Multiple sclerosis (MS) is a chronic demyelinating disorder of the CNS characterized by immune cell infiltration across the brain vasculature into the brain, a process not yet fully understood. We previously demonstrated that the sphingolipid metabolism is altered in MS lesions. In particular, acid sphingomyelinase (ASM), a critical enzyme in the production of the bioactive lipid ceramide, is involved in the pathogenesis of MS; however, its role in the brain vasculature remains unknown. Transmigration of T lymphocytes is highly dependent on adhesion molecules in the vasculature such as intercellular adhesion molecule-1 (ICAM-1). In this article, we hypothesize that ASM controls T cell migration by regulating ICAM-1 function. To study the role of endothelial ASM in transmigration, we generated brain endothelial cells lacking ASM activity using a lentiviral shRNA approach. Interestingly, although ICAM-1 expression was increased in cells lacking ASM activity, we measured a significant decrease in T lymphocyte adhesion and consequently transmigration both in static and under flow conditions. As an underlying mechanism, we revealed that upon lack of endothelial ASM activity, the phosphorylation of ezrin was perturbed as well as the interaction between filamin and ICAM-1 upon ICAM-1 clustering. Functionally this resulted in reduced microvilli formation and impaired transendothelial migration of T cells. In conclusion, in this article, we show that ASM coordinates ICAM-1 function in brain endothelial cells by regulating its interaction with filamin and phosphorylation of ezrin. The understanding of these underlying mechanisms of T lymphocyte transmigration is of great value to develop new strategies against MS lesion formation.

Increasing evidence suggests that alterations in the sphingolipid pathway are involved in the pathogenesis of several neurologic disorders. Sphingolipids represent a class of lipids ubiquitously present in eukaryotic cells, particularly in cell membranes. Until recently, their main role was thought to organize specific domains in the plasma membrane. Nowadays, it becomes increasingly clear that sphingolipids are not just bystander molecules, but they provide an active role in modulating cellular events related to proliferation, differentiation, apoptosis, and inflammation (1, 2). Sphingolipid metabolism alterations were associated with Alzheimer’s, Huntington’s, Parkinson’s, and prion diseases, a process possibly associated with their pathogenesis (37).

Altered sphingolipid metabolism may also contribute to multiple sclerosis (MS). MS is a chronic inflammatory disorder of the CNS characterized by blood–brain barrier dysfunction, immune cell infiltration into the brain, astrogliosis, demyelination, and axonal damage (8, 9). Previously, we reported that a number of enzymes involved in sphingolipid metabolism, in particular the enzymes sphingomyelinases are deregulated in MS. Sphingomyelinases are rate-limiting enzymes involved in ceramide synthesis and have been classified in acid, neutral, or alkaline regarding their pH optimum for activity. Acid sphingomyelinase (ASM) is the acidic form and converts sphingomyelin into ceramide. Depending on its glycosylation pattern, ASM can be found in lysosomes or be secreted (10). It is thought that ASM resides in secretory lysosomes, which are directed to and fused with the plasma membrane upon certain stimuli. For instance, ASM has been shown to be activated by the inflammatory cytokines TNF-α and IL-1, and by oxidative stress (1113). We previously demonstrated that the expression of ASM is upregulated in reactive astrocytes in MS lesions (14). Furthermore, ASM-derived ceramide decreased the barrier function in brain endothelial cells (BECs) (14). In fact, ceramide, an important second lipophilic messenger involved in receptor clustering, signal transduction, and apoptosis, is one of the main effectors of ASM signaling (1517). Furthermore, ceramide is capable of self-aggregation leading to the formation of ceramide-enriched membrane rafts that fuse with each other creating bigger structures or platforms (18). These platforms are thought to trap and cluster plasma membrane receptors facilitating their signaling (16, 17, 1922).

The intercellular adhesion molecule-1 (ICAM-1) regulates the adhesion to and transmigration of T lymphocytes across the brain endothelium (23, 24). Upon binding to its counterpart, LFA-1/macrophage-1 Ag present on T cells, ICAM-1 is clustered into lipid rafts (25). Moreover, filamin, an actin regulator, links ICAM-1 to the actin cytoskeleton upon clustering, required for the formation of so-called docking structures (26), which have been reported to be involved in T cell transmigration (2730).

We have previously shown that exogenously added sphingomyelinase or ceramide increased monocyte transmigration through the brain endothelium (14). However, the mechanism by which sphingolipids regulate this process is not clearly understood. Because ICAM-1 mediates T lymphocyte diapedesis, a process detrimental during MS lesion formation, we hypothesized that ASM-derived ceramide regulates ICAM-1 function and thereby controls T cell transmigration in the brain endothelium.

Brain tissue from nonneurologic control cases and MS patients was obtained at rapid autopsy and immediately frozen in liquid nitrogen or fixed in formalin (in collaboration with the Netherlands Brain Bank, coordinator Dr. Huitinga) (Table I). The Netherlands Brain Bank received permission to perform autopsies for the use of tissue and for access to medical records for research purposes from the Ethical Committee of the VU University Medical Center (Amsterdam, the Netherlands). Tissue samples from control cases without neurologic disease were taken from the subcortical white matter. All control cases, or their next of kin, had given informed consent for autopsy and the use of their brain tissue for research purposes.

Table I.
Clinical data of MS patients and nonneurologic control cases
CasesSexAge (y)PMD (h:m)Type of MSLesion Stage
C1 62 7:20 NA NA 
C2 86 6:30 NA NA 
MS1 77 4:15 RR-SP Active 
MS2 41 7:23 PP Active 
MS3 61 9:15 RR-SP Active 
MS4 66 7:03 PP CA 
MS5 50 9:30 ND CA 
MS6 55 6:20 SP CA 
CasesSexAge (y)PMD (h:m)Type of MSLesion Stage
C1 62 7:20 NA NA 
C2 86 6:30 NA NA 
MS1 77 4:15 RR-SP Active 
MS2 41 7:23 PP Active 
MS3 61 9:15 RR-SP Active 
MS4 66 7:03 PP CA 
MS5 50 9:30 ND CA 
MS6 55 6:20 SP CA 

CA, chronic active; F, female; M, male; NA, not applicable; PMD, postmortem delay; PP, primary progressive MS; RR, relapsing remitting MS; SP, secondary progressive MS.

For immunohistochemical analysis, 5-μm cryosections were fixed in acetone for 10 min, incubated overnight at 4°C with primary Ab against ASM (rabbit; Santa Cruz Biotechnology, Santa Cruz, CA). Subsequently, sections were incubated with EnVision Dual Link (DAKO, Glostrup, Denmark) for 30 min at room temperature. Diaminobenzidine tetrachloride (DAKO, Glostrup, Denmark) was used as the chromogen. Between incubation steps, sections were thoroughly washed with PBS. After a short rinse in tap water, sections were incubated with hematoxylin for 1 min and extensively washed with tap water for 10 min. Finally, sections were dehydrated with ethanol followed by xylol and mounted with Entellan (Merck, Darmstadt, Germany). All Abs were diluted in PBS containing 0.1% BSA (Boehringer-Mannheim, Mannheimm, Germany). For colocalization studies, sections were incubated for 30 min with 10% normal goat serum (NGS), incubated overnight at 4°C with primary Ab against ASM (rabbit; Santa Cruz) after permeabilization with 0.05% Triton X-100, and Goat anti-rabbit Alexa 555 (Molecular Probes, Leiden, the Netherlands) was used as secondary Ab. To visualize endothelial cells, we incubated sections with an Ab directed against CD31 (mouse; DAKO, Glostrup, Denmark), for 1 h at room temperature. Then sections were incubated with secondary goat anti-mouse Alexa 488 (Molecular Probes, Leiden, the Netherlands). After washing, slides were covered with Vectashield (Vector Laboratories, Burlington, CA) supplemented with 0.4% DAPI to stain nuclei. Fluorescence analysis was performed with a Leica DM6000 (Leica Microsystems, Heidelberg, Germany).

