In this study, we describe that the PDZ protein syntenin-1 is a crucial element for the generation of signaling asymmetry during the cellular response to polarized extracellular cues. We analyze the role of syntenin-1 in the control of asymmetry in two independent models of T cell polarization – the migratory response to chemoattractants and the establishment of cognate interactions between T cells and antigen-presenting cells (APCs). A combination of mutant, biochemical and siRNA approaches demonstrate that syntenin-1 is vital for the generation of polarized actin structures such as the leading edge and the contact zone with APCs. We found that the mechanism by which syntenin-1 controls actin polymerization relies on its mandatory role for activation of the small GTPase Rac. Syntenin-1 controls Rac through a specific association with the myosin phosphatase Rho interacting protein (M-RIP), which occurs in response to phosphorylation of syntenin-1 by Src at Tyr4. Our data indicate the key role of syntenin-1 in the generation of functional asymmetry in T cells and provide a novel mechanistic link between receptor activation and actin polymerization and accumulation in response to extracellular stimulation.

T cell migration and activation in response to antigen are characterized by morphological changes dictated by the subcellular redistribution of a number of molecules, including chemokine receptors, adhesion molecules, signaling effectors and cytoskeletal components (Vicente-Manzanares and Sanchez-Madrid, 2004). During T cell migration, the activation of integrins and chemokine receptors triggers many signaling pathways, including the activation of Rho GTPases, which actively induce actin polymerization and reorganization. This results in the acquisition of a polarized morphology (Vicente-Manzanares and Sanchez-Madrid, 2004) with a leading edge and a trailing edge, which is characterized by a pseudopod-like projection, the uropod (Sanchez-Madrid and Serrador, 2009). Similarly, antigen-driven T cell activation involves the formation of a highly organized asymmetric complex of surface receptors and intracellular signaling molecules at the cell-to-cell contact, known as the immunological synapse (IS) (Dustin, 2009). The formation of the IS involves a rapid reorientation of the T cell cytoskeleton, including the formation of a tight structure of polymerized actin at the interface between the T cell and the antigen-presenting cell (APC), clustering of CD3, and relocalization of the microtubule-organizing center (MTOC) towards the contact area (Vicente-Manzanares and Sanchez-Madrid, 2004). This coordinated polarized reorganization is central to the maturation of the immune synapse and subsequent T cell activation (Martín-Cófreces et al., 2008; Dustin, 2009).

Chemotaxis and activation of T cells are both accompanied by polarization of the plasma membrane receptors, cytoskeletal components and intracellular signaling molecules that sense the extracellular chemotactic gradient (Gomez-Mouton and Manes, 2007; Kolsch et al., 2008). Actin dynamics is mainly controlled by signals from Rho GTPases, in which Rac activation leads to branched actin polymerization that is necessary for protrusion of the leading lamella, whereas myosin-dependent contraction is induced by Rho-A-derived signals (Vicente-Manzanares and Sanchez-Madrid, 2004). During antigen recognition, there is a dynamic balance between F-actin polymerization at the peripheral supramolecular activation cluster (p-SMAC) (Dustin, 2009) and myosin-driven waves of contraction that are necessary for the treadmilling of the F-actin filaments towards the center of the synapse, which powers the coalescence of T cell receptor (TCR) microclusters (Ilani et al., 2009). This fine-tuned actin dynamics is essential for the sustained signaling necessary for complete T cell activation.

Although many signals involved in T cell migration and activation have been described, the molecular machinery that organizes signaling molecules and their architecture in response to activation is not completely understood (Shaw and Filbert, 2009). Adaptor proteins containing PDZ domains (named for their occurrence in postsynaptic density protein, Drosophila Discs large and zonula occludens-1) are spatially segregated during T cell migration and formation of the IS (Ludford-Menting et al., 2005). PDZ domains have emerged as central organizers of protein complexes at the plasma membrane, linking extracellular signals to the cytoskeleton and intracellular signaling pathways.

Syntenin-1 (also known as MDA-9) is a cytosolic PDZ-containing protein that was originally identified as a molecule linking syndecan-mediated signaling to the cytoskeleton, and which has since been found to bind many other membrane receptors (Beekman and Coffer, 2008; Sarkar et al., 2008). Syntenin-1 contains two tandemly arranged PDZ domains preceded by a 122-residue N-terminal fragment of unknown structure (Grootjans et al., 1997). Phosphatidylinositol 4,5-bisphosphate [PtdIns(4,5)P2 or PIP2] contributes to the targeting of syntenin-1 to the plasma membrane through its PDZ domains, thereby regulating its availability to associate with membrane receptors (Zimmermann et al., 2005). Syntenin-1 also associates with the tyrosine kinase Src and the cytosolic actin-binding protein Merlin (Kang et al., 2003; Boukerche et al., 2008). Syntenin-1 plays an important role in cellular processes such as intracellular trafficking and cell surface targeting, transcription factor activation, cancer metastasis, neuronal synaptic transmission and axonal outgrowth (Sarkar et al., 2008).

We have studied the role of syntenin-1 in T cell chemotaxis and in the formation of immune conjugates. Our data reveal an unexpected role for this adaptor protein in controlling the targeting of the small GTPase Rac-1 to the plasma membrane. Although the subcellular localization of syntenin-1 is regulated by its PDZ domains, we have defined an ITIM-like motif in syntenin-1 that can be phosphorylated by Src and regulates syntenin-1 binding to myosin phosphatase Rho interacting protein (M-RIP), thereby regulating actin polymerization induced by extracellular cues. Syntenin-1 thus acts as a limiting factor in the signaling cascade linking Src to Rac-1 activation, which controls the asymmetric activation of this pathway.

Fig. 1.

Syntenin-1 is polarized in migrating lymphocytes. (A) PBLs were incubated with or without SDF-1α (100 ng/ml, 5 minutes), fixed, permeabilized and co-stained with anti-syntenin-1 antiserum and anti-ICAM-3. Preparations were analyzed by fluorescence confocal microscopy. Arrow indicates the leading edge (LE) versus the uropod (U). Scale bars: 5 μm. (B) T lymphoblastic HSB2 cells were transfected with GFP–syntenin-1 and monitored at 30 second intervals after stimulation with 100 ng/ml SDF-1α. Representative pseudocolored maximal projections are shown. Scale bar: 5 μm. Arrows indicate the uropod (U) and leading edge (LE).

Fig. 1.

Syntenin-1 is polarized in migrating lymphocytes. (A) PBLs were incubated with or without SDF-1α (100 ng/ml, 5 minutes), fixed, permeabilized and co-stained with anti-syntenin-1 antiserum and anti-ICAM-3. Preparations were analyzed by fluorescence confocal microscopy. Arrow indicates the leading edge (LE) versus the uropod (U). Scale bars: 5 μm. (B) T lymphoblastic HSB2 cells were transfected with GFP–syntenin-1 and monitored at 30 second intervals after stimulation with 100 ng/ml SDF-1α. Representative pseudocolored maximal projections are shown. Scale bar: 5 μm. Arrows indicate the uropod (U) and leading edge (LE).

Syntenin-1 is polarized in migrating lymphocytes

To address the role of syntenin-1 in regulating T cell polarization, we first studied its subcellular localization in lymphocytes. Endogenous syntenin-1 is distributed evenly in non-stimulated primary peripheral blood lymphocytes (PBLs). Upon stimulation with the chemokine CXCL12/SDF-1α, it polarizes to dense clusters at the uropod, showing partial colocalization with uropod markers such as ICAM-3, and to the leading edge with more diffuse staining (Fig. 1A). In polarized T cell lines, syntenin-1 was also found at the uropod, and upon SDF-1α stimulation, partially relocalized to the leading edge within 30 seconds (supplementary material Fig. S1A). Time-lapse confocal microscopy of GFP–syntenin-1 in constitutively polarized T-lymphoblastic cell lines showed accumulation at the leading edge and uropod, sustained throughout cell migration (Fig. 1B and supplementary material Movie 1). Redistribution of syntenin-1 was highly active at the leading edge, which correlated with protrusive activity. This bipolar distribution was similar to that found with a GFP-tagged pleckstrin homology domain of PLCγ that binds PIP2, but different from that observed with other control GFP proteins such as GFP, farnesylated GFP or a different pleckstrin homology domain, BTK–PH–GFP (supplementary material Fig. S1B).

Syntenin-1 is a limiting factor in chemokine-induced actin polymerization and T cell chemotaxis

We next examined the role of this adaptor protein in T cell chemotaxis by suppressing its expression in T cells with two independent siRNA oligonucleotides (supplementary material Fig. S1C). Efficiency of endogenous protein silencing was assessed in every experiment and was greater than 60%. Chemotaxis of primary T lymphoblasts (Fig. 2A) or leukemic CEM T cells (Fig. 2B,C), towards CXCL12/SDF-1α was significantly impaired by both siRNA oligonucleotides. This inhibition could be also observed using a different chemokine such as CCL19/MIP-3β and in basal random migration in the absence of chemotactic stimulus (Fig. 2C). The chemokine SDF-1α induced a rapid increase in polymerized actin in primary T lymphoblasts (Fig. 2D). Knockdown of syntenin-1 almost completely abrogated the SDF-1α-induced actin polymerization response, as determined by flow cytometry in both T cells and primary T lymphoblasts (Fig. 2D,E). The magnitude and progression of actin polymerization induced by the chemokine differs in leukemic cell lines compared with primary PBLs, because primary PBLs display very low basal levels of F-actin.

