiyooka T, Zhang C, Chilian WM. Role of focal adhesion kinase in flow-induced dilation of coronary arterioles. Arterioscler Thromb Vasc Biol 25: 2548–2553, 2005 [Abstract] [Google Scholar]
26. Li Q, Engelhardt JF. Interleukin-1beta induction of NFkappaB is partially regulated by H2O2-mediated activation of NFkappaB-inducing kinase. J Biol Chem 281: 1495–1505, 2006 [Abstract] [Google Scholar]
27. Li S, Kim M, Hu YL, Jalali S, Schlaepfer DD, Hunter T, Chien S, Shyy JY. Fluid shear stress activation of focal adhesion kinase. Linking to mitogen-activated protein kinases. J Biol Chem 272: 30455–30462, 1997 [Abstract] [Google Scholar]
28. Li S, Butler P, Wang Y, Hu Y, Han DC, Usami S, Guan JL, Chien S. The role of the dynamics of focal adhesion kinase in the mechanotaxis of endothelial cells. Proc Natl Acad Sci USA 99: 3546–3551, 2002 [Europe PMC free article] [Abstract] [Google Scholar]
29. Libby P. Inflammation in atherosclerosis. Nature 420: 868–874, 2002 [Abstract] [Google Scholar]
30. Lin T, Zeng L, Liu Y, DeFea K, Schwartz MA, Chien S, Shyy JY. Rho-ROCK-LIMK-cofilin pathway regulates shear stress activation of sterol regulatory element binding proteins. Circ Res 92: 1296–1304, 2003 [Abstract] [Google Scholar]
31. Liu Y, Chen BP, Lu M, Zhu Y, Stemerman MB, Chien S, Shyy JY. Shear stress activation of SREBP1 in endothelial cells is mediated by integrins. Arterioscler Thromb Vasc Biol 22: 76–81, 2002 [Abstract] [Google Scholar]
32. Lowry OH, Rosebrough NJ, Farr AL, Randall RJ. Protein measurement with the Folin phenol reagent. J Biol Chem 193: 265–275, 1951 [Abstract] [Google Scholar]
33. Malek AM, Alper SL, Izumo S. Hemodynamic shear stress and its role in atherosclerosis. JAMA 282: 2035–2042, 1999 [Abstract] [Google Scholar]
34. Mohan S, Mohan N, Sprague EA. Differential activation of NF-κB in human aortic endothelial cells conditioned to specific flow environments. Am J Physiol Cell Physiol 273: C572–C578, 1997 [Abstract] [Google Scholar]
35. Nagai T, Yamakawa N, Aota S, Yamada SS, Akiyama SK, Olden K, Yamada KM. Monoclonal antibody characterization of two distant sites required for function of the central cell-binding domain of fibronectin in cell adhesion, cell migration, and matrix assembly. J Cell Biol 114: 1295–1305, 1991 [Europe PMC free article] [Abstract] [Google Scholar]
36. Nakashima Y, Raines EW, Plump AS, Breslow JL, Ross R. Upregulation of VCAM-1 and ICAM-1 at atherosclerosis-prone sites on the endothelium in the ApoE-deficient mouse. Arterioscler Thromb Vasc Biol 18: 842–851, 1998 [Abstract] [Google Scholar]
37. Okazaki T, Sakon S, Sasazuki T, Sakurai H, Doi T, Yagita H, Okumura K, Nakano H. Phosphorylation of serine 276 is essential for p65 NF-kappaB subunit-dependent cellular responses. Biochem Biophys Res Commun 300: 807–812, 2003 [Abstract] [Google Scholar]
38. Orr AW, Sanders JM, Bevard M, Coleman E, Sarembock IJ, Schwartz MA. The subendothelial extracellular matrix modulates NF-kappaB activation by flow: a potential role in atherosclerosis. J Cell Biol 169: 191–202, 2005 [Europe PMC free article] [Abstract] [Google Scholar]
39. Orr AW, Ginsberg MH, Shattil SJ, Deckmyn H, Schwartz MA. Matrix-specific suppression of integrin activation in shear stress signaling. Mol Biol Cell 17: 4686–4697, 2006 [Europe PMC free article] [Abstract] [Google Scholar]
40. Orr AW, Stockton R, Simmers MB, Sanders JM, Sarembock IJ, Blackman BR, Schwartz MA. Matrix-specific p21-activated kinase activation regulates vascular permeability in atherogenesis. J Cell Biol 176: 719–727, 2007 [Europe PMC free article] [Abstract] [Google Scholar]
41. Orr AW, Hahn C, Blackman BR, Schwartz MA. p21-activated kinase signaling regulates oxidant-dependent NF-kappa B activation by flow. Circ Res 103: 671–679, 2008 [Europe PMC free article] [Abstract] [Google Scholar]
42. 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 158: 773–785, 2002 [Europe PMC free article] [Abstract] [Google Scholar]
43. Parsons JT, Martin KH, Slack JK, Taylor JM, Weed SA. Focal adhesion kinase: a regulator of focal adhesion dynamics and cell movement. Oncogene 19: 5606–5613, 2000 [Abstract] [Google Scholar]
44. Perkins ND. Post-translational modifications regulating the activity and function of the nuclear factor kappa B pathway. Oncogene 25: 6717–6730, 2006 [Abstract] [Google Scholar]
45. Sakurai H, Chiba H, Miyoshi H, Sugita T, Toriumi W. IkappaB kinases phosphorylate NF-kappaB p65 subunit on serine 536 in the transactivation domain. J Biol Chem 274: 30353–30356, 1999 [Abstract] [Google Scholar]