The human brain BEC line hCMEC/D3 (31) was kindly provided by Dr. P.-O. Couraud (Institut Cochin, Université Paris Descartes, Paris, France). BECs were grown in endothelial growth basal medium 2 supplemented with human epidermal growth factor, hydrocortisone, GA-1000, vascular endothelial growth factor, human fibroblastic growth factor B, R3-IGF-1, ascorbic acid, and 2.5% FCS (Lonza, Basel, Switzerland). Human PBLs were recovered after monocyte isolation from buffy coats (remaining fraction) (32). These T cells were activated with 1 mg/ml PHA (Sigma Aldrich, Zwijndrecht, the Netherlands) and 10 ng/ml IL-2 (MACS; Miltenyi Biotec, Bergisch Gladbach, Germany) for 48 h.

For knockdown of ASM (National Center for Biotechnology Information Gene ID: SMPD1), we used a vector-based shRNA delivery system. shRNA expression plasmids from the TRC library (https://www.broadinstitute.org/rnai/trc/lib) targeting human SMPD1 were used to produce recombinant lentiviruses. To this end, subconfluent human embryonic kidney 293T cells were cotransfected with the shRNA lentivirus expression plasmid, packaging plasmids (pMDLg/pRRE and pRSV Rev), and the envelope vector pMD2.G using calcium phosphate as a transfection reagent. Human embryonic kidney 293T cells were cultured in DMEM supplemented with 10% FCS, 1% penicillin/streptomycin, in a 37°C incubator with 5% CO2. Infectious lentiviruses were collected 48 h after transfection, and the supernatant was centrifuged to remove cell debris. BECs were transduced with the lentivirus-containing shRNA. Forty-eight hours postinfection, stable cell lines were selected by puromycin treatment (2 μg/ml). The knockdown efficiency of all five SMPD1 library constructs was tested, and the most effective construct used in subsequent experiments was TRCN0000049014, encoding CCCAATCTGCAAAGGTCTATT targeting nt 458–478 of the NM_000543.4 SMPD1 RefSeq. The TRC SHC002 vector containing a nontargeting control (NTC) sequence of the human genome was used as a negative control.

Cell lysates of control and ASM-deficient cells were prepared with lysis buffer (Cell Signaling, Leiden, the Netherlands), and ASM activity of the supernatants was quantified using the Acid Sphingomyelinase Assay Kit (Echelon Biosciences, Salt Lake City, UT) according to manufacturer’s instructions and using the standard curve as reference. ASM activity was corrected for protein content in each sample after protein measurement (Pierce BCA Protein kit; Thermo Fisher).

mRNA was isolated from BECs using the TRIzol method (Life Technologies, Bleiswijk, the Netherlands), and cDNA was synthesized with the Reverse Transcription System kit (Promega, Leiden, the Netherlands). The following primer sequences were used: SMPD1 forward 5′-GCTGGAGCTGGAATTATTACCG-3′, SMPD1 reverse 5′-CGGCTCAGAGTCTCTTCATTCAT-3′, SMPD2 forward 5′-TGTCCGCATTGACTACGTG-3′, SMPD2 reverse 5′-ACAAACAGAGTAGCCATGAGG-3′, SMPD4 forward 5′-CTCATGGTGTTCCGAGTGG-3′, SMPD4 reverse 5′-AAGCTCCCAGTGAATGTGG-3′, GAPDH forward 5′-CCATGTTCGTCATGGGTGTG-3′, GAPDH reverse 5′-GGTGCTAAGCAGTTGGTGGTG-3′. Oligonucleotides were synthesized by Invitrogen (Bleiswijk, the Netherlands). Quantitative PCRs (qPCRs) were performed in an ABI7900HT sequence detection system using the SYBR Green method (Applied Biosystems, New York, NY). Expression levels were normalized to GAPDH expression levels.

Cells were detached from 24-well culture plates with 1 mg/ml collagenase type I (Sigma Aldrich), washed, and blocked with 5% NGS in PBS/0.1% BSA for 30 min on ice. Subsequently, cells were stained with monoclonal mouse anti–ICAM-1 (Rek-1, 5 μg/ml, a kind gift from the Department of Tumor Immunology, University Medical Center St. Radboud, Nijmegen, the Netherlands) for 30 min on ice. After washing, staining was detected using a goat anti-mouse Alexa 488 (Molecular Probes, Eugene, OR). Omission of primary Abs served as negative control. Fluorescence intensity was measured using a FACSCalibur flow cytometer (Becton Dickinson, San Jose, CA). The mean fluorescence intensity was used as a measure for the expression of ICAM-1.

Cells were plated in collagen-coated μ-slide 8-well slides (Ibidi, Martinsried, Germany) and stimulated with recombinant human TNF-α (10 ng/ml; Peprotech) for 24 h. Ab-coated polystyrene beads (10 μm; Polysciences, Eppelheim, Germany) were pretreated with 8% (v/v) glutaraldehyde for 6 h, washed five times with PBS, and incubated with 200 μg/ml ICAM-1 (Santa Cruz) according to the manufacturer’s protocol. Free glutaraldehyde groups were blocked with 0.2 M ethanolamine and 10 mg/ml BSA. Anti–ICAM-1–coated beads were added for 30 min to the cells. After extensive wash, cells were fixated with 4% formaldehyde (Sigma, St. Louis, MO) in PBS (Life Technologies). Cells were washed and blocked with PBS/0.1% BSA (Sigma) and 5% NGS. Subsequently, cells were incubated with mouse anti-ceramide primary Ab (Alexis, Lausen, Switzerland) in PBS/0.1% BSA overnight and anti–ICAM-1. Cells were washed and incubated with goat anti-mouse Alexa 488 or goat anti-rabbit Alexa 555 secondary Abs (Molecular Probes, Eugene, OR). After washing the cells with PBS, Hoechst (Molecular Probes) was used for nuclear staining. Staining was analyzed using a confocal LSM microscope (Leica).

Control and ASM-deficient BECs were seeded in 96-well plates and stimulated with TNF-α (10 ng/ml) for 24 h after cells reached confluency. Human PBLs were fluorescently labeled with 0.5 μM calcein-AM (Molecular Probes, Eugene OR) and resuspended in RPMI complete medium + 0.5% BSA + 25 mM HEPES to a final concentration of 1 × 106 T cells. A standard curve was made with 0, 12.5, 25, 50, and 100% of this T cell suspension. BECs were washed, and T cells were added to the endothelium for 30 min in a 37°C incubator with 5% CO2. Nonadherent cells were washed and adherent cells were lysed with 0.1M NaOH. Fluorescence intensity was measured (FLUOstar Galaxy; BMG Labtechnologies, Offenburg, Germany; excitation 480 nm, emission 520 nm), and the number of adhered T cells was calculated using the standard curve.

