Skip to main content

Main menu

  • Home
  • Articles
    • Current Issue
    • Next in The JI
    • Archive
    • Brief Reviews
    • Pillars of Immunology
    • Translating Immunology
    • Most Read
    • Top Downloads
    • Annual Meeting Abstracts
  • COVID-19/SARS/MERS Articles
  • Info
    • About the Journal
    • For Authors
    • Journal Policies
    • Influence Statement
    • For Advertisers
  • Editors
  • Submit
    • Submit a Manuscript
    • Instructions for Authors
    • Journal Policies
  • Subscribe
    • Journal Subscriptions
    • Email Alerts
    • RSS Feeds
    • ImmunoCasts
  • More
    • Most Read
    • Most Cited
    • ImmunoCasts
    • AAI Disclaimer
    • Feedback
    • Help
    • Accessibility Statement
  • Other Publications
    • American Association of Immunologists
    • ImmunoHorizons

User menu

  • Subscribe
  • My alerts
  • Log in
  • Log out

Search

  • Advanced search
The Journal of Immunology
  • Other Publications
    • American Association of Immunologists
    • ImmunoHorizons
  • Subscribe
  • My alerts
  • Log in
  • Log out
The Journal of Immunology

Advanced Search

  • Home
  • Articles
    • Current Issue
    • Next in The JI
    • Archive
    • Brief Reviews
    • Pillars of Immunology
    • Translating Immunology
    • Most Read
    • Top Downloads
    • Annual Meeting Abstracts
  • COVID-19/SARS/MERS Articles
  • Info
    • About the Journal
    • For Authors
    • Journal Policies
    • Influence Statement
    • For Advertisers
  • Editors
  • Submit
    • Submit a Manuscript
    • Instructions for Authors
    • Journal Policies
  • Subscribe
    • Journal Subscriptions
    • Email Alerts
    • RSS Feeds
    • ImmunoCasts
  • More
    • Most Read
    • Most Cited
    • ImmunoCasts
    • AAI Disclaimer
    • Feedback
    • Help
    • Accessibility Statement
  • Follow The Journal of Immunology on Twitter
  • Follow The Journal of Immunology on RSS

Antigen Recognition Is Facilitated by Invadosome-like Protrusions Formed by Memory/Effector T Cells

Peter T. Sage, Laya M. Varghese, Roberta Martinelli, Tracey E. Sciuto, Masataka Kamei, Ann M. Dvorak, Timothy A. Springer, Arlene H. Sharpe and Christopher V. Carman
J Immunol April 15, 2012, 188 (8) 3686-3699; DOI: https://doi.org/10.4049/jimmunol.1102594
Peter T. Sage
*Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115
†Department of Medicine, Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Laya M. Varghese
†Department of Medicine, Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Roberta Martinelli
†Department of Medicine, Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Tracey E. Sciuto
‡Department of Pathology, Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215; and
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Masataka Kamei
†Department of Medicine, Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Ann M. Dvorak
‡Department of Pathology, Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215; and
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Timothy A. Springer
§Department of Pathology, Immune Disease Institute, Harvard Medical School, Boston, MA 02115
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Arlene H. Sharpe
*Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Christopher V. Carman
†Department of Medicine, Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF + SI
  • PDF
Loading

Abstract

Adaptive immunity requires that T cells efficiently scan diverse cell surfaces to identify cognate Ag. However, the basic cellular mechanisms remain unclear. In this study, we investigated this process using vascular endothelial cells, APCs that possess a unique and extremely advantageous, planar morphology. High-resolution imaging revealed that CD4 memory/effector T cells dynamically probe the endothelium by extending submicron-scale, actin-rich “invadosome/podosome-like protrusions” (ILPs). The intimate intercellular contacts enforced by ILPs consistently preceded and supported T cell activation in response to endothelial MHC class II/Ag. The resulting calcium flux stabilized dense arrays of ILPs (each enriched in TCR, protein kinase C-θ, ZAP70, phosphotyrosine, and HS1), forming what we term a podo-synapse. Similar findings were made using CD8 CTLs on endothelium. Furthermore, careful re-examination of both traditional APC models and professional APCs suggests broad relevance for ILPs in facilitating Ag recognition. Together, our results indicate that ILPs function as sensory organelles that serve as actuators of immune surveillance.

Adaptive immunity relies on the ability of TCRs expressed on lymphocytes to efficiently recognize peptide Ag bound to MHC molecules (1). During the priming phase, naive lymphocytes must constitutively scan professional APCs within lymph nodes. During the effector phase, memory/effector T cells need to effectively survey an extremely wide range of APCs and potential target cells within peripheral tissues. Fundamental cellular mechanisms for such immune surveillance activities remain incompletely understood.

At the heart of immune surveillance is the requirement for T cells to form extremely intimate (∼14 nm, the total height of the TCR–MHC complex) (2) contacts with apposing cells to sample MHC/Ag. However, the surfaces of all cells are modified by a relatively thick (50–500 nm) gel-like polysaccharide coat termed the glycocalyx (3), which provides a formidable energy barrier (via steric and electrostatic repulsion) to close membrane–membrane encounter (4, 5). In this way, TCR and MHC are effectively shielded and immune recognition is opposed (5–10). Thus, a fundamental question is, how do T cells overcome this energy barrier to engage immune receptors?

It has long been appreciated that as a consequence of Ag recognition, specialized cell–cell interfaces form that involve membrane alignment, cytoskeletal remodeling, clustering/segregation of immune receptors/adhesion molecules, and glycocalyx components (5, 11–15). These “immunological synapses” (ISs) form within minutes of Ag recognition and are thought to amplify and sustain signaling, as well as facilitate exchange of cytokine and/or cytotoxic materials (5, 11–15). However, detailed mechanisms facilitating initial Ag sampling between T cells and APCs, and how these lead to early IS formation, are not completely understood (16, 17). This stems from technical challenges associated with irregular topologies of APC surfaces and poorly controlled orientation of the cell–cell interaction planes, issues that profoundly limit the requisite imaging approaches (16–18).

Investigators have partially circumvented these restrictions by developing planar substrate models (i.e., lipid bilayers and Ab-coated surfaces) that provide optimal spatiotemporal resolution for monitoring Ag response dynamics (19–23). These models have afforded invaluable insights, such as the discovery of TCR microclusters as critical mediators of effective signaling (15, 19–22). However, these systems lack key features of cellular APC surfaces, such as the glycocalyx, topological deformability, and molecular complexity. Therefore, it remains uncertain how to translate such findings to physiologic cell–cell immune surveillance.

In this study, we used vascular endothelial cells (ECs) as a planar APC model to interrogate the details of initial Ag recognition dynamics on a physiologic cellular substrate. ECs represent the interface between the blood circulation and tissue, and play critical roles in regulating immune cell trafficking (1, 24, 25). The discovery that ECs express MHC class I (MHC-I), MHC class II (MHC-II), and a large number of costimulatory molecules (e.g., CD40, LFA-3, ICOSL, 4-1BB, OX40L, TL1A, PD-L1, but not CD80 and CD86) has led to the controversial hypothesis that endothelium can also function as a type of APC (26–28). Indeed, several studies have demonstrated that endothelium can effectively restimulate CD4 and CD8 memory/effector, but not naive, T cells (29–33). Critically, when grown in vitro, ECs form virtually planar cell surfaces that are ideal for high spatiotemporal resolution imaging of topological dynamics (34).

This study provides a detailed investigation of the initial events in CD4 and CD8 T cell scanning for Ag. We previously discovered that lymphocytes actively probe the surface of the endothelium by dynamic insertion and retraction of submicron-scale, actin-rich cylindrical protrusions related to invadosomes (35), termed invadosome/podosome-like protrusions (ILPs) (34). These were demonstrated to function in supporting migratory pathfinding (34, 36). In this study, we found that dynamic ILP probing by CD4 and CD8 memory/effector lymphocytes enforces close T cell–EC apposition, which seems to facilitate Ag recognition and TCR signaling. Moreover, T cell activation is sustained through a novel IS architecture dominated by dense arrays of calcium-stabilized ILPs (each enriched in signaling molecules) that we term a podo-synapse. Complementary studies with model substrates and professional APCs (B cells and dendritic cells [DCs]) suggest that rather than being a unique feature of T cell–EC ISs, ILPs are uniquely revealed in this setting. Together, our results indicate that memory/effector T cells use ILPs to facilitate efficient search for Ag and to help sustain the resulting signaling responses.

Materials and Methods

Cells

Natural human T memory cells (nTmem; CD4+CD45RO+CD45RA−) were isolated via negative selection from peripheral blood using a CD4 T cell isolation kit, followed by CD45RA+ cell depletion (Miltenyi Biotec) to >95% purity. Induced/expanded human T memory cells (iTmem; CD4+CD45RO+) were made by culturing peripheral blood buffy coats for 72 h in 1 μg/ml bacterial superantigens staphylococcal enterotoxin B (SEB) and toxic shock syndrome toxin 1 (TSST; Toxin Technology). CD4 memory cells were enriched by using a CD4 T cell memory negative isolation kit (Miltenyi Biotec). Cells were cultured 3–10 d in 20 ng/ml IL-15 (R&D Systems) to maintain memory phenotype. Mouse CD4 or CD8 T cells were isolated by magnetic separation from either OTII+TCRα−/−CD45.1+/+ or OTI+Thy1.1+/+ mice and activated with irradiated splenocytes in the presence of 30 μg/ml OVA 323–339 (OTII) or 500 ng/ml OVA-SIINFEKL (OTI) supplemented with IL-2 for 3 d. T cells were reselected with MACS beads and rested in culture in the presence of IL-2 for a further 3 d. Human lung microvascular ECs (HLMVECs), human dermal microvascular ECs (HDMVECs), and human umbilical vein ECs (HUVECs) were from Lonza and cultured on fibronectin (10 μg/ml; Invitrogen) in EBM-2 MV media (Lonza). Mouse heart ECs were isolated from C57BL/6 mice by collagenization of heart tissue followed by positive selection with anti-CD31 (MEC13.3)–coated beads (Invitrogen). ECs were cultured for 7 d in media with EC growth supplement (Biomedical Technologies) followed by reselection with beads coupled to anti-CD102. ECs were >95% purity based on expression of CD105+CD102+. ECs were transfected by Amaxa electroporation according to manufacturer’s instructions (Lonza). Palmitoylated YFP (mem-YFP) and soluble monomeric DsRed were from Clontech. Palmitoylated DsRed (mem-DsRed) and ICAM-1–GFP were generated as described previously (34). ECs were activated for 48 h with 100 ng/ml IFN-γ (R&D Systems) and 12 h with 50 ng/ml TNF-α (R&D Systems). Priess B cell line was a gift from Dr. Kai Wucherpfennig. Chinese hamster ovary (CHO)-K1 epithelial cells were from American Type Culture Collection. Murine bone marrow-derived DCs (BMDCs) were prepared from bone marrow cells cultured in 30 ng/ml GM-CSF (PeproTech) for 6 d.

