Journal of Histochemistry and Cytochemistry, Vol. 50, 661-670, May 2002, Copyright © 2002, The Histochemical Society, Inc.


ARTICLE

Cell Type-specific Localization of Human Cardiac S1P Receptors

David Mazuraisa, Philippe Roberta, Bernard Gouta, Isabelle Berrebi–Bertranda, Marie Paule Lavillea, and Thierry Calmelsa
a Laboratoire GlaxoSmithKline, Unité de Biologie Cardiovasculaire, Saint-Grégoire, France

Correspondence to: Thierry Calmels, Bioprojet-biotech, 4 rue du Chesnay Beauregard BP 96205, 35762 Saint-Grégoire cedex, France. E-mail: t.calmels@bioprojet.com.


  Summary
Top
Summary
Introduction
Materials and Methods
Results
Discussion
Literature Cited

Sphingosine 1-phosphate (S1P), which derives from the metabolism of sphingomyelin, is mainly synthesized, stored, and released from platelets after activation by physiological and pathophysiological events. S1P acts in cardiovascular tissues through cell surface G-protein-coupled receptors of the endothelial differentiation gene (EDG) family, i.e., EDG1, EDG3 and EDG5. The aim of the present study was to assess the precise distribution of EDG1, EDG3, and EDG5 receptors expressed in human cardiovascular tissues to investigate their respective physiological implication. When assessed by Northern blots, EDG1, EDG3, and EDG5 displayed wide expression levels in decreasing order, respectively. In particular, EDG3 was mainly detected in the aorta. Detailed analysis by in situ hybridization (ISH) and immunohistochemistry (IHC) revealed strong EDG1 expression in cardiomyocytes and in endothelial cells of cardiac vessels. In cardiomyocytes, the EDG1 receptor is likely to be co-expressed with EDG3 and EDG5, although EDG1 exhibits the most prominent expression pattern. Unlike EDG3 and EDG5, which are expressed in the smooth muscle cell layer of the human aorta, no signal corresponding to EDG1 expression could be detected in the aorta. Moreover, only EDG3 expression was also found in smooth muscle cells of cardiac vessels. The present results provide new insight into the expression pattern of S1P receptors in human cardiovascular tissues, indicating a differential pattern of expression for these receptors in human vessels. (J Histochem Cytochem 50:661–669, 2002)

Key Words: EDG receptor, S1P, blood vessels, cardiomyocytes, localization, heart


  Introduction
Top
Summary
Introduction
Materials and Methods
Results
Discussion
Literature Cited

SPHINGOSINE 1-PHOSPHATE (S1P) is a bioactive lysophospholipid that can stimulate a large range of responses in various cell types intracellularly as second messenger and through its interaction with members of the G-protein-coupled receptor (GPCR) family known by the acronym EDG (endothelial differentiation gene). S1P-specific receptors include EDG1, EDG3, EDG5, EDG6, and EDG8, whereas EDG2, EDG4, and EDG7 receptors are specific for lysophosphatidic acid (LPA) (Hla and Maciag 1990 ; Zondag et al. 1998 ; Spiegel and Milstien 2000 ). Activation by S1P of EDG receptors affects multiple biological processes that can be generally divided into either growth-related or cytoskeletal functions (Postma et al. 1996 ; Kawa et al. 1997 ; Sato et al. 1997 ; Pyne and Pyne 2000 ). These properties led to the view that EDG receptor-associated signaling pathways may have important roles in many developmental physiological and pathological events. All of these aminophospholipid ligands activate GPCRs and related receptor families that are capable of coupling differentially to one or multiple G-proteins and intracellular effectors. Recent biochemical evidence supports this notion, EDG1 interacts with G{alpha}i, whereas EDG3 and EDG5 interact with G{alpha}q and G{alpha}13, as well as G{alpha}i, in a ligand-dependent manner (Pyne and Pyne 2000 ).

Tissue expression of the EDG1, EDG3, and EDG5 receptors in mice indicates that heart and lung have the highest overall expression of these genes (Zhang et al. 1999 ; Ishii et al. 2001 ), whereas EDG6 and EDG8 receptors are mainly expressed in lymphoid (Graler et al. 1998 ) and brain tissues (Im et al. 2000 ), respectively. The intense expression of EDG1, EDG3, and EDG5 receptors in the mouse heart, the essential role played by EDG1 receptor in vessel formation (Liu et al. 2000 ), the release of S1P from activated platelets (Yatomi et al. 1995 ), and its accumulation in certain areas of the myocardium raise the possibility that sphingolipids, and more particularly, S1P may be, through activation of seven specific transmembrane receptors, a key contributor to physiological cardiac regulation. It has recently been proposed that EDG-mediated S1P negative inotropic and cardiotoxic effects may play important roles in acute myocardial ischemia. S1P was shown to increase the influx of extracellular calcium, which then causes calcium overload and impairs cardiac function (Nakajima et al. 2000 ). Interestingly, it was also recently shown in rat that S1P induces cardiomyocyte hypertrophy, mainly via the EDG1 receptor (Robert et al. 2001 ).