Fig. 2.

Syntenin-1 regulates chemokine-induced actin polymerization and T cell chemotaxis. (A) Primary T lymphoblasts were transfected with two independent siRNA oligonucleotides targeted against syntenin-1 or control oligonucleotide and allowed to migrate towards SDF-1α (100 ng/ml). Data are the means ± s.d. of three independent experiments. (B) T cells transfected with two independent siRNAs to knockdown syntenin-1 or control oligonucleotide were allowed to migrate in Transwell inserts towards SDF-1α (100 ng/ml). Data are the means ± s.d. of three independent experiments. (C) Control and syntenin-1-depleted cells were allowed to migrate in Transwell inserts towards SDF-1α (100 ng/ml) or MIP-3β (1 μg/ml). Data are means ± s.e.m. of four independent experiments. (D) Cells treated with SDF-1α (100 ng/ml) were fixed at the indicated times, permeabilized, stained with phalloidin Alexa Fluor 488, and analyzed by flow cytometry. Data are the means ± s.e.m. of five independent experiments. (E) Primary T lymphoblasts were transfected with two independent siRNA oligonucleotides targeted against syntenin-1 or control oligonucleotide and treated with SDF-1α (100 ng/ml), fixed at the indicated times, permeabilized, stained with phalloidin Alexa Fluor 488, and analyzed by flow cytometry (mean ± s.d. of two independent experiments). (F) Transwell assays of migration of T cells transfected with GFP, GFP–syntenin-1 or HA–syntenin-1 towards SDF-1α (100 ng/ml). Data are means ± s.d. of three independent experiments. (G) Cells transfected with GFP or GFP–syntenin-1 were treated with SDF-1α (100 ng/ml), fixed at the indicated times, permeabilized, stained with phalloidin Alexa Fluor 647, and analyzed by flow cytometry. Data are means ± s.d. of two independent experiments. *P<0.05.

Fig. 2.

Syntenin-1 regulates chemokine-induced actin polymerization and T cell chemotaxis. (A) Primary T lymphoblasts were transfected with two independent siRNA oligonucleotides targeted against syntenin-1 or control oligonucleotide and allowed to migrate towards SDF-1α (100 ng/ml). Data are the means ± s.d. of three independent experiments. (B) T cells transfected with two independent siRNAs to knockdown syntenin-1 or control oligonucleotide were allowed to migrate in Transwell inserts towards SDF-1α (100 ng/ml). Data are the means ± s.d. of three independent experiments. (C) Control and syntenin-1-depleted cells were allowed to migrate in Transwell inserts towards SDF-1α (100 ng/ml) or MIP-3β (1 μg/ml). Data are means ± s.e.m. of four independent experiments. (D) Cells treated with SDF-1α (100 ng/ml) were fixed at the indicated times, permeabilized, stained with phalloidin Alexa Fluor 488, and analyzed by flow cytometry. Data are the means ± s.e.m. of five independent experiments. (E) Primary T lymphoblasts were transfected with two independent siRNA oligonucleotides targeted against syntenin-1 or control oligonucleotide and treated with SDF-1α (100 ng/ml), fixed at the indicated times, permeabilized, stained with phalloidin Alexa Fluor 488, and analyzed by flow cytometry (mean ± s.d. of two independent experiments). (F) Transwell assays of migration of T cells transfected with GFP, GFP–syntenin-1 or HA–syntenin-1 towards SDF-1α (100 ng/ml). Data are means ± s.d. of three independent experiments. (G) Cells transfected with GFP or GFP–syntenin-1 were treated with SDF-1α (100 ng/ml), fixed at the indicated times, permeabilized, stained with phalloidin Alexa Fluor 647, and analyzed by flow cytometry. Data are means ± s.d. of two independent experiments. *P<0.05.

Moreover, overexpression of GFP- or HA-tagged syntenin-1 significantly increased chemotaxis (Fig. 2F) and enhanced chemokine-induced actin polymerization (Fig. 2G), suggesting that syntenin-1 acts as a limiting factor in chemokine-induced signaling and migration.

Syntenin-1 regulates activation and translocation of Rac to the plasma membrane

The small Rho GTPase Rac is essential for chemokine-induced actin polymerization at the leading edge (Vicente-Manzanares et al., 2005), and is therefore a candidate syntenin-1 effector. SDF-1α stimulation of control oligonucleotide-transfected cells triggered an increase in GTP-loaded Rac that was abrogated in syntenin-1-depleted cells (Fig. 3A). Confocal analysis in primary T lymphoblasts detected a diffuse staining for Rac-1, which, upon SDF-1α stimulation, became polarized and colocalized with endogenous syntenin-1 at the leading edge (Fig. 3B,C). By contrast, SDF-1α stimulation of syntenin-1-depleted cells did not relocate Rac-1 to the leading edge (Fig. 3B,C). In cell fractionation experiments, translocation of Rac-1 to the membrane fraction was impaired by stimulation with SDF-1α after knockdown of syntenin-1 (Fig. 3D).

Syntenin-1 is a Src family kinase substrate that mediates Src-dependent activation of Rac-1

Rac activity lies downstream of Src activation in several cellular contexts, including cell migration and chemotaxis (Ouyang et al., 2008; Sai et al., 2008), and direct association of syntenin-1 with Src kinase has been recently reported in melanoma cells (Boukerche et al., 2008). Basal tyrosine phosphorylation of syntenin-1 was detected in syntenin-1-immunoprecipitated lysates (Fig. 4A), which was rapidly and transiently increased by stimulation with SDF-1α (peaking between 30 seconds and 1 minute; Fig. 4A). Treatment of T cells with the Src family kinase (SFK) inhibitor PP2, selectively inhibited syntenin-1 phosphorylation (Fig. 4B), indicating that SFKs are required for this process.

Fig. 3.

Knockdown of syntenin-1 inhibits activation of Rac and impairs its relocalization to the leading edge. (A) T cells were transfected with the negative oligonucleotide or siRNA against syntenin-1 and treated with SDF-1α (100 ng/ml) for 2 minutes before lysis. GTP-loaded Rac was pulled down with GST–PAK–CRIB and blot revealed with anti-Rac-1. Content of Rac in total cell lysates is also shown. (B) Syntenin-1-depleted and control primary T lymphoblasts were treated with SDF-1α (100 ng/ml, 5 minutes), fixed, permeabilized and stained for syntenin-1, Rac-1 and F-actin. Summatory projections of the confocal stacks, and merged images are shown. Scale bars: 5 μm. (C) Quantification of the translocation of Rac to the leading edge of primary T lymphoblasts treated and stained as in B. Data correspond to the mean ± s.e.m. of three independent experiments. (D) Western blot analysis of the partition of syntenin-1 and Rac-1 in membrane (m) and cytosolic (c) fractions. Syntenin-1-depleted and control-transfected J77 T-cells were treated with SDF-1α (100 ng/ml) for 2.5 minutes. α-tubulin is shown as a loading and fractionation control. Numbers represent the ratio of m/c of Rac signal normalized with respect to untreated control cells. *P<0.05.

Fig. 3.

Knockdown of syntenin-1 inhibits activation of Rac and impairs its relocalization to the leading edge. (A) T cells were transfected with the negative oligonucleotide or siRNA against syntenin-1 and treated with SDF-1α (100 ng/ml) for 2 minutes before lysis. GTP-loaded Rac was pulled down with GST–PAK–CRIB and blot revealed with anti-Rac-1. Content of Rac in total cell lysates is also shown. (B) Syntenin-1-depleted and control primary T lymphoblasts were treated with SDF-1α (100 ng/ml, 5 minutes), fixed, permeabilized and stained for syntenin-1, Rac-1 and F-actin. Summatory projections of the confocal stacks, and merged images are shown. Scale bars: 5 μm. (C) Quantification of the translocation of Rac to the leading edge of primary T lymphoblasts treated and stained as in B. Data correspond to the mean ± s.e.m. of three independent experiments. (D) Western blot analysis of the partition of syntenin-1 and Rac-1 in membrane (m) and cytosolic (c) fractions. Syntenin-1-depleted and control-transfected J77 T-cells were treated with SDF-1α (100 ng/ml) for 2.5 minutes. α-tubulin is shown as a loading and fractionation control. Numbers represent the ratio of m/c of Rac signal normalized with respect to untreated control cells. *P<0.05.

