Europe PMC

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

Abstract 


Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid that mediates cellular functions by ligation via G protein-coupled S1P receptors. In addition to its extracellular action, S1P also has intracellular effects; however, the signaling pathways modulated by intracellular S1P remain poorly defined. We have previously demonstrated a novel pathway of intracellular S1P generation in human lung endothelial cells (ECs). In the present study, we examined the role of intracellular S1P generated by photolysis of caged S1P on EC barrier regulation and signal transduction. Intracellular S1P released from caged S1P caused mobilization of intracellular calcium, induced activation of MAPKs, redistributed cortactin, vascular endothelial cadherin, and β-catenin to cell periphery, and tightened endothelial barrier in human pulmonary artery ECs. Treatment of cells with pertussis toxin (PTx) had no effect on caged S1P-mediated effects on Ca(2+) mobilization, reorganization of cytoskeleton, cell adherens junction proteins, and barrier enhancement; however, extracellular S1P effects were significantly attenuated by PTx. Additionally, intracellular S1P also activated small GTPase Rac1 and its effector Ras GTPase-activating-like protein IQGAP1, suggesting involvement of these proteins in the S1P-mediated changes in cell-to-cell adhesion contacts. Downregulation of sphingosine kinase 1 (SphK1), but not SphK2, with siRNA or inhibition of SphK activity with an inhibitor 2-(p-hydroxyanilino)-4-(p-chlorophenyl) thiazole (CII) attenuated exogenously administrated S1P-induced EC permeability. Furthermore, S1P1 receptor inhibitor SB649164 abolished exogenous S1P-induced transendothelial resistance changes but had no effect on intracellular S1P generated by photolysis of caged S1P. These results provide evidence that intracellular S1P modulates signal transduction in lung ECs via signaling pathway(s) independent of S1P receptors.

Free full text 


Logo of ajplungLink to Publisher's site
Am J Physiol Lung Cell Mol Physiol. 2011 Jun; 300(6): L840–L850.
Published online 2011 Apr 8. https://doi.org/10.1152/ajplung.00404.2010
PMCID: PMC3119122
PMID: 21478254

Photolysis of caged sphingosine-1-phosphate induces barrier enhancement and intracellular activation of lung endothelial cell signaling pathways

Abstract

Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid that mediates cellular functions by ligation via G protein-coupled S1P receptors. In addition to its extracellular action, S1P also has intracellular effects; however, the signaling pathways modulated by intracellular S1P remain poorly defined. We have previously demonstrated a novel pathway of intracellular S1P generation in human lung endothelial cells (ECs). In the present study, we examined the role of intracellular S1P generated by photolysis of caged S1P on EC barrier regulation and signal transduction. Intracellular S1P released from caged S1P caused mobilization of intracellular calcium, induced activation of MAPKs, redistributed cortactin, vascular endothelial cadherin, and β-catenin to cell periphery, and tightened endothelial barrier in human pulmonary artery ECs. Treatment of cells with pertussis toxin (PTx) had no effect on caged S1P-mediated effects on Ca2+ mobilization, reorganization of cytoskeleton, cell adherens junction proteins, and barrier enhancement; however, extracellular S1P effects were significantly attenuated by PTx. Additionally, intracellular S1P also activated small GTPase Rac1 and its effector Ras GTPase-activating-like protein IQGAP1, suggesting involvement of these proteins in the S1P-mediated changes in cell-to-cell adhesion contacts. Downregulation of sphingosine kinase 1 (SphK1), but not SphK2, with siRNA or inhibition of SphK activity with an inhibitor 2-(p-hydroxyanilino)-4-(p-chlorophenyl) thiazole (CII) attenuated exogenously administrated S1P-induced EC permeability. Furthermore, S1P1 receptor inhibitor SB649164 abolished exogenous S1P-induced transendothelial resistance changes but had no effect on intracellular S1P generated by photolysis of caged S1P. These results provide evidence that intracellular S1P modulates signal transduction in lung ECs via signaling pathway(s) independent of S1P receptors.

Keywords: MAPKs, cortactin, Ca2+, cell-to-cell adhesion

sphingosine-1-phosphate (S1P), a bioactive sphingolipid, secreted by erythrocytes, activated platelets, and other cells, regulates a wide range of biological processes such as cell growth, differentiation, proliferation, motility, and barrier function (10, 26, 31, 37, 38). S1P exerts dual action in cells. It is a natural ligand for five G protein-coupled receptors, S1P receptors1–5 (10, 24, 38, 40, 49), and also acts as an intracellular second messenger (4, 14, 25, 37, 50). The receptors also recognize dihydro-S1P (3, 29, 38). In cells and tissues, formation of S1P from sphingosine (Sph) is catalyzed by SphK1 and SphK2 isoforms (18, 26). S1P, however, can be degraded by S1P phosphatases to Sph and S1P lyase to hexadecenal and ethanolamine phosphate (18, 30, 38, 53). In the vascular endothelium, S1P plays an important role in protecting lungs from agonist- or sepsis-induced pulmonary leak and lung injury, as demonstrated using both in vitro and in vivo models (8, 9, 22, 28, 33). The mechanisms by which S1P regulates the integrity of the endothelial barrier, however, still remain unclear. S1P modulates endothelial cytoskeleton, particularly cortactin, an actin-binding protein, in the formation of a strong cortical actin ring in the cell periphery (8, 10, 17). It stimulates MAPKs and focal adhesion kinase, causing rearrangement of vascular endothelial (VE)-cadherin, paxillin, catenins, and zonula occludens (ZO)-1 in endothelial cells (ECs) (8, 23, 36). Additionally, S1P is a potent agonist for the regulation of intracellular calcium (4, 14, 20, 23, 24, 35, 50), which in turn plays an important role in the activation of several kinases including AKT and formation of stress fibers (41, 42, 45). Finally, S1P activates Rac1, a critical regulator of nonmuscle cytoskeleton, cell-to-cell and cell matrix-tethering forces (12, 36). In addition to its extracellular action, S1P can function as an intracellular signaling molecule. Photolysis of intracellular caged S1P mobilized cytosolic Ca2+ from thapsigargin-sensitive stores that was independent of S1P receptors in HEK-293, SKNMC, and HepG2 cells (25) and also inhibited cell motility in breast cancer cells (47).

Maintenance of EC barrier integrity is critical for vessel wall homeostasis and normal organ function. We recently demonstrated that exogenous S1P was rapidly converted to intracellular S1P in ECs, thereby raising the possibility that a novel pathway(s) may exist (32). In an attempt to understand these pathway(s), we uploaded ECs with photolyzable caged S1P that can be released upon UV illumination directly into the cytoplasm. Our data presented here with caged S1P support a role for intracellular S1P in elevating calcium levels, MAPK and Rac1/IQGAP1 activation, cytoskeleton remodeling, and modulation of barrier function in human pulmonary artery ECs (HPAECs). The responses of released S1P from its cage were pertussis toxin (PTx)-insensitive support of the concept of intracellular actions of S1P that are independent of S1P receptors (25).

MATERIALS AND METHODS

Materials.