Although it is believed that EDG receptor family members could mediate many S1P effects in heart cell function, the cellular mechanisms underlying these effects are still largely unknown. Cardiac tissues are composed of diverse cell types, notably including cardiomyocytes, endothelial cells, fibroblasts, and smooth muscle cells of blood vessels. EDG1, the first S1P receptor cloned from endothelial cells, was suggested to be involved in the differentiation of endothelial cells (Hla and Maciag 1990 ) and was shown to be expressed in rat cardiomyocytes (Nakajima et al. 2000 ; Robert et al. 2001 ). EDG3 and EDG5 mRNA have been detected in rat smooth muscle cell lines (Tamama et al. 2001 ). EDG1, EDG3, and EDG5 messengers were detected in human heart and atrial cardiomyocytes by Northern blotting (An et al. 1998 ) or RT-PCR analysis (Himmel et al. 2000 ), respectively. However, these expression studies used cell cultures or whole-heart homogenates and were not focused on the in situ cell type-specific localization of the different S1P receptors, especially in human cardiac tissues.

The aim of the present work was to assess the precise distribution of EDG1, EDG3, and EDG5 receptors in human cardiovascular tissues to investigate their possible physiological implication. We first compared EDG1, EDG3, and EDG5 mRNA expression patterns in different regions of the heart and in aorta by Northern blotting as well as EDG1, EDG3, and EDG5 protein detection in whole heart, aorta, and smooth muscle by Western blotting analysis. We next determined the precise localization of EDG1, EDG3, and EDG5 expression in heart, aorta, and cardiac vessels by ISH or IHC.


  Materials and Methods
Top
Summary
Introduction
Materials and Methods
Results
Discussion
Literature Cited

Northern Blotting
Human cardiovascular MTN blots (Clontech; Palo Alto, CA) containing blotted RNA from normal human cardiovascular tissues were used: 21 specimens (age 21–30 weeks; cause of death spontaneous abortion) for male fetal heart; four specimens (age 19–54 years; cause of death trauma) for male adult heart; 50 specimens (age 18–75 years; cause of death trauma) for male adult aorta; 15 specimens (age 18–88 years; cause of death trauma) for male adult apex of heart; 39 specimens (age 18–72 years; cause of death trauma) for male adult left atrium; 38 specimens (age 18–72 years; cause of death trauma) for male adult right atrium; 11 specimens (age 18–61 years; cause of death trauma) for male adult left ventricle; 20 specimens (age 18–70 years; cause of death trauma) for male adult right ventricle. The blots were pre-hybridized for 30 min at 65C in 5 ml of ExpressHyb buffer (Clontech), then hybridized for 2 hr at 65C in the same buffer containing denatured {alpha}[32P]-dCTP-labeled probe (EDG1, EDG3, or EDG5) at 2 x 106 cpm/ml. The filters were then washed for 40 min at room temperature (RT) in 50 ml of 2 x SSC, 0.1% SDS, and for 40 min at 50C in 50 ml of 0.2 x SSC, 0.1% SDS. Each blot was stripped and then re-hybridized with glyceraldehyde-3-phosphate dehydrogenase (G3PDH) probe for normalization. The relative quantification of EDG1, EDG3, and EDG5 expression was performed by densitometric analysis of the film using 1D Image Analysis Software (Kodak Digital Science; Rochester, New York). Results were normalized using the ratio of EDG probe levels to G3PDH probe levels.

EDG1-specific Peptide Synthesis and Polyclonal Antibody Production
The protein sequence of the human EDG1 receptor was analyzed for highly antigenic regions using the Jameson–Wolf antigenic index. A synthetic peptide was selected at the C-terminal intracellular tail. A homology search was performed to check for sequence redundancy with other proteins. As expected, the peptide sequences had very high homology with EDG1 receptors from different mammalian species. Homologies with other mammalian proteins were not significant, particularly with other EDG receptor family members. The following synthetic peptide (amino acids 350–367 FSRSKSDNSSHPQKDEGDN) was synthesized, purified and conjugated as described in detail elsewhere (Robert et al. 1994 ). After immunization of rabbits according to a standard protocol, the immunoglobulin fraction from the anti-EDG1 immune serum was obtained by affinity chromatography on protein A–Sepharose.

Western Blotting
For Western blotting, 50 µg of total protein (Protein Medley; Clontech) was used from normal human heart, aorta, and smooth muscle from small intestine. Proteins were extracted by the supplier from whole aortas pooled from 49 males/females (ages 15–65; cause of death trauma or sudden death), small intestine smooth muscle pooled from 16 males/females (ages 16–50; cause of death trauma or sudden death) and whole heart from a 44-year-old white man (cause of death trauma). Western blots were subjected to SDS-PAGE after polyvinylidene difluoride (PVDF) membrane blotting (Biotrace PVDF; Pall–Gelman, Ann Arbor, MI). EDG1 receptor protein was detected using a purified polyclonal antibody (Ab729; 0.5 µg/ml) and an immunogenic synthetic peptide (10 µg/ml) as competitor. EDG3 and EDG5 receptor proteins were analyzed using commercial monoclonal antibodies (MAbs) with high interspecies homology (0.1 µg/ml) (Antibody Solutions; Palo Alto, CA). The molecular weights were estimated by using a prestained protein standard mixture (Biolabs; Beverly, MA). For EDG1 detection, the membrane was developed for only 15 sec using an ECL(+) kit (Amersham–Pharmacia Biotech; Poole, UK), whereas 45 min was necessary for EDG3 and EDG5.