BECs were grown to confluence in 96-well plates and subsequently exposed to TNF-α (10 ng/ml) for 24 h. After extensive washing, human T cells (7.5 × 105 cells/ml) were added to BEC monolayers, and the number of migrated T cells was assessed after 4 h (33). To monitor T cell migration, we placed cocultures in an inverted phase-contrast microscope (Nikon Eclipse TE300; Lijnden, the Netherlands) housed in a temperature-controlled (37°C), 5% CO2 gassed chamber. A field (220 × 220 μm) was randomly selected and recorded for 10 min for 50 times by using a digital video camera using Cell F imaging software (Olympus, Heidelberg, Germany). Diapedesis was assessed by enumerating the number of T cells within the field that had either adhered or migrated through the monolayer. Transmigrated cells (phase-dark) could be readily distinguished from those remaining on the cell surface by their highly refractive (phase-bright) morphology. To test the role of ASM inhibitor on T cell migration, 50 μM Imipramine hydrochloride (imipramine hydrochloride; Sigma Aldrich) was added simultaneously with the T cells to BEC monolayers.

To assess the migration under physiological flow conditions, we cultured BECs to confluence in collagen-coated μ−Slide VI flow chambers (Ibidi). Cells were treated for 24 h with 10 ng/ml TNF-α, connected to a perfusion pump, subjected to the physiological flow speed of 0.8 dyn/cm2, and 5 × 105 T cells were perfused per condition. During this time leukocyte adhesion and transmigration under flow were recorded for 30 min with a Zeiss Axiovert 200 microscope (10× objective) equipped with a motorized stage. Images were recorded with Zeiss Zen 2008 software. Live Imaging was performed at 37°C and 5% CO2. Transmigrated T cells were distinguished from adhered T cells by their transition from bright to dark morphology. Quantification was performed by using the cell-counter plugin in the ImageJ software (v1.38r).

For immunofluorescence, Control (NTC shRNA) and ASM-deficient BECs (ASM shRNA) were grown on collagen-coated Lab-Tek (Thermo Fisher Scientific, Waltham, MA), treated for 24 h with 10 ng/ml TNF-α, fixed, and stained for F-actin and endogenously expressed ICAM-1. mAb against ICAM-1 was purchased from R&D Systems (Minneapolis, MN), F-actin filaments were visualized by using Acti-stain-555 phalloidin, Cytoskeleton (Cytoskeleton, Denver, CO), and secondary Alexa-coupled Abs were purchased from Invitrogen. Imaging was performed using an LSM510 Meta confocal laser-scanning microscope (Carl Zeiss MicroImaging, Jena, Germany) using Zeiss Zen 2008 software. Detailed Z-stacks of BECs were created and combined into a maximum intensity projection image (ImageJ). Microvilli were quantified using ImageJ.

BECs were seeded in collagen-coated eight-well μ-slide (Ibidi) and transfected with 0.5 μg ICAM-1–GFP construct using Dharmafect4 (Thermo Fisher Scientific) as a transfecting reagent. Cells were then stimulated with 10 ng/ml TNF-α 24 h before imaging. Slides were mounted onto a heating block connected to a confocal microscope (Zeiss LSM510). Fluorescent recovery after photobleaching (FRAP) experiments were performed using 30 iterations with 488-nm laser illumination, at maximum power (25 mW). Fluorescence recovery was measured by time-lapse imaging. Image analysis was performed with LSM510 software (Carl Zeiss MicroImaging). The first points before photobleaching were set at 100%, and the first measurement after bleaching was set to 0% to normalize the fluorescence recovery from each window.

Magnetic goat anti-mouse IgG-coated Dynabeads (Life Technologies, Bleiswijk, the Netherlands) were coated with anti–ICAM-1 mAb (R&D Systems, Abingdon, U.K.), according to the manufacturer’s protocol. In short, beads were washed with ice-cold PBS (with 0.1% BSA, 2 mM EDTA) and incubated with 1.6 μg Ab for 45 min at 4°C under constant head-over-head rotation. The beads were subsequently washed twice with PBS (with 0.1% BSA, 2 mM EDTA) to remove any unbound Ab. Endothelial cells were seeded in six-well plates, stimulated with TNF-α, and Ab-coated magnetic beads were incubated for 30 min at 37°C. For subsequent immunoprecipitation, cells were gently washed in cold PBS (supplemented with 1 mM CaCl2, 0.5 mM MgCl2) to remove unbound beads and lysed in cold RIPA buffer (1% Nonidet P-40, 10% glycerol, 100 mM NaCl, 10 mM MgCl2, 50 mM Tris, pH 7.4, 1% deoxycholate, and 0.1% SDS). Beads were extracted from cell lysates using a magnetic holder (PickPen 1M; BioNobile) and subsequently washed five times in a Nonidet P-40–based lysis buffer (1% Nonidet P-40, 10% glycerol, 100 mM NaCl2, 10 mM MgCl2, 50 mM Tris, pH 7.4). Finally, beads were resuspended in 30 μl SDS sample buffer for SDS-PAGE followed by Western blot analysis for ICAM-1 (rabbit polyclonal; Santa Cruz), filaminA (mouse monoclonal; Serotec, Puchheim, Germany), p-ezrin/radixin/moesin (p-ERM), and ezrin (Cell Signaling).

The samples were analyzed by 7.5% SDS-PAGE. Proteins were transferred to 0.45 μm nitrocellulose membrane (Schleicher and Schuell, Munich, Germany) and the blots were first blocked with 5% (w/v) BSA in TBST for 1 h, subsequently incubated at 4°C overnight with the appropriate Abs, followed by 30-min incubation with HRP-conjugated secondary Abs (DAKO, Glostrup, Denmark) at room temperature. Between the various incubation steps, the blots were washed five times with TBST and finally developed with an ECL detection system (GE Healthcare, Amersham, U.K.).

Results are shown as mean values with SEM. Statistical analysis was performed using GraphPad Prism software (v5.01; GraphPad Software, La Jolla, CA) using either unpaired Student t test or one-way ANOVA followed by post hoc Bonferroni correction. All statistical tests are described in the figure legends.

To investigate the regulation of ASM under inflammatory conditions, we exposed BECs to the inflammatory cytokine TNF-α and measured mRNA levels of ASM. The results showed that TNF-α significantly increased mRNA expression of ASM in BEC (p < 0.001; Fig. 1A). The expression of ASM by BEC was also studied in postmortem brain material from MS patients in active and chronic active lesions (Fig. 1B). Immunohistochemical analysis indicated a vascular staining pattern, although no significant difference in ASM expression comparing control subjects with MS patients with active or chronic active lesions was observed (Table I). Using ex vivo brain sections, we underscored the vascular localization of ASM by double-labeling studies of ASM with the vascular marker CD31 (Supplemental Fig. 1).

FIGURE 1.

ASM is expressed by the brain vasculature. (A) BECs were stimulated with 10 ng/ml TNF-α for 24 h, and the relative expression (RE) of ASM was measured by qPCR. TNF stimulation induced an upregulation of ASM RNA levels. Expression values were normalized using GAPDH. (B) Immunohistochemical analysis of postmortem brain tissue from nonneurologic controls, MS patients with active lesions, or MS patients with chronic active lesions showed ASM expression in the brain vasculature (arrows show vascular staining) (original magnification ×200). Data presented are mean triplicate values ± SEM of at least three independent experiments. Statistical analysis was carried out using Student t test: ***p < 0.001.

FIGURE 1.

ASM is expressed by the brain vasculature. (A) BECs were stimulated with 10 ng/ml TNF-α for 24 h, and the relative expression (RE) of ASM was measured by qPCR. TNF stimulation induced an upregulation of ASM RNA levels. Expression values were normalized using GAPDH. (B) Immunohistochemical analysis of postmortem brain tissue from nonneurologic controls, MS patients with active lesions, or MS patients with chronic active lesions showed ASM expression in the brain vasculature (arrows show vascular staining) (original magnification ×200). Data presented are mean triplicate values ± SEM of at least three independent experiments. Statistical analysis was carried out using Student t test: ***p < 0.001.