HPAECs and endothelial basal media (EBM-2) were obtained from Lonza (San Diego, CA). PBS was from Biofluids (Rockville, MD). Glass-bottom Microwell dishes (MatTek, Ashland, MA), Fura-2 AM ester, Fluo-4 AM ester, pluronic acid (F-127), mounting media, BAPTA-AM, and precast Tris-Glycine PAAG (Invitrogen, Eugene, OR), S1P (Avanti Polar Lipids, Alabaster, AL), caged S1P (Toronto Research Chemicals, Toronto, ON, Canada), gadolinium chloride (Sigma, St. Louis, MO), thapsigargin (TG), inhibitors SP600125, SB203580, PD98050, NSC23766, and PTx (Calbiochem, San Diego, CA), SphK inhibitor, compound II, (Cayman Chemical, Ann Arbor, MI), scrambled siRNA and siRNA for SphK1 (Santa Cruz Biotechnology, Santa Cruz, CA), and GeneSilencer (Genlantis, San Diego, CA) were commercially obtained. Adenoviral constructs, vector control, and SphK1-flag (Dn) were generated at the services of the University of Iowa Gene Transfer Vector Core (Iowa City, IA). S1P1 receptor inhibitor SB649146 was kindly provided by GlaxoSmithKline. Cell lysis buffer (Cell Signaling, Danvers, MA) and immunobilon-P, 0.45 μm (Millipore, Bedford, MA), were also purchased.

Antibodies.

Anti-SphK1 antibody was purchased from Abcam (rabbit, cat. no. ab37980, Cambridge, MA). Anti-ERK1 (rabbit, cat. no. sc-93), anti-ERK2 (rabbit, cat. no. sc-154) anti-phosphospecific-ERK (mouse, cat. no. sc-7383), anti-JNK (rabbit, cat. no. sc-571), anti-phosphospecific-JNK (mouse, cat. no. sc-6254), anti-p38 (rabbit, cat. no. sc-535) anti-phosphospecific-p38 (mouse, cat. no. sc-7973), and anti-cortactin (rabbit, cat. no. sc-11408) antibodies were purchased from Santa Cruz Biotechnology. Anti-VE-cadherin (rabbit, cat. no. 160840; Cayman Chemical), anti-β-catenin (mouse, cat. no. 610153), anti-Flag (rabbit, cat. no. F7425; Sigma), secondary antibodies Alexa Fluor 488 (mouse, cat. no. A21202 and rabbit, cat. no. A11034), Alexa Fluor 568 (rabbit, cat. no. A11036) (Invitrogen), goat anti-rabbit- (cat. no. 170-6515) or anti-mouse- (cat. no. 170-6516) IgG (H+L) horseradish peroxidase conjugates (Bio-Rad, Hercules, CA) were commercially obtained.

EC culture.

HPAECs cultured in EBM, were maintained at 37°C and 5% CO2, and grown to contact-inhibited monolayers that revealed typical cobblestone morphology. Cells were then detached with 0.05% trypsin, resuspended in fresh medium, and cultured on gold electrodes for electrical resistance determinations, on glass coverslips for calcium measurement or fluorescent microscopy studies, or on 100-mm culture dishes for immunoblotting.

Loading of ECs with caged S1P.

HPAECs grown on either cell culture dishes, glass-bottom 35-mm dishes, or gold electrodes were preloaded with caged S1P (intracellularly, “in”), or caged S1P was added directly into the media (outside loading, “out”). Time for saturating the cells with caged S1P and UV flashing was established in preliminary experiments. Serum-free and phenol red-free EBM medium was used in all experiments. Intracellular loading was performed in the dark (diffused red light, Littlight lamp) with 1 μM caged S1P for 15 min, and cells were washed thrice. For the extracellular application, 1 μM caged S1P was added into the incubation media before UV stimulation as indicated. The dishes or electrodes were then flashed with UV light (254 nm) for 20 s at 5-cm distance, the shortest and most efficient time period and distance as determined for the release of S1P by a Spectroline ENF-240-C (Spectronics, New York) and a UV-light source (0.2 A).

Measurement of intracellular Ca2+ concentration.

HPAECs were grown in complete EBM as monolayers on glass coverslips or glass-bottom 35-mm dishes using a basic phenol red-free medium (in mM): 116 NaCl, 5.37 KCl, 26.2 NaHCO3, 1.8 CaCl2, 0.81 MgSO4, 1.02 NaHPO4, 5.5 glucose, 10 HEPES/HCl pH 7.40. Cells were loaded for 15 min with 5 μM Fura-2 AM (spectrofluorimetry) or with Fluo-4 AM (confocal microscopy) in the above media, in the presence of 0.1% BSA and 0.03% pluronic acid F-127 at 37°C in a cell-culture incubator as recommended by the manufacturer. Intracellular calcium was monitored with an Aminco-Bowman Series 2 luminescence spectrometer (SLM/Aminco, Urbana, IL) at excitation wavelengths of 340 and 380 nm and emission wavelength of 510 nm. Confocal microscopy imaging experiments were performed in real time using a Leica DMI6000 and ×63 (NA 1.40) objective on a SP5 resonant scanner laser scanning confocal microscope (Leica Microsystems, Wetzlar, Germany). Photo excitation of Fluo-4 was achieved by illumination at 488 nm (Ar laser), and emitted light was acquired at 510–540 nm. Data acquisition (5 Hz) was performed with Leica Confocal software. Data analysis was performed with NIH ImageJ software (27).

Preparation of cell lysates and immunoblotting.

ECs were grown on 100-mm culture dishes, and 18 h before the experiment the cells were transferred to serum-free medium. After treatment with caged S1P, dishes were rinsed with ice-cold PBS in the presence of 1 mM sodium orthovanadate. ECs were lysed in 1 ml of lysis buffer containing 1% phosphatase inhibitor cocktail, scraped off the dishes, sonicated on ice with a probe sonicator (15 s), and centrifuged at 5,000 g in a microfuge (4°C for 5 min), and protein concentrations of the supernatants were determined using Pierce protein assay kit. The supernatants, adjusted to 0.5–1.0 mg protein/ml (cell lysates) were denatured by boiling in 2× SDS sample buffer for 5 min and analyzed on 10% SDS-PAGE gels. Protein bands were transferred overnight (25 V, 4°C) on the PVDF (Millipore) membrane, probed with primary and secondary antibodies, and immunodetected by enhanced chemiluminescence (ECL Kit, Amersham). The blots were scanned (UMAX Power Lock II) and quantified by ImageJ software (27).

Immunofluorescence microscopy.

HPAECs grown on slide chambers were fixed with 3.7% paraformaldehyde in PBS for 10 min and permeabilized for 4 min in 3.7% paraformaldehyde containing 0.25% Triton X-100. In some experiments aimed at Rac1, permeabilization was performed by methanol treatment for 4 min at −20°C. Cells were then rinsed and incubated for 30 min in TBS with Tween (TBST) blocking buffer containing 1% BSA followed by incubation with primary antibodies (1:200 dilution in blocking buffer, 1 h). After being thoroughly rinsed with TBST, cells were then stained with Alexa Fluor secondary antibodies (1:200 dilutions in blocking buffer, 1 h). The washed slides were prepared using mounting media and examined with a Nikon TE 2000-S fluorescence microscope and Hamamatsu digital camera (Japan) using a ×60 oil-immersion objective lens and MetaVue software (Universal Imaging, West Chester, PA).

Infection and transfection of HPAECs.

HPAECs grown to ~80% confluence were infected with 5 pfu/ml purified adenoviral empty vector and adenoviral vector containing cDNA for SphK1-flag dominant negative. After infection (24 h) the virus-containing medium was replaced with EBM, and the experiments were carried out. In separate experiments HPAECs grown to ~50% confluence were transfected with 50 nM scrambled siRNA and SphK1 siRNA in serum-free EBM-2 medium according to the manufacturer's recommendation. After 3 h posttransfection, complete EGM-2 medium containing 10% FBS was added, and the cells were cultured for an additional 72 h.

RNA isolation and real-time RT-PCR.