To directly determine whether syntenin-1 regulates Rac-1 activation downstream of Src, we examined the four potentially phosphorylatable Tyr residues contained in the syntenin-1 N-terminal domain (Beekman and Coffer, 2008). These residues occur in sequences consistent with Tyr-based domains described for functional immunoreceptors and are prototypical target sequences for Src family tyrosine kinases: one ITAM motif [YxxI/Lx(6–12)YxxI/L], which includes an additional hemi-ITAM, and one ITIM motif (S/I/V/LxYxxI/V/L) (Fig. 4C) (Beekman and Coffer, 2008). We generated mutant versions of GFP–syntenin-1, in which these tyrosine residues were substituted by phenylalanine: Y3, in which the three tyrosine residues included in the ITAM-like domain were mutated, and Y1, which had a mutation in the single Tyr in the ITIM-like domain (Fig. 4C). When expressed on polarized CEM T-cells, these mutants showed a distribution that was similar to that of GFP-tagged wild-type syntenin-1 during SDF-1α-induced migration (Fig. 4D and supplementary material Fig. S1D and Movie 2). When we analyzed the phosphorylation of the mutants upon treatment with SDF-1α, we observed that Y1 mutation almost completely abrogated syntenin-1 phosphorylation, suggesting that Tyr4 in the ITIM-like motif is the main substrate residue for Src kinases in syntenin-1 (Fig. 4E).

Fig. 4.

Syntenin-1 is tyrosine phosphorylated by Src family kinases. (A) T cells were treated with SDF-1α (100 ng/ml) for the times indicated, lysates were immunoprecipitated with anti-syntenin-1 and blots were sequentially probed with anti-phosphotyrosine and anti-syntenin-1 antibodies. Numbers represent quantitative densitometry analysis of syntenin-1 phosphorylation in the experiment shown (corrected for total syntenin-1 in each lane and normalized to the untreated sample). (B) T cells were treated with SDF-1α (100 ng/ml) for the times indicated after preincubation with PP2 (10 mM). Total cell lysates were sequentially probed with anti-phosphotyrosine and anti-syntenin-1. Numbers correspond to quantitative densitometry analysis of syntenin-1 phosphorylation in the experiment shown (corrected for total syntenin-1 in each lane and normalized to the untreated sample). (C) Sequence alignment of human and mouse syntenin-1 N-terminal regions, showing the positions of tyrosine residues in the ITIM and ITAM motifs. Y-F mutations introduced into GFP–syntenin-1 protein are indicated. (D) Cells transfected with wild-type or mutant versions of GFP–syntenin-1 were stimulated with SDF-1α, and analyzed by confocal time-lapse microscopy. A representative image is shown for each sample. Arrows indicate the uropod (U). Scale bar: 5 μm. (E) Cells transfected with GFP–syntenin-1 or the different mutants were treated with SDF-1α (100 ng/ml) for 2 minute, lysed and immunoprecipitated for GFP. Blots were sequentially probed with anti-phosphotyrosine and anti-GFP antibodies. Numbers correspond to quantitative densitometry analysis of tyrosine phosphorylation corrected for GFP signal in each lane and normalized to the untreated GFP–syntenin-1 sample.

Fig. 4.

Syntenin-1 is tyrosine phosphorylated by Src family kinases. (A) T cells were treated with SDF-1α (100 ng/ml) for the times indicated, lysates were immunoprecipitated with anti-syntenin-1 and blots were sequentially probed with anti-phosphotyrosine and anti-syntenin-1 antibodies. Numbers represent quantitative densitometry analysis of syntenin-1 phosphorylation in the experiment shown (corrected for total syntenin-1 in each lane and normalized to the untreated sample). (B) T cells were treated with SDF-1α (100 ng/ml) for the times indicated after preincubation with PP2 (10 mM). Total cell lysates were sequentially probed with anti-phosphotyrosine and anti-syntenin-1. Numbers correspond to quantitative densitometry analysis of syntenin-1 phosphorylation in the experiment shown (corrected for total syntenin-1 in each lane and normalized to the untreated sample). (C) Sequence alignment of human and mouse syntenin-1 N-terminal regions, showing the positions of tyrosine residues in the ITIM and ITAM motifs. Y-F mutations introduced into GFP–syntenin-1 protein are indicated. (D) Cells transfected with wild-type or mutant versions of GFP–syntenin-1 were stimulated with SDF-1α, and analyzed by confocal time-lapse microscopy. A representative image is shown for each sample. Arrows indicate the uropod (U). Scale bar: 5 μm. (E) Cells transfected with GFP–syntenin-1 or the different mutants were treated with SDF-1α (100 ng/ml) for 2 minute, lysed and immunoprecipitated for GFP. Blots were sequentially probed with anti-phosphotyrosine and anti-GFP antibodies. Numbers correspond to quantitative densitometry analysis of tyrosine phosphorylation corrected for GFP signal in each lane and normalized to the untreated GFP–syntenin-1 sample.

Expression of the Y3 mutant increased the chemotactic response to SDF-1α, although to a lesser extent than overexpression of wild-type syntenin-1 (Fig. 5A). By contrast, the Y1 mutation completely abolished the enhancing effect on cell migration, with Y1-transfected cells migrating similarly to control GFP-transfected cells (Fig. 5A). The effects of the syntenin-1 Tyr to Phe substitution on the ITIM-like motif on T cell chemotaxis were related to an effect on SDF-1α-induced actin polymerization. Overexpression of the Y1 mutant did not enhance F-actin polymerization to levels above that in control cells transfected with GFP upon treatment with SDF-1α (Fig. 5B). Moreover, Y1 overexpression in syntenin-1-depleted cells did not rescue the inhibition of chemokine-induced actin polymerization produced by silencing of syntenin-1 (Fig. 5C).

To directly determine whether syntenin-1 phosphorylation affects Rac-1 activation, we analyzed antibody-stained GTP-Rac by flow cytometry after SDF-1α stimulation. GTP-Rac staining increased with time in SDF-1α-stimulated GFP-transfected cells (Fig. 5D). This effect was greatly enhanced in cells expressing GFP–syntenin-1 and the Y3 mutant. Rac-1 activation was inhibited to below control levels in Y1-transfected cells (Fig. 5D). In classical pull-down experiments, the Y1 mutant was ineffective in the rescue of Rac activation after syntenin-1 depletion (Fig. 5E), whereas transfection of wild-type syntenin-1 increased Rac activity in both resting and stimulated conditions. Together, our results suggest that syntenin-1, through a mechanism requiring phosphorylation of its ITIM motif, regulates Src-induced Rac-dependent actin polymerization during T cell migration.

The syntenin-1 ITIM-like motif regulates Rac activation in T cells by binding to M-RIP

To investigate how Src-phosphorylated syntenin-1 mediates Rac-1 regulation, we conducted a proteomic screen for proteins that bind wild-type GFP–syntenin-1 but not the Y1 mutant. SDF-1α-stimulated J77 T cells transiently expressing GFP–syntenin-1 or GFP–Y1-mutant were immunoprecipitated with anti-GFP antibody. Proteomic comparisons identified specific binding of GFP–syntenin-1 to regulators of Rho-GTPases, including Rho-GAP, obscurin (a Rho GEF) and myosin phosphatase Rho interacting protein (M-RIP) (supplementary material Table S1). M-RIP, which is also called p116RIP is a multidomain scaffold protein that binds to Rho-A and the myosin-binding subunit (MBS) of myosin light chain phosphatase (Surks et al., 2003), inhibiting Rho-A stimulated contractility (Gebbink et al., 1997; Koga and Ikebe, 2005). The interaction of wild-type syntenin-1 with M-RIP was confirmed by immunoblotting of GFP immunoprecipitates of GFP–syntenin-1 (Fig. 6A). Binding of M-RIP to the Y1 mutant could not be detected. Inducible association of endogenous syntenin-1 was observed in immunoprecipitates of endogenous M-RIP (Fig. 6B), which showed that this interaction peaked at 1–5 minutes of SDF-1α stimulation. We found that endogenous M-RIP had a partially submembranous distribution (supplementary material Fig. S1E) that relocalized to the leading edge upon SDF-1α stimulation, where it colocalized with Rac-1 and F-actin (Fig. 6C). Pull-down assays with GST-fusion proteins of different M-RIP domains showed that syntenin-1 binds the second coiled-coil domain (residues 728–878), overlapping with the reported binding site for MBS (Surks et al., 2003) (Fig. 6D and supplementary material Fig. S1F). Although M-RIP binding to RhoA through an adjacent RBD site has been reported (Surks et al., 2003), the possible relationship with Rac-1 GTPase has not been explored. We detected binding of Rac-1 to the first N-terminal half of M-RIP (residues 1–528), comprising the two PH domains (Fig. 6D).

Fig. 5.

Syntenin-1 phosphorylation is required for T-cell chemotaxis and chemokine-induced actin polymerization. (A) T cells transfected with wild-type or mutant versions of GFP–syntenin-1 or GFP alone were allowed to migrate in Transwell chambers to SDF-1α (100 ng/ml). Data are the means ± s.e.m. of four independent experiments. (B) Cells transfected as in A were treated with SDF-1α (100 ng/ml) for 1 min, fixed, permeabilized, stained with phalloidin Alexa Fluor 647 and analyzed by flow cytometry in the GFP+ population. Data are means ± s.d. of three independent experiments. (C) Cells were transfected with the negative oligonucleotide or siRNA to knockdown syntenin-1 alone or in combination with GFP, GFP–syntenin-1 or the Y1 mutant, treated with SDF-1α (100 ng/ml) for the indicated times, fixed, permeabilized, stained with phalloidin Alexa Fluor 647 and analyzed by flow cytometry. (D) Cells transfected with wild-type or mutant versions of GFP–syntenin-1 or GFP alone were treated with SDF-1α (100 ng/ml) for the indicated times, fixed, permeabilized and stained with anti-Rac-GTP. The percentage of positive cells in the GFP+ population was analyzed by flow cytometry. (E) T cells transfected with the negative oligonucleotide or siRNA against syntenin-1 in combination with GFP–syntenin-1 or the Y1 mutant were treated with SDF-1α (100 ng/ml) for 2 minutes before lysis. GTP-loaded Rac was pulled down with GST–PAK–CRIB and blot revealed with anti-Rac-1. Content of Rac in total cell lysates is also shown. *P<0.05.

