Skip to main content
Neoplasia (New York, N.Y.) logoLink to Neoplasia (New York, N.Y.)
. 2007 Dec;9(12):1038–1045. doi: 10.1593/neo.07643

Differential Gene Expression of Primary Cultured Lymphatic and Blood Vascular Endothelial Cells1,2

Gregory M Nelson *, Timothy P Padera *, Igor Garkavtsev *, Toshi Shioda , Rakesh K Jain *
PMCID: PMC2137938  PMID: 18084611

Abstract

Blood vascular endothelial cells (BECs) and the developmentally related lymphatic endothelial cells (LECs) create complementary, yet distinct vascular networks. Each endothelial cell type interacts with flowing fluid and circulating cells, yet each vascular system has evolved specialized gene expression programs and thus both cell types display different phenotypes. BECs and LECs express distinct genes that are unique to their specific vascular microenvironment. Tumors also take advantage of the molecules that are expressed in these vascular systems to enhance their metastatic potential. We completed transcriptome analyses on primary cultured LECs and BECs, where each comparative set was isolated from the same individual. Differences were resolved in the expression of several major categories, such as cell adhesion molecules (CAMs), cytokines, and cytokine receptors. We have identified new molecules that are associated with BECs (e.g., claudin-9, CXCL11, neurexin-1, neurexin-2, and the neuronal growth factor regulator-1) and LECs (e.g., claudin-7, CD58, hyaluronan and proteoglycan link protein 1 (HAPLN1), and the poliovirus receptor-related 3 molecule) that may lead to novel therapeutic treatments for diseases of lymphatic or blood vessels, including metastasis of cancer to lymph nodes or distant organs.

Keywords: Lymphangiogenesis, microarray, LEC, BEC, endothelial

Introduction

The lymphatic system maintains tissue-fluid equilibrium while organizing the surveillance for foreign antigens by immune cells. Its smallest vessels are initial lymphatic vessels that collect interstitial fluid and immune cells from the surrounding tissue. These initial lymphatics consist of lymphatic endothelial cells (LECs) that sit on a discontinuous basement membrane and are tethered to the extracellular matrix (ECM) through anchoring filaments. The protein-rich lymph and extravasated immune cells enter the initial lymphatic vessels through intercellular junctions, are transported to a series of lymph nodes, and are then returned to the blood primarily through the thoracic duct [1]. Initial lymphatics are different from blood vessel capillaries, which are composed of blood vascular endothelial cells (BECs) encircled by pericytes and basement membrane. It is of great interest to distinguish the molecular differences between the similar but different LECs and BECs.

The lymphatic system also plays a role in pathologic states [2]. Lymphatic vessels act as a conduit for metastatic tumor cells to escape from the primary tumor. Cells can be imaged in lymphatic vessels in transit between the primary tumor and sentinel lymph node in animals [3] and sentinel lymph node biopsies positive for cancer cells serve as important determinants of cancer prognosis and for the subsequent course of therapy [4,5]. The molecular pathways involved with the development and growth of the lymphatic system are beginning to be understood. Molecules such as vascular endothelial growth factor (VEGF)-C [6,7], VEGF-D [8], VEGF-A [9,10], hepatocyte growth factor [11,12], and platelet-derived growth factor-BB [13] have been shown to be lymphangiogenic. VEGF-A and VEGF-C can also create hyperplastic tumor margin lymphatic vessels when overexpressed [9,14]. The enlarged vessels allow a greater number of metastatic cells to leave the primary tumor, thus promoting metastasis [3,15].

In addition to aiding metastasis, the lymphatic system plays a role in diseases involving lymphedema. Hereditary or primary lymphedema occurs as a result of problems with the formation of a functional lymphatic system. Some forms of primary lymphedema have been linked to gene mutations in VEGFR-3, i.e., the receptor for VEGF-C found on LECs [16]. Lymphedema can also occur as a result of an infection or injury to the lymphatic systems, preventing lymphatic vessels from collecting lymph and returning to the blood. These secondary forms of lymphedema are commonly caused by filarial infections or lymphatic vessel transection during surgery [17,18].

Research on lymphatic vessels and LECs has lagged behind that of BECs and blood vessels. With the identification of molecules that can distinguish LECs from BECs, the field has grown tremendously. Yong et al. [19] used a spotted oligonucleotide array to determine the gene expression changes that occurred when LECs were subjected to VEGF-C. In contrast to this study, previous comparisons of the transcriptional expression of LECs and BECs [20–22] did not have a full genome array available and did not make the full data sets accessible. One recent study compared cultured and uncultured LECs and BECs and found that differences in the handling of the cells can change the differential expression of the LECs and BECs [23].

Here we compare LECs and BECs with the largest possible gene array to identify differentially expressed genes that will strengthen our understanding of lymphatic and blood vascular biology and potentially uncover targets for therapeutic intervention in lymphedema and lymphatic metastasis. We used the Affymetrix GeneChip Human Genome U133 Plus 2.0 Array. Our analysis yields targets that may enhance our understanding of lymphatic and blood vascular biology.