Close modal

ASM has been shown to convert sphingomyelin into ceramide at the plasma membrane. To investigate whether ceramide generation at the plasma membrane occurs at sites where ICAM-1 is present, we used beads that were coated with anti–ICAM-1 Abs to mimic T cell–induced clustering of ICAM-1, a prerequisite for T cell transmigration (27). Upon attachment, we found ceramide to accumulate at the plasma membrane around anti–ICAM-1 Ab–coated beads, suggesting recruitment of ceramide to sites of bead-induced ICAM-1 clustering (Fig. 2A, Supplemental Fig. 2).

FIGURE 2.

Ceramide colocalizes with ICAM-1 in the plasma membrane upon its clustering. (A) BECs were stimulated with 10 ng/ml TNF-α for 24 h. Anti–ICAM-1–coated beads were added for 30 min. After extensive washing to remove unattached beads, cells were fixed and stained for ceramide and ICAM-1 (upper panel) or ceramide only (lower panel). Colocalization of ceramide with ICAM-1 was assessed by confocal microscopy (original magnification ×400). Arrows show colocalization of ceramide with ICAM-1 at the plasma membrane. (B) To determine the function of ASM during T cell transmigration, we stimulated BECs with 10 ng/ml TNF-α for 24 h, and 50 μM imipramine was added during the migration experiment. T cell migration was assessed by time-lapse microscopy. Data presented are mean values ± SEM of three independent experiments (four wells per condition). Statistical analysis was carried out using one-way ANOVA: *p < 0.05, ***p < 0.001.

FIGURE 2.

Ceramide colocalizes with ICAM-1 in the plasma membrane upon its clustering. (A) BECs were stimulated with 10 ng/ml TNF-α for 24 h. Anti–ICAM-1–coated beads were added for 30 min. After extensive washing to remove unattached beads, cells were fixed and stained for ceramide and ICAM-1 (upper panel) or ceramide only (lower panel). Colocalization of ceramide with ICAM-1 was assessed by confocal microscopy (original magnification ×400). Arrows show colocalization of ceramide with ICAM-1 at the plasma membrane. (B) To determine the function of ASM during T cell transmigration, we stimulated BECs with 10 ng/ml TNF-α for 24 h, and 50 μM imipramine was added during the migration experiment. T cell migration was assessed by time-lapse microscopy. Data presented are mean values ± SEM of three independent experiments (four wells per condition). Statistical analysis was carried out using one-way ANOVA: *p < 0.05, ***p < 0.001.

Close modal

To further understand the importance of endothelial ceramide during T cell transmigration, we determined the effect of impaired ASM activity on T cell trafficking across the brain vasculature. Upon addition of the specific ASM inhibitor imipramine, a significant reduction of transendothelial migration of T cells was detected (p < 0.001; Fig. 2B). However, upon ASM blockade in the migration experiments, we could observe a decreased motility of the T cells (Supplemental Video 1). Therefore, to specifically assess the function of endothelial ASM in T cell trafficking, we reduced ASM expression in BECs using a lentiviral approach. The knockdown of ASM resulted in a decreased RNA (p < 0.001; Fig. 3A) and protein expression (p < 0.01; Fig. 3B) when compared with NTC. Furthermore, this downregulation was confirmed by a significant decrease in specific ASM activity in ASM-deficient cells (p < 0.01; Fig. 3C). Importantly, silencing of ASM did not affect the expression of neutral sphingomyelinase (Fig. 3D).

FIGURE 3.

Lentiviral knockdown of ASM in BECs. (A) BECs were transduced with lentiviruses containing shRNA against ASM. Protein knockdown levels were assessed by qPCR after puromycin selection. (B) Knockdown was confirmed by protein levels and by (C) decreased ASM activity in cell lysates. (D) ASM knockdown did not affect the expression of neutral sphingomyelinase (SMPD2 and SMPD4 genes) as determined by qPCR. Data presented are mean triplicate values ± SEM of at least three independent experiments. Statistical analysis was carried out using Student t test: **p < 0.01, ***p < 0.001.

FIGURE 3.

Lentiviral knockdown of ASM in BECs. (A) BECs were transduced with lentiviruses containing shRNA against ASM. Protein knockdown levels were assessed by qPCR after puromycin selection. (B) Knockdown was confirmed by protein levels and by (C) decreased ASM activity in cell lysates. (D) ASM knockdown did not affect the expression of neutral sphingomyelinase (SMPD2 and SMPD4 genes) as determined by qPCR. Data presented are mean triplicate values ± SEM of at least three independent experiments. Statistical analysis was carried out using Student t test: **p < 0.01, ***p < 0.001.

Close modal

Subsequent transmigration experiments showed a significant decrease in transmigration of T cells across ASM-deficient endothelial cells when compared with control cells (p < 0.05; Fig. 4A). To address whether the decreased transmigration observed in ASM-deficient cells was due to a defect in cell adhesion, we performed adhesion experiments in both control and ASM-deficient cells. We measured a minimal but significant decrease in adhesion of immune cells to ASM-deficient endothelium (p < 0.05; Fig. 4B). Interestingly, TNF-α–induced expression of ICAM-1 and VCAM-1 (data not shown) was slightly but significantly increased in ASM-deficient endothelium (p < 0.01; Fig. 4C). To mimic T cell migration under physiological conditions, we performed transendothelial migration assays under physiological flow. Strikingly, the total number of T cells that adhered and transmigrated was significantly reduced in ASM-deficient cells (p < 0.05; Fig. 4D, Supplemental Video 2). In line with this observation, fewer transmigrated T cells also were observed (p < 0.01; Fig. 4E). However, from the fraction of T cells that is able to adhere under flow in the absence of ASM, the migration capacity is not affected (Fig. 4F). These data indicate that loss of ASM mainly affects the adhesion of T cells to the endothelium.

FIGURE 4.

Cell migration is decreased in ASM-deficient endothelial monolayers. (A) T cell migration was drastically decreased through ASM-deficient cells when compared with control cells. Migration was determined in static conditions by time-lapse microscopy after 10 ng/ml TNF-α pretreatment of the endothelium for 24 h. (B) To determine T cell adhesion, we stimulated BECs for 24 h with 10 ng/ml TNF-α and T cells were added to the confluent monolayers for 30 min. T cell adhesion to ASM-deficient BECs was also decreased when compared with control cells. BECs transduced with an NTC shRNA and not exposed to TNF-α were set as 100% in both migration and adhesion analysis. Data presented are mean values ± SEM of three independent experiments (four wells per condition). (C) ICAM-1 expression in ASM-deficient cells was assessed after 24-h exposure to 10 ng/ml TNF-α by flow cytometry. For the migration under physiological flow conditions, BECs were cultured to confluence in collagen-coated μ-Slide VI flow chambers. BECs were treated for 24 h with 10 ng/ml TNF-α, connected to a perfusion pump, subjected to the physiological flow speed of 0.8 dyn/cm2, and 5 × 105 T cells were perfused per condition. Adhesion and transmigration under flow was followed for 30 min. Transendothelial migration was characterized by a change in appearance of T cells from bright to dim. (D) Graph shows the total number of T cells (both adhered and transmigrated) per mm2. (E) Graph shows the number of transmigrated T cells per mm2. (F) Graph shows the percentage of TEM (migrated T cells/adhered + migrated T cells). Statistical analysis was carried out using t test or one-way ANOVA: *p < 0.05, **p < 0.01, ***p < 0.001.

FIGURE 4.