Fig. 5.

Syntenin-1 phosphorylation is required for T-cell chemotaxis and chemokine-induced actin polymerization. (A) T cells transfected with wild-type or mutant versions of GFP–syntenin-1 or GFP alone were allowed to migrate in Transwell chambers to SDF-1α (100 ng/ml). Data are the means ± s.e.m. of four independent experiments. (B) Cells transfected as in A were treated with SDF-1α (100 ng/ml) for 1 min, fixed, permeabilized, stained with phalloidin Alexa Fluor 647 and analyzed by flow cytometry in the GFP+ population. Data are means ± s.d. of three independent experiments. (C) Cells were transfected with the negative oligonucleotide or siRNA to knockdown syntenin-1 alone or in combination with GFP, GFP–syntenin-1 or the Y1 mutant, treated with SDF-1α (100 ng/ml) for the indicated times, fixed, permeabilized, stained with phalloidin Alexa Fluor 647 and analyzed by flow cytometry. (D) Cells transfected with wild-type or mutant versions of GFP–syntenin-1 or GFP alone were treated with SDF-1α (100 ng/ml) for the indicated times, fixed, permeabilized and stained with anti-Rac-GTP. The percentage of positive cells in the GFP+ population was analyzed by flow cytometry. (E) T cells transfected with the negative oligonucleotide or siRNA against syntenin-1 in combination with GFP–syntenin-1 or the Y1 mutant were treated with SDF-1α (100 ng/ml) for 2 minutes before lysis. GTP-loaded Rac was pulled down with GST–PAK–CRIB and blot revealed with anti-Rac-1. Content of Rac in total cell lysates is also shown. *P<0.05.

To demonstrate that M-RIP provides a molecular link between Src-mediated syntenin-1 phosphorylation and the downstream regulation of Rac, we suppressed the expression of this adaptor protein in T cells by RNAi (Fig. 6E) and quantified the Rac-GTP content (Fig. 6F) after stimulation with SDF-1α. Silencing of M-RIP inhibited Rac activation (Fig. 6F) and actin polymerization after SDF-1α stimulation (Fig. 6G). The same pattern of inhibition was observed in chemotaxis experiments in both cell lines and primary T lymphoblasts (Fig. 6H,I). No additive effect was observed in any of these functional readouts when both syntenin-1 and M-RIP molecules were simultaneously silenced, further demonstrating that they act on the same signaling pathway.

Syntenin-1 modulates CD3-induced actin polymerization

Rac-1 is activated during antigen presentation (Arrieumerlou et al., 2000; Yu et al., 2001), a context in which dynamic asymmetric actin polymerization is also crucial for proper functioning of the immune synapse (Billadeau et al., 2007; Gordón-Alonso et al., 2010). However, Src activation occurs early after TCR stimulation (Rudd, 1990). We therefore assessed whether the syntenin-1–M-RIP axis could be also involved in TCR-induced activation of Rac. We first observed that both endogenous syntenin-1 and M-RIP polarized to the contact area in cognate conjugates of T cells and APCs (Fig. 7A,B and supplementary material Movie 3). Knockdown of either syntenin-1 or M-RIP, impaired actin polymerization and Rac activation in response to TCR crosslinking (Fig. 7C,D and supplementary material Fig. S2A). Moreover, syntenin-1 knockdown reduced Rac translocation to the plasma membrane (supplementary material Fig. S2B). Overexpression of GFP-tagged syntenin-1 was able to increase TCR-stimulated actin polymerization, and this effect was abolished by mutation of the tyrosine residue in the ITIM-like motif (supplementary material Fig. S2C).

When we analyzed the conjugates formed by syntenin-1-depleted cells with superantigen-loaded APCs, we observed no effect on the number of conjugates formed or on the translocation of the MTOC to the intercellular contact (supplementary material Fig. S2D). However, antigen-driven actin polymerization at the IS was locally reduced (supplementary material Fig. S2E), and as a consequence, CD3 congregation at the central SMAC was significantly impaired (Fig. 7E,F). When signals derived from antigen stimulation were analyzed, lower levels of phosphorylation of Vav-1, PLC-γ and ERK1/2 were detected in syntenin-1-silenced cells that were exposed to superantigen-loaded B cells at late time points of activation (10 minutes, Fig. 7G and supplementary material Fig. S2F).

This study reports the involvement of syntenin-1 in Rac-1 activation and actin polymerization downstream of SFK during both chemokine-induced T cell migration and cognate interactions with APCs. In both phenomena, cell polarity plays a crucial role, determining the front-to-rear axis during chemotaxis, and reorienting signaling and adhesion receptors towards the APC contact site in cognate interactions. Our data indicate that syntenin-1 combines a polarized subcellular localization, dictated by its binding to PIP2 (Zimmermann et al., 2002), with a Src-dependent scaffold activity. Thus, phosphorylated syntenin-1, through binding to the adaptor protein M-RIP, leads to polar translocation of Rac-1 to the plasma membrane and subsequent asymmetric actin polymerization. Syntenin-1-depleted cells have impaired Rac-1 activation and membrane targeting, which resulted in almost complete abrogation of stimuli-induced actin polymerization.

Syntenin-1 was recently shown to bind directly to Src (Boukerche et al., 2008), and Src acts in parallel with PI3K during CXCR2-directed neutrophil chemotaxis (Sai et al., 2008). The functional relationship between Src and Rac activities is complex. Rac activation in response to PDGF has been shown to depend on Src kinase activity (Ouyang et al., 2008). However, PDGF-induced Src activity is not polarized, whereas the consequent Rac activity is clearly localized to the leading edge. It is therefore conceivable that chemokine- or CD3-induced Src activation triggers a polarized signal through its binding to polarized syntenin-1. Syntenin-1 has also been reported to act upstream of the related GTPase Cdc42 in a non-canonical Wnt signaling pathway (Luyten et al., 2008); however, the implications of syntenin-1 phosphorylation were not analyzed.

The functional analysis of Y to F point mutations in the syntenin-1 ITAM-like and ITIM-like domains revealed that mutation of Tyr4 (Y1), in the ITIM-like domain, abolished syntenin-1 phosphorylation and the enhancing effect of syntenin-1 overexpression on chemotaxis, actin polymerization and Rac-1 activation. Overexpression of the Y1 mutant might even have a dominant-negative effect on Rac activation as suggested by the flow cytometry results in which the GFP-positive population can be separately analyzed. Moreover, this mutant, in contrast to wild-type GFP-tagged syntenin-1 was not able to rescue silencing of syntenin-1 in chemokine-induced Rac activation and actin polymerization, confirming syntenin-1 as a link in Src-induced Rac-1 activation in T cells. This tyrosine residue is crucial for the binding to M-RIP in T cells. Mutation of the tyrosine residues in the ITAM-like sequence did not statistically affect syntenin-1 function, suggesting that the effects might be due to small conformational changes. Although ITAM (Ivashkiv, 2009) and ITIM motifs (Daëron et al., 2008) have been extensively studied in immune cell recognition receptors, where they integrate signals emanating from different extracellular stimuli, other cytosolic adaptor proteins, such as ERMs (ezrin–radixin–moesin proteins), also contain functional Tyr-based motifs (Urzainqui et al., 2002). Thus, by harboring equivalent signaling motifs, membrane actin adaptors might share some of the signaling cascades elicited by the receptors themselves. It is intriguing that an inhibitory motif, such as an ITIM, which is potentially bound to phosphatases, can actually positively regulate Rac activity. However, positive regulation of Ras or Src has been previously reported to occur downstream of SHP-2 activation (Barrow and Trowsdale, 2006).

The mutation of phosphorylatable tyrosine residues did not affect the subcellular polarization of syntenin-1, suggesting that this is governed by the two PDZ domains, which is consistent with the finding that deletion of these domains abolishes the enhancing effect of syntenin-1 upon tumor cell invasion (Meerschaert et al., 2007). Syntenin-1 PDZ domains have been demonstrated to interact with the ERM protein merlin (Kang et al., 2003) and with PIP2 (Zimmermann et al., 2002). These interactions probably contribute to plasma membrane anchoring of syntenin-1 at lamellipodia and the T-cell–APC contact area, which are sites of active PIP2 production. Other transmembrane receptors, such as CD6 or tetraspanins (Gimferrer et al., 2005; Latysheva et al., 2006), might also contribute to subcellular localization of syntenin-1 in T cells. The fact that syntenin-1 overexpression enhances chemotaxis and actin polymerization suggests that it acts as a limiting factor in the pathway that links Src activity with Rac-1 activation. This effect is crucial for the polarization of the signal, ensuring that Rac-1 activation and subsequent actin polymerization will be optimal only at specific subcellular localizations containing syntenin-1.