In Situ Hybridization
Probe Synthesis. RNA probe synthesis was carried out with an RNA transcription Kit (Stratagene; La Jolla, CA). Plasmids (pBluescript; Stratagene) containing EDG1, EDG3, and EDG5 cDNA were linearized to give rise to the antisense and sense probes. One µg of the linearized DNA template was incubated for 2 hr at 37C in a solution containing 1 x transcription buffer, dithiothreitol (30 mM), rATP (0.4 mM), rGTP (0.4 mM), rCTP (0.4 mM), {alpha}[35S]-UTP (5 µCi/µl), RNase inhibitor (1.6 U/µl), and RNA polymerase (0.4 U/µl) T7 (sense) and T3 (antisense). The DNA templates were then digested with RQ-1 DNase (10 U) for 15 min at 37C. After incubation, 10 µg yeast tRNA was added to the sample. Probe isolation was achieved on a Sephadex G50 column. After precipitation the probe was dissolved in hybridization mix (50% formamide, 0.3 M NaCl, 20 mM Tris-HCl, pH 8.5, 5 mM EDTA, 10% dextran sulfate, 1 x Denhardt's solution, 0.5 µg/µl yeast tRNA, and 10 mM DTT) at a final concentration of 2.104 cpm/µl and stored at -70C until hybridization.

Slide Treatment. Paraffin sections were obtained from Novagen (Hybrid Ready Tissues; Madison, WI). Human adult normal tissue sections originated from male donor aorta and heart tissues, ages 26–36 (cause of death trauma). Tissues were removed, fixed in 4% paraformaldehyde, embedded in paraffin, sectioned at 5 µm (manufacturer's protocol), and stored at RT. Radioactive ISH was performed as previously described (Mazurais et al. 1999 ). Comparisons between EDG1, EDG3, and EDG5 were performed on tissues from the same donor. After treatment with xylene (three times for 5 min) to remove paraffin and rehydration through an ethanol series, sections were rinsed in 0.85% NaCl and phosphate buffer (PBS 0.1 M, pH 7.4), postfixed in 4% paraformaldehyde, and then treated with hydrochloric acid (0.2 M) and proteinase K (40 µg/ml) diluted in TE buffer (50 mM Tris-HCl, 5 mM EDTA, pH 8). After a rinse in PBS, the sections were refixed in 4% paraformaldehyde to stop proteinase K activity and then rinsed in PBS before acetylation with acetic anhydride (0.25% in triethanolamine 0.1 M, pH 8). Finally, sections were rinsed in distilled water before dehydration through an ethanol series. After air-drying, tissue sections were hybridized under coverslips with radioactive probe diluted in hybridization buffer (2.104 cpm/µl) overnight at 55C in a humid chamber. After hybridization, coverslips were removed in 5 x standard saline citrate (1 x SSC = trisodium citrate 15 mM, NaCl 150 mM, pH 7), 10 mM DTT at 55C for 15 min and the slides were washed (30 min) with 2 x SSC, 50% formamide, 10 mM DTT at 65C. After a rinse (10 min) in NTE buffer (10 mM Tris-HCl, 0.5 mM NaCl, 5 mM EDTA, pH 8), sections were treated with RNase A (20 µg/ml in NTE) for 30 min at 37C. Slides were than washed in NTE for 15 min at RT and incubated for 30 min in 2 x SSC, 50% formamide, 10 mM DTT at 65C. Before autoradiography, the tissues were rinsed in 2 x SSC and 0.1 x SSC at RT and dehydrated in an ethanol series containing 0.3 M ammonium acetate. Autoradiography was performed by dipping the slides in Ilford K5 nuclear track emulsion and exposing the slides in the dark for 14 days at 4C. After development, the sections were counterstained with toluidine blue and mounted in Pertex (Microm; Francheville, France). The photographs presented in Fig 3 Fig 4 Fig 5 Fig 6 Fig 7 are representative of three independent experiments.



View larger version (43K):
[in this window]
[in a new window]
 
Figure 1. (A) Northern blotting analysis of EDG1 (top), EDG3 (middle), and EDG5 (bottom) mRNA in human cardiovascular tissues. The multiple-tissue Northern blots were obtained from Clontech, and contain 2 µg of poly (A)+ RNA from indicated adult human tissues in each lane. The membrane filters were hybridized with full-length human cDNA for EDG1, EDG3, and EDG5, washed, and then exposed for 5 hr. A single band corresponding to each mRNA can be detected in each tissue, 3 kb, 4.6 kb, and 4.2 kb for EDG1, EDG3 and EDG5, respectively. Normalization with corresponding G3PDH expression was performed in each tissue. (B) The relative quantification of EDG1, EDG3 and EDG5 expression was performed by densitometric analysis of the film using 1D Image Analysis Software (Kodak Digital Science).

Figure 2. Detection of EDG1, EDG3, and EDG5 proteins in human whole heart, aorta and smooth muscle by Western blotting. Analysis of EDG1, EDG3, and EDG5 was performed using 50 µg of total proteins (Protein Medley; Clontech) of human heart, aorta, and smooth muscle. For EDG1 detection, the membrane was developed for only 15 sec using an ECL(+) kit (Amersham–Pharmacia Biotech), whereas 45 min was necessary for EDG3 and EDG5.



View larger version (47K):
[in this window]
[in a new window]
 
Figure 3. EDG1 expression in cardiomyocytes of the left ventricle. (A–D) Light microscopy of ISH with the EDG1 probe. Brightfield (A,C) and darkfield (B,D) illumination showing the specific signal with the antisense probe (A,B). Note the high density of EDG1 messengers around the cardiomyocyte nuclei (arrows). Control sections incubated with the sense probe do not reveal significant signal (C,D). (E,F) Sections of the left ventricle illustrating EDG1-immunoreactive cardiomyocytes. (E) Detection of EDG1 protein in cardiomyocytes of human left ventricle. (F) Results obtained in human left ventricle section using anti-EDG1 antibody preincubated with EDG1-immunoreactive peptide. Bars = 20 µm.