Cell migration is decreased in ASM-deficient endothelial monolayers. (A) T cell migration was drastically decreased through ASM-deficient cells when compared with control cells. Migration was determined in static conditions by time-lapse microscopy after 10 ng/ml TNF-α pretreatment of the endothelium for 24 h. (B) To determine T cell adhesion, we stimulated BECs for 24 h with 10 ng/ml TNF-α and T cells were added to the confluent monolayers for 30 min. T cell adhesion to ASM-deficient BECs was also decreased when compared with control cells. BECs transduced with an NTC shRNA and not exposed to TNF-α were set as 100% in both migration and adhesion analysis. Data presented are mean values ± SEM of three independent experiments (four wells per condition). (C) ICAM-1 expression in ASM-deficient cells was assessed after 24-h exposure to 10 ng/ml TNF-α by flow cytometry. For the migration under physiological flow conditions, BECs were cultured to confluence in collagen-coated μ-Slide VI flow chambers. BECs were treated for 24 h with 10 ng/ml TNF-α, connected to a perfusion pump, subjected to the physiological flow speed of 0.8 dyn/cm2, and 5 × 105 T cells were perfused per condition. Adhesion and transmigration under flow was followed for 30 min. Transendothelial migration was characterized by a change in appearance of T cells from bright to dim. (D) Graph shows the total number of T cells (both adhered and transmigrated) per mm2. (E) Graph shows the number of transmigrated T cells per mm2. (F) Graph shows the percentage of TEM (migrated T cells/adhered + migrated T cells). Statistical analysis was carried out using t test or one-way ANOVA: *p < 0.05, **p < 0.01, ***p < 0.001.

Close modal

Several reports have indicated that ICAM-1–rich microvilli are involved in leukocyte adhesion and migration (29, 34, 35). Therefore, we analyzed the formation of microvilli in ASM-deficient BECs and control cells. Whereas under control conditions (NTC shRNA), TNF-α stimulation induced the formation of ICAM-1–rich F-actin+ microvilli, we observed that in ASM-deficient BECs (ASM shRNA), the formation of microvilli was reduced with a rather dispersed ICAM-1 localization (Fig. 5A, Supplemental Fig. 3). A significant reduction in microvilli-positive cells (p < 0.001; Fig. 5B) is observed as well in ASM-deficient BECs. In addition, if ASM-deficient cells express microvilli, the number of microvilli per cell is diminished as well (p < 0.01; Fig. 5C). These data demonstrated the importance of ASM for proper microvilli formation. Importantly, Oh and coworkers (34) showed that ezrin is required for the induction of these ICAM-1–rich microvilli structures. Depletion of ezrin perturbed the formation of ICAM-1–rich microvilli and reduced T cell adhesion. In addition, Kanters and coworkers (26) showed that ICAM-1 clustering resulted in the recruitment of the actin adapter filamin. Interestingly, the presence of endothelial filamin was shown to be required for ICAM-1–mediated T cell adhesion and transmigration (36). Therefore, we tested whether ASM may interfere with the association of ICAM-1 with filamin and ezrin-mediated ICAM-1–rich microvilli.

FIGURE 5.

ASM-deficient BECs express less ICAM-1+ microvilli. (A) Control (NTC shRNA) and ASM-deficient BECs (ASM shRNA) were treated for 24 h with 10 ng/ml TNF-α, fixed, and stained for F-actin and endogenously expressed ICAM-1 (original magnification ×630). (B) Bar graph shows the number of microvilli-positive cells and (C) the number of both ICAM-1 and F-actin+ microvilli per microvillus-positive cell. Statistical analysis was carried out using the Student t test: **p < 0.01, ***p < 0.001.

FIGURE 5.

ASM-deficient BECs express less ICAM-1+ microvilli. (A) Control (NTC shRNA) and ASM-deficient BECs (ASM shRNA) were treated for 24 h with 10 ng/ml TNF-α, fixed, and stained for F-actin and endogenously expressed ICAM-1 (original magnification ×630). (B) Bar graph shows the number of microvilli-positive cells and (C) the number of both ICAM-1 and F-actin+ microvilli per microvillus-positive cell. Statistical analysis was carried out using the Student t test: **p < 0.01, ***p < 0.001.

Close modal

Clustering of ICAM-1 using the Ab-coated beads in ASM-deficient endothelial cells showed impaired recruitment of filamin to ICAM-1 compared with control endothelial cells (Fig. 6A). In addition, ICAM-1 clustering induced the phosphorylation of ezrin, in line with data reported by Oh and colleagues (34). Our data showed that ezrin that is precipitated in the ICAM-1 fraction is specifically phosphorylated. We did not observe increased phosphorylation of ezrin in the total cell lysate samples, indicating that only a specific ezrin pool is phosphorylated upon ICAM-1 clustering. However, in ASM-deficient endothelial cells, phosphorylation of this specific ezrin pool downstream from ICAM-1 clustering was reduced compared with the control cells (Fig. 6A).

FIGURE 6.

Lack of ASM in BECs impairs ICAM-1 association to actin-binding proteins. (A) ASM-deficient and control BECs were stimulated for 24 h with 10 ng/ml TNF-α after which anti–ICAM-1–coated magnetic beads were added for 30 min. After cross-linking, cells were washed and ICAM-1 was pulled out using a magnetic holder. Western blot of both total cell lysates and samples where ICAM-1 was immunoprecipitated showed a decreased association of filaminA in ASM-deficient endothelial cells as well as decreased p-ERM. (B) FRAP was performed in control and ASM-deficient BECs transfected with ICAM-1–GFP, and the fluorescence recovery of GFP was recorded for 5 min (n = 8 fields of view). The initial fluorescence intensity was set at 100% and the fluorescence right after bleaching at 0%.

FIGURE 6.

Lack of ASM in BECs impairs ICAM-1 association to actin-binding proteins. (A) ASM-deficient and control BECs were stimulated for 24 h with 10 ng/ml TNF-α after which anti–ICAM-1–coated magnetic beads were added for 30 min. After cross-linking, cells were washed and ICAM-1 was pulled out using a magnetic holder. Western blot of both total cell lysates and samples where ICAM-1 was immunoprecipitated showed a decreased association of filaminA in ASM-deficient endothelial cells as well as decreased p-ERM. (B) FRAP was performed in control and ASM-deficient BECs transfected with ICAM-1–GFP, and the fluorescence recovery of GFP was recorded for 5 min (n = 8 fields of view). The initial fluorescence intensity was set at 100% and the fluorescence right after bleaching at 0%.

Close modal

We additionally tested whether the dynamics of ICAM-1 have changed in the absence of ASM activity. Therefore, we transfected ASM-deficient endothelial cells with ICAM-1–GFP and studied ICAM-1 mobility using FRAP. The results showed that there was a slight decrease in ICAM-1–GFP recovery in the absence of ASM activity, although this was not significant (Fig. 6B). These data indicate that ASM activity is required for proper ICAM-1 function to remodel the actin cytoskeleton to facilitate T cell adhesion and consequently transmigration.

Sphingolipids are becoming increasingly recognized for their involvement in neurodegeneration and neuroinflammation. However, the exact role of sphingolipids and, in particular, ASM in MS pathogenesis is still not fully understood. The development of MS lesions is largely dependent on immune cell infiltration into the brain. Upon adhesion of lymphocytes, the endothelial cells that line the lumen of brain vessels become activated and, through endothelial cell adhesion molecules such as ICAM-1 and VCAM-1, transmigrate across the blood–brain barrier. Although much is known about ICAM-1 function, not all processes concerning its regulation are fully understood. In this study, we demonstrate how proper ICAM-1 clustering in BECs is dependent on a functional ASM and its product ceramide.