Fig. 6.

M-RIP binds to syntenin-1 and Rac-1 and regulates Rac activation and actin polymerization in T cells. (A) J77 T cells transfected with GFP, GFP–syntenin-1 or GFP–Y1 mutant were treated with SDF-1α (100 ng/ml). Lysates were immunoprecipitated with anti-GFP, and blots sequentially probed with anti-M-RIP and anti-GFP antibodies. Numbers show densitometric analysis of M-RIP binding (corrected for total GFP in each lane and normalized to untreated GFP). (B) T cells were treated with SDF-1α (100 ng/ml), lysates immunoprecipitated with anti-M-RIP, and blots sequentially probed with anti-M-RIP and anti-syntenin-1 antibodies. Numbers show densitometric analysis of M-RIP/syntenin-1 coimmunoprecipitation (corrected for M-RIP immunoprecipitated protein in each lane and normalized to the untreated sample). (C) Primary PBLs were incubated with SDF-1α (100 ng/ml, 5 minutes). Samples were fixed, permeabilized and co-stained for Rac-1, M-RIP and F-actin. Arrow indicates the leading edge (LE). Scale bar: 5 μm. (D) Primary T lymphoblast lysates were used in pull-down assays with GST fusion proteins of M-RIP domains (Nterm, 1–538; RBD, 545–823; CC1, 672–707; CC2, 728–878; CC3, 900–974). Blots were probed with anti-syntenin-1 and anti-Rac-1 antibodies. (E) Western blot analysis of M-RIP and syntenin-1 expression in lysates of T-cells transfected with siRNA against M-RIP, syntenin-1 or both, or with control oligonucleotide. Tubulin is shown as a loading control. Numbers indicate relative protein expression (corrected by tubulin signal and normalized to negative oligo transfected cells). (F) T cells transfected with siRNA against M-RIP or control oligonucleotide were treated with SDF-1α (100 ng/ml) for 5 minutes, fixed, permeabilized, stained with anti-Rac-GTP and analyzed by flow cytometry. Data are means ± s.e.m. of four independent experiments. (G) Cells transfected with siRNA against M-RIP, syntenin-1 or both or with negative control oligonucleotide were treated with SDF-1α (100 ng/ml), stained with phalloidin Alexa 488, and analyzed by flow cytometry. Data are means ± s.e.m. of three independent experiments. (H) T cells were allowed to migrate in Transwell inserts towards SDF-1α (100 ng/ml). Data are the means ± s.e.m. of four independent experiments. (I) Primary T lymphoblasts were allowed to migrate in Transwell inserts towards SDF-1α (100 ng/ml). Data are the means ± s.d. of a representative experiment performed in triplicate. *P<0.05; **P<0.01.

Fig. 6.

M-RIP binds to syntenin-1 and Rac-1 and regulates Rac activation and actin polymerization in T cells. (A) J77 T cells transfected with GFP, GFP–syntenin-1 or GFP–Y1 mutant were treated with SDF-1α (100 ng/ml). Lysates were immunoprecipitated with anti-GFP, and blots sequentially probed with anti-M-RIP and anti-GFP antibodies. Numbers show densitometric analysis of M-RIP binding (corrected for total GFP in each lane and normalized to untreated GFP). (B) T cells were treated with SDF-1α (100 ng/ml), lysates immunoprecipitated with anti-M-RIP, and blots sequentially probed with anti-M-RIP and anti-syntenin-1 antibodies. Numbers show densitometric analysis of M-RIP/syntenin-1 coimmunoprecipitation (corrected for M-RIP immunoprecipitated protein in each lane and normalized to the untreated sample). (C) Primary PBLs were incubated with SDF-1α (100 ng/ml, 5 minutes). Samples were fixed, permeabilized and co-stained for Rac-1, M-RIP and F-actin. Arrow indicates the leading edge (LE). Scale bar: 5 μm. (D) Primary T lymphoblast lysates were used in pull-down assays with GST fusion proteins of M-RIP domains (Nterm, 1–538; RBD, 545–823; CC1, 672–707; CC2, 728–878; CC3, 900–974). Blots were probed with anti-syntenin-1 and anti-Rac-1 antibodies. (E) Western blot analysis of M-RIP and syntenin-1 expression in lysates of T-cells transfected with siRNA against M-RIP, syntenin-1 or both, or with control oligonucleotide. Tubulin is shown as a loading control. Numbers indicate relative protein expression (corrected by tubulin signal and normalized to negative oligo transfected cells). (F) T cells transfected with siRNA against M-RIP or control oligonucleotide were treated with SDF-1α (100 ng/ml) for 5 minutes, fixed, permeabilized, stained with anti-Rac-GTP and analyzed by flow cytometry. Data are means ± s.e.m. of four independent experiments. (G) Cells transfected with siRNA against M-RIP, syntenin-1 or both or with negative control oligonucleotide were treated with SDF-1α (100 ng/ml), stained with phalloidin Alexa 488, and analyzed by flow cytometry. Data are means ± s.e.m. of three independent experiments. (H) T cells were allowed to migrate in Transwell inserts towards SDF-1α (100 ng/ml). Data are the means ± s.e.m. of four independent experiments. (I) Primary T lymphoblasts were allowed to migrate in Transwell inserts towards SDF-1α (100 ng/ml). Data are the means ± s.d. of a representative experiment performed in triplicate. *P<0.05; **P<0.01.

Fig. 7.

Syntenin-1–M-RIP regulates F-actin polymerization at the immune synapse. (A) Antigen-induced conjugates were stained for syntenin-1 and CD3 or for M-RIP and F-actin. Maximal projections of the confocal image stacks are shown, together with the corresponding DIC images. Scale bars: 5 μm. (B) Relocalization of endogenous syntenin-1 and M-RIP, and CD45 (as negative control), in conjugates formed by primary T lymphoblasts was quantified. Data shown correspond to means ± s.e.m. of 120 conjugates from three independent experiments for each condition. (C) J77 T cells transfected with siRNA against M-RIP, syntenin-1 or both or with negative control oligonucleotide were treated with anti-CD3 (HIT3a, 10 μg/ml), fixed at the times indicated, permeabilized, stained with phalloidin Alexa Fluor 488 and analyzed by flow cytometry. Data are means ± s.e.m. of five independent experiments. (D) T cells transfected with siRNA against M-RIP, syntenin-1 siRNA or both or with negative control oligonucleotide were treated with anti-CD3 (HIT3a, 10 μg/ml) for 5 minutes, fixed, permeabilized, stained with anti-Rac-GTP and analyzed by flow cytometry. Data are means ± s.d. of two independent experiments. (E) T cells transfected with siRNA against syntenin-1 or control oligonucleotide were conjugated with CMAC-labeled SEE-loaded Raji cells for 30 minutes, fixed and stained for CD3. Corresponding CMAC (blue) and DIC images are also shown. Scale bar: 10 μm. (F) CD3 relocalization in conjugates formed as in C (means ± s.e.m. of 180 conjugates from three independent experiments for each condition). (G) T cells transfected with siRNA against syntenin-1 or control oligonucleotide were conjugated with SEE-loaded Raji cells (5:1 ratio) for the indicated times. Total cell lysates were analyzed by western blot for phosphorylation of Vav-1, PLC-γ1 and ERK1/2. **P<0.01.

Fig. 7.

Syntenin-1–M-RIP regulates F-actin polymerization at the immune synapse. (A) Antigen-induced conjugates were stained for syntenin-1 and CD3 or for M-RIP and F-actin. Maximal projections of the confocal image stacks are shown, together with the corresponding DIC images. Scale bars: 5 μm. (B) Relocalization of endogenous syntenin-1 and M-RIP, and CD45 (as negative control), in conjugates formed by primary T lymphoblasts was quantified. Data shown correspond to means ± s.e.m. of 120 conjugates from three independent experiments for each condition. (C) J77 T cells transfected with siRNA against M-RIP, syntenin-1 or both or with negative control oligonucleotide were treated with anti-CD3 (HIT3a, 10 μg/ml), fixed at the times indicated, permeabilized, stained with phalloidin Alexa Fluor 488 and analyzed by flow cytometry. Data are means ± s.e.m. of five independent experiments. (D) T cells transfected with siRNA against M-RIP, syntenin-1 siRNA or both or with negative control oligonucleotide were treated with anti-CD3 (HIT3a, 10 μg/ml) for 5 minutes, fixed, permeabilized, stained with anti-Rac-GTP and analyzed by flow cytometry. Data are means ± s.d. of two independent experiments. (E) T cells transfected with siRNA against syntenin-1 or control oligonucleotide were conjugated with CMAC-labeled SEE-loaded Raji cells for 30 minutes, fixed and stained for CD3. Corresponding CMAC (blue) and DIC images are also shown. Scale bar: 10 μm. (F) CD3 relocalization in conjugates formed as in C (means ± s.e.m. of 180 conjugates from three independent experiments for each condition). (G) T cells transfected with siRNA against syntenin-1 or control oligonucleotide were conjugated with SEE-loaded Raji cells (5:1 ratio) for the indicated times. Total cell lysates were analyzed by western blot for phosphorylation of Vav-1, PLC-γ1 and ERK1/2. **P<0.01.