Figure 4. EDG3 and EDG5 expression is detected in cardiomyocytes of the left ventricle (arrows). Light microscopy of ISH with EDG3 (A,B) or EDG5 probe (E,F). The signals are specific because control sections incubated with the respective sense probes do not reveal significant signal (C,D,G,H). Bars = 10 µm.



View larger version (95K):
[in this window]
[in a new window]
 
Figure 5. EDG1 expression in coronary arteries of the left ventricle. (A–D) Light microscopy of ISH with the EDG1 probe. Brightfield (A,C) and dark field (B,D) photomicrographs of a left ventricle section illustrating the distribution of EDG1 messengers in cardiomyocytes (cm) and endothelial cell layer (ec) of vessel walls. The specificity of the labeling obtained with the antisense probe (A,B) is validated with control sections incubated with the sense probe (C,D). This pattern of EDG1 expression is confirmed by immunohistological studies (E,F). (E) Detection of EDG1 protein in cardiomyocytes (cm) and endothelial cells of arterioles (ec). Anti-EDG1 antibodies preincubated with EDG1 peptides in control section (F) reveal no appreciable staining. Bars = 20 µm.

Figure 6. Differential patterns of S1P receptor expression in smooth muscle cells of human aorta. Light microscopy results of ISH with EDG1, EDG3, and EDG5 probes on adjacent aortic tissue sections are represented in A,B, C,D, and E,F, respectively. Brightfield (A) and darkfield (B) photomicrographs indicate no expression of EDG1 in the smooth muscle (sm) cells of aorta. Brightfield (C) and darkfield (D) photomicrographs illustrate the expression of EDG3 (black and white arrowheads) in the smooth muscle cells of the aorta. Brightfield (E) and darkfield (F) photomicrographs reveal specific expression of EDG5 in the smooth muscle cells of the aorta (black and white arrowheads). EDG3 and EDG5 signals are specific because control sections incubated with the respective sense probes do not reveal significant signal (data not shown). Bars = 20 µm.

Figure 7. Differential patterns of S1P receptor expression in coronary vessel walls. Light microscopy results of ISH with EDG1, EDG3, and EDG5 probes on adjacent sections of ventricle are represented in A–C, D–F, and G–I, respectively. (A–C) Brightfield (A,C) and darkfield (B) photomicrographs illustrate strong expression of EDG1 in the endothelial cell layer (ec) of the coronary (white arrow). No significant signal can be detected in the smooth muscle cell layer. (D–F) Brightfield (D,F) and darkfield (E) photomicrographs illustrate expression of EDG3 in the smooth muscle cells (sm) of the coronary (black arrow). (G–I) Brightfield (G) and darkfield (H) photomicrographs do not reveal significant specific expression of EDG5 in the vessel walls in comparison with sense control (I). C and F show higher magnifications of A and D, respectively. Bars = 20 µm.

Immunohistochemistry
Human paraffin sections were obtained from Novagen (Hybrid-Ready Tissues). Normal human adult tissue sections originated from donor heart tissues, ages 20–65 (cause of death trauma). Tissues were removed, fixed in 4% paraformaldehyde, embedded in paraffin, sectioned at 5 µm (manufacturer's protocol), and stored at RT. After treatment with xylene and rehydratation through an ethanol series and PBS, the specimens were heated in target retrieval solution, pH 6 (Dako; Glastrup, Denmark), 40 min at 95C and then cooled for 20 min on the bench. After two washes of 2 min in TBS solution (Dako), sections were incubated for 1 hr at RT with anti-EDG1 primary antibody at 2 µg/µl. Use of commercially available EDG3 and EDG5 antibodies (Antibody Solutions) did not obtain suitable results. For control slides, competition was performed by overnight incubation of the primary antibody with a related peptide used to generate the anti-EDG1 rabbit polyclonal antibody. After washes in PBS, the goat anti-rabbit secondary antibody and the tyramide signal amplification peroxidase IHC detection kit (Dako) were used according to the manufacturer's instructions with diaminobenzidine as a substrate. After hematoxylin counterstaining, sections were analyzed on a Nikon (Eclipse E800, Sony camera DXC-950P) microscope. The photographs in Fig 3 and Fig 5 are representative of three independent experiments.


  Results
Top
Summary
Introduction
Materials and Methods
Results
Discussion
Literature Cited

EDG1, EDG3, and EDG5 Receptor Expression in Heart and Aorta
To determine the comparative mRNA expression patterns of EDG1, EDG3, and EDG5 receptor genes in different regions of human heart, Northern blots were probed at high stringency with EDG1, EDG3, and EDG5 cDNA probes corresponding to their respective coding regions (Fig 1). The 3.0-kb EDG1 mRNA was strongly and equally expressed in the different compartments (right and left ventricle, right and left atrium, apex) of the adult human heart, in the whole fetal heart, and in aorta. In contrast, the ~4.6-kb EDG3 mRNA was more abundant in the aorta than in heart regions (Fig 1B). Finally, EDG5 receptor transcript is weakly and equally expressed in the different regions of the heart and in aorta.

Western blotting experiments were performed to detect EDG1, EDG3, and EDG5 proteins in adult human heart, aorta, and smooth muscle of small intestine (Fig 2). It is worth noting that a long film exposure of 45 min was necessary to visualize EDG3 and EDG5 signals from total proteins loaded. Conversely, only 15 sec was sufficient to detect the EDG1 receptor. Considering that each antibody exhibits proper affinity, it is difficult to compare relative abundance at the protein level of each receptor in different tissues. The 38-kD EDG1 protein was detected at a high level in aorta and heart as well as in smooth muscle of small intestine. Similarly to results obtained by Northern blotting, the EDG3 45-kD protein was abundant in the aorta and smooth muscle compared to adult human heart. EDG5 receptor protein was equally present in whole heart, aorta, and smooth muscle.