Total RNA was isolated from HPAECs grown on 35-mm dishes using Trizol reagent according to the manufacturer's instruction. iQ SYBR Green Supermix was used to do the real-time measurements using iCycler by Bio-Rad. 18S (sense, 5′-GTAACCCGTTGAACCCCATT-3′, and antisense, 5′- CCATCCAATCGGTAGTAGCG-3′) was used as a housekeeping gene to normalize expression. The reaction mixture consisted of 0.3 μg of total RNA (target gene) or 0.03 μg of total RNA (18S rRNA), 12.5 μl of iQ SYBR Green, 2 μl of cDNA, 1.5 μM target primers, or 1 μM 18S rRNA primers, in a total volume of 25 μl. For all samples, reverse transcription was carried out at 25°C for 5 min, followed by cycling to 42°C for 30 min and 85°C for 5 min with iScript cDNA synthesis kit. Amplicon expression in each sample was normalized to its 18S rRNA content.

Measurement of transendothelial cell electrical resistance.

HPAECs were seeded on gold electrodes (8 wells, 10 electrodes/well) to ~95% confluence, electrodes were treated with caged S1P as described above, and transendothelial electrical resistance (TER) was measured across the EC monolayer. To estimate differences between cell-to-cell and cell-to-matrix components, total TER was resolved into values reflecting resistance to current flow beneath the cell layer (α) and resistance to current flow between adjacent cells (Rb), utilizing the method of Giaever and Keese (11), which models the endothelial monolayer mathematically.

Statistical analysis.

ANOVA with Student-Newman-Keuls test was used to compare means of clearance rates of two or more different treatment groups. The level of significance is P < 0.05 unless otherwise stated. Data are expressed as means ± SE.

RESULTS

Intracellular S1P modulates Ca2+ signaling in HPAECs.

It is well established that agonist-mediated calcium signaling is initiated through PLC/IP3 mechanisms, which induces calcium release from the endoplasmic reticulum and it triggers activation of store-operated calcium entry, resulting in Ca2+ influx from extracellular media (51). As S1P is a potent modulator of calcium signaling (23, 35, 41, 42), we examined the effect of S1P on mobilization of intracellular calcium release. There was a dose-dependent release of intracellular calcium by S1P. S1P at 10, 100, 500 and 1,000 nM corresponded to [Ca2+]i at 99 ± 11, 237 ± 14, 334 ± 19, and 365 ± 21 nM, respectively. Treatment of cells with BAPTA (25 μM), an intracellular Ca2+ chelator, for 1 h completely abolished changes in [Ca2+]i induced by S1P [control with no BAPTA - 100%; with BAPTA (25 μM) plus S1P: (1 μM), 66 ± 10%; (10 μM), 23 ± 11%; (25 μM), 9 ± 6%]. Pretreatment of HPAECs with gadolinium (Gd3+), a specific blocker of store-operated calcium channels (14, 50, 51), partially attenuated S1P-induced [Ca2+]i response (compare control - 100%, vs. Gd3+ plus S1P - 65 ± 4%), whereas treatment of cells with 5 μM TG released Ca2+ from endoplasmic reticulum, and subsequent addition of S1P caused no further change in [Ca2+]i. Furthermore, exogenous addition of S1P or its release from caged S1P (out) into incubation media or release intracellularly from loaded caged S1P (in) caused rapid and significant increase in [Ca2+]i (Fig. 1, AC). UV light alone did not cause any changes in [Ca2+]i (Fig. 1B). Pretreatment of HPAECs with TG (Fig. 1D), BAPTA (Fig. 1G), or in the presence of Gd3+- or Ca-free media (Fig. 1, E and F) attenuated intracellularly released S1P-induced changes in [Ca2+]i. It is well known that most S1P effects on cell signaling act through G protein-coupled receptors (10, 24, 29, 38, 49). Pretreatment of HPAECs with PTx, a Gi-protein receptor blocker, in a time-dependent manner, attenuated extracellular S1P-induced changes in [Ca2+]i (Fig. 2A) but had no effect on [Ca2+]i induced by intracellular S1P (Fig. 2B). These data suggest that intracellular S1P directly induces Ca release from the endoplasmic reticulum and modulates [Ca2+]i in a Gi-independent manner in HPAECs.

An external file that holds a picture, illustration, etc.
Object name is zh50061158850001.jpg

Effect of sphingosine-1-phosphate (S1P) and caged S1P on [Ca2+]i. Endothelial cells (ECs) grown on 35-mm glass-bottom dishes were loaded with calcium fluorescent indicator Fluor-4 AM, and intracellular ionic Ca2+ was monitored by confocal microscopy as described in materials and methods. A: ECs were challenged with S1P (1 μM). B: caged S1P (1 μM) was added to the incubation media. C: ECs were preloaded with caged S1P for 15 min, cells were washed, and intracellular calcium was monitored. D: same as C, but cells were treated with 5 μM thapsigargin (TG) before UV flash. EG: same as C, but cells were pretreated with Ga3+ (1 μM, 15 min) (E), or incubated in Ca-free media (F), or pretreated with BAPTA (25 μM, 1 h) (G) before UV flash. As expected, treatment with TG resulted in the depletion of intracellular Ca2+ stores, and further exposure to UV had no additional effect, demonstrating that intracellular S1P targeted intracellular stored [Ca2+]i. Representative tracings from 3 independent experiments are shown.

An external file that holds a picture, illustration, etc.
Object name is zh50061158850002.jpg

Effect of pertussis toxin (PTx) on S1P-mediated [Ca2+]i. A: ECs were pretreated with PTx (100 ng/ml) for different time intervals, loaded with Fura-2 AM, and treated with S1P (1 μM), and changes in intracellular Ca2+ were monitored by spectrofluorimetry. B: ECs were pretreated with PTx (100 ng/ml, 6 h), loaded with Fura-2 AM, and then stimulated with 1 μM S1P or caged S1P as indicated, and intracellular Ca2+ was monitored. Values are means ± SE. *Significantly different from control (P < 0.05).

Intracellular S1P activates MAPKs in HPAECs.

On the basis of earlier results that S1P activates MAPKs (5, 43, 48, 49), we next investigated the effect of intracellular S1P on MAPK activation. As shown in Fig. 3, exogenously added or released S1P as well as S1P released from caged S1P inside the cell stimulated phosphorylation of JNK, p38 MAPK, and ERK. Pretreatment of cells with PTx significantly attenuated exogenously added S1P-induced MAPK activation but had no effect on intracellularly released S1P from caged S1P on phosphorylation of JNK, p38 MAPK, and ERK (Fig. 3, A and B). We and others previously demonstrated that intracellular calcium is critical for phosphorylation of cellular proteins mediated by protein kinases (41, 42, 45). Furthermore, pretreatment of HPAECs with 25 μM BAPTA significantly attenuated MAPK phosphorylation mediated by intracellular S1P released from caged S1P (Fig. 4, A and B). These data suggest that both extracellular and intracellular S1P-induced MAPK activation is dependent on intracellular calcium.

An external file that holds a picture, illustration, etc.
Object name is zh50061158850003.jpg

Effect of PTx on S1P- and caged S1P-mediated tyrosine phosphorylation of MAPKs. ECs grown on 100-mm dishes were preincubated with PTx (100 ng/ml, for 6 h) and treated with 1 μM S1P or caged S1P as indicated. Cell lysates (20–40 μg of protein) were subjected to 10% SDS-PAGE and probed with anti-phospho-JNK or total JNK, anti-phospho-p38 MAPK and total p38, or anti-phospho-ERK and total ERK antibodies. Fold change in phospho-JNK/JNK, phospho-p38/p38, or phospho-ERK/ERK MAPKs determined from the respective Western blots by image analysis were normalized to total JNK, p38 MAPK, or ERK. Representative blots from 3 different experiments are shown. Values are means ± SE. *Significantly different from vehicle control (P < 0.05); **significantly different from PTx-treated cells for caged S1P added outside and S1P (P < 0.05).