Materials and Methods

Cell Maintenance and Separation

Three independent lots of CD31+ human dermal microvascular endothelial cells (HDMECs; PromoCell, Heidelberg, Germany) isolated from different male individuals were maintained on fibronectin-coated tissue culture plates in endothelial cell growth medium MV low serum (PromoCell) according to the manufacturer's instructions. The isolation of cells from separate individuals allowed us to compare LECs and BECs from the same person, reducing the intrasample variability. The cells were immunomagnetically separated with mouse anti-human podoplanin IgG (a generous gift of Dr. Dontscho Kerjaschki [24]). Cells were washed with PBS/2% BSA solution and incubated in a magnetic particle concentrator (MPC-1; Invitrogen, Carlsbad, CA). Bead-bound cells (LECs) were washed five times to remove all unbound cells. Unbound cells in the wash buffer were collected and centrifuged to pellets (900g for 3 minutes at room temperature). Unbound cells (BECs) were resuspended in wash buffer and placed on the magnetic particle concentrator to remove bead-bound cells; the process was repeated five times. Pure populations of LECs (CD31+/podoplanin+) and BECs (CD31+/podoplanin-) were cultured in HDMEC media and grown for two to four passages. Cells were used in passage four to nine for microarray analysis and harvested at approximately the same confluence.

RNA Isolation, Complementary RNA Preparation, and Microarray Experiment

Total RNA was isolated from subconfluent LECs and BECs with TRIzol reagent followed by a Qiagen RNA cleanup procedure (RNeasy; Qiagen, Valencia, CA). Five micrograms of total RNA was used from each of the three independent LEC and BEC preparations from cells isolated from three separate individuals. Samples were submitted to the Biopolymer Institute, a microarray core facility (Harvard Medical School, Boston, MA). Total RNA (2.5–5 µg) was used in the generation of the biotinylated complementary RNA as recommended by the manufacturer using the One-Cycle Eukaryotic Target Labeling Assay (Affymetrix, Santa Clara, CA). Biotinylated targets were hybridized to an Affymetrix GeneChip Human Genome U133 Plus 2.0 Array with the Gene Chip Hybridization Oven 640 (Affymetrix) and the Gene Chip Fluidics Station 450 (Affymetrix). The hybridized array was scanned with the Gene Chip Scanner 3000 7G (Affymetrix). Affymetrix GeneChip Operating Software (GCOS) performed a post run normalization by scaling the data to the global mean of 500.

Quantitative Reverse Transcription-Polymerase Chain Reaction (qRT-PCR)

Single-strand cDNA synthesis from total RNA was prepared using TaqMan Reverse Transcription Reagents as recommended by the manufacturer (Applied Biosystems, Foster City, CA). TaqMan Assays-On-Demand were purchased from Applied Biosystems for HoxA5 (Hs00430330_m1), HoxA10 (Hs00538183_m1), Podoplanin (Hs00366764_m1), and Prox1 (Hs00160463_m1). The genes ESX1 and BEX1 were assayed using Power SYBR Green-I (Applied Biosystems). Primers for the assay are as follows: ESX1, forward primer 5′-CTCAATATCCCGACGTTGTGG-3′ and reverse primer 5′-CCAAACCTGCACTCTGTCTTCA-3′; BEX1, forward primer 5′-GATAGGCCCAGGAGTAATGGAG-3′ and reverse primer 5′-CTTGGTTGGCATTTTCCATG-3′; and human glyceraldehyde-3-phosphate dehydrogenase, forward primer 5′-CATGAGAAGTATGACAACAGCCT-3′ and reverse primer 5′-AGTCCTTCCACGATACCAAAGT-3′. Each qRT-PCR performed was normalized to the endogenous glyceraldehyde-3-phosphate dehydrogenase levels and calibrated to the relative BEC signal for each gene using the ΔΔCT method as previously described [25]. All qRT-PCR analyses were performed in triplicate with ABI PRISM 7300 (Applied Biosystems).

Microarray Data Analysis

The unfiltered microarray data were subjected to X–Y plot analysis using GCOS. Three independently obtained LEC-BEC data pairs were superimposed on a single, logscale X–Y plot (X-axis: BEC mRNA expression; Y-axis: LEC mRNA expression). Gene expression calls (Present/Absent) were calculated using the default setting of GCOS and indicated by color of each datum point: red, present in both BEC and LEC; blue, absent in either BEC or LEC; yellow, absent in both BEC and LEC. Datum points for representative BEC-and LEC-specific genes were also indicated on the plot.

For gene ontology analysis, unfiltered microarray data were uploaded into GeneSifter (www.genesifter.net), a webbased analysis program with BEC data sets selected as control. Normalized data was log2-transformed followed by applying a fold change filter of ≥ 2.0 and a statistical t test with a P < .05 cutoff (two-tailed unpaired Student's t test assuming equal variance). Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis was performed on the resultant probe sets and scored for significance. A z-score of absolute value greater than 2 indicated whether a pathway occurs more or less frequently than expected. Extreme positive numbers (greater than 2) indicated that the term occurred more frequently than expected, whereas an extreme negative number (less than -2) indicated that the term occurred less frequently than expected.

The data were additionally filtered by expression level, selecting probe sets whose ratios were the result of having at least one of the two comparisons (LECs or BECs) having a log2-transformed expression level greater than 6.644 (this number equals a scaled raw expression value of > 100) in the numerator, i.e., LECs/BECs or BECs/LECs.

Two-dimensional hierarchical clustering analysis of 312 informative genes was performed using Cluster software and visualized with TreeView software (Lawrence Berkeley National Laboratories, Berkeley, CA) [26]. Complete linkage clustering analysis was performed using an uncentered correlation as a similarity metric for both genes and array data sets.