46. Schlaepfer DD, Hauck CR, Sieg DJ. Signaling through focal adhesion kinase. Prog Biophys Mol Biol 71: 435–478, 1999 [Abstract] [Google Scholar]
47. Schreck R, Rieber P, Baeuerle PA. Reactive oxygen intermediates as apparently widely used messengers in the activation of the NF-kappa B transcription factor and HIV-1. EMBO J 10: 2247–2258, 1991 [Europe PMC free article] [Abstract] [Google Scholar]
48. Shen TL, Park AY, Alcaraz A, Peng X, Jang I, Koni P, Flavell RA, Gu H, Guan JL. Conditional knockout of focal adhesion kinase in endothelial cells reveals its role in angiogenesis and vascular development in late embryogenesis. J Cell Biol 169: 941–952, 2005 [Europe PMC free article] [Abstract] [Google Scholar]
49. Srinivasan S, Hatley ME, Reilly KB, Danziger EC, Hedrick CC. Modulation of PPARalpha expression and inflammatory interleukin-6 production by chronic glucose increases monocyte/endothelial adhesion. Arterioscler Thromb Vasc Biol 24: 851–857, 2004 [Abstract] [Google Scholar]
50. Sulciner DJ, Irani K, Yu ZX, Ferrans VJ, Goldschmidt-Clermont P, Finkel T. rac1 regulates a cytokine-stimulated, redox-dependent pathway necessary for NF-kappaB activation. Mol Cell Biol 16: 7115–7121, 1996 [Europe PMC free article] [Abstract] [Google Scholar]
51. Tai LK, Zheng Q, Pan S, Jin ZG, Berk BC. Flow activates ERK1/2 and endothelial nitric oxide synthase via a pathway involving PECAM1, SHP2, and Tie2. J Biol Chem 280: 29620–29624, 2005 [Abstract] [Google Scholar]
52. Tilghman RW, Slack-Davis JK, Sergina N, Martin KH, Iwanicki M, Hershey ED, Beggs HE, Reichardt LF, Parsons JT. Focal adhesion kinase is required for the spatial organization of the leading edge in migrating cells. J Cell Sci 118: 2613–2623, 2005 [Abstract] [Google Scholar]
53. Tzima E, del Pozo MA, Shattil SJ, Chien S, Schwartz MA. Activation of integrins in endothelial cells by fluid shear stress mediates Rho-dependent cytoskeletal alignment. EMBO J 20: 4639–4647, 2001 [Europe PMC free article] [Abstract] [Google Scholar]
54. Tzima E, Del Pozo MA, Kiosses WB, Mohamed SA, Li S, Chien S, Schwartz MA. Activation of Rac1 by shear stress in endothelial cells mediates both cytoskeletal reorganization and effects on gene expression. EMBO J 21: 6791–6800, 2002 [Europe PMC free article] [Abstract] [Google Scholar]
55. Tzima E, Kiosses WB, del Pozo MA, Schwartz MA. Localized cdc42 activation, detected using a novel assay, mediates microtubule organizing center positioning in endothelial cells in response to fluid shear stress. J Biol Chem 278: 31020–31023, 2003 [Abstract] [Google Scholar]
56. Tzima E, Irani-Tehrani M, Kiosses WB, Dejana E, Schultz DA, Engelhardt B, Cao G, DeLisser H, Schwartz MA. A mechanosensory complex that mediates the endothelial cell response to fluid shear stress. Nature 437: 426–431, 2005 [Abstract] [Google Scholar]
57. Wang H, Joseph JA. Quantifying cellular oxidative stress by dichlorofluorescein assay using microplate reader. Free Radic Biol Med 27: 612–616, 1999 [Abstract] [Google Scholar]
58. Weis SM, Lim ST, Lutu-Fuga KM, Barnes LA, Chen XL, Gothert JR, Shen TL, Guan JL, Schlaepfer DD, Cheresh DA. Compensatory role for Pyk2 during angiogenesis in adult mice lacking endothelial cell FAK. J Cell Biol 181: 43–50, 2008 [Europe PMC free article] [Abstract] [Google Scholar]
59. Yang F, Tang E, Guan K, Wang CY. IKK beta plays an essential role in the phosphorylation of RelA/p65 on serine 536 induced by lipopolysaccharide. J Immunol 170: 5630–5635, 2003 [Abstract] [Google Scholar]

Articles from American Journal of Physiology - Cell Physiology are provided here courtesy of American Physiological Society

Citations & impact 


Impact metrics

Jump to Citations
Jump to Data

Citations of article over time

Smart citations by scite.ai
Smart citations by scite.ai include citation statements extracted from the full text of the citing article. The number of the statements may be higher than the number of citations provided by EuropePMC if one paper cites another multiple times or lower if scite has not yet processed some of the citing articles.
Explore citation contexts and check if this article has been supported or disputed.
https://scite.ai/reports/10.1152/ajpcell.00226.2009

Supporting
Mentioning
Contrasting
6
75
1

Article citations


Go to all (48) article citations

Data 


Data behind the article

This data has been text mined from the article, or deposited into data resources.

Similar Articles 


To arrive at the top five similar articles we use a word-weighted algorithm to compare words from the Title and Abstract of each citation.


Funding 


Funders who supported this work.

NHLBI NIH HHS (2)