An external file that holds a picture, illustration, etc.
Object name is zh50061158850004.jpg

Effect of BAPTA on S1P- and caged S1P-mediated tyrosine phosphorylation of MAPKs. ECs grown on 100-mm dishes were preincubated with BAPTA (25 μM) for 1 h and treated with 1 μM S1P or caged S1P as indicated. Cell lysates (20–40 μg of protein) were subjected to 10% SDS-PAGE, blotted, and probed with anti-phospho-JNK or total JNK, anti-phospho-p38 MAPK and total p38, or anti-phospho-ERK and total ERK antibodies. Fold changes in phospho-JNK/JNK, phospho-p38/p38 MAPK, or phospho-ERK/ERK were calculated from the respective Western blots by image analysis, and data were normalized to total JNK, p38 MAPK, or ERK. Representative blots from 3 different experiments are shown. Values are means ± SE. *Significantly different from vehicle control (P < 0.05); **significantly different from BAPTA untreated cells with caged S1P added outside (P < 0.05).

Intracellular S1P modulates cytoskeletal rearrangement in HPAECs.

As exogenous S1P is known to induce cytoskeletal rearrangement of proteins such as cortactin (8, 22, 34), we examined the effect of intracellular S1P released from caged S1P on cortactin reorganization. As shown in Fig. 5, control cells showed a diffused cortactin distribution, with little localization near the cell periphery. Treatment of HPAECs with extracellular S1P or intracellular S1P released from caged S1P induced translocation of cortactin to regions near the cell periphery and areas of membrane ruffle (Fig. 5, top). Pretreatment of cells with PTx attenuated cortactin redistribution by exogenously added S1P or S1P released from caged S1P outside but had no effect on intracellular S1P-mediated cortactin redistribution (Fig. 5, bottom). These data demonstrate for the first time the ability of intracellular S1P to modulate cytoskeletal reorganization in HPAECs.

An external file that holds a picture, illustration, etc.
Object name is zh50061158850005.jpg

Effect of PTx on S1P- and caged S1P-mediated cortactin redistribution. ECs grown on coverslips were preincubated with PTx (100 ng/ml, 6 h) and then stimulated with 1 μM S1P or caged S1P as indicated. Cortactin reorganization was visualized by immunocytochemistry. Shown are representative immunofluorescence images from several independent experiments.

Intracellular S1P modulates focal adhesion and adherens junction proteins in HPAECs.

Earlier studies have demonstrated the ability of S1P to stimulate redistribution of focal adhesion and adherens junction proteins to cell periphery and membrane ruffles, which in turn regulates endothelial barrier function (8, 17, 23, 36). Having established that intracellular S1P modulates cortactin, we next investigated the effect of intracellular S1P on redistribution of focal adhesion and adherens junction proteins. As shown in Fig. 6A, HPAECs that were exposed to UV flash showed a somewhat diffused localization pattern of VE-cadherin, β-catenin, and ZO-1; however, intracellular S1P promoted reorganization of VE-cadherin and β-catenin toward cell periphery and changes in ZO-1 localization, which were similar to the redistribution of adherens and tight junction proteins by S1P applied outside the cells. These results provide strong evidence for redistribution of adherens junction proteins by intracellular S1P in HPAECs.

An external file that holds a picture, illustration, etc.
Object name is zh50061158850006.jpg

Intracellular S1P mediates reorganization of adherens and tight junction proteins and activation Rac1 and IQGAP1. Human pulmonary artery ECs (HPAECs) were preloaded with caged S1P (1 μM), and S1P was released by UV flash. Alternately, cells were stimulated with S1P added outside. Reorganization of vascular endothelial (VE)-cadherin, β-catenin, and zonula occludens (ZO)-1 (A) and activation of Rac1 and IQGAP1 (B) was analyzed by immunocytochemistry. Representative immunofluorescence images from several independent experiments are shown.

Intracellular S1P activates Rac1 and IQGAP1 in HPAECs.

Earlier, it has been shown that exogenous S1P activated Rac1, cytoskeletal rearrangement, and barrier enhancement via S1PR1 in ECs (12, 23, 36). As intracellular S1P modulates [Ca2+]i and activates JNK, p38 MAPK, and ERK that is PTx independent, we next evaluated the role of intracellular S1P on Rac1 and IQGAP1 activation. As shown in Fig. 6B, in control-untreated cells, Rac1 and IQGAP1 were dispersed mainly in the cytosol with minor quantities localized to plasma membrane. However, S1P released from caged S1P induced redistribution of Rac1 and IQGAP1 to cell periphery, which was similar to the S1P applied outside (Fig. 6B). These results demonstrate that S1P generated inside HPAECs from caged S1P activates Rac1 and IQGAP1.

Intracellular S1P modulates endothelial permeability.

To further investigate intracellular S1P effects on endothelial barrier function, HPAECs were grown on gold microelectrodes, preloaded with 1 μM caged S1P that was released inside the cell by UV flash. The resulting changes in TER were monitored as an index of endothelial permeability. As shown in Fig. 7A, the release of intracellular S1P resulted in a rapid and significant increase in TER, a reflection of a proportional decrease in EC permeability. This effect is similar to S1P action applied exogenously as described previously by us and others (8, 10, 14, 34). Caged S1P alone added to the incubation media without exposure to UV flash had no effect on TER, suggesting that release of S1P from the cage is critical for its EC barrier enhancement. S1P released into incubation media by UV light demonstrated similar effect as that of intracellular S1P (Fig. 7A). Exposure of cells to UV flash alone did not result in any significant changes in the TER. Additionally, our study demonstrates that the major alteration in TER after S1P treatment, both extracellular and intracellular, was attributable to changes in Rb (cell-to-cell contacts) through increased cell-to-cell adhesion (Fig. 7B). In contrast to Rb, very little change to α (cell-matrix contacts) occurred, which is a measure of the distance between the endothelial plasma membrane and the surface of the electrode. The above results indicated for the first time that intracellular S1P modulates barrier function through the changes in cell-to-cell adhesion contacts.

An external file that holds a picture, illustration, etc.
Object name is zh50061158850007.jpg

Intracellular S1P induces barrier function in ECs through cell-to-cell adhesion contacts. HPAECs grown on gold electrodes were loaded with caged S1P so as to generate intracellular S1P after photolysis with UV (A) or treated directly with extracellular caged S1P and transendothelial electrical resistance (TER) measured. Cell-to-cell (Rb) and cell-matrix (α) components (B) were resolved from A using manufacturer's software. Shown are representative tracings from 3 independent experiments.

Downregulation of Sph kinase 1 expression or activity attenuates S1P-induced intracellular calcium and barrier enhancement.