Transendothelial migration

Lymphocyte transendothelial migration assays were conducted in both static and shear conditions as described previously (34). In some cases, T cells were added to 8-μm pore transwell inserts that were preceded with ECs and then collected from the bottom chamber 6 h later and analyzed by flow cytometry.

Live-cell imaging and analysis

Live-cell imaging was performed on an Axiovert S200 microscope with Axiovision software (Zeiss). For calcium imaging, T cells were preincubated with 1 μg/ml Fura-2–AM or Fluo-4–AM (Invitrogen) for 30 min. For Fura-2, standard Fura-2 filters were used according to manufacturer’s instructions (Chroma) and ratiometric calcium flux was calculated (340nmEX-510nmEM-background/380nmEX-510nmEM-background) for each cell using Axiovision. For Fluo-4, green fluorescence filters were used and basal fluorescence signal was established by imaging cells on uncoated chambers. T cell polarity was calculated from bright-field images as the ratio of the greatest edge-to-edge dimension in the x–y plane (i.e., the long axis) divided by the perpendicular axis. Migration velocity was measured for cells using the Axiovision Cell Tracking Module. Podo-print/ILP lifetimes were assessed using podo-print appearance and disappearance on mem-dsRed– or mem-YFP–transfected ECs as a readout for the presence of ILPs (34). Podo-print/ILP lifetimes were calculated as follows: the last time point when an individual podo-print/ILP was visible − the time point when that podo-print/ILP first appeared. Podo-print/ILP lateral translocation was assessed by determining the distance between the T cell centroid and the first and last positions of an individual podo-print/ILP during its lifetime. Podo-print/ILP index, a measure of the total number of ILPs per cell, was calculated at 5 min after addition of T cells. “Offset time” was used to describe relative time between first podo-print/ILP formation and calcium flux rise above background. Temporal resolution was limited to 10 s, the maximal achievable acquisition frame rate of this analysis. Data was then sorted into 10-s interval bins.

For selected studies, lymphocytes were preincubated, or lymphocyte and APCs coincubated, with select pharmaceutical agents, including latrunculin A (1 μg/ml; Sigma), BAPTA-AM (40 μM, Sigma), BTP2 (20 μM; Calbiochem), thapsigargin (1 μM Sigma), and calcium-calmodulin–dependent kinase II (CAMKII) inhibitor CK59 (50 μM; EMD Biosciences) as indicated. For calcium blockade conditions, T cells were pretreated with BAPTA-AM and BTP2 in imaging buffer containing high (45 mM) potassium. T cells were added to ECs and imaged with additional BTP2 for the duration of imaging.

Fixed cell imaging

Fixed cell samples were imaged on an LSM 510 confocal microscope (Zeiss). Samples were fixed and stained for αL integrin (TS2/4), ICAM-1 (IC1/11), F-actin (phalloidin), MHC-II (WR18; Abcam), protein kinase C (PKC)-θ, talin (rabbit anti-talin, a gift from Dr. Keith Burridge), CD3 (OKT3; eBioscience), CD43 (L60; BD Biosciences), CD45 (HI30 or UCHL1; eBioscience), hematopoietic lineage cell-specific protein 1 (HS1; clone 9 [BD Biosciences] or rabbit anti-HS1 [Cell Signaling]), ZAP70 (Santa Cruz), phosphotyrosine (4G10 platinum; Millipore), or NFAT (BD Biosciences). Transmigration was analyzed by confocal microscopy as described previously (37).

CTL killing assay

For specific killing assay, murine ECs were labeled with either 0.6 or 0.02 μM CFSE (CFSEhi and CFSElow, respectively). The CFSEhi population was pulsed with 1 μg/ml SIINFEKL for 20 min. Both CFSEhi and CFSElow populations were plated together in a 1:1 ratio. Activated OTI CD8 T cells were rested for 5 h and added to monolayers in a 3:1 T cell/target cell ratio. Cultures were stained for CD105 to differentiate the ECs from T cells and analyzed by flow cytometry. Specific lysis was calculated by the following equation: 1 − ([CFSEhi/(CFSEhi + CFSElow)]/[control CFSEhi/CFSElow])*100.

Planar coated-glass APC model

Coated glass IS models were prepared as described previously (20, 38). In brief, imaging chambers were coated with ICAM-1–Fc (10 μg/ml; R&D Systems) alone or together with anti-CD3, anti-CD43, anti-CD45, or control IgG Ab (10 μg/ml). Fluo-4–labeled lymphocytes were added and imaged by fluorescence and interference reflection microscopy (IRM).

Coated cell APC model

CHO-K1 cells were transfected with ICAM-1-GFP and soluble dsRed via electroporation. Cells were then coated with Abs via a biotin/streptavidin (SA)-based approach (39). Cells were subjected to primary amine-targeted cell surface biotinylation for 15 min with biotin-NHS-ester (0.5 μg/ml; Thermo Electron). Cells were incubated with SA at 10 μg/ml for 15 min. Cells were then incubated with biotinylated anti-CD3, -CD43, or -CD45 (eBioscience) at 10 μg/ml for 15 min.

T cell activation by CD3 cross-linking

Fluo-4–labeled T cells were settled onto uncoated glass chambers, 10 μg/ml anti-CD3 and -CD28 were added to the imaging chambers, and calcium was monitored.

Transmission electron microscopy

Transmission electron microscopy of T cell–APC complexes was performed as described previously (34). Fifty randomly selected micrographs were analyzed by enumerating zones of membrane apposition (<20 nm) and correlating them with respect to ILP location.

Statistical analysis

Error bars represent SE or SD as indicated. The p values were calculated via unpaired Student t tests in GraphPad Prism. Statistical significance is indicated with p values as follows: ***p < 0.0005, **p < 0.005, *p < 0.05.

Results

Endothelial-presented Ag promotes activation of CD4 memory T cells

We wanted to elucidate the mechanisms by which memory/effector T cells probe for Ag using ECs as model APCs. Therefore, we isolated natural CD4 memory-like lymphocytes (nTmem) and generated induced/expanded CD4 memory-like lymphocytes (iTmem) from human peripheral blood. The isolated nTmem were CD4+, CD45RO+, and contained both central memory and effector memory T cells (∼78 and ∼22%, respectively) based on CD62L staining (Supplemental Fig. 1A). The iTmem were slightly more polarized toward the effector memory T cell subtype (∼69 versus ∼31%; Supplemental Fig. 1B). Monolayers of HLMVECs, HDMVECs, and HUVECs were used as models for the vasculature. In vivo endothelial MHC-II expression is dependent on IFN-γ (26). This was recapitulated in vitro by culturing ECs with exogenous IFN-γ (Supplemental Fig. 1C). Endothelium was additionally activated with TNF-α to promote an inflamed phenotype.

Next, we set up live-cell analysis to concomitantly monitor T cell activation (i.e., calcium flux) and migration on endothelium pulsed with Ag (i.e., bacterial superantigen, a widely used model Ag) (38, 40, 41). In the absence of Ag, nTmem and iTmem fluxed little calcium (Fig. 1A), remained polarized (Fig. 1B), and underwent continuous lateral (Fig. 1C) and transendothelial migration (i.e., “diapedesis”; Fig. 1D). On HLMVECs pulsed with Ag (1 μg/ml SEB and TSST), T cells rapidly fluxed calcium, lost polarity, and arrested migration (Fig. 1A–C). Similar results were found with alternate Ag (e.g., SEE and MAM) presented by HDMVECs or HUVECs (Supplemental Fig. 1D and data not shown). In addition, in the absence of Ag, the majority of iTmem transmigrated within 30 min, whereas in the presence of Ag, diapedesis was delayed by ∼30–60 min (Fig. 1D). Thus, the migratory stop signal was transient. ITmem also exhibited Ag-dependent transient migration arrest under conditions of laminar fluid shear flow conditions similar to those found in microvasculature in vivo (2 dyne/cm2; Fig. 1E–G, Supplemental Video 1).

FIGURE 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 1.

Human microvascular ECs are capable of Ag-specific stimulation of CD4 memory-like T cells. (A) Human CD4 nTmem (circles) or iTmem (triangles) were loaded with Fura-2 and incubated on Ag-pulsed HLMVECs. Calcium flux at 5 min (left) or at maximal value over a 30-min duration (right) were plotted. (B and C) iTmem were incubated on HLMVEC pulsed with Ag and T cell polarity (length/width ratio) at 5 min, and velocity was calculated over a 30-min duration. (D) iTmem transendothelial migration was scored on Ag pulsed ECs as described in Materials and Methods. (E) iTmem were infused over Ag-pulsed HLMVEC at 2 dyne/cm2 and imaged. Depicted is an end-point image with migration tracks of apical (blue) and transmigrated (red) lymphocytes. See also Supplemental Video 1. (F and G) Lateral migration velocities for T cells from (E) during both prediapedesis and postdiapedesis phases of migration over 30 min (F) and percentage of iTmem transmigrated were measured (G). (H) Representative images of nTmem incubated on Ag-pulsed HLMVEC and staining for actin (green), NFAT (red), and nucleus (blue). (I) nTmem were incubated on activated HLMVEC. Nuclear NFAT translocation was scored according to line scan profiles (left). (J) Samples were acquired as in (H), and representative transmigrating or transmigrated (‘Under’) lymphocytes are shown. Data represent mean ± SD (B, C) or SEM (D, F, G, I). Scale bars, 5 μm. *p < 0.05, **p < 0.005, ***p < 0.0005.

During T cell activation, the transcription factor NFAT translocates from the cytoplasm to the nucleus. Incubation of nTmem on Ag-pulsed ECs promoted robust nuclear translocation of NFAT (Fig. 1H, 1I). Significantly, transmigrating nTmem and iTmem retained nuclear NFAT, demonstrating that these cells remained activated (Fig. 1J). NTmem on endothelium that was pulsed with Ag but not pretreated with IFN-γ (and therefore lacked strong MHC-II expression) exhibited little NFAT translocation (Fig. 1I), confirming that the responses to Ag-pulsed endothelium were MHC-II dependent.