Localization of EDG1, EDG3, and EDG5 Transcripts and Proteins in Human Heart
Full-length riboprobes of human EDG1, EDG3, and EDG5 receptors were used for ISH on normal human tissue sections. In parallel, IHC using an anti-EDG1 antibody was performed. Commercially available anti-EDG3 and anti-EDG5 antibodies (Antibody Solutions) did not allow successful IHC studies.

In the left ventricle, EDG1 mRNA was expressed in cardiomyocytes (Fig 3A and Fig 3B) which also homogeneously displayed EDG1-immunoreactive signal (Fig 3E). It is interesting to note that the strong density of labeling (Fig 3B) was detected around cardiomyocyte nuclei because of the larger amounts of mRNA in this area. The expression pattern was remarkably consistent from one experiment to the other, and the specificity of the signal was confirmed by the absence of signal on the control slides, either with sense probe (Fig 3C and Fig 3D) or polyclonal antibody preincubated with immunogenic EDG1 peptide as competitor (Fig 3F). ISH experiments using EDG3 (Fig 4A and Fig 4B) and EDG5 (Fig 4E and Fig 4F) probes indicated lower expression of the respective messengers in cardiomyocytes, compared to EDG1. Absence of any significant signal on the control slides demonstrated the specificity of this labeling (Fig 4C, Fig 4D, Fig 4G, and Fig 4H). Because cardiomyocytes are densely packed, it was difficult to ascertain whether all cells were labeled, but a large majority displayed silver grains with the three different probes. As a consequence, EDG1, EDG3, and EDG5 are most likely co-expressed in a large number of cardiomyocytes. Both in atrium and septum, expression of EDG1, EDG3, and EDG5 could also be detected in cardiomyocytes (data not shown). Although it is difficult to estimate a precise distribution between different areas of the myocardium, such as inner, middle, outer myocardium, interstitium, or endocardium, by ISH or immunocytochemistry, no differences in gene expression or protein detection were identified for each receptor independently. In addition, localization of EDG1, EDG3, and EDG5 expression by ISH and IHC were revealed at the base of a growing tricuspid valve. These experiments revealed the expression of EDG1, EDG3, and EDG5 mRNA in the myocardium but absence of labeling in the fibrosa (data not shown).

Strong radioactive labeling corresponding to EDG1 mRNA localization was detected in the endothelial cells of coronary arteries in the left ventricle (Fig 5A and Fig 5B). The nature of the endothelial cell layer was confirmed by IHC analysis using an antibody raised against factor VIII as an endothelial cell lineage marker (data not shown). IHC study confirmed the localization of EDG1 protein limited to the endothelial cell layer of vessels (Fig 5E). No specific signal could be detected in the media and adventitia of vessels. The signal specificity was confirmed by the absence of signal on the control slides, either with the sense probe (Fig 5C and Fig 5D) or with the anti-EDG1 antibody preincubated with immunogenic EDG1 peptide as competitor (Fig 5F). Endothelial cells of atrial and septal vessels were shown by ICC and ISH to express the EDG1 receptor (data not shown). Special attention was focused on the differential expression of EDG1, EDG3, and EDG5 mRNA in aorta and in cardiac vessels. In agreement with the Northern blotting experiment, EDG3 and EDG5 were expressed in the smooth muscle cells of the aortic media layer (Fig 6A–6F), whereas expression of EDG1 was not detected in smooth muscle cells of human aorta. In addition, microscopic analysis of EDG1, EDG3, and EDG5 expression was performed on serial tissue sections at the level of a small left ventricular coronary (Fig 7). This study showed that, unlike EDG1, which is expressed in the endothelial cells (Fig 7A–7C), EDG3 mRNA was largely distributed in the smooth muscle cells of the coronary vessel (Fig 7D–7F). No specific EDG3 signal could be detected in the endothelial cells. The specificity of the signal was confirmed by the absence of labeling on the control slides using the sense probe (data not shown). Regarding EDG5, in contrast to the smooth muscle cell layer of human aorta, no significantly positive cells could be detected in the different layers of the coronary vessel walls (Fig 7G–7I).


  Discussion
Top
Summary
Introduction
Materials and Methods
Results
Discussion
Literature Cited

This study documents the precise distribution and compares the relative expression patterns of EDG1, EDG3, and EDG5 genes in normal human cardiovascular tissues. Our data provide, for the first time, detailed information on the nature and the localization of the cardiac cells that express the S1P receptor at both the mRNA and the protein level.

First, Northern blots from human subjects reveal that EDG1, EDG3, and EDG5 receptors are expressed in both adult and fetal heart. More precisely, they are expressed in all anatomically distinct subregions of the adult heart, i.e., the apex, left and right ventricles, left and right atria, and the aorta. For each EDG independently, Northern blotting analysis reveals additional information on the relative amount of each receptor transcript in those cardiac regions. Whereas EDG1 mRNA is equally and strongly expressed in different parts of the heart and in aorta, the EDG3 transcript is more abundant in aorta than in the different compartments of the heart. Finally, EDG5 transcript is ubiquitously but weakly expressed in the heart and aorta in comparison with EDG3 and EDG1, except in fetal samples. These results are confirmed at the protein level by Western blotting experiments. However, it is difficult to compare the respective protein amounts because each antibody has a specific affinity and different times of exposure for detection were necessary. Taken together, Northern and Western blotting data provide new insight into previous reports indicating expression of EDG1, EDG3, and EDG5 transcripts in human (An et al. 1998 ) and mouse (Zhang et al. 1999 ) heart.