We found that ASM was expressed by the inflamed brain vasculature. Next to our earlier reports on the presence of ASM in reactive astrocytes in MS lesions (14), ASM was also detected in the vasculature, although we did not detect a significant change in ASM expression compared with nonneurologic controls. This may be because of its secretion from the brain endothelium (37). We could observe that under inflammatory conditions, ASM activity in BEC supernatants is increased (M.A. Lopes Pinheiro, unpublished data), which may be due to its increased secretion. This result is in line with other studies that have demonstrated that endothelial cells are high secretors of ASM, both in naive and in inflamed conditions (37). However, it remains to be established whether the secreted form of ASM plays a role in membrane dynamics through the production of ceramide.

To further investigate the importance of ASM in ICAM-1 function, we first determined whether ceramide and ICAM-1 colocalize upon triggering. It is believed that ASM generates ceramide in the outer leaflet of the plasma membrane. Indeed, we observed that ICAM-1 colocalized with ceramide upon clustering. These results are in line with a previous study demonstrating the importance of ceramide for the endocytosis of ICAM-1–coated carriers (38). However, no functional relation has been made between ASM activity/ceramide and ICAM-1 function concerning cell migration. By blocking ASM via a selective inhibitor (imipramine) or through a lentiviral knockdown approach, we demonstrated that T cell migration was deficient in these conditions. Importantly, this was mainly due to reduced adhesion of T cells in ASM-deficient BECs. T cell diapedesis is highly dependent on adhesion molecules in the endothelium and it is well established that ICAM-1 plays a major role in this process (39). For proper intracellular signaling, ICAM-1 requires clustering and association to the actin cytoskeleton through actin adaptor proteins such as filamin (25). In this study, we demonstrate that the decreased adhesion and migration of T lymphocytes across ASM-deficient BECs is due to dysfunctional ICAM-1. Indeed, our data show that ASM-deficient BECs have reduced number of microvilli. Importantly, filamin recruitment to ICAM-1 is decreased in cells that lack ASM activity. Furthermore, ERM proteins, which are essential for microvilli and docking structure formation, were also less phosphorylated upon ICAM-1 clustering in ASM-deficient cells. Interestingly, it has been shown that phosphorylation of ERM proteins can be regulated by ASM and ceramide (40). In that study, increased ASM or ceramide was associated with increased ERM dephosphorylation, rendering these proteins to an inactive state. However, we observe that a decrease of ASM and ultimately ceramide does not change the total p-ERM pool in the cell, but are, in fact, less associated with ICAM-1. This discrepancy may be because of the type of cells used, because the effect of sphingolipids was analyzed in cancer cells, which are highly proliferative in comparison with confluent monolayers of BECs. Furthermore, we studied specific protein–protein interactions as opposed to membrane fractions, because these interactions are more informative and are related to their particular function.

Thus, based on these data, we put forward the idea that ASM activity controls proper ICAM-1 clustering to provide a perfect actin network across which T cells can adhere and transmigrate. If this delicate basis is affected, for example, by a deficient recruitment of filamin or ERM, this results in an improper association of proteins necessary for docking structure formation, ultimately slowing down or even inhibiting T cell transmigration.

Proper membrane structure is essential for the function of adhesion molecules such as VCAM-1 and ICAM-1. In fact, it has been proposed that ICAM-1 is already organized in the plasma membrane in endothelial adhesive platforms (41). In contrast, ceramide has been shown to displace cholesterol from rafts with a consequent alteration of its structure (42). Therefore, we believe that ASM-derived ceramide rearrange raft composition, possibly bringing together endothelial adhesive platforms for optimal ICAM-1 function. ASM would then be a modulator of raft dynamics and membrane structure via ceramide. Likewise ICAM-1, CD40, CD95, death receptor 5, FcγRII, and platelet-activating factor receptor are some examples of receptors for which activation is dependent on ceramide-enriched membrane platforms (16, 17, 1922).

Our data implicate that ASM plays a role in the transmigration process of T cells into the CNS under inflammatory conditions, like seen during MS. Previous data revealed that inhibitors of ASM, such as fluoxetine (43), attenuate the development of a severe form of the mouse model of MS, experimental autoimmune encephalomyelitis (44). Similar phenotype was observed in ASM-deficient mice that develop a milder clinical course of experimental autoimmune encephalomyelitis together with decreased immune cell infiltrates in the spinal cord (45). This phenotype may be caused by the function of ASM in the release of cytotoxic granules by CD8+ T cells (46) or decreased inflammatory status in glial cells (14, 45). In line with our present findings, we postulate that ASM activity in the brain endothelium also contributes to the process of deficient transmigration capacity of immune cells into the brain.

In summary, we provide evidence for the involvement of ASM and ceramide in the process of T cell migration via ICAM-1 function. ICAM-1 triggering in the plasma membrane, possibly because of ligation to VLA-4 or LFA-1, activates ASM, where ceramide rearranges the lipid structure, providing the conditions for proper ICAM-1 clustering, association with actin-binding proteins, and intracellular signaling. The understanding of processes that mediate cell migration into the brain will open new avenues for the development of new therapeutic approaches to block this process.

We thank Dr. Jos van Rijssel for some technical support in ICAM-1 immunoprecipitation assays.

This work was supported by Dutch MS Research Foundation Grant 09–358 (to M.A.L.P.) and Dutch Heart Foundation Grants 2005T039 (to J.D.v.B.) and Grant 2005T3901 (to J.K.).

The online version of this article contains supplemental material.

Abbreviations used in this article:

ASM

acid sphingomyelinase

BEC

brain endothelial cell

ERM

ezrin/radixin/moesin

FRAP

fluorescent recovery after photobleaching

ICAM-1

intercellular adhesion molecule-1

MS

multiple sclerosis

NGS

normal goat serum

NTC

nontargeting control

qPCR

quantitative PCR.