To show that T cells achieved complete activation, we conducted studies using a transwell system, in which T cells loaded into an upper chamber migrated across an EC monolayer to reach the lower chamber. Flow cytometry revealed that after migrating across Ag-pulsed ECs, iTmem showed increased surface expression of CD69 and a reduction in CD62L (Supplemental Fig. 1E). In addition, in these nonpolarizing conditions, a subset of the iTmem upregulated IFN-γ expression (Supplemental Fig. 1F, 1G) and increased proliferation (Supplemental Fig. 1H). Collectively, these results demonstrate that endothelium is able to present model Ag in an MHC-II–dependent manner to CD4 memory/effectorlymphocytes, which induces a transient delay in diapedesis and T cell activation.

ILP arrays dominate the T cell–EC interface during activation

Next, we investigated the cellular and molecular basis for T cell activation in our endothelial APC model. Previously, we used fluorescent membrane markers (mem-YFP or mem-DsRed) expressed in the ECs to detect topological changes in the plasma membrane (34). In this way, we demonstrated that ∼0.5-μm fluorescent rings that formed dynamically on the endothelium under migrating T cells corresponded to cylinder-shaped cell surface invaginations (i.e., “podo-prints”) induced by lymphocyte ILPs. Thus, podo-prints formed on the EC surface served as an indirect but sensitive readout for T cell ILPs (34). In this study, we similarly observed in control settings that podo-prints formed and disappeared continuously (with lifetimes of tens of seconds) under the leading edge lamellipodia of migrating iTmem (Fig. 2A, 2C, Supplemental Video 2). Strikingly, when ECs were pulsed with Ag, iTmem rapidly formed dense arrays of podo-prints largely localized at the periphery of the T cell–EC interface under symmetrical lamellipodia (Fig. 2B, example 1, and Supplemental Video 3). A minority (∼20–30%) of the iTmem cells formed rosette-type podo-print arrays that lacked bias for the cell periphery (Fig. 2B, example 2, and Supplemental Video 4). In both cases, podo-prints/ILPs were significantly stabilized (lifetimes of ∼18 min; Fig. 2C) and exhibited limited lateral translocation (0.31 ± 1.5 μm; Fig. 2D–F). After ∼30 min of contact, ILPs began to disappear and T cells initiated transmigration (Supplemental Video 4 and data not shown).

FIGURE 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 2.

CD4 memory-like T cells form stabilized arrays of ILPs on Ag-presenting endothelium. (A and B) HLMVECs were transfected with mem-YFP or mem-DsRed, activated, and pulsed without (A) or with (B) Ag (TSST/SEB). iTmem were imaged live on addition to ECs. Individual frames at selected time intervals are shown. Arrows indicate fluorescent rings formed on endothelium under adherent lymphocytes. The corresponding control (A) and Ag (b1 and b2) videos are provided as Supplemental Videos 2–4, respectively. (C) Podo-print/ILP lifetimes from imaging studies as in (A) and (B). Data represent mean ± SEM of at least 80 ILPs formed by at least 25 cells from ≥3 separate experiments. (D) Schematic representation of podo-print/ILP lateral translocation analysis. Gray “T cell outline” represents a T cell–EC contact area with the “T cell centroid” indicated as a blue circle and individual podo-prints indicated as black rings. The linear distance between the centroid and podo-print was measured at both the first and last appearance of an individual podo-print/ILP during its lifetime. (E and F) Podo-print/ILP distances were measured as in (D). Distances from the cell centroid at time of formation and disappearance were plotted for individual podo-prints (E). Data from (E) was further processed to report change in distance (F). Scale bars, 5 μm. ***p < 0.0005.

As confirmation that the earlier observations reflected three-dimensional podo-prints in response to T cell ILPs, we cotransfected endothelium with mem-DsRed together with soluble GFP as a marker for cytoplasmic volume (34). This showed individual fluorescent membrane rings of each podo-print, in fact, represented cytosol-displacing invaginations into the EC surface (Fig. 3A). Furthermore, confocal imaging showed that podo-prints were ICAM-1–enriched cylindrical EC invaginations into which T cell ILPs extended (Fig. 3B, Supplemental Video 5). ILPs were enriched in LFA-1 (Fig. 3B), F-actin (Fig. 3C), and talin (Fig. 3D), similarly to invadosomes (35, 36). Analogous structures formed under physiologic shear flow (Supplemental Fig. 2A), with diverse ECs (including HLMVECs [Fig. 3A, 3C, 3D], HUVECs [Fig. 3B], HDMVECs [data not shown]), with nTmem (Supplemental Fig. 2B), and with alternate Ag (Fig. 3B, Supplemental Fig. 2D). Similar structures also formed when previously activated murine OT-II CD4+ T cells were incubated on heart microvascular ECs pulsed with OVA 323–339 peptide Ag (Supplemental Fig. 2E).

FIGURE 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 3.

Ag-stabilized ILPs exhibit a discrete three-dimensional architecture. (A) Ag-stabilized T cell ILPs protrude into the EC surface. Imaging was conducted as in Fig. 2B on HLMVECs coexpressing soluble, cytoplasmic GFP (green) and mem-DsRed (red). (B) Three-dimensional reconstruction from confocal imaging of podo-prints and ILPs. iTmem were incubated for 20 min on activated Ag-pulsed (SEE) HUVECs and then fixed, stained for ICAM-1 (green) and LFA-1(red), and imaged by confocal microscopy. Sections were digitally reconstructed and projected as three-dimensional renderings. (b) Magnified view of the ILP arrays. (b′) Orthogonal cross section. See also Supplemental Video 5. (C and D) Ag-stabilized ILPs are enriched in actin and talin. Mem-YFP–transfected (C; green) or mem-DsRed–transfected (D; red) HLMVECs were pulsed with Ag (TSST/SEB) and incubated with iTmem for 5 min and stained for F-actin (C; red) or talin (D; green). (E) Quantitation of ultrastructural depth and width of T cell ILPs. Samples as in (C) were imaged by electron microscopy and ILPs were measured. Data represent mean ± SEM from at least 100 ILPs from at least 20 representative micrographs per condition. (F and G) ILPs enforce close T cell–EC membrane apposition in the absence and presence of Ag. T cell and endothelial nuclei are indicated with red and green overlays, respectively. Arrows and (a) highlight regions of extremely close lymphocyte–EC membrane apposition enforced at the ILP tips. Scale bars, 5 μm (A–D); 500 nm (F, G). *p < 0.05. ns, Not significant.

To assess the T cell–EC interaction in greater detail, we used transmission electron microscopy. In the absence of Ag, ILPs averaged 430 ± 34 nm in depth and 348 ± 33 nm in width (Fig. 3E, 3F). In the presence of Ag, ILPs were similar in morphology and size (depth = 437 nm, width = 277 nm), but tended to form in denser clusters (Fig. 3E–G). An important feature of the ILPs, whether in the absence or presence of Ag, was the existence of zones of extremely close T cell–EC apposition, typically at the tips of the ILPs as if driven by ILP extension (Fig. 3F, 3G, arrows). Although not exclusive to these locations, quantitative analysis revealed that intercellular contacts of <20 nm were 9-fold greater at ILP tips compared with other regions. The idea that ILPs can exert significant force is supported by their ability to both drive transcellular diapedesis (34) and to displace/deform organelles such as the nucleus (Fig. 3F).

Ag-stabilized ILPs share features of traditional TCR signaling microclusters

To address how these ILP-dominated cell–cell interfaces relate to TCR signaling, we stained for traditional IS markers (15, 20, 21). Confocal microscopy revealed that podo-prints on endothelium were modestly, but consistently, enriched in MHC-II compared with MHC-I or mem-YFP (Fig. 4A, 4B, Supplemental Fig. 3A), whereas T cell ILPs were enriched in CD3 (Fig. 4C, Supplemental Fig. 3B). Comparable enrichment formed under physiologic shear flow (Fig. 4I, 4J). In <5% of T cells, CD3 was distributed into a central supramolecular activation cluster-like cluster rather than in peripheral ILPs (Supplemental Fig. 3C).

FIGURE 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 4.

Ag-stabilized ILPs are foci for immune signaling. ITmem cells were incubated with activated, Ag-pulsed (TSST/SEB) HLMVEC for 5 min, fixed, and stained as indicated and imaged by confocal microscopy. (A) Ag-stabilized ILPs (F-actin, green) protrude into MHC-II (red)–enriched podo-prints. Arrows indicate MHC-II–enriched podo-prints. (B) Samples as in (A) were stained for MHC-II (HLA-DR/DP/DQ; red) and MHC-I (HLA A/B/C; green). Schematic of gated regions of interest is shown on the left and included a region outside of the IS (I), the ILP-rich region of the IS (II), and the central region of the IS (III). Right panels show pixel fluorescence intensity histograms for regions I–III. (C) ILPs (F-actin, blue) protruding into podo-prints (mem-YFP, green) are enriched in CD3 (red). Within individual ILPs, CD3 is predominantly focused at the tip (b1) and, to a lesser extent, the edge (b2). (D) PKC-θ (red) is enriched with CD3 (green) in ILPs. (E–G) ILPs (F-actin, blue) colocalize with ZAP70 (E; red), phosphotyrosine (F; red), and HS1 (G; red). (H) HS1 (red) is highly enriched in ILPs. Cross-sectional views from serial-section confocal microscopy are shown of a lymphocyte adherent to the endothelium-presenting Ag. See three-dimensional rotation in Supplemental Video 6. (I and J) ILP arrays were allowed to form as above with the additional presence of physiologic laminar fluid shear flow (2.0 dyne/cm2; arrow indicates direction). (I) MHC-II (red) and F-actin (green) are shown. (J) CD3 (red) and ICAM-1 (green) are shown. Scale bars, 5 μm.

Molecules implicated in TCR signaling, including PKC-θ (Fig. 4D, Supplemental Fig. 3D), ZAP70 (Fig. 4E), and phosphotyrosine (Fig. 4F) were also enriched in ILP cores, suggesting active signaling at these sites. The cortactin homolog HS1, a known regulator of both podosomes and ISs (38, 42–44), showed particularly strong enrichment (Fig. 4G, 4H, Supplemental Video 6). Alternatively, the inhibitory molecules CD43 and CD45 localized primarily to the region outside of the T cell–EC interface (Supplemental Fig. 3B). These observations suggest that ILPs may serve as discrete loci for TCR signaling with general features similar to TCR signaling microclusters and “multifocal” synapses (15, 21, 22, 45–47).

Collectively, these experiments show that CD4 iTmem activation caused by EC MHC-II/Ag is coupled to cell–cell interfaces dominated by arrays of ILPs that are enriched in TCR signaling molecules. We term this previously undescribed architecture, which shares features of both podosome belts/rosettes and multifocal ISs, a “podo-synapse.”