To further understand the role of intracellular S1P, we investigated whether downregulation of Sph kinase 1 (SphK1) expression or activity modulates extracellular action of S1P. Infection of HPAECs with SphK1-flag, (Dn) significantly increased expression of the protein that attenuated S1P-induced intracellular Ca2+ (Fig. 8A). This suggests a role for intracellular S1P generated by SphK1 in [Ca2+]i. Next, we investigated the role of SphK1 on S1P-induced increases in endothelial permeability. Transfection of HPAECs with SphK1 siRNA knocked down SphK1 protein expression as evidenced by real-time PCR and Western blotting (Fig. 8B) and reduced ability of cells to convert Sph to S1P (2). Furthermore, downregulation of SphK1 by siRNA attenuated S1P-induced increase of TER compared with scrambled siRNA (Fig. 8B). However, knockdown of SphK2 had no effect on S1P-induced TER (data not shown). Similarly, inhibiting SphK activity with SphK inhibitor, CII, also prevented S1P-mediated TER changes (Fig. 8B). These results suggest a role for intracellular S1P generated by SphK1 in exogenous S1P mediated [Ca2+]i and endothelial barrier enhancement.

An external file that holds a picture, illustration, etc.
Object name is zh50061158850008.jpg

Downregulation of sphingosine kinase 1 (SphK1) attenuates S1P-induced intracellular calcium and transendothelial barrier enhancement. A: HPAECs grown on glass coverslips infected with 5 pfu/ml of vector control or SphK1-flag (Dn) for 24 h and intracellular Ca2+ were monitored by spectrofluorimetry as described in materials and methods. Cell lysates were probed by Western blotting as indicated. B: HPAECs were transfected with 50 nM scrambled siRNA or SphK1 siRNA for 72 h, and cell lysates were analyzed for SphK1 expression by real-time PCR and immunoblotting (IB) as indicated. HPAECs grown on gold electrodes or 35-mm dishes were transfected with 50 nM scrambled siRNA or SphK1 siRNA for 72 h or pretreated with 10 μM CII (SphK1 inhibitor) as indicated and were then challenged with S1P (1 μM), and TER was measured. Values are mean ± SE. *Significantly different from vector control or control (P < 0.05). **Significantly different from S1P-treated cells in scrambled RNA (P < 0.05).

The role of S1P1 receptor and Rac1 on intracellular S1P-mediated endothelial barrier enhancement.

Several signaling pathways including activation of G protein-coupled S1P receptors have been implicated in S1P-induced endothelial barrier function. Having established that intracellularly generated S1P from caged S1P modulates endothelial signaling and that barrier function was PTx independent, we next examined the role of S1P1 receptor and Rac1 activation in intracellular S1P-mediated endothelial barrier regulation. HPAECs were pretreated with SB649146, a S1P1 receptor inhibitor, or NSC23766, a Rac1 inhibitor, and S1P-induced TER changes were measured. Treatment of cells with SB649146 attenuated TER mediated by S1P released outside the cell from caged S1P (control: caged S1P, out - 312 ± 46; SB649146 + caged-S1P, out - 57 ± 17) and had no effect, however, on TER mediated by S1P released inside the cell from caged S1P (Fig. 9A). In contrast to SB649146, treatment of cells with NSC23766, an inhibitor of Rac1, blocked TER changes mediated by extracellular S1P released from caged S1P (data not shown) or intracellular S1P generated from caged S1P (Fig. 9B). These results suggest that intracellular S1P generated from caged S1P modulates endothelial barrier function independent of S1P1 receptor but requires Rac1.

An external file that holds a picture, illustration, etc.
Object name is zh50061158850009.jpg

The role of S1P1 receptor and Rac1 activation in the intracellular S1P-mediated barrier function in ECs. HPAECs grown on gold electrodes were pretreated with S1P1 receptor inhibitor SB649146 (1 μM, 3 h) (A) or Rac1 inhibitor NSC23766 (50 μM, 30 min) (B) and were then loaded with caged S1P so as to generate intracellular S1P after photolysis with UV and TER measured. Values are means ± SE. *Significantly different from control (P < 0.05).

DISCUSSION

S1P is a naturally occurring bioactive lipid that has both extracellular and intracellular effects in mammalian cells. Extracellular action of S1P is through its G protein-coupled S1PRs that trigger subsequent activation of downstream targets such as Rho-GTPases, cytoskeletal reorganization, adherens and tight-junction assembly, and focal adhesion formation (8, 21, 29, 37). Also, there is some evidence for intracellular S1P-mediated effects; however, the targets and underlying mechanisms are yet to be fully defined. In the cell, S1P is generated by phosphorylation of Sph catalyzed by SphK1 and SphK2 (18). S1P is catabolized by S1P phosphatases to Sph or by S1P lyase to ethanolamine phosphate and hexadecenal (3, 18, 26, 29, 38, 53). The accumulation of S1P in cells is a natural balance between its synthesis and catabolism. Whereas some of the earlier studies have used inhibitors of SphKs to determine intracellular action of S1P, we have taken the approach of generating intracellular S1P by uploading the cells with caged S1P followed by UV photolysis (32). In the present study, using such an approach, we show that intracellular S1P 1) increases intracellular calcium [Ca2+]i, 2) activates MAPKs, JNK, p38 MAPK and ERK, which are PTx insensitive, 3) induces cortical actin redistribution to cell periphery that is also PTx insensitive, 4) stimulates rearrangement of focal adhesion and tight junction proteins VE-cadherin, β-catenin, and ZO-1, 5) activates small GTPase Rac1 and its effector IQGAP1, and 6) decreases EC permeability or tightens the barrier through the changes in cell-to-cell adhesion contacts.

S1P is a potent modulator of intracellular calcium release in ECs (23, 35, 41, 42). Exogenous S1P mobilizes Ca2+ from intracellular stores either through an IP3-dependent or -independent mechanism (4, 20, 24, 37). Furthermore, S1P-mediated Ca2+ mobilization was independent of mitochondrial Ca2+ stores as evidenced by studies with uptake inhibitor, CCCP, or release inhibitor, cyclosporine (35). Use of caged S1P to evaluate potential effects of intracellular S1P on DNA synthesis has been previously reported (32). Our results demonstrate that photolysis of caged S1P raised intracellular free Ca2+ levels in human lung ECs, which are in agreement with an earlier report that also used caged S1P in SKNMC and HepG2 cells (25). In the present study, caged S1P altered Ca2+ mobilization in HPAECs, which were sensitive to TG and BAPTA. Treatment of cells with TG released Ca2+ from endoplasmic reticulum, and subsequent photolysis of caged S1P inside the cell did not alter [Ca2+]i . However, gadolinium (Gd3+), a specific blocker of store-operated calcium channels, or Ca-free media abolished calcium influx from extracellular space but did not alter Ca2+ release from the endoplasmic reticulum. These data support the idea that S1P released intracellularly directly induces Ca2+ release from the endoplasmic reticulum. Additionally, we show that the caged S1P-induced Ca2+ mobilization was PTx insensitive compared with exogenous S1P-mediated Ca2+ release that was PTx sensitive. In mammalian cells, intracellular S1P is generated by phosphorylation of Sph catalyzed by SphK1 and 2. In the present study, a role for intracellular S1P in [Ca2+]i and endothelial permeability was demonstrated by modulating SphK1 with dominant-negative SphK1, SphK1 siRNA, or SphK1 inhibitor (Fig. 8). Interestingly, inhibition of S1P1 receptor with SB649146 had no effect on endothelial permeability mediated by intracellularly generated S1P from caged S1P inside the cell (Fig. 9A), suggesting a S1P1 receptor-independent mechanism in barrier regulation by intracellular S1P.