1
El Alwani
M.
,
Wu
B. X.
,
Obeid
L. M.
,
Hannun
Y. A.
.
2006
.
Bioactive sphingolipids in the modulation of the inflammatory response.
Pharmacol. Ther.
112
:
171
183
.
2
Piccinini
M.
,
Scandroglio
F.
,
Prioni
S.
,
Buccinnà
B.
,
Loberto
N.
,
Aureli
M.
,
Chigorno
V.
,
Lupino
E.
,
DeMarco
G.
,
Lomartire
A.
, et al
.
2010
.
Deregulated sphingolipid metabolism and membrane organization in neurodegenerative disorders.
Mol. Neurobiol.
41
:
314
340
.
3
Ceccom
J.
,
Loukh
N.
,
Lauwers-Cances
V.
,
Touriol
C.
,
Nicaise
Y.
,
Gentil
C.
,
Uro-Coste
E.
,
Pitson
S.
,
Maurage
C. A.
,
Duyckaerts
C.
, et al
.
2014
.
Reduced sphingosine kinase-1 and enhanced sphingosine 1-phosphate lyase expression demonstrate deregulated sphingosine 1-phosphate signaling in Alzheimer’s disease.
Acta Neuropathol. Commun.
2
:
12
21
.
4
Couttas
T. A.
,
Kain
N.
,
Daniels
B.
,
Lim
X. Y.
,
Shepherd
C.
,
Kril
J.
,
Pickford
R.
,
Li
H.
,
Garner
B.
,
Don
A. S.
.
2014
.
Loss of the neuroprotective factor Sphingosine 1-phosphate early in Alzheimer’s disease pathogenesis.
Acta Neuropathol. Commun.
2
:
9
21
.
5
Desplats
P. A.
,
Denny
C. A.
,
Kass
K. E.
,
Gilmartin
T.
,
Head
S. R.
,
Sutcliffe
J. G.
,
Seyfried
T. N.
,
Thomas
E. A.
.
2007
.
Glycolipid and ganglioside metabolism imbalances in Huntington’s disease.
Neurobiol. Dis.
27
:
265
277
.
6
Herrero
M. T.
,
Kastner
A.
,
Perez-Otaño
I.
,
Hirsch
E. C.
,
Luquin
M. R.
,
Javoy-Agid
F.
,
Del Río
J.
,
Obeso
J. A.
,
Agid
Y.
.
1993
.
Gangliosides and parkinsonism.
Neurology
43
:
2132
2134
.
7
Ohtani
Y.
,
Tamai
Y.
,
Ohnuki
Y.
,
Miura
S.
.
1996
.
Ganglioside alterations in the central and peripheral nervous systems of patients with Creutzfeldt-Jakob disease.
Neurodegeneration
5
:
331
338
.
8
Alvarez
J. I.
,
Cayrol
R.
,
Prat
A.
.
2011
.
Disruption of central nervous system barriers in multiple sclerosis.
Biochim. Biophys. Acta
.
1812
:
252
264
.
9
Frohman
E. M.
,
Racke
M. K.
,
Raine
C. S.
.
2006
.
Multiple sclerosis--the plaque and its pathogenesis.
N. Engl. J. Med.
354
:
942
955
.
10
Schissel
S. L.
,
Keesler
G. A.
,
Schuchman
E. H.
,
Williams
K. J.
,
Tabas
I.
.
1998
.
The cellular trafficking and zinc dependence of secretory and lysosomal sphingomyelinase, two products of the acid sphingomyelinase gene.
J. Biol. Chem.
273
:
18250
18259
.
11
Hofmeister
R.
,
Wiegmann
K.
,
Korherr
C.
,
Bernardo
K.
,
Krönke
M.
,
Falk
W.
.
1997
.
Activation of acid sphingomyelinase by interleukin-1 (IL-1) requires the IL-1 receptor accessory protein.
J. Biol. Chem.
272
:
27730
27736
.
12
Li
X.
,
Gulbins
E.
,
Zhang
Y.
.
2012
.
Oxidative stress triggers Ca-dependent lysosome trafficking and activation of acid sphingomyelinase.
Cell. Physiol. Biochem.
30
:
815
826
.
13
Schütze
S.
,
Potthoff
K.
,
Machleidt
T.
,
Berkovic
D.
,
Wiegmann
K.
,
Krönke
M.
.
1992
.
TNF activates NF-kappa B by phosphatidylcholine-specific phospholipase C-induced “acidic” sphingomyelin breakdown.
Cell
71
:
765
776
.
14
van Doorn
R.
,
Nijland
P. G.
,
Dekker
N.
,
Witte
M. E.
,
Lopes-Pinheiro
M. A.
,
van het Hof
B.
,
Kooij
G.
,
Reijerkerk
A.
,
Dijkstra
C.
,
van van der Valk
P.
, et al
.
2012
.
Fingolimod attenuates ceramide-induced blood-brain barrier dysfunction in multiple sclerosis by targeting reactive astrocytes.
Acta Neuropathol.
124
:
397
410
.
15
Verheij
M.
,
Bose
R.
,
Lin
X. H.
,
Yao
B.
,
Jarvis
W. D.
,
Grant
S.
,
Birrer
M. J.
,
Szabo
E.
,
Zon
L. I.
,
Kyriakis
J. M.
, et al
.
1996
.
Requirement for ceramide-initiated SAPK/JNK signalling in stress-induced apoptosis.
Nature
380
:
75
79
.
16
Grassmé
H.
,
Bock
J.
,
Kun
J.
,
Gulbins
E.
.
2002
.
Clustering of CD40 ligand is required to form a functional contact with CD40.
J. Biol. Chem.
277
:
30289
30299
.
17
Abdel Shakor
A. B.
,
Kwiatkowska
K.
,
Sobota
A.
.
2004
.
Cell surface ceramide generation precedes and controls FcgammaRII clustering and phosphorylation in rafts.
J. Biol. Chem.
279
:
36778
36787
.
18
Holopainen
J. M.
,
Subramanian
M.
,
Kinnunen
P. K.
.
1998
.
Sphingomyelinase induces lipid microdomain formation in a fluid phosphatidylcholine/sphingomyelin membrane.
Biochemistry
37
:
17562
17570
.
19
Dumitru
C. A.
,
Gulbins
E.
.
2006
.
TRAIL activates acid sphingomyelinase via a redox mechanism and releases ceramide to trigger apoptosis.
Oncogene
25
:
5612
5625
.
20
Göggel
R.
,
Winoto-Morbach
S.
,
Vielhaber
G.
,
Imai
Y.
,
Lindner
K.
,
Brade
L.
,
Brade
H.
,
Ehlers
S.
,
Slutsky
A. S.
,
Schütze
S.
, et al
.
2004
.
PAF-mediated pulmonary edema: a new role for acid sphingomyelinase and ceramide.
Nat. Med.
10
:
155
160
.
21
Grassme
H.
,
Jekle
A.
,
Riehle
A.
,
Schwarz
H.
,
Berger
J.
,
Sandhoff
K.
,
Kolesnick
R.
,
Gulbins
E.
.
2001
.
CD95 signaling via ceramide-rich membrane rafts.
J. Biol. Chem.
276
:
20589
20596
.
22
Grassmé
H.
,
Jendrossek
V.
,
Bock
J.
,
Riehle
A.
,
Gulbins
E.
.
2002
.
Ceramide-rich membrane rafts mediate CD40 clustering.
J. Immunol.
168
:
298
307
.
23
Abadier
M.
,
Haghayegh Jahromi
N.
,
Cardoso Alves
L.
,
Boscacci
R.
,
Vestweber
D.
,
Barnum
S.
,
Deutsch
U.
,
Engelhardt
B.
,
Lyck
R.
.
2015
.
Cell surface levels of endothelial ICAM-1 influence the transcellular or paracellular T-cell diapedesis across the blood-brain barrier.
Eur. J. Immunol.
45
:
1043
1058
.
24
Reiss
Y.
,
Hoch
G.
,
Deutsch
U.
,
Engelhardt
B.
.
1998
.
T cell interaction with ICAM-1-deficient endothelium in vitro: essential role for ICAM-1 and ICAM-2 in transendothelial migration of T cells.
Eur. J. Immunol.
28
:
3086
3099
.
25
Tilghman
R. W.
,
Hoover
R. L.
.
2002
.
E-selectin and ICAM-1 are incorporated into detergent-insoluble membrane domains following clustering in endothelial cells.
FEBS Lett.
525
:
83
87
.
26
Kanters
E.
,
van Rijssel