ILPs support efficient Ag recognition and sustained signaling

To determine the functional relationship between ILP formation and T cell activation, we concomitantly monitored calcium flux and ILP dynamics. In the presence of Ag, calcium flux was evident shortly after the first appearance of ILPs. It then peaked several minutes later and gradually decayed over the following 5–60 min (Fig. 5A, 5B, Supplemental Video 7). Calcium flux always occurred subsequent to (or in the same 10-s interval as) the appearance of at least one ILP, with an average offset time of ∼25 s (Fig. 5C). In turn, stabilized ILP arrays and symmetric lamellipodia became evident in the ∼30–60 s following the increase in calcium (Fig. 5A, 5B, Supplemental Video 7).

FIGURE 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 5.

T cell ILP formation precedes and supports efficient Ag recognition. (A) ITmem were labeled with Fura-2 and imaged live (at a maximal temporal resolution of 10 s) during migration on mem-DsRed–transfected, Ag-pulsed HLMVECs. Upper panels show mem-DsRed. Arrows indicate initial ILP formation. Middle panels indicate calcium flux values on a rainbow scale. Lower panels provide a schematic representation of newly formed (green) and previously formed (in relation to previous field; red) podo-prints. (a) Frame of initial ILP/podo-print. (b) Frame when calcium flux rises above background. (c) Frame when the peripheral ILP array is stabilized. Note this correlates with peak calcium flux. See also Supplemental Video 7. (B) Graphical representation of calcium flux with frames a–c noted. (C) Quantitation of ILP-calcium flux offset time. Live-cell imaging was as in (A) and offset time (time from when first ILP forms until calcium flux rises above background) was calculated. Data were binned into 10-s intervals, and average ± SEM is shown for 35 individual T cells from 3 separate experiments. (D) Imaging was performed as in (A) with additional pretreatment of T cells with latrunculin A before addition to Ag-pulsed EC monolayers. (E) Podo-print/ILP index (average number of podo-prints/ILPs per cell) and average calcium flux at 5 min was calculated. Both analyses are pooled mean ± SEM from three separate experiments. (F) ITmem were labeled with Fluo-4 and imaged live with anti-CD3/CD28 cross-linking with or without latrunculin A pretreatment. Left panels show resting T cells. Right panels show activated T cells imaged 60 s after addition of cross-linking Abs. Arrows indicate de novo formation of micron-scale T cell protrusions. (G) Average calcium flux was calculated from three separate experiments as in (F). Scale bars, 5 μm. Data represent mean ± SEM. *p < 0.05, ***p < 0.0005.

These observations suggested that the close intercellular contacts driven by ILPs may promote initial Ag recognition and TCR triggering. Testing this hypothesis is challenging because both ILPs and TCR signaling are fundamentally dependent on F-actin assembly and many of the same actin regulatory pathways (34, 35, 38, 42–44, 48). Thus, we simply compared effects of F-actin inhibition (via latrunculin A) on T cell activation through an Ag-pulsed APC versus direct TCR cross-linking. When stimulated by Ag-pulsed ECs, iTmem pretreatment with latrunculin A caused 100% blockade of both ILP formation and calcium flux during the first 5 min of coincubation (Fig. 5D, 5E). By contrast, when TCRs were stimulated directly by anti-CD3/CD28 cross-linking, significant (although attenuated by ∼60%), immediate calcium flux was elicited in the presence of latrunculin A (Fig. 5F, 5G), as shown previously (49). From this we conclude that the total latrunculin A-induced blockade of early response to Ag-pulsed ECs reflects a defect in initial Ag recognition/TCR triggering (Fig. 5F, 5G). We speculate that ILP formation may represent the latrunculin A-sensitive process behind this defect.

Calcium flux is necessary and sufficient for Ag-mediated ILP stabilization

Next, we investigated the transition from dynamic ILP probing to formation of stable ILP arrays. Given the correlation between increase in calcium and appearance of ILP arrays, we hypothesized that calcium may be key for stabilizing ILPs. To test this, we pretreated T cells with the calcium chelator BAPTA and the CRAC channel inhibitor BTP2. On Ag-pulsed ECs, this greatly inhibited calcium flux in iTmem, which correlated with strong reduction in the number of stabilized ILPs (Fig. 6A–C). To determine whether calcium flux was sufficient to stabilize ILPs, iTmem were incubated on ECs in the absence of Ag, followed by application of thapsigargin to directly increase intracellular calcium. This caused iTmem already engaged in dynamic ILP probing to arrest migration and stabilize ILP clusters (Fig. 6D–G). Thus, whereas dynamic ILP probing of ECs by iTmem seems to facilitate initial Ag recognition, the resulting increase in calcium is both necessary and sufficient to drive accumulation/stabilization of ILPs into podo-synapses.

FIGURE 6.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 6.

Calcium flux is necessary and sufficient for ILP stabilization. (A) ITmem were Fura-2 labeled, pretreated with the calcium chelator BAPTA and the CRAC channel inhibitor BTP2, and imaged live on Ag-pulsed ECs. Arrows indicate a few sporadically formed podo-prints. (B and C) Podo-print/ILP index (B) and average calcium flux (C) were calculated at 5 min for experiments as in (A). (D) Live-cell imaging was conducted in the absence of Ag before and after addition of the calcium ionophore thapsigargin (at time = ‘0:00’). Arrows indicate ILPs/podo-prints. (E) Correlation of calcium flux with ILP number after addition of thapsigargin. (F) Quantitation of the number of newly formed ILPs in the 2 min before and after addition of thapsigargin. (G) Quantitation of the lifetime of ILPs with or without addition of thapsigargin. (H and I) Imaging and analysis was performed as in (A)–(C) except T cells were pretreated with the CAMKII inhibitor CK59. Data represent mean ± SEM. Scale bars, 5 μm. **p < 0.005, ***p < 0.0005.

The earlier findings point to a positive feedback loop for sustaining TCR signaling. To break this loop, we sought to block proximal signaling downstream of calcium flux, specifically targeting CAMKII via the inhibitor CK59 (50). Treatment with CK59 attenuated Ag-mediated ILP stabilization by ∼50% (Fig. 6H), which was coupled to a proportional decrease in calcium flux (Fig. 6I). This result supports the interdependence between ILPs and calcium flux.

CD8 CTLs use ILPs to probe for Ag

The endothelium has been shown to initiate MHC-I/Ag-dependent activation of CD8 CTLs leading to direct killing of ECs (31, 32). To investigate these responses, we incubated previously activated murine OT-I CD8 T cells on murine heart microvascular ECs pulsed with SIINFEKL peptide Ag. Ag-pulsed ECs initiated a rapid calcium flux (Fig. 7A) that was coupled to progressive specific lysis of ECs over a 4-h duration (∼50% lysis ∼2 h; Fig. 7B). Live-cell imaging showed that in the absence of Ag, CTLs avidly probed the endothelium with dynamic ILPs while migrating without fluxing calcium (Fig. 7C, Supplemental Video 8). In the presence of Ag, initial ILPs were rapidly followed by calcium flux (offset time = 32.8 ± 6.6 s), which then led to migrational arrest and formation of peripheral ILP arrays similar to those formed by CD4 effector T cells (Fig. 7D, 7E). Thus, T cell ILPs seem to be a general feature of Ag sensing on ECs.

FIGURE 7.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 7.

Murine CD8 CTLs use ILPs to sense Ag-bound MHC-I on endothelium. (A) Previously activated murine OTI CD8 T cells (CTLs) were imaged on mem-YFP–transfected, mouse heart microvascular ECs pulsed with SIINFEKL. Individual ratiometric calcium flux values at 5 min were plotted for ±Ag conditions. (B) CTLs were added to mixed cultures of Ag-pulsed/unpulsed ECs, and percentage of specific lysis is plotted at indicated time points. (C) CTLs were imaged during EC probing in the absence of Ag. Differential interference contrast and mem-YFP are shown in the upper and lower panels, respectively. Dashed line represents the outline of the migrating CTL under which transient ILPs are continuously formed. See also corresponding Supplemental Video 8. (D) Imaging was conducted as in (C) on ECs pulsed with SIINFEKL. Red arrow indicates initial ILP formation preceding calcium flux. (E) Average offset time for calcium flux relative to initial ILP formation. Data represent mean ± SEM. Scale bars, 5 μm.

A planar-coated cell model for Ag recognition

We next considered whether ILPs either represent unique features of T cell-endothelial Ag recognition or may be more broadly relevant and uniquely revealed by endothelium. We speculated that the rigidity of coated substrate models may frustrate/mask ILP formation, whereas orientation/resolution issues might obscure detection of ILPs with traditional cellular APCs (16–18).

First, we simply asked whether evidence consistent with ILPs could be detected in a coated glass model (Fig. 8A). ITmem plated on glass coated with ICAM-1 and anti-CD3 (but not anti-CD43/CD45) Ab rapidly fluxed calcium (Supplemental Fig. 4A–C). IRM revealed that initial “microcontacts” (∼0.2- to 1-μm dots consistent with T cell ILPs and/or microvilli) always preceded calcium flux (Supplemental Fig. 4B, 4C; offset time = 21.4 s). New microcontacts also continued to form after T cell spreading (Fig. 8B, Supplemental Fig. 4D, 4E, Supplemental Video 9).

FIGURE 8.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 8.

ILPs facilitate Ag recognition on professional APCs. (A) Schematic of the “coated-glass model” using ICAM-1– and Ab-coated glass. (b) Magnified view of the interaction surface. (B) iTmem were added to an ICAM-1–Fc– and anti-CD3–coated glass chamber and imaged by differential interference contrast and IRM. Arrows indicate IRM-detected “microcontacts.” (C) Schematic of the “planar coated-cell APC model.” CHO-K1 cells expressing ICAM-1–GFP and soluble DsRed were surface “coated” with Abs against CD3, CD43, or CD45 using a biotin/SA capture approach (see also Supplemental Fig. 4F). (d) Magnified view of the interaction surface. (D) Cells prepared as in (C) coated with anti-CD3 Ab were imaged live on addition of Fura-2–labeled iTmem. Panels demonstrate relatively early and later phases of interaction as indicated. Arrows indicate podo-prints/ILPs. See also Supplemental Video 10. (E and F) Imaging experiments were conducted as in (D), but T cells were pretreated with latrunculin A. Podo-print/ILP index (E) and calcium flux (F) were calculated. (G) ITmem were incubated with Ag-pulsed Priess B cells for 5 min and stained for LFA-1 and actin. Image represents a three-dimensional projection of T cell settled on top of the B cells with the IS forming more nearly parallel to the x–y imaging plane. See also Supplemental Video 11. (H) Transmission electron micrograph of T cell–B cell IS as in (G). The T cell is identified by a red overlay. Arrows indicate close intercellular contacts formed by T cell ILPs. (I) Schematic (upper left panel) shows both lateral (a) and various en face (b) interfaces typically formed in vitro between T cells and the highly irregular surfaces of DCs. (1) and (2) show previously activated murine CD4 OT-II lymphocytes interacting with Ag-pulsed mem-YFP–transfected DCs (green, 2). Red outlines depict T cell perimeter based on DIC imaging (1). (3) is identical to (2) but shows three boxed regions (i–iii) expanded on the left. (3i) shows a lateral interaction in which the T cell has pushed small finger-like invagination into the side of the BMDC putatively formed by ILPs (arrows). (3ii) and (3iii) show en face interactions whereby ILPs seem to be extending normal to the imaging plane, giving rise to the typical ring-shaped podo-print appearance observed in endothelium (i.e., Fig. 2). (J) DCs were transfected with soluble monomeric DsRed and mem-YFP, pulsed with Ag, and CD4 OT-II T cells were added. Panels show cytoplasm displacing podo-prints/ILP (arrows), which precede calcium flux and are stabilized after calcium flux. (K) Transmission electron micrograph of T cell–DC IS as in (J) fixed after 5 min. The T cell is identified by a red overlay. Arrows indicate close intercellular contacts formed by T cell ILPs. (L) Calcium flux of previously activated CD4 OTII T cells 8 min after interaction with C57BL/6 (WT) or Ciita−/− Ag-pulsed DCs with or without latrunculin A pretreatment. Data represent mean ± SEM. Scale bars, 5 μm. ***p < 0.0005.