With the aim of investigating the respective roles played by EDG1, EDG3, and EDG5 receptors in cardiovascular tissues, it was important to determine the nature of the cells that express the respective messengers or proteins. For this purpose, ISH and IHC studies were performed at the cellular level. Our data indicate that EDG1 mRNA and protein are strongly expressed in ventricular, septal, and atrial cardiomyocytes as well as in the endothelial cell layer of cardiac vessels, whereas EDG3 mRNA is highly expressed in the smooth muscle cell layer of aorta and cardiac vessels and weakly expressed in cardiomyocytes from both atria and ventricles. EDG5 transcript is detected in cardiomyocytes and in smooth muscle cells of the aortic medial layer and is absent in cardiac coronary microvessels. Regarding the abundance of respective messengers in cardiomyocytes, the intensity of ISH signals obtained with each EDG-specific probe confirms the expression pattern obtained by Northern blotting described above. It was important to clarify EDG1, EDG3, and EDG5 localization in human cardiomyocytes because interspecies discrepancies between mouse (Liu and Hla 1997 ; Zhang et al. 1999 ), rat (Lado et al. 1994 ), and human (An et al. 1998 ) were previously observed for some S1P/EDG receptors in several tissues. Perinuclear labeling obtained with the EDG1 probe is much more abundant than that obtained with EDG3 and EDG5 probes. This result complements previous data obtained by RT-PCR in human atrial myocytes (Himmel et al. 2000 ) and by Western blotting in rat neonatal cardiac myocytes (Robert et al. 2001 ). Strong expression of EDG1 mRNA in human cardiomyocytes is also in good agreement with the presence of EDG1 protein in adult ventricular myocytes of rat papillary muscle (Nakajima et al. 2000 ). The high expression of EDG1 gene in adult and neonatal cardiomyocytes suggests its potential involvement in S1P-related cardiac functions at all stages of heart development. To support this hypothesis, developmental studies in zebrafish have recently indicated that S1P signaling via an EDG-like receptor, i.e., Mil, is essential for heart development (Kupperman et al. 2000 ). On the other hand, recent works have demonstrated that S1P induces cardiomyocyte hypertrophy (Robert et al. 2001 ) and calcium deregulation (Nakajima et al. 2000 ), mainly via the EDG1 receptor in rat. This result raises the hypothesis that EDG1 might be involved in cardiac pathophysiology. However, as shown by ISH, the lower expression of EDG3 and EDG5 receptors in cardiomyocytes does not rule out the possibility that these Gq-coupled receptors may also mediate S1P function, notably the cardiac calcium influx regulation as previously suggested (Nakajima et al. 2000 ). EDG3 expression in human cardiomyocytes is also in line with a recent study suggesting that S1P could activate the acetylcholine-stimulated inward rectifier K+ current via EDG3 receptors in human atrial myocytes and then participate in the modulation of heart rhythm (Himmel et al. 2000 ). Co-localization of EDG1, EDG3, and EDG5 in cardiomyocytes may result in distinct responses to S1P stimulation, because these receptors are capable of coupling differentially to one or multiple G-proteins and intracellular effectors. Whereas EDG1 was demonstrated to be coupled to Gi/o-protein and can utilize the Rho-dependent signaling pathway, EDG3 and EDG5 signal not only via Gi but also through G{alpha}q and G{alpha}12/{alpha}13 (Pyne and Pyne 2000 ).

The relatively high expression of EDG3 in different compartments of the heart, as revealed by Northern blotting and the faint signal obtained by ISH in cardiomyocytes, suggested that the EDG3 receptor might be expressed in other cardiac cell types. In agreement with this hypothesis, EDG3 protein was shown to be strongly detected in whole rat heart homogenates but not in purified ventricular cardiomyocytes (Robert et al. 2001 ). Our ISH experiments demonstrated intense expression of EDG3 receptor gene in the medial layer of human aorta and cardiac vessels composed of smooth muscle cells. These data are in good agreement with results obtained in rat (Tamama et al. 2001 ) showing the expression of EDG3 receptor mRNA in isolated primary aortic smooth muscle cells. EDG3 expression in vessels is confirmed by our Western blotting analysis indicating a higher level of protein in aorta than in heart, probably due to the higher relative percentage of smooth muscle cells in the aortic tissue samples compared to heart samples. We were unable to detect significant expression of EDG5 in human cardiac microvessels, although the EDG5 transcript was strongly detected in aortic smooth muscle cells. This is in good agreement with the work of Tamama and co-workers 2001 , who have demonstrated that rat aortic smooth muscle cells express EDG5 mRNA. Such discrete EDG5 expression in smooth muscle cells originating from different cardiovascular vessels might be explained by a differential and vessel size-specific gene expression. Moreover, unlike what has been previously shown in cultures of smooth muscle cells either in rodent or human (Hla et al. 1999 ; Rakhit et al. 1999 ; Tamama et al. 2001 ), our ISH amd IHC studies did not enable us to detect EDG1 mRNA or EDG1-immunoreactive cells in the smooth muscle cell layer of adult human aorta and cardiac vessels. This result suggests that expression of EDG1 in the smooth muscle cells of adult vessels, if any, is likely to be very weak. The use of very sensitive experiments, such as RT-PCR, to detect EDG1 expression in most of the previous studies (Hla and Maciag 1990 ; Rakhit et al. 1999 ) appears to confirm this hypothesis, which questions the physiological role played by this weak EDG1 expression.