Results

Analysis of Microarray Data

We analyzed six individual Affymetrix gene arrays, three from CD31+/podoplanin-cell types and three from CD31+/podoplanin+ cell types, hereafter referred to as BECs and LECs, respectively. Each BEC and LEC cell line was isolated from a mixed population of CD31+ HDMECs taken from the same individual person, so each of the three sets was genetically identical. The mixed HDMEC population was immunomagnetically sorted into podoplanin-positive and -negative cells with thorough washing. The cultures were compared for podoplanin gene expression by measuring the amount of podoplanin mRNA in each cell type by qRT-PCR. LECs had podoplanin mRNA expression values ranging from 100- to 1500-fold higher than cultured BECs (data not shown).

To examine microarray data quality, scaled data were subjected to scatterplot analysis of LECs versus BECs (Figure 1). The overall shape of the plotted data showed a narrow, symmetrical triangular contour pointing to the right upper corner, which is typically observed when significantly similar data sets are compared by this method. On the X–Y plot for three superimposed BEC-LEC pair data sets, reproducible detection of differential expression of several known LEC-specific marker genes (i.e., podoplanin [PDPN], Prox-1, and mannose receptor C, type I [MRC1]) and BEC-specific marker genes (i.e., interferon, alpha-inducible protein 27 [IFI27], STAT6, selectin E [SELE], and von Willebrand Factor [vWF]) are demonstrated. These results strongly support the notion that the LEC and BEC cell preparations used in the present study appropriately represented gene expression characteristics for these cell types and that the microarray analysis of differential gene expression was properly performed.

Figure 1.

Figure 1

Gene expression characteristics of LEC and BEC cell types. Microarray data were superimposed onto an X–Y plot and typical marker genes were highlighted for each cell type. Prox1, prospero-related homeobox 1; MRC1, mannose receptor C, type I; PDPN, podoplanin; SELE, selectin E; vWF, von Willebrand factor; STAT6, signal transducer and activator of transcription 6; IFI27, interferon, alpha-inducible protein 27.

To further validate our gene array data, we choose representative genes with altered gene expression in LECs and BECs. Podoplanin and Prox-1 gene expression were measured because they are recognized as typical LEC-associated molecules and were differentially expressed in LECs. These genes were shown to have more abundant expression in LECs than BECs using TaqMan and SYBR Green qRT-PCR (Figure 2). Four other genes, ESX1, BEX1, HOXA5, and HOXA10, were chosen for verification because they comprised a range of expression levels and they had not been previously been associated with LECs. ESX1 and BEX1 exhibited high levels of gene expression. ESX1 was more abundant than podoplanin using qRT-PCR (Figure 2). HOXA5 and HOXA10 had similar relevant expression to Prox-1 (Figure 2). This small subset of genes helps verify the original array data and gives more confidence in the validity of the data.

Figure 2.

Figure 2

qRT-PCR expression analysis. Error bars are the relative quantification minimum and maximum.

To obtain objective evidence, we performed hierarchical clustering analysis of genes differentially expressed between LECs and BECs (Figure W1). The clustering algorithm clearly separated the triplicate LEC and BEC data sets (Figure W1A), providing preliminary lists of LEC/BEC cell type-discriminating marker genes. Because the number of genes that were expressed stronger in BECs (Figure W1B) and in LECs (Figure W1C) were comparable, the clustering analysis was unlikely to reflect nonspecific differences in cell viability or lower RNA quality for one of these cell types.

Data from the scaled microarray results were subjected to selection criteria and used to create a list of differentially expressed genes. The initial selection criteria (1.5-fold differential expression; t test P value < .05) identified 2287 probe sets; 1197 probe sets had a higher expression in LECs and 1090 probe sets had higher expression in BECs. This data set was used for KEGGpathway analysis.We applied a more stringent selection criteria by removing all probe sets with both LEC and BEC log2-transformed intensity values less than 6.644 (< 100 of a nontransformed scaled intensity value), which is generally not detectable by qRT-PCR. There were 1262 probe sets that passed this additional selection (see Table W1); 699 for LECs and 563 for BECs. This data set was used to generate the top 25 genes that demonstrated the highest expression differences between LECs and BECs (Table 1).

Table 1.

List of 25 Highly Expressed Genes in LECs and BECs.