We hypothesized that these microcontacts at least partly reflected ILP activity that was mechanically frustrated by the rigid substrate. To test this, we designed a “coated-cell APC” model whereby the earlier Ab-coated glass substrate was recapitulated on the surface of a deformable CHO-K1 cell (Fig. 8C, Supplemental Fig. 4F). In this setting, clear three-dimensional ILP arrays were readily detected that were coupled to calcium flux (Fig. 8D, Supplemental Video 10). As with iTmem on ECs, initial ILPs always preceded (Supplemental Fig. 4G; offset time ∼56 s) and seemed to functionally support (Fig. 8E, 8F) initiation of calcium flux. These findings suggest a general tendency of memory/effector T cells to use ILPs to probe diverse substrates for recognition and response to Ag (as modeled in Fig. 9).

ILPs are involved in recognition of Ag presented by professional APCs

Next, we re-examined Ag recognition with professional APCs. Thus, iTmem were incubated with Priess B cells pulsed with Ag and imaged by confocal microscopy. Equatorial cross sections recapitulate classic views (12) of the T cell–B cell IS whereby T cell LFA-1 shows enrichment in peripheral-supramolecular activation cluster–like peripheral membrane bulges (Supplemental Fig. 4H, inset 2, arrows). Cross sections taken at the edge of the IS additionally suggest presence of LFA-1–rich finger-like protrusions (Supplemental Fig. 4H, insets 1a–d) similar to ILPs formed on ECs (Fig. 3Bb). More compelling imaging of putative ILPs can be seen in occasional examples where the T cells settle on top of B cells such that the IS aligns with the optimal imaging plane (18) (Fig. 8G, Supplemental Video 11). Ultrastructural views, evident in a subset of micrographs, further support the presence of ILPs (Fig. 8H).

Finally, we investigated murine OTII CD4 T cells incubated with BMDCs coexpressing mem-YFP and soluble DsRed. T cells interacted with DCs both laterally (Fig. 8I, subset a) and to a lesser extent through en face contacts (Fig. 8I, subset b). In the former, side views of DC invaginations were readily evident that were similar to podo-prints/ILPs seen on endothelium (Fig. 8I, panel 3i). En face interactions also revealed discrete circular podo-prints/ILPs (Fig. 8I, panels 3ii, 3iii), although these were generally less obvious than those formed on endothelium because of the highly active and irregular surface topology of DCs. Dynamic imaging showed ILP probing preceded calcium flux, which, in turn, was coupled to peripheral ILP array formation (Fig. 8J). Ultrastructural confirmation of ILP formation could also be obtained in a subset of electron micrographs (Fig. 8K). Finally, treatment of T cells with latrunculin A strongly inhibited both ILPs and initial calcium responses to Ag presented by DCs (Fig. 8L).

Discussion

Basic cellular mechanisms by which lymphocytes effectively scan for peptide Ag on apposing cells and how this leads to IS formation have remained mysterious. Our results investigating endothelium as a model APC indicate potential roles for ILPs in these processes. Our further studies with diverse Ag recognition settings suggest broad, previously unappreciated functions for ILPs as actuators of immune surveillance.

Vascular ECs represent intriguing and understudied APCs/target cells for adaptive immune responses. These MHC-I–, MHC-II–, and costimulator-expressing cells are strategically positioned at the blood–tissue interface to serve as unique sentinels for the immune system (26). Previous studies demonstrated that ECs can effectively restimulate memory/effector, but not naive, T cells (26–33). Thus, ECs have been hypothesized to serve as peripheral ancillary or “semiprofessional” APCs that contribute to the effector phase of adaptive immune responses. Although overall roles remain controversial, studies suggest that Ag presentation by ECs can influence T cell activation, differentiation, trafficking, and memory (26–28, 51). In this study, we demonstrate that CD4 and CD8 memory/effector T cells exhibited a transient arrest in migration coupled, respectively, to Th1 and cytotoxic responses on ECs presenting MHC/Ag, consistent with previous findings (31, 52, 53).

Importantly, ECs exhibit a virtually planar cell surface in vitro, which we previously established to be ideal for high spatiotemporal resolution imaging of topological dynamics (34). We reasoned that ECs might, therefore, serve as physiologic “planar APC/target cell” models uniquely suited to fill two key gaps in our understanding of immune surveillance mechanisms: 1) how initial MHC/Ag-sampling contacts form between T cells and APCs, and 2) how these lead to formation of productive ISs. Although planar APC substrates (i.e., lipid bilayers, Ab-coated glass) provide excellent imaging, which has led to critical insights to these dynamics (15, 19–22), such models are inherently limiting. Alternatively, details of physiologic cell–cell scanning dynamics are profoundly obscured by orientation-related imaging issues (16–18). The imaging afforded with our planar endothelial APC model in this study revealed unexpected involvement of ILPs in Ag recognition and response activities.

ILPs (“invadosome-like protrusions”) are lymphocyte equivalents (36) to podosomes (formed largely by myeloid lineage cells) and invadopodia (formed by transformed cells), collectively termed “invadosomes” (35). Invadosomes and ILPs are actin-dependent cylindrical, protrusive organelles (∼200–1000 nm in diameter and depth) that form on the ventral surface of migratory cells (35). Distinct from invadosomes, characterized to form on matrix, bone, glass/plastic substrates, T cell ILPs have only been seen on cellular substrates, as shown in a range of in vitro (34, 54, 55) and in vivo studies (reviewed in Ref. 56). In such settings, dynamic ILP probing seems to function in migratory pathfinding, apparently serving as biomechanical sensors (34, 36, 54, 55, 57). These studies suggest that ILPs may also facilitate biochemical or “informational” scanning of diverse cell surfaces.

We speculated that the ILP scanning seen on ECs was uniquely revealed by this setting, rather than a unique feature of it. IRM imaging of a rudimentary APC model (anti-CD3–coated glass) showed that T cells form a series of microcontacts with the substrate that are consistent with either ILP or microvilli interactions. Similar microcontacts were previously shown to be enriched in F-actin, WASP, and TCR, and hypothesized to represent frustrated podosomes (20, 22). When we tested this hypothesis by recapitulating the same anti-CD3 activation stimuli on a pliable cell surface, we, indeed, found that the microcontacts transitioned into three-dimensional ILPs and podo-prints. Moreover, our studies using approaches optimized for imaging professional APCs (i.e., B cells and DCs) readily detected ILPs probing in these settings. Importantly, although dynamics, molecular composition, and three-dimensional architectures were not defined, extensive (largely EM-based) studies previously evidenced similar T cell protrusions extending against diverse APCs and target cell surfaces (21, 43, 58–63). Collectively, this suggests broad relevance for memory/effector T cells probing their cellular environment with ILPs. The important question of whether naive T cells similarly use ILPs remains to be addressed.

One possible role for ILP probing may be in enhancing the efficiency of Ag recognition. The act of Ag detection requires T cell and APC/target membranes to come within ∼14 nm of each other (2, 64). Yet, all cell membranes are extensively shielded by ∼50- to 500-nm-thick glycocalyces (3–5) that physically oppose close contact and negatively modulate immune recognition (5–10) (Fig. 9, inset 2b). In the periphery, patrolling effector/memory lymphocytes must effectively scan the surfaces of widely varying cell types (indeed, essentially any host cell could become a target), each with glycocalyces of different thicknesses and physicochemical properties. Forces provided by ILP (which form independently of Ag during lateral migration) (34) are sufficient to deform nuclear lamina, displace and distort cytoskeleton and intracellular organelles, and promote transcellular diapedesis (34). More than sufficient energy would, therefore, seem to be available at the tips of ILPs (Fig. 9, inset 2a) to overcome repulsion provided by the glycocalyx (Fig. 9, inset 2c).

FIGURE 9.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 9.

Hypothetical model for ILP function in Ag recognition and response. Schematic depicts top-down and side views of a memory/effector T cell interacting with an APC/target cell. Lymphocytes initiate lateral migration (Step 1) and begin to dynamically drive ILPs against the apposing cell (Step 2, 2a). Close interactions between T and APC/target cells, which are partially opposed by the cell glycocalyces (2b), form preferentially (but not exclusively) at ILP tips (2c). We hypothesize that TCR/MHC interactions may be facilitated in these zones. Initial calcium released on Ag recognition (green overlay/arrows; Step 3) seems to be coupled to stabilization/accumulation of ILP arrays (“podo-synapses”; Step 4), which we hypothesize could, in turn, help sustain/enhance signaling (Step 5).

Previous studies showed that lamellipodia of migrating lymphocytes are zones of heightened Ag recognition efficiency (65–67). In both our previous (34) and current work, we show that lamellipodia are also the predominant areas of ILP formation. Moreover, in our experimental system, ILPs, but not necessarily lamellipodia, consistently preceded calcium flux. Thus, although migration behaviors are necessary for immune surveillance, ILPs may function more directly in promoting intimate contacts, and thereby ensuring efficient Ag sampling. Our studies with latrunculin A are consistent with (although do not prove) such functional roles. It is also interesting to note that forces and membrane bending associated with ILPs are consistent with the hypothesized force-based mechanism for formal triggering of TCR signaling (22, 68–71). ILPs are probably not absolutely required for either TCR signaling or forming intimate T cell–APC/target contacts, but rather act to ensure efficiency and fidelity of immune surveillance.