Syntenin-1 has also been reported to be associated with membranes in intracellular compartments and to regulate intracellular trafficking of associated receptors (Sarkar et al., 2008). In T cells, however, most endogenous staining was submembranous. We observed no alterations to the basal expression of chemokine receptors or CD3 at the plasma membrane of syntenin-1-depleted cells (data not shown). However, it is also conceivable that vesicular syntenin-1 contributes to Rac activation during recycling (Palamidessi et al., 2008).

Our proteomic analyses aimed to search for proteins that could interact with wild-type syntenin-1 but not with mutant syntenin-1. In this screening, some other regulators of the Rho family of GTPases were also identified; however, only the association with M-RIP was assessed further and confirmed by immunoprecipitation and pull-down analyses. Because of the nature of M-RIP as a scaffolding protein, some of these other molecules might derive from the interaction between syntenin-1 and M-RIP. Further studies will be needed to address the hierarchies and kinetics of these interactions.

Our data identify M-RIP as a crucial local regulator of the Rho–Rac balance during chemotaxis and antigen recognition. Syntenin-1 binds to the same M-RIP domain reported to be essential for the binding to the MBS of myosin light chain phosphatase (Surks et al., 2003). Therefore, syntenin-1 binding might compete with MBS, shifting the balance towards Rac-induced actin polymerization, rather than Rho-mediated contraction. Myosin function in lymphocytes is not restricted to regulation of cell contractility during migration, but also regulates immune synapse maturation (Ilani et al., 2009), cytotoxicity (Andzelm et al., 2007) and chemokine receptor internalization (Rey et al., 2007). MLC phosphorylation is detected at the IS (Ilani et al., 2009) and at the leading edge of migrating polarized lymphocytes (Vicente-Manzanares et al., 2002), both sites of active Rac-induced actin polymerization.

Upon antigen presentation, actin treadmilling driven by myosin-IIA has been shown to be responsible for aggregation of CD3 at the c-SMAC in the IS (Ilani et al., 2009). Thus, syntenin-1 silencing only mildly attenuates the initial signaling events in antigen-driven cognate interactions, and MTOC translocation occurs normally. However, actin dynamics is perturbed, CD3 remains dispersed on the plasma membrane and the activation magnitude and duration are not maintained.

The involvement of syntenin-1 in T cell migration is in agreement with other observations indicating that syntenin-1 might be an important determinant of the malignant phenotype of epithelial cancers (Helmke et al., 2004; Sarkar et al., 2004; Boukerche et al., 2005). Moreover, M-RIP silencing has been reported to inhibit cell invasivity (Ono et al., 2008) and to lie downstream of TGF-β during breast cancer dissemination (Giampieri et al., 2009), so the syntenin-1–M-RIP axis might also be relevant for tumor cell migration. The homing of tumor cells to specific metastatic sites under the influence of chemokines and their receptors is a hallmark of tumorigenesis (Ben-Baruch, 2006), although the SDF–CXCR4 axis acts in conjunction with other pro-malignancy mechanisms. Overexpression of syntenin-1 might be one of the amplifiers of SDF–CXCR4 signals, enhancing the metastatic potential of tumor cells, and in malignancies of the haematopoietic system.

Cells, antibodies and reagents

Lymphoblastoid cell lines of the T (CEM, J77, CH7C17, HSB2) and B (Raji, Hom2) lineages were cultured in RPMI 1640 (Flow Laboratories, Irvine, UK) supplemented with 10% fetal calf serum (FCS, Flow Laboratories). Human PBLs were obtained from buffy coats from healthy volunteer blood donors and derived to T-lymphoblasts by treatment with PHA/IL-2 (Sala-Valdes et al., 2006).

Anti-CD3 (T3b) and anti-ICAM-3 (HP2/19) monoclonal antibodies were obtained as previously described (Mittelbrunn et al., 2002). For detection of Tyr phosphorylation, we used a combination of 4G10 and PY72 antibodies (kindly provided by Balbino Alarcón, Centro de Biologia Molecular Severo Ochoa, Madrid, Spain). Antibodies againt phosphorylated Vav-1 and total Vav-1 were kindly provided by Xose Bustelo (Centro de investigacion del Cancer, Salamanca, Spain). Rabbit anti-syntenin-1 was purchased from Synaptic Systems (Göttingen, Germany), anti-α-tubulin and anti-M-RIP from Sigma, anti-Rac-1 from Millipore (Billerica, MA), anti-Rac-GTP from NewEastBio (Malvern, PA). p-ERK, total ERK, p-ZAP70, ZAP70, p-PLCγ1 and PLCγ1 antibodies were from Cell Signaling (Danvers, MA) and p-Src and Src antibodies from Invitrogen (Carlsbad, CA). Anti-GFP was purchased from Clontech and GFP-trap monobodies from Chromotek (Planegg-Martinsried).

SDF-1α and MIP-3β were purchased from Peprotech (London, UK). PP2 was purchased from Sigma. Jurkat cells were activated with HIT3a antibody (10 mg/ml) (Biolegend, San Diego, CA).

Recombinant DNA constructs

Psrα-HAGFP-syntenin-1 was generated by cloning cDNA encoding syntenin-1, obtained from EcoRI-digested PMT2–HA–syntenin-1 (Gimferrer et al., 2005), into psrα-HAGFP. Single point mutations of GFPHA–syntenin-1 were made by directed mutagenesis using the Quikchange Multi-Site Directed Mutagenesis Kit from Stratagene (La Jolla, CA) with the following sequences: Y3 mutation, 5′-GATGGAAATCTGTTTCCTAAACTGTTTCCAGAGCTCTCTCATTCATGGGACTGTC-3′; Y1 mutation, 5′-GGAATTCATGTCTCTTTTTCCATCTCTTGAAGATTTGAAGGTAGAC-3′.

GST constructs of M-RIP (Surks et al., 2003) were provided by Michael Mendelsohn (Tufts Medical Center, Boston, MA). GST–PAK–CRIB was donated by John Collard (The Netherlands Cancer Institute, Amsterdam, The Netherlands). GFP-tagged pleckstrin domains were provided by Tamas Balla (National Institutes of Health, Bethesda, MA).

Cell transfection

CEM/J77 cells (2×107) were transiently transfected with 20 mg of plasmids in OPTIMEM medium (Invitrogen) by electroporation at 250 V and 1200 mF (Gene Pulser II, Bio-Rad). To selectively knockdown the expression of endogenous syntenin-1, two target sequences were used: oligo 1 and oligo 2 (reference numbers J-008270-08 and J-008270-06, Dharmacon Research, Lafayette, CO). For rescue experiments, oligo 1 (which pairs at the non-coding region) was used. For silencing of M-RIP, Dharmacon reference J-014102 siRNA oligonucleotide was used. RNA duplexes (2 μM per sample) or a negative control oligonucleotide (Eurogentec, Seraing, Belgium) were transfected by electroporation or by nucleofection using the Nucleofector kit T (program T-23) (Amaxa, Cologne, Germany). Efficiency of knockdown was routinely assessed by western blot to be greater than 50% for each transfection.

Migration assay

Migration assays were performed 24 hours after transfection with GFP-tagged constructs and 48 hours after transfection with siRNA oligonucleotides, either on primary T lymphoblasts or CEM T cells. Cells (5×105/100 ml) were placed in the upper chamber of 5 mm pore-size Transwell inserts (Costar, Cambridge, MA). The lower chambers contained 600 ml complete RPMI medium, with or without SDF-1α (100 nM) or MIP3β (1 μM) (Peprotech, London, UK). After 2–3 hours at 37°C, migrated cells were recovered from the lower chamber and counted by flow cytometry.

Actin polymerization

Cells (primary T lymphoblasts or J77 T cells) were stimulated, fixed with 4% formaldehyde, permeabilized with 0.5% Triton X-100 (5 minutes) and stained with Phalloidin Alexa Fluor 647 or Phalloidin Alexa Fluor 488 (Invitrogen). Mean fluorescence intensity of F-actin staining was analyzed in a FACS CANTO cytofluorometer (BD Biosciences).

Immunoprecipitation, pull down and western blot

Total cell lysates were obtained in Tris-buffered saline containing 1% Nonidet P-40, 2 mM CaCl2, 2 mM MgCl2, phosphatase inhibitors (1 mM sodium pyrophosphate, 1 mM sodium orthovanadate, 10 mM sodium fluoride), and protease inhibitors (Complete; Roche Applied Science). For immunoprecipitation, cell lysates were incubated with the indicated antibodies coupled to Protein-G–Sepharose. After washing six times with lysis buffer, proteins bound to Sepharose beads were eluted by boiling in sample buffer. Pull-down experiments with GST proteins were performed as described (Sala-Valdes et al., 2006).

Target proteins were detected by chemiluminescence with a LAS3000 luminescent imager (Fuji Photo Film, Elmsford, NY). Quantitative densitometric analysis of immunoblots was performed with Image Gauge v.346 software (Fujifilm).