The distinguishable spatial expression patterns of EDG1, EDG3, and EDG5 receptors suggest distinct biological roles played by each receptor in the different vessel layers. EDG3 and EDG5 expression in smooth muscle cells layers suggests that this receptor might be important for the S1P-induced mobilization of Ca2+ in human smooth muscle (Bornfeldt et al. 1995 ) and could then participate in regulation of the proliferation and migration of vascular smooth muscle cells, which are important events in the development of vascular disorders such as atherosclerosis and restenosis (Ross 1999 ). High expression of the EDG1 receptor in endothelial cells of cardiac vessels confirms its potential role in the regulation of endothelial function under physiological blood flow conditions (Takada et al. 1997 ). Detection of EDG1 expression in endothelial cells is also consistent with the fact that EDG1 might be involved in the phorbol ester-induced tube formation of human umbilical vein endothelial cells (HUVECs) (Hla and Maciag 1990 ; Lee et al. 1998 ). In fact, it has been demonstrated that inhibition of expression of EDG1 by specific antisense oligonucleotides abolishes the effects of S1P on capillary-like network formation (Lee et al. 1999 ). Moreover, the presence of functional EDG1 protein in endothelial cells is in good agreement with downstream events involving the recruitment of smooth muscle cells by S1P-stimulated endothelial cells (Liu et al. 2000 ). Working with EDG1 knockout mice, Liu et al. 2000 have shown in embryos that the EDG1 receptor is necessary for vascular formation via smooth muscle investment during vessel maturation. Indeed, aortic sections of EDG1-/- mouse embryos demonstrated the lethal absence of smooth muscle cells at the dorsal surface of the aorta. The absence of EDG1 expression in smooth muscle cells of adult human cardiac vessels, unlike embryonic mouse vessels, suggests a potentially differential expression of the EDG1 receptor during development. In agreement with this hypothesis, recent results obtained in rat demonstrated that pupintimal vascular smooth muscle cells express higher levels of EDG1 mRNA than adult medial vascular smooth muscle cells (Kluk and Hla 2001 ). To confirm this latter hypothesis, it would be necessary to look for potential expression of the EDG1 receptor in smooth muscle cells of human embryonic cardiac vessels.

In conclusion, this study provides new insights into the expression pattern of S1P/EDG receptors in adult human cardiovascular tissues. Although further studies will be necessary to better delineate the S1P mechanisms of action in human heart and vessels, the present data suggest that the receptors of the EDG family may represent good targets for a therapeutic strategy aimed at modulating the transduction machineries involved in human cardiovascular diseases.


  Acknowledgments

We thank Dr V. Paumier and Dr P. Noret for help in histological analysis, and Jean-Claude Camelin and Sabine Rouanet for their contribution to this project.

Received for publication July 20, 2001; accepted December 5, 2001.


  Literature Cited
Top
Summary
Introduction
Materials and Methods
Results
Discussion
Literature Cited

An S, Goetzl E, Lee H (1998) Signaling mechanisms and molecular characteristics of G protein-coupled receptors for lysophosphatidic acid and sphingosine 1-phosphate. J Cell Biochem 31:147-157

Bornfeldt KE, Graves LM, Rainse EW, Lgarashi Y, Wayman G, Yamamura S, Yatomi Y, Sidhu JS, Krebs EG, Hakomori S, Ross R (1995) Sphingosine-1-phosphate inhibits PDGF-induced chemotaxis of human arterial smooth muscle cells: spatial and temporal modulation of PDGF chemotactic signal transduction. J Cell Biol 130:193-206[Abstract]

Gräler MH, Brenhardt G, Lipp M (1998) EDG6, a novel G-protein-coupled receptor related to receptors for bioactive lysophospholipids, is specifically expressed in lymphoid tissue. Genomics 53:164-169[Medline]

Himmel HM, Meyer Zu Heringdorf DM, Graf E, Dobrev D, Kortner A, Schueler S, Jakobs KH, Ravens U (2000) Evidence for EDG3 receptor-mediated activation of IK.Ach by sphingosine-1-phosphate in human atrial cardiomyocytes. Mol Pharmacol 58:449-454[Abstract/Free Full Text]

Hla T, Maciag T (1990) An abundant transcript induced in differentiating human endothelial cells encodes a polypeptide with structural similarities to G-protein-coupled receptors. J Biol Chem 265:9308-9313[Abstract/Free Full Text]

Hla T, Lee MJ, Ancellin N, Liu CH, Thangada S, Thompson BD, Kluk M (1999) Sphingosine-1-phosphate: extracellular mediator or intracellular second messenger? Biochem Pharmacol 58:201-217[Medline]

Im DS, Heise CE, Ancellin N, O'Dowd BF, Shei GJ, Heavens RP, Rigby MR, Hla T, Mandala S, McAllister G, George SR, Lynch KR (2000) Characterization of a novel sphingosine 1-phosphate receptor, EDG8. J Biol Chem 275:14281-14286[Abstract/Free Full Text]

Ishii I, Friedman B, Ye X, Kawamura S, McGiffert C, Contos JJA, Kingsbury MA, Zhang G, Brown JH, Chun J (2001) Selective loss of S1P signaling with no obvious phenotypic abnormality in mice lacking its G protein-coupled receptor, LPB3/EDG3. J Biol Chem 276:33697-33704[Abstract/Free Full Text]

Kawa S, Kimura S, Hakomori S, Igarashi Y (1997) Inhibition of chemotactic motility and trans-endothelial migration of human neutrophils by sphingosine 1-phosphate. FEBS Lett 420:196-200[Medline]