Another possible function for ILPs may be to support signal amplification and sustenance. Our studies reveal dense arrays of calcium-stabilized ILPs that dominate the IS after Ag recognition. Individual ILPs showed enrichment in TCR and molecules suggestive of active signaling. This feature is generally not unlike TCR signaling microclusters defined using planar APC model substrates (14, 15, 20–22, 38, 72, 73), with the key distinction that ILPs have a discrete three-dimensional architecture. The well-developed concept that microclusters may function as “signalosomes” to amplify and sustain signaling by concentrating important molecules/activities might then be extended to include limited “reaction volumes” formed within ILPs (17, 74–78). In addition, the stable peripheral ILP arrays are strikingly similar to osteoclast “podosome-belts” (79, 80) that form sealing zones for directed secretion of bone-degrading enzymes (81). Thus, it may be hypothesized that peripheral podo-synapses may function analogously for directed secretion of cytokines or cytotoxic materials.

These studies collectively support the hypothesis that ILPs may serve as newly appreciated sensory organelles that facilitate Ag recognition and responses. By virtue of physical force exertion, ILPs literally allow T cells to get a deeper understanding of their local cellular environment. Such proactive “informational scanning” might ensure robust sampling of MHC/Ag on diverse cell types. It is tempting to speculate that stabilized TCR-enriched ILPs resulting from Ag recognition represent a clearer, more physiologic view of TCR microclusters and “multifocal” ISs characterized to date.

Disclosures

The authors have no financial conflicts of interest.

Footnotes

  • This work was supported by the National Institutes of Health (Grants T32 AI070085 to P.T.S., AI078897 to A.H.S., and HL04006 to C.V.C.) and the American Heart Association, the Arthritis Foundation, and the Roche Organ Transplant Research Foundation (to C.V.C.).

  • The online version of this article contains supplemental material.

  • Abbreviations used in this article:

    BMDC
    bone marrow-derived dendritic cell
    CAMKII
    calcium-calmodulin–dependent kinase II
    CHO
    Chinese hamster ovary
    DC
    dendritic cell
    EC
    endothelial cell
    HDMVEC
    human dermal microvascular EC
    HLMVEC
    human lung microvascular EC
    HS1
    hematopoietic lineage cell-specific protein 1
    HUVEC
    human umbilical vein EC
    ILP
    invadosome/podosome-like protrusion
    IRM
    interference reflection microscopy
    IS
    immunological synapse
    iTmem
    induced/expanded human T memory cell
    mem-DsRed
    palmitoylated DsRed
    mem-YFP
    palmitoylated YFP
    MHC-I
    MHC class I
    MHC-II
    MHC class II
    nTmem
    natural human T memory cell
    PKC
    protein kinase C
    SA
    streptavidin
    SEB
    staphylococcal enterotoxin B
    TSST
    toxic shock syndrome toxin 1.

  • Received September 14, 2011.
  • Accepted February 13, 2012.
  • Copyright © 2012 by The American Association of Immunologists, Inc.