Probe Set ID Gene Description Ratio Gene Identifier Chromosome
LECs
204879_at PDPN Podoplanin* 428.1 NM_006474 1
205523_at HAPLN1 Hyaluronan and proteoglycan link protein 1* 148.4 U43328 5
04438_at MRC1 Mannose receptor, C type 1 71.0 NM_002438 10
212560_at SORL1 Sortilin-related receptor, L(DLR class) A repeats-containing 20.3 AV728268 11
218332_at BEX1 Brain-expressed, X-linked 1 18.3 NM_018476 X
1552445_a_at ESX1 Extraembryonic, spermatogenesis, homeobox 1 homolog (mouse) 17.9 NM_153448 X
205015_s_at TGFA Transforming growth factor, alpha 17.4 NM_003236 2
229029_at CAMK4 Calcium/calmodulin-dependent protein kinase IV 17.3 AI745230 5
213436_at CNR1 Cannabinoid receptor 1 (brain) 16.8 U73304 6
204301_at KBTBD11 Kelch repeat and BTB (POZ) domain containing 11 15.1 NM_014867 8
1559975_at BTG1 B-cell translocation gene 1, antiproliferative 14.8 BC009050 12
213974_at ADAMTSL3 ADAMTS-like 3 14.4 AB033059 15
205965_at BATF Basic leucine zipper transcription factor, ATF-like 13.3 NM_006399 14
218741_at CENPM Centromere protein M 13.1 NM_024053 22
223631_s_at C19orf33 Chromosome 19 open reading frame 33 12.7 AF213678 19
211959_at IGFBP5 Insulin-like growth factor binding protein 5 12.6 AW007532 2
220318_at EPN3 Epsin 3 12.6 NM_017957 17
1557196_a_at - cDNA DKFZp547B198 (from clone DKFZp547B198) 12.4 AL831886 X
229081_at SLC25A13 Solute carrier family 25, member 13 (citrin) 12.2 AW268880 7
208161_s_at ABCC3 ATP-binding cassette, subfamily C (CFTR/MRP), member 3 12.0 NM_020037 17
204393_s_at ACPP Acid phosphatase, prostate 11.5 NM_001099 3
231805_at PRLHR Prolactin releasing hormone receptor 11.5 AL563031 10
207024_at CHRND Cholinergic receptor, nicotinic, delta 11.5 NM_000751 2
238529_at - CDNA clone IMAGE:6342029 10.3 AA573088 1
236817_at DEADC1 Deaminase domain containing 1 10.3 AI336346 6
BECs
202411_at IFI27 Interferon, alpha-inducible protein 27 274.53 NM_005532 14
206211_at SELE Selectin E (endothelial adhesion molecule 1) 104.42 NM_000450 1
207173_x_at CDH11 Cadherin 11, type 2, OB-cadherin (osteoblast) 76.01 D21254 16
209994_s_at ABCB1 ATP-binding cassette, subfamily B (MDR/TAP), member 1 51.57 AF016535 7
210163_at CXCL11 Chemokine (C-X-C motif) ligand 11 48.73 AF030514 4
212554_at CAP2 CAP, adenylate cyclase-associated protein, 2 (yeast) 34.81 N90755 6
203881_s_at DMD Dystrophin (muscular dystrophy, Duchenne and Becker types) 33.64 NM_004010 X
244204_at PRR3 Proline-rich 3 19.89 W87300 6
209074_s_at FAM107A Family with sequence similarity 107, member A 18.51 AF089853 3
228617_at BIRC4BP XIAP-associated factor-1 17.7 AA142842 17
205174_s_at QPCT Glutaminyl-peptide cyclotransferase (glutaminyl cyclase) 16.07 NM_012413 2
201332_s_at STAT6 Signal transducer and activator of transcription 6, interleukin-4 induced 15.37 BC004973 12
1552902_a_at FOXP2 Forkhead box P2 15.32 NM_148898 7
209474_s_at ENTPD1 Ectonucleoside triphosphate diphosphohydrolase 15.16 AV717590 10
230109_at PDE7B Phosphodiesterase 7B 14.99 AI638433 6
226281_at DNER Delta/notch-like EGF repeat containing 14.38 BF059512 2
1566161_at MEIS2 Meis1, myeloid ecotropic viral integration site 1 homolog 2 (mouse) 14.33 AA340499 15
230741_at - Full-length insert cDNA clone YX74D05 13.76 AI655467 12
210882_s_at TRO Trophinin 13.6 U04811 X
226992_at NOSTRIN Nitric oxide synthase trafficker 13.58 AK002203 2
228948_at EPHA4 EPH receptor A4 13.07 T15545 2
203687_at CX3CL1 Chemokine (C-X3-C motif) ligand 1 13.03 NM_002996 16
202718_at IGFBP2 Insulin-like growth factor-binding protein 2, 36 kDa 12.72 NM_000597 2
203153_at IFIT1 Interferon-induced protein with tetratricopeptide repeats 1 12.66 NM_001548 10
239669_at HIST1H3D Histone cluster 1, H3d 12.54 AW006409 6
*

This is the highest representative of multiple probe sets.

Podoplanin had the highest fold change overall suggesting it is an appropriate molecule to use to separate BECs and LECs. The next highest expressed gene in LECs was the hyaluronan and proteoglycan link protein 1 (HAPLN1), an ECM-linking protein that acts as a stabilizer of the interaction between versican and hyaluronan in various tissues [27]. Also included in the top 25 LEC genes were transcription factors (BATF and ESX1), receptors (MRC1, SORL1, CNR1, PRLHR, and CHRND), and several open reading frames of unknown function or lacking description.

BECs also had a wide variety of genes expressed at higher levels than in LECs. The interferon, alpha-inducible protein 27 (IFI27) was the highest differentially expressed gene for BECs. The only cell-surface receptor in the BEC top 25 list was EPH receptor A4 and was accompanied by the cell adhesion molecule (CAM), selectin E, which was expressed as expected [28]. Critical for extracellular signaling, the chemokine ligands CXCL11 and CX3CL1 were represented.

We employed KEGG pathways analysis to group the differentially expressed genes into known functional or biochemical pathways. Table 2 lists the pathways identified for LECs and BECs that were scored as significant (> |2| z-score). Each pathway listed in Table 2 has the associated genes listed in Tables W2 and W3. LECs had high z-scores for the cell cycle, pyrimidine metabolism, and one carbon pool by folate pathways, indicating optimal growth media used in culturing the LECs compared to BECs. The calcium signaling pathway also scored significantly for LECs.