Kluk MJ, Hla T (2001) Role of the sphingosine 1-phosphate receptor EDG-1 in vascular smooth muscle cell proliferation and migration. Circ Res 89:496-502[Abstract/Free Full Text]

Kupperman E, An S, Osborne N, Waldron S, Stainier DY (2000) A sphingosine 1-phosphate receptor regulates cell migration during vertebrate heart development. Nature 406:192-195[Medline]

Lado DC, Browe CS, Gaskin AA, Borden JM, McLennan AJ (1994) Cloning of the rat EDG1 immediate-early gene: expression pattern suggests diverse functions. Gene 149:331-336[Medline]

Lee MJ, Thangada S, Claffey KP, Ancellin N, Liu CH, Kluk M, Volpi M, Sha'afi RI, Hla T (1999) Vascular endothelial cell adherens junction assembly and morphogenesis induced by sphingosine-1-phosphate. Cell 99:301-312[Medline]

Lee MJ, Van BJ, Thangada S, Liu CH, Hand AR, Menzeleev R, Spiegel S, Hla T (1998) Sphingosine-1-phosphate as a ligand for the G protein-coupled receptor EDG1. Science 279:1552-1555[Abstract/Free Full Text]

Liu CH, Hla T (1997) The mouse gene for the inducible G-protein-coupled receptor EDG1. Genomics 43:15-24[Medline]

Liu Y, Wada R, Yamashita T, Mi Y, Deng CX, Hobson JP, Rosenfeldt HM, Nava VE, Chae SS, Lee MJ, Liu CH, Hla T, Spiegel S, Proia RL (2000) EDG1, the G protein-coupled receptor for sphingosine-1-phosphate, is essential for vascular maturation. J Clin Invest 106:951-961[Abstract/Free Full Text]

Mazurais D, Brierley I, Anglade I, Drew J, Randall C, Bromage N, Michel D, Kah O, Williams LM (1999) Central melatonin receptors in the rainbow trout: comparative distribution of ligand binding and gene expression. J Comp Neurol 409:313-324[Medline]

Nakajima N, Cavalli AL, Biral D, Gembotski CC, Donough PM MC, Ho PD, Betto R, Sandona D, Palade PT, Dettbarn CA, Klepper RE, Sabbadini RA (2000) Expression and characterization of EDG1 receptors in rat cardiomyocytes calcium deregulation in response to sphingosine 1-phosphate. Eur J Biochem 267:5679-5686[Abstract/Free Full Text]

Postma FR, Jalink K, Hengeveld T, Moolenaar WH (1996) Sphingosine-1-phosphate rapidly induces rho-dependent neurite retraction: action through a specific cell surface receptor. EMBO J 15:2388-2392[Abstract]

Pyne S, Pyne NJ (2000) Sphingosine 1-phosphate signalling in mammalian cells. Biochem J 349:385-402[Medline]

Rakhit S, Conway AM, Tate R, Bower T, Pyne NJ, Pyne S (1999) Sphingosine 1-phosphate stimulation of the p42/p44 mitogen-activated protein kinase pathway in airway smooth muscle. Role of endothelial differentiation gene 1,(c-Src tyrosine kinase and phosphoinositide 3-kinase. Biochem J 338):643-649

Robert P, Troalen F, Bellet D, Bousfield GR, Bidart JM (1994) Immunochemical mapping of human lutropin: I. Delineation of a conformational antigenic determinant. Mol Cell Endocrinol 101:11-20[Medline]

Robert P, Tsui P, Laville MP, Livi GP, Sarau HM, Bril A, Berrebi–Bertrand I (2001) EDG1 receptor stimulation leads to cardiac hypertrophy in rat neonatal myocytes. J Mol Cell Cardiol 33:1589-1606[Medline]

Ross R (1999) Atherosclerosis—an inflammatory disease. N Engl J Med 340:115-126[Free Full Text]

Sato K, Tomura H, Igarashi Y, Ui M, Okajima F (1997) Exogenous sphingosine-1-phosphate induces neurite retraction possibly through a cell surface receptor in PC12 cells. Biochem Biophys Res Commun 240:329-334[Medline]

Spiegel S, Milstien S (2000) Functions of a new family of sphingosine-1-phosphate receptors. Biochim Biophys Acta 1484:107-116[Medline]

Takada Y, Kato C, Kondo S, Korenaga R, Ando J (1997) Cloning of cDNAs encoding G protein-coupled receptor expressed in human endothelial cells exposed to fluid shear stress. Biochem Biophys Res Commun 240:737-741[Medline]

Tamama K, Kon J, Sato K, Tomura H, Kuwabara A, Kimura T, Kanda T, Ohta H, Ui M, Kobayashi I, Okajima F (2001) Extracellular mechanism through the EDG family of receptors might be responsible for sphingosine-1-phosphate-induced regulation of DNA synthesis and migration of rat aortic smooth-muscle cells. Biochem J 353:139-146[Medline]

Yatomi Y, Ruan FQ, Hakomori SI, Igarashi Y (1995) Sphingosine-1-phosphate: a platelet-activating sphingolipid released from agonist-stimulated human platelets. Blood 86:193-202[Abstract/Free Full Text]

Zhang G, Contos JJA, Weiner JA, Fukushima N, Chun J (1999) Comparative analysis of three murine G-protein coupled receptors activated by sphingosine-1-phosphate. Gene 227:89-99[Medline]

Zondag GC, Postma FR, Etten IV, Verlaan I, Moolenaar WH (1998) Sphingosine 1-phosphate signaling through the G-protein-coupled receptor EDG1. Biochem J 330:605-609[Medline]