References

  1. ↵
    1. von Andrian U. H.,
    2. C. R. Mackay
    . 2000. T-cell function and migration. Two sides of the same coin. N. Engl. J. Med. 343: 1020–1034.
    OpenUrlCrossRefPubMed
  2. ↵
    1. Rudolph M. G.,
    2. R. L. Stanfield,
    3. I. A. Wilson
    . 2006. How TCRs bind MHCs, peptides, and coreceptors. Annu. Rev. Immunol. 24: 419–466.
    OpenUrlCrossRefPubMed
  3. ↵
    1. Weinbaum S.,
    2. J. M. Tarbell,
    3. E. R. Damiano
    . 2007. The structure and function of the endothelial glycocalyx layer. Annu. Rev. Biomed. Eng. 9: 121–167.
    OpenUrlCrossRefPubMed
  4. ↵
    1. Bell G. I.,
    2. M. Dembo,
    3. P. Bongrand
    . 1984. Cell adhesion. Competition between nonspecific repulsion and specific bonding. Biophys. J. 45: 1051–1064.
    OpenUrlCrossRefPubMed
  5. ↵
    1. Springer T. A.
    1990. Adhesion receptors of the immune system. Nature 346: 425–434.
    OpenUrlCrossRefPubMed
    1. Gubbels J. A.,
    2. M. Felder,
    3. S. Horibata,
    4. J. A. Belisle,
    5. A. Kapur,
    6. H. Holden,
    7. S. Petrie,
    8. M. Migneault,
    9. C. Rancourt,
    10. J. P. Connor,
    11. M. S. Patankar
    . 2010. MUC16 provides immune protection by inhibiting synapse formation between NK and ovarian tumor cells. Mol. Cancer 9: 11.
    OpenUrlCrossRefPubMed
    1. Komatsu M.,
    2. L. Yee,
    3. K. L. Carraway
    . 1999. Overexpression of sialomucin complex, a rat homologue of MUC4, inhibits tumor killing by lymphokine-activated killer cells. Cancer Res. 59: 2229–2236.
    OpenUrlAbstract/FREE Full Text
    1. Tsuboi S.,
    2. M. Fukuda
    . 2001. Roles of O-linked oligosaccharides in immune responses. Bioessays 23: 46–53.
    OpenUrlCrossRefPubMed
    1. van de Wiel-van Kemenade E.,
    2. M. J. Ligtenberg,
    3. A. J. de Boer,
    4. F. Buijs,
    5. H. L. Vos,
    6. C. J. Melief,
    7. J. Hilkens,
    8. C. G. Figdor
    . 1993. Episialin (MUC1) inhibits cytotoxic lymphocyte-target cell interaction. J. Immunol. 151: 767–776.
    OpenUrlAbstract
  6. ↵
    1. Manjunath N.,
    2. M. Correa,
    3. M. Ardman,
    4. B. Ardman
    . 1995. Negative regulation of T-cell adhesion and activation by CD43. Nature 377: 535–538.
    OpenUrlCrossRefPubMed
  7. ↵
    1. Shaw A. S.,
    2. M. L. Dustin
    . 1997. Making the T cell receptor go the distance: a topological view of T cell activation. Immunity 6: 361–369.
    OpenUrlCrossRefPubMed
  8. ↵
    1. Monks C. R.,
    2. B. A. Freiberg,
    3. H. Kupfer,
    4. N. Sciaky,
    5. A. Kupfer
    . 1998. Three-dimensional segregation of supramolecular activation clusters in T cells. Nature 395: 82–86.
    OpenUrlCrossRefPubMed
    1. Delon J.,
    2. S. Stoll,
    3. R. N. Germain
    . 2002. Imaging of T-cell interactions with antigen presenting cells in culture and in intact lymphoid tissue. Immunol. Rev. 189: 51–63.
    OpenUrlCrossRefPubMed
  9. ↵
    1. Brossard C.,
    2. V. Feuillet,
    3. A. Schmitt,
    4. C. Randriamampita,
    5. M. Romao,
    6. G. Raposo,
    7. A. Trautmann
    . 2005. Multifocal structure of the T cell - dendritic cell synapse. Eur. J. Immunol. 35: 1741–1753.
    OpenUrlCrossRefPubMed
  10. ↵
    1. Dustin M. L.
    2009. The cellular context of T cell signaling. Immunity 30: 482–492.
    OpenUrlCrossRefPubMed
  11. ↵
    1. Balagopalan L.,
    2. E. Sherman,
    3. V. A. Barr,
    4. L. E. Samelson
    . 2011. Imaging techniques for assaying lymphocyte activation in action. Nat. Rev. Immunol. 11: 21–33.
    OpenUrlCrossRefPubMed
  12. ↵
    1. Cebecauer M.,
    2. M. Spitaler,
    3. A. Sergé,
    4. A. I. Magee
    . 2010. Signalling complexes and clusters: functional advantages and methodological hurdles. J. Cell Sci. 123: 309–320.
    OpenUrlAbstract/FREE Full Text
  13. ↵
    1. Oddos S.,
    2. C. Dunsby,
    3. M. A. Purbhoo,
    4. A. Chauveau,
    5. D. M. Owen,
    6. M. A. Neil,
    7. D. M. Davis,
    8. P. M. French
    . 2008. High-speed high-resolution imaging of intercellular immune synapses using optical tweezers. Biophys. J. 95: L66–L68.
    OpenUrlCrossRefPubMed
  14. ↵
    1. Grakoui A.,
    2. S. K. Bromley,
    3. C. Sumen,
    4. M. M. Davis,
    5. A. S. Shaw,
    6. P. M. Allen,
    7. M. L. Dustin
    . 1999. The immunological synapse: a molecular machine controlling T cell activation. Science 285: 221–227.
  15. ↵
    1. Bunnell S. C.,
    2. D. I. Hong,
    3. J. R. Kardon,
    4. T. Yamazaki,
    5. C. J. McGlade,
    6. V. A. Barr,
    7. L. E. Samelson
    . 2002. T cell receptor ligation induces the formation of dynamically regulated signaling assemblies. J. Cell Biol. 158: 1263–1275.
    OpenUrlAbstract/FREE Full Text
  16. ↵
    1. Yokosuka T.,
    2. K. Sakata-Sogawa,
    3. W. Kobayashi,
    4. M. Hiroshima,
    5. A. Hashimoto-Tane,
    6. M. Tokunaga,
    7. M. L. Dustin,
    8. T. Saito
    . 2005. Newly generated T cell receptor microclusters initiate and sustain T cell activation by recruitment of Zap70 and SLP-76. Nat. Immunol. 6: 1253–1262.
    OpenUrlCrossRefPubMed
  17. ↵
    1. Seminario M. C.,
    2. S. C. Bunnell
    . 2008. Signal initiation in T-cell receptor microclusters. Immunol. Rev. 221: 90–106.
    OpenUrlCrossRefPubMed
  18. ↵
    1. Dustin M. L.
    2009. Supported bilayers at the vanguard of immune cell activation studies. J. Struct. Biol. 168: 152–160.
    OpenUrlCrossRefPubMed
  19. ↵
    1. Springer T. A.
    1994. Traffic signals for lymphocyte recirculation and leukocyte emigration: the multistep paradigm. Cell 76: 301–314.
    OpenUrlCrossRefPubMed
  20. ↵
    1. Butcher E. C.
    1991. Leukocyte-endothelial cell recognition: three (or more) steps to specificity and diversity. Cell 67: 1033–1036.
    OpenUrlCrossRefPubMed
  21. ↵
    1. Choi J.,
    2. D. R. Enis,
    3. K. P. Koh,
    4. S. L. Shiao,
    5. J. S. Pober
    . 2004. T lymphocyte-endothelial cell interactions. Annu. Rev. Immunol. 22: 683–709.
    OpenUrlCrossRefPubMed
    1. Pober J. S.,
    2. W. C. Sessa
    . 2007. Evolving functions of endothelial cells in inflammation. Nat. Rev. Immunol. 7: 803–815.
    OpenUrlCrossRefPubMed
  22. ↵
    1. Marelli-Berg F. M.,
    2. K. Okkenhaug,
    3. V. Mirenda
    . 2007. A two-signal model for T cell trafficking. Trends Immunol. 28: 267–273.
    OpenUrlCrossRefPubMed
  23. ↵
    1. Marelli-Berg F. M.,
    2. R. E. Hargreaves,
    3. P. Carmichael,
    4. A. Dorling,
    5. G. Lombardi,
    6. R. I. Lechler
    . 1996. Major histocompatibility complex class II-expressing endothelial cells induce allospecific nonresponsiveness in naive T cells. J. Exp. Med. 183: 1603–1612.
    OpenUrlAbstract/FREE Full Text
    1. Ma W.,
    2. J. S. Pober
    . 1998. Human endothelial cells effectively costimulate cytokine production by, but not differentiation of, naive CD4+ T cells. J. Immunol. 161: 2158–2167.
    OpenUrlAbstract/FREE Full Text
  24. ↵
    1. Perez V. L.,
    2. L. Henault,
    3. A. H. Lichtman
    . 1998. Endothelial antigen presentation: stimulation of previously activated but not naïve TCR-transgenic mouse T cells. Cell. Immunol. 189: 31–40.
    OpenUrlCrossRefPubMed
  25. ↵
    1. Epperson D. E.,
    2. J. S. Pober
    . 1994. Antigen-presenting function of human endothelial cells. Direct activation of resting CD8 T cells. J. Immunol. 153: 5402–5412.
    OpenUrlAbstract
  26. ↵
    1. Shiao S. L.,
    2. N. C. Kirkiles-Smith,
    3. B. R. Shepherd,
    4. J. M. McNiff,
    5. E. J. Carr,
    6. J. S. Pober
    . 2007. Human effector memory CD4+ T cells directly recognize allogeneic endothelial cells in vitro and in vivo. J. Immunol. 179: 4397–4404.
    OpenUrlAbstract/FREE Full Text
  27. ↵
    1. Carman C. V.,
    2. P. T. Sage,
    3. T. E. Sciuto,
    4. M. A. de la Fuente,
    5. R. S. Geha,
    6. H. D. Ochs,
    7. H. F. Dvorak,
    8. A. M. Dvorak,
    9. T. A. Springer
    . 2007. Transcellular diapedesis is initiated by invasive podosomes. Immunity 26: 784–797.
    OpenUrlCrossRefPubMed
  28. ↵
    1. Linder S.
    2009. Invadosomes at a glance. J. Cell Sci. 122: 3009–3013.
    OpenUrlFREE Full Text
  29. ↵
    1. Carman C. V.
    2009. Mechanisms for transcellular diapedesis: probing and pathfinding by ‘invadosome-like protrusions’. J. Cell Sci. 122: 3025–3035.
    OpenUrlAbstract/FREE Full Text
  30. ↵
    1. Carman C. V.,
    2. T. A. Springer
    . 2004. A transmigratory cup in leukocyte diapedesis both through individual vascular endothelial cells and between them. J. Cell Biol. 167: 377–388.
    OpenUrlAbstract/FREE Full Text
  31. ↵
    1. Gomez T. S.,
    2. S. D. McCarney,
    3. E. Carrizosa,
    4. C. M. Labno,
    5. E. O. Comiskey,
    6. J. C. Nolz,
    7. P. Zhu,
    8. B. D. Freedman,
    9. M. R. Clark,
    10. D. J. Rawlings,
    11. et al
    . 2006. HS1 functions as an essential actin-regulatory adaptor protein at the immune synapse. Immunity 24: 741–752.
    OpenUrlCrossRefPubMed
  32. ↵
    1. Sarkar D.,
    2. P. K. Vemula,
    3. G. S. Teo,
    4. D. Spelke,
    5. R. Karnik,
    6. Y. Wee,
    7. J. M. Karp
    . 2008. Chemical engineering of mesenchymal stem cells to induce a cell rolling response. Bioconjug. Chem. 19: 2105–2109.
    OpenUrlCrossRefPubMed
  33. ↵
    1. Antón I. M.,
    2. M. A. de la Fuente,
    3. T. N. Sims,
    4. S. Freeman,
    5. N. Ramesh,
    6. J. H. Hartwig,
    7. M. L. Dustin,
    8. R. S. Geha
    . 2002. WIP deficiency reveals a differential role for WIP and the actin cytoskeleton in T and B cell activation. Immunity 16: 193–204.
    OpenUrlCrossRefPubMed
  34. ↵
    1. Wülfing C.,
    2. A. Bauch,
    3. G. R. Crabtree,
    4. M. M. Davis
    . 2000. The vav exchange factor is an essential regulator in actin-dependent receptor translocation to the lymphocyte-antigen-presenting cell interface. Proc. Natl. Acad. Sci. USA 97: 10150–10155.
    OpenUrlAbstract/FREE Full Text
  35. ↵
    1. Billadeau D. D.,
    2. J. C. Nolz,
    3. T. S. Gomez
    . 2007. Regulation of T-cell activation by the cytoskeleton. Nat. Rev. Immunol. 7: 131–143.
    OpenUrlCrossRefPubMed
  36. ↵
    1. Butler B.,
    2. D. H. Kastendieck,
    3. J. A. Cooper
    . 2008. Differently phosphorylated forms of the cortactin homolog HS1 mediate distinct functions in natural killer cells. Nat. Immunol. 9: 887–897.
    OpenUrlCrossRefPubMed
  37. ↵
    1. Klos Dehring D. A.,
    2. F. Clarke,
    3. B. G. Ricart,
    4. Y. Huang,
    5. T. S. Gomez,
    6. E. K. Williamson,
    7. D. A. Hammer,
    8. D. D. Billadeau,
    9. Y. Argon,
    10. J. K. Burkhardt
    . 2011. Hematopoietic lineage cell-specific protein 1 functions in concert with the Wiskott-Aldrich syndrome protein to promote podosome array organization and chemotaxis in dendritic cells. J. Immunol. 186: 4805–4818.
    OpenUrlAbstract/FREE Full Text
  38. ↵
    1. Fooksman D. R.,
    2. S. Vardhana,
    3. G. Vasiliver-Shamis,
    4. J. Liese,
    5. D. A. Blair,
    6. J. Waite,
    7. C. Sacristán,
    8. G. D. Victora,
    9. A. Zanin-Zhorov,
    10. M. L. Dustin
    . 2010. Functional anatomy of T cell activation and synapse formation. Annu. Rev. Immunol. 28: 79–105.
    OpenUrlCrossRefPubMed
    1. Trautmann A.,
    2. S. Valitutti
    . 2003. The diversity of immunological synapses. Curr. Opin. Immunol. 15: 249–254.
    OpenUrlCrossRefPubMed
  39. ↵
    1. Bunnell S. C.
    2010. Multiple microclusters: diverse compartments within the immune synapse. Curr. Top. Microbiol. Immunol. 340: 123–154.
    OpenUrlCrossRefPubMed
  40. ↵
    1. Dovas A.,
    2. D. Cox
    . 2011. Signaling networks regulating leukocyte podosome dynamics and function. Cell. Signal. 23: 1225–1234.
    OpenUrlCrossRefPubMed
  41. ↵
    1. Arrieumerlou C.,
    2. C. Randriamampita,
    3. G. Bismuth,
    4. A. Trautmann
    . 2000. Rac is involved in early TCR signaling. J. Immunol. 165: 3182–3189.
    OpenUrlAbstract/FREE Full Text
  42. ↵
    1. Hughes K.,
    2. S. Edin,
    3. A. Antonsson,
    4. T. Grundström
    . 2001. Calmodulin-dependent kinase II mediates T cell receptor/CD3- and phorbol ester-induced activation of IkappaB kinase. J. Biol. Chem. 276: 36008–36013.
    OpenUrlAbstract/FREE Full Text
  43. ↵
    1. Turesson C.
    2004. Endothelial expression of MHC class II molecules in autoimmune disease. Curr. Pharm. Des. 10: 129–143.
    OpenUrlCrossRefPubMed
  44. ↵
    1. Manes T. D.,
    2. S. L. Shiao,
    3. T. J. Dengler,
    4. J. S. Pober
    . 2007. TCR signaling antagonizes rapid IP-10-mediated transendothelial migration of effector memory CD4+ T cells. J. Immunol. 178: 3237–3243.
    OpenUrlAbstract/FREE Full Text
  45. ↵
    1. Manes T. D.,
    2. J. S. Pober
    . 2008. Antigen presentation by human microvascular endothelial cells triggers ICAM-1-dependent transendothelial protrusion by, and fractalkine-dependent transendothelial migration of, effector memory CD4+ T cells. J. Immunol. 180: 8386–8392.
    OpenUrlAbstract/FREE Full Text
  46. ↵
    1. Shulman Z.,
    2. V. Shinder,
    3. E. Klein,
    4. V. Grabovsky,
    5. O. Yeger,
    6. E. Geron,
    7. A. Montresor,
    8. M. Bolomini-Vittori,
    9. S. W. Feigelson,
    10. T. Kirchhausen,
    11. et al
    . 2009. Lymphocyte crawling and transendothelial migration require chemokine triggering of high-affinity LFA-1 integrin. Immunity 30: 384–396.
    OpenUrlCrossRefPubMed
  47. ↵
    1. Gérard A.,
    2. R. A. van der Kammen,
    3. H. Janssen,
    4. S. I. Ellenbroek,
    5. J. G. Collard
    . 2009. The Rac activator Tiam1 controls efficient T-cell trafficking and route of transendothelial migration. Blood 113: 6138–6147.
    OpenUrlAbstract/FREE Full Text
  48. ↵
    1. Sage P. T.,
    2. C. V. Carman
    . 2009. Settings and mechanisms for trans-cellular diapedesis. Front. Biosci. 14: 5066–5083.
    OpenUrlCrossRefPubMed
  49. ↵
    1. Albiges-Rizo C.,
    2. O. Destaing,
    3. B. Fourcade,
    4. E. Planus,
    5. M. R. Block
    . 2009. Actin machinery and mechanosensitivity in invadopodia, podosomes and focal adhesions. J. Cell Sci. 122: 3037–3049.
    OpenUrlAbstract/FREE Full Text
  50. ↵
    1. Bunnell S. C.,
    2. A. L. Singer,
    3. D. I. Hong,
    4. B. H. Jacque,
    5. M. S. Jordan,
    6. M. C. Seminario,
    7. V. A. Barr,
    8. G. A. Koretzky,
    9. L. E. Samelson
    . 2006. Persistence of cooperatively stabilized signaling clusters drives T-cell activation. Mol. Cell. Biol. 26: 7155–7166.
    OpenUrlAbstract/FREE Full Text
    1. Wetzel S. A.,
    2. T. W. McKeithan,
    3. D. C. Parker
    . 2002. Live-cell dynamics and the role of costimulation in immunological synapse formation. J. Immunol. 169: 6092–6101.
    OpenUrlAbstract/FREE Full Text
    1. Barcia C.,
    2. A. Gomez,
    3. V. de Pablos,
    4. E. Fernández-Villalba,
    5. C. Liu,
    6. K. M. Kroeger,
    7. J. Martín,
    8. A. F. Barreiro,
    9. M. G. Castro,
    10. P. R. Lowenstein,
    11. M. T. Herrero
    . 2008. CD20, CD3, and CD40 ligand microclusters segregate three-dimensionally in vivo at B-cell-T-cell immunological synapses after viral immunity in primate brain. J. Virol. 82: 9978–9993.
    OpenUrlAbstract/FREE Full Text
    1. Barcia C.,
    2. N. S. Sanderson,
    3. R. J. Barrett,
    4. K. Wawrowsky,
    5. K. M. Kroeger,
    6. M. Puntel,
    7. C. Liu,
    8. M. G. Castro,
    9. P. R. Lowenstein
    . 2008. T cells’ immunological synapses induce polarization of brain astrocytes in vivo and in vitro: a novel astrocyte response mechanism to cellular injury. PLoS ONE 3: e2977.
    OpenUrlCrossRefPubMed
    1. Barcia C., Jr..,
    2. A. Gómez,
    3. J. M. Gallego-Sanchez,
    4. A. Perez-Vallés,
    5. M. G. Castro,
    6. P. R. Lowenstein,
    7. C. Barcia Sr..,
    8. M. T. Herrero
    . 2009. Infiltrating CTLs in human glioblastoma establish immunological synapses with tumorigenic cells. Am. J. Pathol. 175: 786–798.
    OpenUrlCrossRefPubMed
  51. ↵
    1. Davis D. M.
    2009. Mechanisms and functions for the duration of intercellular contacts made by lymphocytes. Nat. Rev. Immunol. 9: 543–555.
    OpenUrlCrossRefPubMed
  52. ↵
    1. Rudd P. M.,
    2. T. Elliott,
    3. P. Cresswell,
    4. I. A. Wilson,
    5. R. A. Dwek
    . 2001. Glycosylation and the immune system. Science 291: 2370–2376.
  53. ↵
    1. Valitutti S.,
    2. M. Dessing,
    3. K. Aktories,
    4. H. Gallati,
    5. A. Lanzavecchia
    . 1995. Sustained signaling leading to T cell activation results from prolonged T cell receptor occupancy. Role of T cell actin cytoskeleton. J. Exp. Med. 181: 577–584.
    OpenUrlAbstract/FREE Full Text
    1. Negulescu P. A.,
    2. T. B. Krasieva,
    3. A. Khan,
    4. H. H. Kerschbaum,
    5. M. D. Cahalan
    . 1996. Polarity of T cell shape, motility, and sensitivity to antigen. Immunity 4: 421–430.
    OpenUrlCrossRefPubMed
  54. ↵
    1. Wei X.,
    2. B. J. Tromberg,
    3. M. D. Cahalan
    . 1999. Mapping the sensitivity of T cells with an optical trap: polarity and minimal number of receptors for Ca(2+) signaling. Proc. Natl. Acad. Sci. USA 96: 8471–8476.
    OpenUrlAbstract/FREE Full Text
  55. ↵
    1. Ma Z.,
    2. K. A. Sharp,
    3. P. A. Janmey,
    4. T. H. Finkel
    . 2008. Surface-anchored monomeric agonist pMHCs alone trigger TCR with high sensitivity. PLoS Biol. 6: e43.
    OpenUrlCrossRefPubMed
    1. Ma Z.,
    2. P. A. Janmey,
    3. T. H. Finkel
    . 2008. The receptor deformation model of TCR triggering. FASEB J. 22: 1002–1008.
    OpenUrlAbstract/FREE Full Text
    1. Groves J. T.
    2007. Bending mechanics and molecular organization in biological membranes. Annu. Rev. Phys. Chem. 58: 697–717.
    OpenUrlCrossRefPubMed
  56. ↵
    1. Xu C.,
    2. E. Gagnon,
    3. M. E. Call,
    4. J. R. Schnell,
    5. C. D. Schwieters,
    6. C. V. Carman,
    7. J. J. Chou,
    8. K. W. Wucherpfennig
    . 2008. Regulation of T cell receptor activation by dynamic membrane binding of the CD3epsilon cytoplasmic tyrosine-based motif. Cell 135: 702–713.
    OpenUrlCrossRefPubMed
  57. ↵
    1. Dustin M. L.,
    2. S. Y. Tseng,
    3. R. Varma,
    4. G. Campi
    . 2006. T cell-dendritic cell immunological synapses. Curr. Opin. Immunol. 18: 512–516.
    OpenUrlCrossRefPubMed
  58. ↵
    1. Saito T.,
    2. T. Yokosuka
    . 2006. Immunological synapse and microclusters: the site for recognition and activation of T cells. Curr. Opin. Immunol. 18: 305–313.
    OpenUrlCrossRefPubMed
  59. ↵
    1. Burbach B. J.,
    2. R. B. Medeiros,
    3. K. L. Mueller,
    4. Y. Shimizu
    . 2007. T-cell receptor signaling to integrins. Immunol. Rev. 218: 65–81.
    OpenUrlCrossRefPubMed
    1. Perez-Moreno M.,
    2. C. Jamora,
    3. E. Fuchs
    . 2003. Sticky business: orchestrating cellular signals at adherens junctions. Cell 112: 535–548.
    OpenUrlCrossRefPubMed
    1. Harwood N. E.,
    2. F. D. Batista
    . 2008. New insights into the early molecular events underlying B cell activation. Immunity 28: 609–619.
    OpenUrlCrossRefPubMed
    1. Vicente-Manzanares M.,
    2. F. Sánchez-Madrid
    . 2004. Role of the cytoskeleton during leukocyte responses. Nat. Rev. Immunol. 4: 110–122.
    OpenUrlCrossRefPubMed
  60. ↵
    1. Miranti C. K.,
    2. J. S. Brugge
    . 2002. Sensing the environment: a historical perspective on integrin signal transduction. Nat. Cell Biol. 4: E83–E90.
    OpenUrlCrossRefPubMed
  61. ↵
    1. Luxenburg C.,
    2. D. Geblinger,
    3. E. Klein,
    4. K. Anderson,
    5. D. Hanein,
    6. B. Geiger,
    7. L. Addadi
    . 2007. The architecture of the adhesive apparatus of cultured osteoclasts: from podosome formation to sealing zone assembly. PLoS ONE 2: e179.
    OpenUrlCrossRefPubMed
  62. ↵
    1. Destaing O.,
    2. A. Sanjay,
    3. C. Itzstein,
    4. W. C. Horne,
    5. D. Toomre,
    6. P. De Camilli,
    7. R. Baron
    . 2008. The tyrosine kinase activity of c-Src regulates actin dynamics and organization of podosomes in osteoclasts. Mol. Biol. Cell 19: 394–404.
    OpenUrlAbstract/FREE Full Text
  63. ↵
    1. Saltel F.,
    2. O. Destaing,
    3. F. Bard,
    4. D. Eichert,
    5. P. Jurdic
    . 2004. Apatite-mediated actin dynamics in resorbing osteoclasts. Mol. Biol. Cell 15: 5231–5241.
    OpenUrlAbstract/FREE Full Text
View Abstract
PreviousNext
Back to top