Table 2.

KEGG Pathway Summary for LECs and BECs.

Total z Score


List LEC BEC Array LEC BEC
(A) LEC KEGG Pathways
Cell cycle 26 22 4 112 7.98 -0.26
Pyrimidine metabolism 13 10 3 90 3.14 -0.35
One carbon pool by folate 4 3 1 16 2.80 0.45
Calcium signaling pathway 26 15 11 174 2.77 1.56
Circadian rhythm 3 3 0 18 2.54 -0.87
Glycosphingolipid biosynthesis-neo-lactoseries 4 3 1 21 2.21 0.17
Insulin signaling pathway 2 1 1 134 -2.10 -1.97
Diterpenoid biosynthesis 1 1 0 4 2.01 -0.41
Styrene degradation 2 1 1 4 2.01 2.13
(B) BEC KEGG Pathways
Carbazole degradation 1 0 1 1 -0.21 4.87
2,4-Dichlorobenzoate degradation 2 0 2 5 -0.48 4.08
Cell adhesion molecules 19 5 14 127 -0.26 4.06
Fluorene degradation 1 0 1 2 -0.30 3.30
Histidine metabolism 6 1 5 38 -0.53 2.86
Glycosaminoglycan degradation 4 1 3 17 0.30 2.85
Parkinson's disease 4 1 3 18 0.24 2.72
Glycan structures-degradation 5 1 4 29 -0.25 2.67
1,1,1-Trichloro-2,2-bis(4-chlorophenyl)ethane degradation 1 0 1 3 -0.37 2.58
Oxidative phosphorylation 1 1 0 121 -1.95 -2.29
Styrene degradation 2 1 1 4 2.01 2.13
Prion disease 3 1 2 13 0.58 2.08

Cell Adhesion Molecules

BECs scored high (> 4) for the carbazole degradation, 2,4-dichlorobenzoate degradation, and CAM pathways (Table 2). The carbazole degradation and 2,4-dichlorobenzoate degradation pathways had few members within the array, possibly because the pathway is new or still in development. Cell adhesion molecules are of great importance to the structure and function of blood and lymphatic vessels. Table 3 lists the members of the CAM pathway identified by KEGG analysis.

Table 3.

Cell Adhesion Molecules' KEGG Pathway-Associated Genes.

BECs LECs
Claudin-5 CD58 molecule
Claudin-9 Claudin-7
Inducible T-cell costimulator ligand (ICOSL) Major histocompatibility complex, class I, C
Intercellular adhesion molecule 1 (CD54) Major histocompatibility complex, class II, DP alpha 1
Major histocompatibility complex, class II, DO alpha Poliovirus receptor - related 3
Major histocompatibility complex, class II, DR alpha
Neurexin-2
Neurexin-3
Neuronal growth regulator 1
Poliovirus receptor
Programmed cell death 1 ligand 2
Protein tyrosine phosphatase, receptor type, M
Selectin E
Selectin P

Claudin-5 [25], intercellular adhesion molecule 1 [29], inducible T-cell costimulator ligand (ICOSL) [30], CD58 [31], poliovirus receptor [32], protein tyrosine phosphatase, receptor type, M [33,34], selectin E [28], and selectin P [35] have been previously identified with vascular endothelial cell-cell adhesion and were expected. Surprisingly, several genes had not been reported as associated with BECs, such as claudin-9, the major histocompatibility complex class IIDO alpha and DR alpha, neurexin-2, neurexin-3, and neuronal growth regulator-1. Nevertheless, all the CAM-associated genes in Table 3 were novel for LEC expression.

Cytokines and Cytokine Receptors

Cytokines and their receptors are important features of endothelial cells. BECs in this study express the following genes that have not been previously reported for this endothelial cell type. Chemokine (C-C motif) ligand 15 was expressed nine-fold higher in BECs than in LECs. Chemokine (C-C motif) ligand 19 was expressed five-fold higher in BECs. Chemokine (C-X-C motif) ligand 11, expressed in endothelial cells and hepatocytes [36,37], was expressed in BECs at high levels (> 48-fold) compared to LECs in this study.

NOTCH2, a member of the Notch transmembrane protein family and usually expressed in BECs [38], was expressed 3.32-fold higher in LECs, as was HEY2 (9.9-fold), a transcriptional product of downstream Notch signaling [39].

Discussion

Our data show that LECs have a transcriptional program distinct from that of blood vascular endothelial cells. LECs and BECs perform similar functions but in a different micro-environment. Blood vascular endothelial cells line the lumen of capillary tubular structures assisted by perivascular cells and continuous basement membrane. Blood cells and fluid pass through these tubes at high velocity and under considerable pressure, which changes depending on the current state of the physiology of the organism. Protein and fluid are lost from the blood vascular system, bathing the surrounding cells with extracellular fluid. Protein- and lipid-rich fluid enters the initial lymphatic vessels along with immune cells en route to the lymph nodes and eventually reenter the circulatory system. LECs, the major component of the initial lymphatic system, in contrast to BECs, have much slower fluid dynamics. Lymph enters the lymphatic system through pressureinduced fluid transport. The two endothelial cell types are part of a vascular system, but with widely divergent physiological needs and requirements.