It is well recognized that increased vascular EC permeability and edema are hallmarks of many lung inflammatory diseases (21). During acute lung injury, acute respiratory distress syndrome, or thrombocytopenia, increased capillary permeability accelerates fluid and protein extravagation. The effect is reversible with infusion of platelets or platelet-released product, S1P. S1P and its analogs have proven effective in reducing pulmonary leak and barrier dysfunction as shown in in vivo and in vitro models (6, 21, 28, 33). Infusion of S1P in murine and canine models significantly reduced LPS-induced microvascular permeability, inflammation, and phagocyte infiltration (28). Ligation of S1P to S1P1/S1P3 receptors in human lung ECs leads to reorganization of cytoskeletal proteins, enhanced junctional integrity, and tightened endothelial barrier (22). Our data with caged S1P show that intracellularly generated S1P, similar to exogenous S1P, mediates redistribution of VE-cadherin, β-catenin, ZO-1, and Rac1/IQGAP1 to cell periphery (Fig. 6). Unlike previous studies (35), we show here that intracellular S1P-mediated stimulation of [Ca2+]i is PTx insensitive, suggesting a novel underlying mechanism utilized by intracellular S1P as opposed to that mediated by extracellular S1P bound to its cell surface receptors. There are only a few studies that describe calcium release from the endoplasmic reticulum by intracellular S1P (4, 25). However, there are a number of reports on sphingolipid-mediated [Ca2+]i changes, which cannot be attributed to G protein-coupled receptors (4, 25, 50, 51). The presence of a “sphingolipid-calcium release-mediated protein of the ER” was postulated as the Ca channels that appear to be a radically different to either InsP3R or Ryanodine receptors (15, 16, 19, 50).

Several studies have shown that extracellular S1P activates MAPKs, especially p42/p44 MAPK, through Gi-coupled S1P receptors, which are sensitive to PTx (31, 48). Here, we have demonstrated for the first time that S1P released inside the cell from caged S1P stimulated phosphorylation of JNK, p38 MAPK, and ERK. Furthermore, caged S1P-mediated phosphorylation of MAPKs was insensitive to PTx, whereas activation of MAPKs by exogenous S1P was PTx sensitive. Activation of MAPKs by extracellular S1P regulates cellular responses such as DNA synthesis, cell migration, and endothelial capillary tube formation (5). Our results show that inhibition of MAPK by pharmacological inhibitors such as SP600125 (JNK), SB203580 (p38), and PD98050 (ERK) has no effect on S1P-induced redistribution of focal adhesion, tight junction proteins, and Rac1/IQGAP1 redistribution (Supplemental Figs. S1 and S2; supplemental material for this article is available online at the American Journal of Physiology Lung Cellular and Molecular Physiology website). Thus, although the data suggest that both extracellular S1P and intracellular S1P stimulate phosphorylation of JNK, p38 MAPK and ERK, the MAPK signaling pathway is not involved in S1P-induced barrier enhancement in HPAECs.

Although much is known on signaling pathways and extracellular action of S1P via its G protein-coupled receptors, very little is known on the role of intracellular S1P and its targets in mammalian cells. Recent studies suggest potential interaction between S1P and histone acetylase 2 in breast cancer cells (13) and S1P as a cofactor for E3 ubiquitin ligase TRAF2 in HEK-293 cells (1). We have recently demonstrated that intracellularly generated S1P offers protection against LPS-induced lung injury and inflammation in a murine model of acute lung injury (52). Furthermore, EC motility mediated by extracellular S1P was dependent on intracellular S1P production, which was regulated by SphK1 and S1P lyase (2). Our results show that downregulation of SphK1 expression or activity and Rac1, but not S1P1 receptor, attenuated S1P-induced endothelial barrier enhancement (Figs. 8B and and9,9, A and B), further supporting a role for intracellular S1P in modulating signaling pathways independent of S1P receptors in the endothelium. Mechanisms of intracellular action of S1P are yet to be completely characterized (39); however, the action of phosphatidic acid (PA) generated from phospholipase D signal transduction in OVCAR-3 cells seems to act as a membrane anchor of Rac1 with the COOH-terminal polybasic motif of Rac1 (7) being responsible for the direct interaction with PA. Therefore, it is possible that S1P, like PA, may directly bind to target proteins such as Rac1 (7) and induce dissociation of the guanine nucleotide inhibitor from Rac1 for activation and redistribution to cell periphery (44, 46).

In summary, our results presented here demonstrate a role for intracellular S1P in the elevation of intracellular calcium levels, activation of MAPKs and Rac1/IQGAP1, redistribution of cytoskeletal, focal adhesion and tight junction proteins, and modulation of endothelial barrier function. These results support the notion that intracellular S1P can modulate signaling pathways independent of S1P receptors in the endothelium.

GRANTS

This work was supported by grants from National Institutes of Health R37 HL 079396 and P01 HL 098050 to V. Natarajan.

DISCLOSURES

No conflicts of interest, financial or otherwise are declared by the authors.

Supplementary Material

Supplemental Figures:

REFERENCES

1. Alvarez SE, Harikumar KB, Hait NC, Allegood J, Strub GM, Kim EY, Maceyka M, Jiang H, Luo C, Kordula T, Milstien S, Spiegel S. Sphingosine-1-phosphate is a missing cofactor for the E3 ubiquitin ligase TRAF2. Nature 465: 1084–1088 [Europe PMC free article] [Abstract] [Google Scholar]
2. Berdyshev EV, Gorshkova I, Usatyuk P, Kalari S, Zhao Y, Pyne NJ, Pyne S, Sabbadini RA, Garcia JG, Natarajan V. Intracellular S1P generation is essential for S1P-induced motility of human lung endothelial cells: role of sphingosine kinase 1 and S1P lyase. PLos One 6: e16571. [Europe PMC free article] [Abstract] [Google Scholar]
3. Berdyshev EV, Gorshkova IA, Usatyuk P, Zhao Y, Saatian B, Hubbard W, Natarajan V. De novo biosynthesis of dihydrosphingosine-1-phosphate by sphingosine kinase 1 in mammalian cells. Cell Signal 18: 1779–1792, 2006 [Abstract] [Google Scholar]
4. Blom T, Slotte JP, Pitson SM, Tornquist K. Enhancement of intracellular sphingosine-1-phosphate production by inositol 1,4,5-trisphosphate-evoked calcium mobilisation in HEK-293 cells: endogenous sphingosine-1-phosphate as a modulator of the calcium response. Cell Signal 17: 827–836, 2005 [Abstract] [Google Scholar]
5. Bogatcheva NV, Dudek SM, Garcia JG, Verin AD. Mitogen-activated protein kinases in endothelial pathophysiology. J Investig Med 51: 341–352, 2003 [Abstract] [Google Scholar]
6. Camp SM, Bittman R, Chiang ET, Moreno-Vinasco L, Mirzapoiazova T, Sammani S, Lu X, Sun C, Harbeck M, Roe M, Natarajan V, Garcia JG, Dudek SM. Synthetic analogs of FTY720 [2-amino-2-(2-[4-octylphenyl]ethyl)-1,3-propanediol] differentially regulate pulmonary vascular permeability in vivo and in vitro. J Pharmacol Exp Ther 331: 54–64, 2009 [Europe PMC free article] [Abstract] [Google Scholar]
7. Chae YC, Kim JH, Kim KL, Kim HW, Lee HY, Heo WD, Meyer T, Suh PG, Ryu SH. Phospholipase D activity regulates integrin-mediated cell spreading and migration by inducing GTP-Rac translocation to the plasma membrane. Mol Biol Cell 19: 3111–3123, 2008 [Europe PMC free article] [Abstract] [Google Scholar]
8. Dudek SM, Jacobson JR, Chiang ET, Birukov KG, Wang P, Zhan X, Garcia JG. Pulmonary endothelial cell barrier enhancement by sphingosine 1-phosphate: roles for cortactin and myosin light chain kinase. J Biol Chem 279: 24692–24700, 2004 [Abstract] [Google Scholar]
9. English D, Welch Z, Kovala AT, Harvey K, Volpert OV, Brindley DN, Garcia JG. Sphingosine 1-phosphate released from platelets during clotting accounts for the potent endothelial cell chemotactic activity of blood serum and provides a novel link between hemostasis and angiogenesis. FASEB J 14: 2255–2265, 2000 [Abstract] [Google Scholar]
10. Garcia JG, Liu F, Verin AD, Birukova A, Dechert MA, Gerthoffer WT, Bamberg JR, English D. Sphingosine 1-phosphate promotes endothelial cell barrier integrity by Edg-dependent cytoskeletal rearrangement. J Clin Invest 108: 689–701, 2001 [Europe PMC free article] [Abstract] [Google Scholar]
11. Giaever I, Keese CR. A morphological biosensor for mammalian cells. Nature 366: 591–592, 1993 [Abstract] [Google Scholar]
12. Gorshkova I, He D, Berdyshev E, Usatuyk P, Burns M, Kalari S, Zhao Y, Pendyala S, Garcia JG, Pyne NJ, Brindley DN, Natarajan V. Protein kinase C-epsilon regulates sphingosine 1-phosphate-mediated migration of human lung endothelial cells through activation of phospholipase D2, protein kinase C-zeta, and Rac1. J Biol Chem 283: 11794–11806, 2008 [Europe PMC free article] [Abstract] [Google Scholar]
13. Hait NC, Allegood J, Maceyka M, Strub GM, Harikumar KB, Singh SK, Luo C, Marmorstein R, Kordula T, Milstien S, Spiegel S. Regulation of histone acetylation in the nucleus by sphingosine-1-phosphate. Science 325: 1254–1257, 2009 [Europe PMC free article] [Abstract] [Google Scholar]
14. Itagaki K, Yun JK, Hengst JA, Yatani A, Hauser CJ, Spolarics Z, Deitch EA. Sphingosine 1-phosphate has dual functions in the regulation of endothelial cell permeability and Ca2+ metabolism. J Pharmacol Exp Ther 323: 186–191, 2007 [Abstract] [Google Scholar]
15. Kim S, Lakhani V, Costa DJ, Sharara AI, Fitz JG, Huang LW, Peters KG, Kindman LA. Sphingolipid-gated Ca2+ release from intracellular stores of endothelial cells is mediated by a novel Ca(2+)-permeable channel. J Biol Chem 270: 5266–5269, 1995 [Abstract] [Google Scholar]
16. Kindman LA, Kim S, McDonald TV, Gardner P. Characterization of a novel intracellular sphingolipid-gated Ca(2+)-permeable channel from rat basophilic leukemia cells. J Biol Chem 269: 13088–13091, 1994 [Abstract] [Google Scholar]
17. Lee JF, Ozaki H, Zhan X, Wang E, Hla T, Lee MJ. Sphingosine-1-phosphate signaling regulates lamellipodia localization of cortactin complexes in endothelial cells. Histochem Cell Biol 126: 297–304, 2006 [Abstract] [Google Scholar]
18. Maceyka M, Sankala H, Hait NC, Le Stunff H, Liu H, Toman R, Collier C, Zhang M, Satin LS, Merrill AH, Jr, Milstien S, Spiegel S. SphK1 and SphK2, sphingosine kinase isoenzymes with opposing functions in sphingolipid metabolism. J Biol Chem 280: 37118–37129, 2005 [Abstract] [Google Scholar]
19. Mao C, Kim SH, Almenoff JS, Rudner XL, Kearney DM, Kindman LA. Molecular cloning and characterization of SCaMPER, a sphingolipid Ca2+ release-mediating protein from endoplasmic reticulum. Proc Natl Acad Sci USA 93: 1993–1996, 1996 [Europe PMC free article] [Abstract] [Google Scholar]
20. Mattie M, Brooker G, Spiegel S. Sphingosine-1-phosphate, a putative second messenger, mobilizes calcium from internal stores via an inositol trisphosphate-independent pathway. J Biol Chem 269: 3181–3188, 1994 [Abstract] [Google Scholar]
21. McVerry BJ, Garcia JG. In vitro and in vivo modulation of vascular barrier integrity by sphingosine 1-phosphate: mechanistic insights. Cell Signal 17: 131–139, 2005 [Abstract] [Google Scholar]
22. McVerry BJ, Peng X, Hassoun PM, Sammani S, Simon BA, Garcia JG. Sphingosine 1-phosphate reduces vascular leak in murine and canine models of acute lung injury. Am J Respir Crit Care Med 170: 987–993, 2004 [Abstract] [Google Scholar]
23. Mehta D, Konstantoulaki M, Ahmmed GU, Malik AB. Sphingosine 1-phosphate-induced mobilization of intracellular Ca2+ mediates rac activation and adherens junction assembly in endothelial cells. J Biol Chem 280: 17320–17328, 2005 [Abstract] [Google Scholar]
24. Meyer zu Heringdorf D, Lass H, Alemany R, Laser KT, Neumann E, Zhang C, Schmidt M, Rauen U, Jakobs KH, van Koppen CJ. Sphingosine kinase-mediated Ca2+ signalling by G-protein-coupled receptors. EMBO J 17: 2830–2837, 1998 [Europe PMC free article] [Abstract] [Google Scholar]
25. Meyer zu Heringdorf D, Liliom K, Schaefer M, Danneberg K, Jaggar JH, Tigyi G, Jakobs KH. Photolysis of intracellular caged sphingosine-1-phosphate causes Ca2+ mobilization independently of G-protein-coupled receptors. FEBS Lett 554: 443–449, 2003 [Abstract] [Google Scholar]
26. Olivera A, Spiegel S. Sphingosine kinase: a mediator of vital cellular functions. Prostaglandins Other Lipid Mediat 64: 123–134, 2001 [Abstract] [Google Scholar]
27. O'Neill RR, Mitchell LG, Merril CR, Rasband WS. Use of image analysis to quantitate changes in form of mitochondrial DNA after x-irradiation. Appl Theor Electrophor 1: 163–167, 1989 [Abstract] [Google Scholar]