In this issue

The Journal of Immunology: 188 (8)
The Journal of Immunology
Vol. 188, Issue 8
15 Apr 2012
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Advertising (PDF)
  • Back Matter (PDF)
  • Editorial Board (PDF)
  • Front Matter (PDF)
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word about The Journal of Immunology.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Antigen Recognition Is Facilitated by Invadosome-like Protrusions Formed by Memory/Effector T Cells
(Your Name) has forwarded a page to you from The Journal of Immunology
(Your Name) thought you would like to see this page from the The Journal of Immunology web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Antigen Recognition Is Facilitated by Invadosome-like Protrusions Formed by Memory/Effector T Cells
Peter T. Sage, Laya M. Varghese, Roberta Martinelli, Tracey E. Sciuto, Masataka Kamei, Ann M. Dvorak, Timothy A. Springer, Arlene H. Sharpe, Christopher V. Carman
The Journal of Immunology April 15, 2012, 188 (8) 3686-3699; DOI: 10.4049/jimmunol.1102594

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Antigen Recognition Is Facilitated by Invadosome-like Protrusions Formed by Memory/Effector T Cells
Peter T. Sage, Laya M. Varghese, Roberta Martinelli, Tracey E. Sciuto, Masataka Kamei, Ann M. Dvorak, Timothy A. Springer, Arlene H. Sharpe, Christopher V. Carman
The Journal of Immunology April 15, 2012, 188 (8) 3686-3699; DOI: 10.4049/jimmunol.1102594
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like

Jump to section

  • Article
    • Abstract
    • Materials and Methods
    • Results
    • Discussion
    • Disclosures
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF + SI
  • PDF

Related Articles

Cited By...

More in this TOC Section

  • Innate Immunity Together with Duration of Antigen Persistence Regulate Effector T Cell Induction
  • Regulatory Roles of IL-2 and IL-4 in H4/Inducible Costimulator Expression on Activated CD4+ T Cells During Th Cell Development
  • Induction of CD4+ T Cell Apoptosis as a Consequence of Impaired Cytoskeletal Rearrangement in UVB-Irradiated Dendritic Cells
Show more CELLULAR IMMUNOLOGY AND IMMUNE REGULATION

Similar Articles

Navigate

  • Home
  • Current Issue
  • Next in The JI
  • Archive
  • Brief Reviews
  • Pillars of Immunology
  • Translating Immunology

For Authors

  • Submit a Manuscript
  • Instructions for Authors
  • About the Journal
  • Journal Policies
  • Editors

General Information

  • Advertisers
  • Subscribers
  • Rights and Permissions
  • Accessibility Statement
  • Public Access
  • Privacy Policy
  • Disclaimer

Journal Services

  • Email Alerts
  • RSS Feeds
  • ImmunoCasts
  • Twitter

Copyright © 2021 by The American Association of Immunologists, Inc.

Print ISSN 0022-1767        Online ISSN 1550-6606