The transcriptome snapshot of LECs and BECs helps us to understand the differences and the similarities each cell type has. Each is tethered to the ECM. Many more ECM proteins are specifically required for proper BEC functioning. Endothelial cells of blood vessels need to actively interact with molecules displayed on other circulating cells, such as immune surveillance cells. It is not unusual to discover that some genes expressed in BECs are associated with cell-to-cell junction communication, such as claudin-9, which appears to be a developmentally expressed claudin. Before this report, only expression of claudin-9 in neonatal proximal tubule endothelial cells forming tight junctions within the mouse kidney was published [40]. The BECs isolated from neonatal HDMECs taken from infant foreskin for this study express claudin-9, but may not be expressed in adult BECs.

There is mounting evidence that the vascular and nervous systems use common protein machinery to establish the placement of vessels and nerves [41,42]. Neurexin-1, neurexin-2, and the neuronal growth factor regulator-1 were differentially expressed in BECs compared to LECs, providing another example of common usage in nerve and blood vessels patterning.

LECs expressed several genes that have not been previously associated with the lymphatic system.CD58 costimulates CD4(+) T-cells through the CD2–CD58 system by promoting lipid raft formation [31]. The expression of CD58 by LECs may indicate another location previously unknown that T-cell activation can occur, although this study does not indicate whether the expression of CD58 occurs on the luminal side or on the abluminal side of the initial lymphatic vessels.

The role of programmed cell death 1 ligand 2 (PDL2) was examined when Zhang et al. [43] generated knockout mice. The heterozygous -/- animals were healthy; antigen-presenting cells from Pdl2-/- mice induced stronger T-cell proliferation than was expected compared to wild-type cells in vitro and in vivo. Zhang et al. also concluded that PDL2 negatively regulated T-cells and played an essential role in immune tolerance.

Of interest, the poliovirus receptor-related 3 protein may have cell-adhesive functions, but this has not been extensively studied. However, poliovirus receptor-related 2 is thought to participate in the regulation of hematopoietic/endothelial homophilic cellular functions and act as an intercellular junction protein [44]. Thus, poliovirus receptor-related 3 protein may be a new CAM expressed on LECs that helps LECs form cell-cell junctions conducive to lymph formation and immune cell intravasation. In the same class of proteins, LECs express claudin-7, which is associated with tight junctions, which exclude water and solutes across endothelial cells. Claudin-7 also excludes the lateral transfer of lipids and membrane proteins in epithelial cells that defines their apical and basolateral compartments. Claudin-7 may perform the same function in LECs and maintain their polarity.

Cell signaling is an essential characteristic of BECs. Many of the chemokine and chemokine receptors that were identified in this study have been associated with the blood vascular system. These molecules may be significant as many tumor cells express receptors for chemokines and home to a metastatic niche. We have identified several chemokines that have not previously been associated with BECs. For example, chemokine (C-C) ligand 15 (CCL15) [45] can act as a signal for endothelial cell mobilization and, according to this study, BECs can also produce it, which may be context-dependent.

Dendritic cells (DCs) express chemokine (C-C) ligand 19 (CCL19), which regulates DC trafficking and the recruiting of naïve T-cells to the area of DC activity [46]. Expression of CCL19 in BECs indicates that the blood vascular endothelium may take an active role in the recruiting of DCs to areas of activity.

CXCL11 is known to be involved in the negative regulation of angiogenesis [47] through the CXCR3 receptors during interferon-γ induction. Activated T-cells, usually of the Th1 subtype, internalize CXCR3 when bound to their cognate ligand CXCL11 [48]. The expression of CXCL11 on LECs indicates another possible mechanism to attract activated T-cells to lymphatic vessels. Tumor cells may take advantage of this attracting mechanism to actively home the lymphatic network.

The expression of NOTCH2, a transcription factor, and its downstream transcriptional product, HEY2 indicates active Notch signaling in LECs. Notch signaling molecules are known to determine the cell fate of many cell types and developmental processes of several organs, including the nervous system. Notch may act as one of the cell-fate determinates for LECs.

In conclusion, this report confirms previous reports of molecules that are important determinants for cellular function of LECs and BECs. We also report several molecules that are newly associated with either LECs or BECs. Cell signaling is very important for BECs, as they express many more chemokine ligands and receptors, which may not be as important for the LECs due to their different microenvironment and need for cellular recruitment. The molecules identified may also be tested as possible ligands that attract tumor cells as they metastasize.

Extracellular adhesion molecules are more necessary for the BECs to handle the stress and strain of rapid fluid flow and ever changing blood vessel pressure. However, several molecules were discovered that are expressed by LECs that may lead to further characterization of LEC function, especially CD58 and the poliovirus receptor-related 3 molecules. This study reveals additional molecular markers that may be used as targets to modify either the lymphatic or the blood vascular endothelium in pathologic states.

Supplementary Material

Supplementary Figures and Tables
neo0912_1038SD1.pdf (530KB, pdf)

Acknowledgements

We thank Angera Kuo and Johanna Lahdenranta for critical reading of the manuscript. We appreciate the superb technical skill and assistance from Trent Rector and Sarah Annis of the Biopolymers Institute for microarray processing. Special thanks to Dontscho Kerjaschki for the gift of the podoplanin antibody.