28. Peng X, Hassoun PM, Sammani S, McVerry BJ, Burne MJ, Rabb H, Pearse D, Tuder RM, Garcia JG. Protective effects of sphingosine 1-phosphate in murine endotoxin-induced inflammatory lung injury. Am J Respir Crit Care Med 169: 1245–1251, 2004 [Abstract] [Google Scholar]
29. Pyne NJ, Pyne S. Sphingosine 1-phosphate, lysophosphatidic acid and growth factor signaling and termination. Biochim Biophys Acta 1781: 467–476, 2008 [Abstract] [Google Scholar]
30. Pyne S, Long JS, Ktistakis NT, Pyne NJ. Lipid phosphate phosphatases and lipid phosphate signalling. Biochem Soc Trans 33: 1370–1374, 2005 [Abstract] [Google Scholar]
31. Pyne S, Pyne NJ. Sphingosine 1-phosphate signalling and termination at lipid phosphate receptors. Biochim Biophys Acta 1582: 121–131, 2002 [Abstract] [Google Scholar]
32. Qiao L, Kozikowski AP, Olivera A, Spiegel S. Synthesis and evaluation of a photolyzable derivative of sphingosine 1-phosphate–caged SPP. Bioorg Med Chem Lett 8: 711–714, 1998 [Abstract] [Google Scholar]
33. Sammani S, Moreno-Vinasco L, Mirzapoiazova T, Singleton PA, Chiang ET, Evenoski CL, Wang T, Mathew B, Husain A, Moitra J, Sun X, Nunez L, Jacobson JR, Dudek SM, Natarajan V, Garcia JG. Differential Effects of S1P Receptors on Airway and Vascular Barrier Function in the Murine Lung. Am J Respir Cell Mol Biol 43: 394–402, 2009 [Europe PMC free article] [Abstract] [Google Scholar]
34. Schaphorst KL, Chiang E, Jacobs KN, Zaiman A, Natarajan V, Wigley F, Garcia JG. Role of sphingosine-1 phosphate in the enhancement of endothelial barrier integrity by platelet-released products. Am J Physiol Lung Cell Mol Physiol 285: L258–L267, 2003 [Abstract] [Google Scholar]
35. Seol GH, Kim MY, Liang GH, Kim JA, Kim YJ, Oh S, Suh SH. Sphingosine-1-phosphate-induced intracellular Ca2+ mobilization in human endothelial cells. Endothelium 12: 263–269, 2005 [Abstract] [Google Scholar]
36. Singleton PA, Dudek SM, Chiang ET, Garcia JG. Regulation of sphingosine 1-phosphate-induced endothelial cytoskeletal rearrangement and barrier enhancement by S1P1 receptor, PI3 kinase, Tiam1/Rac1, and alpha-actinin. FASEB J 19: 1646–1656, 2005 [Abstract] [Google Scholar]
37. Spiegel S, Milstien S. Exogenous and intracellularly generated sphingosine 1-phosphate can regulate cellular processes by divergent pathways. Biochem Soc Trans 31: 1216–1219, 2003 [Abstract] [Google Scholar]
38. Spiegel S, Milstien S. Sphingosine-1-phosphate: an enigmatic signalling lipid. Nat Rev Mol Cell Biol 4: 397–407, 2003 [Abstract] [Google Scholar]
39. Strub GM, Maceyka M, Hait NC, Milstien S, Spiegel S. Extracellular and intracellular actions of sphingosine-1-phosphate. Adv Exp Med Biol 688: 141–155 [Europe PMC free article] [Abstract] [Google Scholar]
40. Tauseef M, Kini V, Knezevic N, Brannan M, Ramchandaran R, Fyrst H, Saba J, Vogel SM, Malik AB, Mehta D. Activation of sphingosine kinase-1 reverses the increase in lung vascular permeability through sphingosine-1-phosphate receptor signaling in endothelial cells. Circ Res 103: 1164–1172, 2008 [Europe PMC free article] [Abstract] [Google Scholar]
41. Tiruppathi C, Ahmmed GU, Vogel SM, Malik AB. Ca2+ signaling, TRP channels, and endothelial permeability. Microcirculation 13: 693–708, 2006 [Abstract] [Google Scholar]
42. Tiruppathi C, Minshall RD, Paria BC, Vogel SM, Malik AB. Role of Ca2+ signaling in the regulation of endothelial permeability. Vascul Pharmacol 39: 173–185, 2002 [Abstract] [Google Scholar]
43. Tolan D, Conway AM, Rakhit S, Pyne N, Pyne S. Assessment of the extracellular and intracellular actions of sphingosine 1-phosphate by using the p42/p44 mitogen-activated protein kinase cascade as a model. Cell Signal 11: 349–354, 1999 [Abstract] [Google Scholar]
44. Ugolev Y, Berdichevsky Y, Weinbaum C, Pick E. Dissociation of Rac1(GDP) RhoGDI complexes by the cooperative action of anionic liposomes containing phosphatidylinositol 3,4,5-trisphosphate, Rac guanine nucleotide exchange factor, and GTP. J Biol Chem 283: 22257–22271, 2008 [Europe PMC free article] [Abstract] [Google Scholar]
45. Usatyuk PV, Fomin VP, Shi S, Garcia JG, Schaphorst K, Natarajan V. Role of Ca2+ in diperoxovanadate-induced cytoskeletal remodeling and endothelial cell barrier function. Am J Physiol Lung Cell Mol Physiol 285: L1006–L1017, 2003 [Abstract] [Google Scholar]
46. Usatyuk PV, Gorshkova IA, He D, Zhao Y, Kalari SK, Garcia JG, Natarajan V. Phospholipase D-mediated activation of IQGAP1 through Rac1 regulates hyperoxia-induced p47phox translocation and reactive oxygen species generation in lung endothelial cells. J Biol Chem 284: 15339–15352, 2009 [Europe PMC free article] [Abstract] [Google Scholar]
47. Wang F, Van Brocklyn JR, Edsall L, Nava VE, Spiegel S. Sphingosine-1-phosphate inhibits motility of human breast cancer cells independently of cell surface receptors. Cancer Res 59: 6185–6191, 1999 [Abstract] [Google Scholar]
48. Wang L, Cummings R, Usatyuk P, Morris A, Irani K, Natarajan V. Involvement of phospholipases D1 and D2 in sphingosine 1-phosphate-induced ERK (extracellular-signal-regulated kinase) activation and interleukin-8 secretion in human bronchial epithelial cells. Biochem J 367: 751–760, 2002 [Europe PMC free article] [Abstract] [Google Scholar]
49. Wu J, Spiegel S, Sturgill TW. Sphingosine 1-phosphate rapidly activates the mitogen-activated protein kinase pathway by a G protein-dependent mechanism. J Biol Chem 270: 11484–11488, 1995 [Abstract] [Google Scholar]
50. Young KW, Nahorski SR. Intracellular sphingosine 1-phosphate production: a novel pathway for Ca2+ release. Semin Cell Dev Biol 12: 19–25, 2001 [Abstract] [Google Scholar]
51. Young KW, Nahorski SR. Sphingosine 1-phosphate: a Ca2+ release mediator in the balance. Cell Calcium 32: 335–341, 2002 [Abstract] [Google Scholar]
52. Zhao Y, Gorshkova IA, Berdyshev E, He D, Fu P, Ma W, Su Y, Usatyuk PV, Pendyala S, Oskouian B, Saba JD, Garcia JG, Natarajan V. Protection of LPS-induced murine acute lung injury by sphingosine-1-phosphate lyase suppression. Am J Respir Cell Mol Biol. In Press. [Europe PMC free article] [Abstract] [Google Scholar]
53. Zhao Y, Kalari SK, Usatyuk PV, Gorshkova I, He D, Watkins T, Brindley DN, Sun C, Bittman R, Garcia JG, Berdyshev EV, Natarajan V. Intracellular generation of sphingosine 1-phosphate in human lung endothelial cells: role of lipid phosphate phosphatase-1 and sphingosine kinase 1. J Biol Chem 282: 14165–14177, 2007 [Europe PMC free article] [Abstract] [Google Scholar]

Articles from American Journal of Physiology - Lung Cellular and Molecular Physiology are provided here courtesy of American Physiological Society

Citations & impact 


Impact metrics

Jump to Citations

Citations of article over time

Smart citations by scite.ai
Smart citations by scite.ai include citation statements extracted from the full text of the citing article. The number of the statements may be higher than the number of citations provided by EuropePMC if one paper cites another multiple times or lower if scite has not yet processed some of the citing articles.
Explore citation contexts and check if this article has been supported or disputed.
https://scite.ai/reports/10.1152/ajplung.00404.2010

Supporting
Mentioning
Contrasting
3
24
0

Article citations


Go to all (27) article citations

Data 


Data behind the article

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

Similar Articles 


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


Funding 


Funders who supported this work.

NHLBI NIH HHS (4)