Rac activity

GTP-Rac loading was measured by classical pull-down experiments with GST–PAK–CRIB (Vicente-Manzanares et al., 2005). Alternatively, cells were fixed, permeabilized and stained with anti-Rac-GTP antibody and analyzed by flow cytometry.

Cell fractionation

Membrane and cytosolic protein extracts were prepared with the Proteo Extract Subcellular Proteome Extraction Kit (Calbiochem, San Diego, CA), which is based on TX114 solubilization, according to the manufacturer’s instructions.

T cell and B cell conjugate formation

For antigen-specific stimulation, Hom-2 APCs (5×106) were incubated at 37°C for 3 hours with HA307-319 peptide (200 μg/ml) and mixed with a HA-specific T cell clone (CH7C17) at a ratio of 1:3. For superantigen stimulation, Raji B cells were labelled with the fluorescence cell trackers CMAC or CM-TMR (Invitrogen), and then incubated for 20 minutes at 37°C with 1 μg/ml SEE (Superantigen E). Antigen-driven conjugates were processed for immunofluorescence and superantigen stimulation used for western blot analysis of signaling molecule activation.

Immunofluorescence, confocal and time-lapse microscopy

T cell and B cell cognate conjugates, PBLs or T lymphoblasts (seeded onto 50mg/mL fibronectin) were stained as described (Mittelbrunn et al., 2002; Sala-Valdes et al., 2006). Samples were fixed with 4% formaldehyde and permeabilized for 5 minutes with 0.5% Triton X-100, stained and mounted with Prolong (Invitrogen). Confocal images were obtained with a Leica TCS-SP5 confocal scanning laser microscope with an HCX PL APO lambda blue 63×/1.4 oil immersion objective and analyzed with Leica LAS confocal image processing software. For time-lapse confocal fluorescence microscopy, cells were maintained at 37°C in a 5% CO2 atmosphere.

The synapsemeasure plug-in for ImageJ software was used to analyze the redistribution of fluorescent signal to the T-APC contact area (Calabia-Linares et al., 2011). Quantification of Rac translocation was calculated as the ratio of the signal at the leading edge membrane region versus the cytosolic signal in four confocal planes for each cell.

Statistical analysis

Statistical significance was calculated using the Student’s t-test or ANOVA, and significant differences are indicated by *P<0.05 and **P<0.01.

The authors wish to thank P. Zimmermann for help and advice with silencing of syntenin-1, Soraya López-Martín for technical assistance, Maria-Rosa Sarrias for helpful discussions and S. Bartlett for English editing.

Funding

This work was supported by the Instituto de Salud Carlos III [grant numbers PI080794 and PI11/01645] to M.Y.-M.; the Comunidad de Madrid [grant number INSINET-0159/2006] to F.S.M; the Fundación para la Investigación y la Prevención del SIDA en España [grant number FIPSE 36658/07 to] F.S.M. M.S.-V. is funded by a Carlos III Ayuda Predoctoral de Formación en Investigación fellowship [FI05/00238], M.G.-A. by RECAVA. A.I. is recipient of fellowships from the Departament d’Universitats Recerca i Societat de la Informació (DURSI).