Abbreviations

BEC

blood vascular endothelial cell

CAM

cell adhesion molecule

DC

dendritic cell

ECM

extracellular matrix

HDMEC

human dermal microvascular endothelial cell

KEGG

Kyoto Encyclopedia of Genes and Genomes

LEC

lymphatic endothelial cell

qRT-PCR

quantitative reverse transcription-polymerase chain reaction

VEGF

vascular endothelial growth factor

1

This work was supported by National Institutes of Health grants T32 CA73479 (G.N.) and R01 CA85140 (R.J.).

2

This article refers to supplementary materials, which are designated by Tables W1, W2, and W3 and Figure W1 and are available online at www.neoplasia.com.

References

  • 1.Schmid-Schonbein GW. Microlymphatics and lymph flow. Physiol Rev. 1990;70(4):987–1028. doi: 10.1152/physrev.1990.70.4.987. [DOI] [PubMed] [Google Scholar]
  • 2.Alitalo K, Carmeliet P. Molecular mechanisms of lymphangiogenesis in health and disease. Cancer Cell. 2002;1(3):219–227. doi: 10.1016/s1535-6108(02)00051-x. [DOI] [PubMed] [Google Scholar]
  • 3.Hoshida T, Isaka N, Hagendoorn J, di Tomaso E, Chen YL, Pytowski B, Fukumura D, Padera TP, Jain RK. Imaging steps of lymphatic metastasis reveals that vascular endothelial growth factor-C increases metastasis by increasing delivery of cancer cells to lymph nodes: therapeutic implications. Cancer Res. 2006;66(16):8065–8075. doi: 10.1158/0008-5472.CAN-06-1392. [DOI] [PubMed] [Google Scholar]
  • 4.Veronesi U, Paganelli G, Viale G, Luini A, Zurrida S, Galimberti V, Intra M, Veronesi P, Robertson C, Maisonneuve P, et al. A randomized comparison of sentinel-node biopsy with routine axillary dissection in breast cancer. N Engl J Med. 2003;349(6):546–553. doi: 10.1056/NEJMoa012782. [DOI] [PubMed] [Google Scholar]
  • 5.Morton DL, Thompson JF, Cochran AJ, Mozzillo N, Elashoff R, Essner R, Nieweg OE, Roses DF, Hoekstra HJ, Karakousis CP, et al. Sentinel-node biopsy or nodal observation in melanoma. N Engl J Med. 2006;355(13):1307–1317. doi: 10.1056/NEJMoa060992. [DOI] [PubMed] [Google Scholar]
  • 6.Kukk E, Lymboussaki A, Taira S, Kaipainen A, Jeltsch M, Joukov V, Alitalo K. VEGF-C receptor binding and pattern of expression with VEGFR-3 suggests a role in lymphatic vascular development. Development. 1996;122(12):3829–3837. doi: 10.1242/dev.122.12.3829. [DOI] [PubMed] [Google Scholar]
  • 7.Joukov V, Pajusola K, Kaipainen A, Chilov D, Lahtinen I, Kukk E, Saksela O, Kalkkinen N, Alitalo K. A novel vascular endothelial growth factor, VEGF-C, is a ligand for the Flt4 (VEGFR-3) and KDR (VEGFR-2) receptor tyrosine kinases. EMBO J. 1996;15(2):290–298. [PMC free article] [PubMed] [Google Scholar]
  • 8.Achen MG, Jeltsch M, Kukk E, Makinen T, Vitali A, Wilks AF, Alitalo K, Stacker SA. Vascular endothelial growth factor D (VEGF-D) is a ligand for the tyrosine kinases VEGF receptor 2 (Flk1) and VEGF receptor 3 (Flt4) Proc Natl Acad Sci USA. 1998;95(2):548–553. doi: 10.1073/pnas.95.2.548. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Nagy JA, Vasile E, Feng D, Sundberg C, Brown LF, Detmar MJ, Lawitts JA, Benjamin L, Tan X, Manseau EJ, et al. Vascular permeability factor/vascular endothelial growth factor induces lymphangiogenesis as well as angiogenesis. J Exp Med. 2002;196(11):1497–1506. doi: 10.1084/jem.20021244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Hirakawa S, Kodama S, Kunstfeld R, Kajiya K, Brown LF, Detmar M. VEGF-A induces tumor and sentinel lymph node lymphangiogenesis and promotes lymphatic metastasis. J Exp Med. 2005;201(7):1089–1099. doi: 10.1084/jem.20041896. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Kajiya K, Hirakawa S, Ma B, Drinnenberg I, Detmar M. Hepatocyte growth factor promotes lymphatic vessel formation and function. EMBO J. 2005;24(16):2885–2895. doi: 10.1038/sj.emboj.7600763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Cao R, Bjorndahl MA, Gallego MI, Chen S, Religa P, Hansen AJ, Cao Y. Hepatocyte growth factor is a lymphangiogenic factor with an indirect mechanism of action. Blood. 2006;107(9):3531–3536. doi: 10.1182/blood-2005-06-2538. [DOI] [PubMed] [Google Scholar]
  • 13.Cao Y. Direct role of PDGF-BB in lymphangiogenesis and lymphatic metastasis. Cell Cycle. 2005;4(2):228–230. doi: 10.4161/cc.4.2.1419. [DOI] [PubMed] [Google Scholar]
  • 14.Isaka N, Padera TP, Hagendoorn J, Fukumura D, Jain RK. Peritumor lymphatics induced by vascular endothelial growth factor-C exhibit abnormal function. Cancer Res. 2004;64(13):4400–4404. doi: 10.1158/0008-5472.CAN-04-0752. [DOI] [PubMed] [Google Scholar]