J.
,
Hensbergen
P. J.
,
Hondius
D.
,
Mul
F. P.
,
Deelder
A. M.
,
Sonnenberg
A.
,
van Buul
J. D.
,
Hordijk
P. L.
.
2008
.
Filamin B mediates ICAM-1-driven leukocyte transendothelial migration.
J. Biol. Chem.
283
:
31830
31839
.
27
van Buul
J. D.
,
Allingham
M. J.
,
Samson
T.
,
Meller
J.
,
Boulter
E.
,
García-Mata
R.
,
Burridge
K.
.
2007
.
RhoG regulates endothelial apical cup assembly downstream from ICAM1 engagement and is involved in leukocyte trans-endothelial migration.
J. Cell Biol.
178
:
1279
1293
.
28
Barreiro
O.
,
Yanez-Mo
M.
,
Serrador
J. M.
,
Montoya
M. C.
,
Vicente-Manzanares
M.
,
Tejedor
R.
,
Furthmayr
H.
,
Sanchez-Madrid
F.
.
2002
.
Dynamic interaction of VCAM-1 and ICAM-1 with moesin and ezrin in a novel endothelial docking structure for adherent leukocytes.
J. Cell Biol.
157
:
1233
1245
.
29
Carman
C. V.
,
Springer
T. A.
.
2004
.
A transmigratory cup in leukocyte diapedesis both through individual vascular endothelial cells and between them.
J. Cell Biol.
167
:
377
388
.
30
Schnoor
M.
,
Lai
F. P.
,
Zarbock
A.
,
Kläver
R.
,
Polaschegg
C.
,
Schulte
D.
,
Weich
H. A.
,
Oelkers
J. M.
,
Rottner
K.
,
Vestweber
D.
.
2011
.
Cortactin deficiency is associated with reduced neutrophil recruitment but increased vascular permeability in vivo.
J. Exp. Med.
208
:
1721
1735
.
31
Weksler
B. B.
,
Subileau
E. A.
,
Perrière
N.
,
Charneau
P.
,
Holloway
K.
,
Leveque
M.
,
Tricoire-Leignel
H.
,
Nicotra
A.
,
Bourdoulous
S.
,
Turowski
P.
, et al
.
2005
.
Blood-brain barrier-specific properties of a human adult brain endothelial cell line.
FASEB J.
19
:
1872
1874
.
32
van Doorn
R.
,
Lopes Pinheiro
M. A.
,
Kooij
G.
,
Lakeman
K.
,
van het Hof
B.
,
van der Pol
S. M.
,
Geerts
D.
,
van Horssen
J.
,
van der Valk
P.
,
van der Kam
E.
, et al
.
2012
.
Sphingosine 1-phosphate receptor 5 mediates the immune quiescence of the human brain endothelial barrier.
J. Neuroinflammation
9
:
133
.
33
Van der Goes
A.
,
Wouters
D.
,
Van Der Pol
S. M.
,
Huizinga
R.
,
Ronken
E.
,
Adamson
P.
,
Greenwood
J.
,
Dijkstra
C. D.
,
De Vries
H. E.
.
2001
.
Reactive oxygen species enhance the migration of monocytes across the blood-brain barrier in vitro.
FASEB J.
15
:
1852
1854
.
34
Oh
H. M.
,
Lee
S.
,
Na
B. R.
,
Wee
H.
,
Kim
S. H.
,
Choi
S. C.
,
Lee
K. M.
,
Jun
C. D.
.
2007
.
RKIKK motif in the intracellular domain is critical for spatial and dynamic organization of ICAM-1: functional implication for the leukocyte adhesion and transmigration.
Mol. Biol. Cell
18
:
2322
2335
.
35
van Buul
J. D.
,
van Rijssel
J.
,
van Alphen
F. P.
,
Hoogenboezem
M.
,
Tol
S.
,
Hoeben
K. A.
,
van Marle
J.
,
Mul
E. P.
,
Hordijk
P. L.
.
2010
.
Inside-out regulation of ICAM-1 dynamics in TNF-α-activated endothelium.
PLoS One
5
:
e11336
.
36
Schaefer
A.
,
Te Riet
J.
,
Ritz
K.
,
Hoogenboezem
M.
,
Anthony
E. C.
,
Mul
F. P.
,
de Vries
C. J.
,
Daemen
M. J.
,
Figdor
C. G.
,
van Buul
J. D.
,
Hordijk
P. L.
.
2014
.
Actin-binding proteins differentially regulate endothelial cell stiffness, ICAM-1 function and neutrophil transmigration.
J. Cell Sci.
127
:
4470
4482
.
37
Marathe
S.
,
Schissel
S. L.
,
Yellin
M. J.
,
Beatini
N.
,
Mintzer
R.
,
Williams
K. J.
,
Tabas
I.
.
1998
.
Human vascular endothelial cells are a rich and regulatable source of secretory sphingomyelinase. Implications for early atherogenesis and ceramide-mediated cell signaling.
J. Biol. Chem.
273
:
4081
4088
.
38
Serrano
D.
,
Bhowmick
T.
,
Chadha
R.
,
Garnacho
C.
,
Muro
S.
.
2012
.
Intercellular adhesion molecule 1 engagement modulates sphingomyelinase and ceramide, supporting uptake of drug carriers by the vascular endothelium.
Arterioscler. Thromb. Vasc. Biol.
32
:
1178
1185
.
39
van Buul
J. D.
,
Hordijk
P. L.
.
2004
.
Signaling in leukocyte transendothelial migration.
Arterioscler. Thromb. Vasc. Biol.
24
:
824
833
.
40
Canals
D.
,
Jenkins
R. W.
,
Roddy
P.
,
Hernández-Corbacho
M. J.
,
Obeid
L. M.
,
Hannun
Y. A.
.
2010
.
Differential effects of ceramide and sphingosine 1-phosphate on ERM phosphorylation: probing sphingolipid signaling at the outer plasma membrane.
J. Biol. Chem.
285
:
32476
32485
.
41
Barreiro
O.
,
Zamai
M.
,
Yáñez-Mó
M.
,
Tejera
E.
,
López-Romero
P.
,
Monk
P. N.
,
Gratton
E.
,
Caiolfa
V. R.
,
Sánchez-Madrid
F.
.
2008
.
Endothelial adhesion receptors are recruited to adherent leukocytes by inclusion in preformed tetraspanin nanoplatforms.
J. Cell Biol.
183
:
527
542
.
42
Megha
,
London
E.
.
2004
.
Ceramide selectively displaces cholesterol from ordered lipid domains (rafts): implications for lipid raft structure and function.
J. Biol. Chem.
279
:
9997
10004
.
43
Kornhuber, J., M. Muehlbacher, S. Trapp, S. Pechmann, A. Friedl, M. Reichel, C. Mühle, L. Terfloth, T. W. Groemer, and G. M. Spitzer, et al. 2011. Identification of novel functional inhibitors of acid sphingomyelinase. PloS One. 6: e23852. doi:10.1371/journal.pone.0023852
.
44
Yuan
X. Q.
,
Qiu
G.
,
Liu
X. J.
,
Liu
S.
,
Wu
Y.
,
Wang
X.
,
Lu
T.
.
2012
.
Fluoxetine promotes remission in acute experimental autoimmune encephalomyelitis in rats.
Neuroimmunomodulation
19
:
201
208
.
45
Verderio
C.
,
Muzio
L.
,
Turola
E.
,
Bergami
A.
,
Novellino
L.
,
Ruffini
F.
,
Riganti
L.
,
Corradini
I.
,
Francolini
M.
,
Garzetti
L.
, et al
.
2012
.
Myeloid microvesicles are a marker and therapeutic target for neuroinflammation.
Ann. Neurol.
72
:
610
624
.
46
Herz
J.
,
Pardo
J.
,
Kashkar
H.
,
Schramm
M.
,
Kuzmenkina
E.
,
Bos
E.
,
Wiegmann
K.
,
Wallich
R.
,
Peters
P. J.
,
Herzig
S.
, et al
.
2009
.
Acid sphingomyelinase is a key regulator of cytotoxic granule secretion by primary T lymphocytes.
Nat. Immunol.
10
:
761
768
.

The authors have no financial conflicts of interest.