Andzelm
M. M.
,
Chen
X.
,
Krzewski
K.
,
Orange
J. S.
,
Strominger
J. L.
(
2007
).
Myosin IIA is required for cytolytic granule exocytosis in human NK cells
.
J. Exp. Med.
204
,
2285
2291
.
Arrieumerlou
C.
,
Randriamampita
C.
,
Bismuth
G.
,
Trautmann
A.
(
2000
).
Rac is involved in early TCR signaling
.
J. Immunol.
165
,
3182
3189
.
Barrow
A.
,
Trowsdale
J.
(
2006
).
You say ITAM and I say ITIM, let’s call the whole thing off: the ambiguity of immunoreceptor signalling
.
Eur. J. Immunol.
36
,
1646
1653
.
Beekman
J. M.
,
Coffer
P. J.
(
2008
).
The ins and outs of syntenin, a multifunctional intracellular adaptor protein
.
J. Cell Sci.
121
,
1349
1355
.
Ben-Baruch
A.
(
2006
).
The multifaceted roles of chemokines in malignancy
.
Cancer Metastasis Rev.
25
,
357
371
.
Billadeau
D. D.
,
Nolz
J. C.
,
Gomez
T. S.
(
2007
).
Regulation of T-cell activation by the cytoskeleton
.
Nat. Rev. Immunol.
7
,
131
143
.
Boukerche
H.
,
Su
Z. Z.
,
Emdad
L.
,
Baril
P.
,
Balme
B.
,
Thomas
L.
,
Randolph
A.
,
Valerie
K.
,
Sarkar
D.
,
Fisher
P. B.
(
2005
).
mda-9/Syntenin: a positive regulator of melanoma metastasis
.
Cancer Res.
65
,
10901
10911
.
Boukerche
H.
,
Su
Z. Z.
,
Prevot
C.
,
Sarkar
D.
,
Fisher
P. B.
(
2008
).
mda-9/Syntenin promotes metastasis in human melanoma cells by activating c-Src
.
Proc. Natl. Acad. Sci. USA
105
,
15914
15919
.
Calabia-Linares
C.
,
Robles-Valero
J.
,
de la Fuente
H.
,
Perez-Martinez
M.
,
Martin-Cofreces
N.
,
Alfonso-Perez
M.
,
Gutierrez-Vazquez
C.
,
Mittelbrunn
M.
,
Ibiza
S.
,
Urbano-Olmos
F. R.
, et al.
. (
2011
).
Endosomal clathrin drives actin accumulation at the immunological synapse
.
J. Cell Sci.
124
,
820
830
.
Daëron
M.
,
Jaeger
S.
,
Du Pasquier
L.
,
Vivier
E.
(
2008
).
Immunoreceptor tyrosine-based inhibition motifs: a quest in the past and future
.
Immunol. Rev.
224
,
11
43
.
Dustin
M. L.
(
2009
).
The cellular context of T cell signaling
.
Immunity
30
,
482
492
.
Gebbink
M. F.
,
Kranenburg
O.
,
Poland
M.
,
van Horck
F. P.
,
Houssa
B.
,
Moolenaar
W. H.
(
1997
).
Identification of a novel, putative Rho-specific GDP/GTP exchange factor and a RhoA-binding protein: control of neuronal morphology
.
J. Cell Biol.
137
,
1603
1613
.
Giampieri
S.
,
Manning
C.
,
Hooper
S.
,
Jones
L.
,
Hill
C.
,
Sahai
E.
(
2009
).
Localised and reversible TGFβ signalling switched breast cancer cells from cohesive to single cell motility
.
Nat. Cell Biol.
11
,
1287
1296
.
Gimferrer
I.
,
Ibanez
A.
,
Farnos
M.
,
Sarrias
M. R.
,
Fenutria
R.
,
Rosello
S.
,
Zimmermann
P.
,
David
G.
,
Vives
J.
,
Serra-Pages
C.
, et al.
. (
2005
).
The Lymphocyte Receptor CD6 Interacts with Syntenin-1, a Scaffolding Protein Containing PDZ Domains
.
J. Immunol.
175
,
1406
1414
.
Gomez-Mouton
C.
,
Manes
S.
(
2007
).
Establishment and maintenance of cell polarity during leukocyte chemotaxis
.
Cell Adh. Migr.
1
,
69
76
.
Gordón-Alonso
M.
,
Veiga
E.
,
Sánchez-Madrid
F.
(
2010
).
Actin dynamics in the immunological synapse
.
Cell Health Cytoskel.
2
,
33
47
.
Grootjans
J. J.
,
Zimmermann
P.
,
Reekmans
G.
,
Smets
A.
,
Degeest
G.
,
Durr
J.
,
David
G.
(
1997
).
Syntenin, a PDZ protein that binds syndecan cytoplasmic domains
.
Proc. Natl. Acad. Sci. USA
94
,
13683
13688
.
Helmke
B. M.
,
Polychronidis
M.
,
Benner
A.
,
Thome
M.
,
Arribas
J.
,
Deichmann
M.
(
2004
).
Melanoma metastasis is associated with enhanced expression of the syntenin gene
.
Oncol. Rep.
12
,
221
228
.
Ilani
T.
,
Vasiliver-Shamis
G.
,
Vardhana
S.
,
Bretscher
A.
,
Dustin
M. L.
(
2009
).
T cell antigen receptor signaling and immunological synapse stability require myosin IIA
.
Nat. Immunol.
10
,
531
539
.
Ivashkiv
L. B.
(
2009
).
Cross-regulation of signaling by ITAM-associated receptors
.
Nat. Immunol.
10
,
340
347
.
Kang
B. S.
,
Cooper
D. R.
,
Jelen
F.
,
Devedjiev
Y.
,
Derewenda
U.
,
Dauter
Z.
,
Otlewski
J.
,
Derewenda
Z. S.
(
2003
).
PDZ tandem of human syntenin: crystal structure and functional properties
.
Structure
11
,
459
468
.
Koga
Y.
,
Ikebe
M.
(
2005
).
p116Rip decreases myosin II phosphorylation by activating myosin light chain phosphatase and by inactivating RhoA
.
J. Biol. Chem.
280
,
4983
4991
.
Kolsch
V.
,
Charest
P. G.
,
Firtel
R. A.
(
2008
).
The regulation of cell motility and chemotaxis by phospholipid signaling
.
J. Cell Sci.
121
,
551
559
.
Latysheva
N.
,
Muratov
G.
,
Rajesh
S.
,
Padgett
M.
,
Hotchin
N. A.
,
Overduin
M.
,
Berditchevski
F.
(
2006
).
Syntenin-1 is a new component of tetraspanin-enriched microdomains: mechanisms and consequences of the interaction of syntenin-1 with CD63
.
Mol. Cell. Biol.
26
,
7707
7718
.
Ludford-Menting
M. J.
,
Oliaro
J.
,
Sacirbegovic
F.
,
Cheah
E. T.
,
Pedersen
N.
,
Thomas
S. J.
,
Pasam
A.
,
Iazzolino
R.
,
Dow
L. E.
,
Waterhouse
N. J.
, et al.
. (
2005
).
A network of PDZ-containing proteins regulates T cell polarity and morphology during migration and immunological synapse formation
.
Immunity
22
,
737
748
.
Luyten
A.
,
Mortier
E.
,
Van Campenhout
C.
,
Taelman
V.
,
Degeest
G.
,
Wuytens
G.
,
Lambaerts
K.
,
David
G.
,
Bellefroid
E.
,
Zimmermann
P.
(
2008
).
The Postsynaptic Density 95/Disc-Large/Zona Occludens Protein Syntenin Directly Interacts with Frizzled 7 and Supports Noncanonical Wnt Signaling
.
Mol. Biol. Cell
19
,
1594
1604
.
Martín-Cófreces
N.
,
Robles-Valero
J.
,
Cabrero
J.
,
Mittelbrunn
M.
,
Gordón-Alonso
M.
,
Sung
C.
,
Alarcón
B.
,
Vázquez
J.
,
Sánchez-Madrid
F.
(
2008
).
MTOC translocation modulates IS formation and controls sustained T cell signaling
.
J. Cell Biol.
182
,
951
962
.
Meerschaert
K.
,
Bruyneel
E.
,
De Wever
O.
,
Vanloo
B.
,
Boucherie
C.
,
Bracke
M.
,
Vandekerckhove
J.
,
Gettemans
J.
(
2007
).
The tandem PDZ domains of syntenin promote cell invasion
.
Exp. Cell Res.
313
,
1790
1804
.
Mittelbrunn
M.
,
Yanez-Mo
M.
,
Sancho
D.
,
Ursa
A.
,
Sanchez-Madrid
F.
(
2002
).
Cutting edge: dynamic redistribution of tetraspanin CD81 at the central zone of the immune synapse in both T lymphocytes and APC
.
J. Immunol.
169
,
6691
6695
.
Ono
R.
,
Matsuoka
J.
,
Yamatsuji
T.
,
Naomoto
Y.
,
Tanaka
N.
,
Matsui
H.
,
Matsushita
M.
(
2008
).
M-RIP, a novel target of JNK signaling and a requirement for human cancer cell invasion
.
Int. J. Mol. Med.
22
,
199
203
.
Ouyang
M.
,
Sun
J.
,
Chien
S.
,
Wang
Y.
(
2008
).
Determination of hierarchical relationship of Src and Rac at subcellular locations with FRET biosensors
.
Proc. Natl. Acad. Sci. USA
105
,
14353
14358
.
Palamidessi
A.
,
Frittoli
E.
,
Garré
M.
,
Faretta
M.
,
Mione
M.
,
Testa
I.
,
Diaspro
A.
,
Lanzetti
L.
,
Scita
G.
,
Di Fiore
P.
(
2008
).
Endocytic trafficking of Rac is required for the spatial restriction of signaling in cell migration
.
Cell
134
,
135
147
.
Rey
M.
,
Valenzuela-Fernandez
A.
,
Urzainqui
A.
,
Yanez-Mo
M.
,
Perez-Martinez
M.
,
Penela
P.
,
Mayor
F.
Jr
,
Sanchez-Madrid
F.
(
2007
).
Myosin IIA is involved in the endocytosis of CXCR4 induced by SDF-1alpha
.
J. Cell Sci.
120
,
1126
1133
.
Rudd
C. E.
(
1990
).
CD4, CD8 and the TCR-CD3 complex: a novel class of protein-tyrosine kinase receptor
.
Immunol. Today
11
,
400
406
.
Sai
J.
,
Raman
D.
,
Liu
Y.
,
Wikswo
J.
,
Richmond
A.
(
2008
).
Parallel phosphatidylinositol 3-kinase (PI3K)-dependent and Src-dependent pathways lead to CXCL8-mediated Rac2 activation and chemotaxis
.
J. Biol. Chem.
283
,
26538
26547
.
Sala-Valdes
M.
,
Ursa
A.
,
Charrin
S.
,
Rubinstein
E.
,
Hemler
M. E.
,
Sanchez-Madrid
F.
,
Yanez-Mo
M.
(
2006
).
EWI-2 and EWI-F Link the Tetraspanin Web to the Actin Cytoskeleton through Their Direct Association with Ezrin-Radixin-Moesin Proteins
.
J. Biol. Chem.
281
,
19665
19675
.
Sanchez-Madrid
F.
,
Serrador
J. M.
(
2009
).
Bringing up the rear: defining the roles of the uropod
.
Nat. Rev. Mol. Cell Biol.
10
,
353
359
.
Sarkar
D.
,
Boukerche
H.
,
Su
Z. Z.
,
Fisher
P. B.
(
2004
).
mda-9/syntenin: recent insights into a novel cell signaling and metastasis-associated gene
.
Pharmacol. Ther.
104
,
101
115
.
Sarkar
D.
,
Boukerche
H.
,
Su
Z. Z.
,
Fisher
P. B.
(
2008
).
mda-9/Syntenin: more than just a simple adapter protein when it comes to cancer metastasis
.
Cancer Res.
68
,
3087
3093
.
Shaw
A. S.
,
Filbert
E. L.
(
2009
).
Scaffold proteins and immune-cell signalling
.
Nat. Rev. Immunol.
9
,
47
56
.
Surks
H. K.
,
Richards
C. T.
,
Mendelsohn
M. E.
(
2003
).
Myosin phosphatase-Rho interacting protein. A new member of the myosin phosphatase complex that directly binds RhoA
.
J. Biol. Chem.
278
,
51484
51493
.
Urzainqui
A.
,
Serrador
J. M.
,
Viedma
F.
,
Yanez-Mo
M.
,
Rodriguez
A.
,
Corbi
A. L.
,
Alonso-Lebrero
J. L.
,
Luque
A.
,
Deckert
M.
,
Vazquez
J.
, et al.
. (
2002
).
ITAM-based interaction of ERM proteins with Syk mediates signaling by the leukocyte adhesion receptor PSGL-1
.
Immunity
17
,
401
412
.
Vicente-Manzanares
M.
,
Sanchez-Madrid
F.
(
2004
).
Role of the cytoskeleton during leukocyte responses
.
Nat. Rev. Immunol.
4
,
110
122
.
Vicente-Manzanares
M.
,
Cabrero
J. R.
,
Rey
M.
,
Perez-Martinez
M.
,
Ursa
A.
,
Itoh
K.
,
Sanchez-Madrid
F.
(
2002
).
A role for the Rho-p160 Rho coiled-coil kinase axis in the chemokine stromal cell-derived factor-1alpha-induced lymphocyte actomyosin and microtubular organization and chemotaxis
.
J. Immunol.
168
,
400
410
.
Vicente-Manzanares
M.
,
Cruz-Adalia
A.
,
Martin-Cofreces
N. B.
,
Cabrero
J. R.
,
Dosil
M.
,
Alvarado-Sanchez
B.
,
Bustelo
X. R.
,
Sanchez-Madrid
F.
(
2005
).
Control of lymphocyte shape and the chemotactic response by the GTP exchange factor Vav
.
Blood
105
,
3026
3034
.
Yu
H.
,
Leitenberg
D.
,
Li
B.
,
Flavell
R.
(
2001
).
Deficiency of small GTPase Rac2 affects T cell activation
.
J. Exp. Med.
194
,
915
926
.
Zimmermann
P.
,
Meerschaert
K.
,
Reekmans
G.
,
Leenaerts
I.
,
Small
J. V.
,
Vandekerckhove
J.
,
David
G.
,
Gettemans
J.
(
2002
).
PIP(2)-PDZ domain binding controls the association of syntenin with the plasma membrane
.
Mol. Cell
9
,
1215
1225
.
Zimmermann
P.
,
Zhang
Z.
,
Degeest
G.
,
Mortier
E.
,
Leenaerts
I.
,
Coomans
C.
,
Schulz
J.
,
N’Kuli
F.
,
Courtoy
P. J.
,
David
G.
(
2005
).
Syndecan recycling is controlled by syntenin-PIP2 interaction and Arf6
.
Dev. Cell
9
,
377
388
.

Supplementary information