  • 15.Padera TP, Kadambi A, di Tomaso E, Carreira CM, Brown EB, Boucher Y, Choi NC, Mathisen D, Wain J, Mark EJ, et al. Lymphatic metastasis in the absence of functional intratumor lymphatics. Science. 2002;296(5574):1883–1886. doi: 10.1126/science.1071420. [DOI] [PubMed] [Google Scholar]
  • 16.Karkkainen MJ, Ferrell RE, Lawrence EC, Kimak MA, Levinson KL, McTigue MA, Alitalo K, Finegold DN. Missense mutations interfere with VEGFR-3 signalling in primary lymphoedema. Nat Genet. 2000;25(2):153–159. doi: 10.1038/75997. [DOI] [PubMed] [Google Scholar]
  • 17.Golshan M, Smith B. Prevention and management of arm lymphedema in the patient with breast cancer. J Support Oncol. 2006;4(8):381–386. [PubMed] [Google Scholar]
  • 18.Ottesen EA. Lymphatic filariasis: treatment, control and elimination. Adv Parasitol. 2006;61:395–441. doi: 10.1016/S0065-308X(05)61010-X. [DOI] [PubMed] [Google Scholar]
  • 19.Yong C, Bridenbaugh EA, Zawieja DC, Swartz MA. Microarray analysis of VEGF-C responsive genes in human lymphatic endothelial cells. Lymphat Res Biol. 2005;3(4):183–207. doi: 10.1089/lrb.2005.3.183. [DOI] [PubMed] [Google Scholar]
  • 20.Podgrabinska S, Braun P, Velasco P, Kloos B, Pepper MS, Skobe M. Molecular characterization of lymphatic endothelial cells. Proc Natl Acad Sci USA. 2002;99(25):16069–16074. doi: 10.1073/pnas.242401399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Petrova TV, Makinen T, Makela TP, Saarela J, Virtanen I, Ferrell RE, Finegold DN, Kerjaschki D, Yla-Herttuala S, Alitalo K. Lymphatic endothelial reprogramming of vascular endothelial cells by the Prox-1 homeobox transcription factor. EMBO J. 2002;21(17):4593–4599. doi: 10.1093/emboj/cdf470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Hirakawa S, Hong YK, Harvey N, Schacht V, Matsuda K, Libermann T, Detmar M. Identification of vascular lineage-specific genes by transcriptional profiling of isolated blood vascular and lymphatic endothelial cells. Am J Pathol. 2003;162(2):575–586. doi: 10.1016/S0002-9440(10)63851-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Wick N, Saharinen P, Saharinen J, Gurnhofer E, Steiner CW, Raab I, Stokic D, Giovanoli P, Buchsbaum S, Burchard A, et al. Transcriptomal comparison of human dermal lymphatic endothelial cells ex vivo and in vitro. Physiol Genomics. 2007;28(2):179–192. doi: 10.1152/physiolgenomics.00037.2006. [DOI] [PubMed] [Google Scholar]
  • 24.Breiteneder-Geleff S, Soleiman A, Kowalski H, Horvat R, Amann G, Kriehuber E, Diem K, Weninger W, Tschachler E, Alitalo K, et al. Angiosarcomas express mixed endothelial phenotypes of blood and lymphatic capillaries: podoplanin as a specific marker for lymphatic endothelium. Am J Pathol. 1999;154(2):385–394. doi: 10.1016/S0002-9440(10)65285-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Morita K, Sasaki H, Furuse K, Furuse M, Tsukita S, Miyachi Y. Expression of claudin-5 in dermal vascular endothelia. Exp Dermatol. 2003;12(3):289–295. doi: 10.1034/j.1600-0625.2003.120309.x. [DOI] [PubMed] [Google Scholar]
  • 26.Eisen MB, Spellman PT, Brown PO, Botstein D. Cluster analysis and display of genome-wide expression patterns. Proc Natl Acad Sci USA. 1998;95(25):14863–14868. doi: 10.1073/pnas.95.25.14863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Shi S, Grothe S, Zhang Y, O'Connor-McCourt MD, Poole AR, Roughley PJ, Mort JS. Link protein has greater affinity for versican than aggrecan. J Biol Chem. 2004;279(13):12060–12066. doi: 10.1074/jbc.M310091200. [DOI] [PubMed] [Google Scholar]
  • 28.Silverman MD, Babra B, Pan Y, Planck SR, Rosenbaum JT. Differential E-selectin expression by iris versus retina microvascular endothelial cells cultured from the same individuals. Microvasc Res. 2005;70(1–2):32–42. doi: 10.1016/j.mvr.2005.07.001. [DOI] [PubMed] [Google Scholar]
  • 29.Clark PR, Manes TD, Pober JS, Kluger MS. Increased ICAM-1 expression causes endothelial cell leakiness, cytoskeletal reorganization and junctional alterations. Invest Dermatol. 2007;127(4):762–774. doi: 10.1038/sj.jid.5700670. [DOI] [PubMed] [Google Scholar]
  • 30.Khayyamian S, Hutloff A, Buchner K, Grafe M, Henn V, Kroczek RA, Mages HW. ICOS-ligand, expressed on human endothelial cells, costimulates Th1 and Th2 cytokine secretion by memory CD4+ T cells. Proc Natl Acad Sci USA. 2002;99(9):6198–6203. doi: 10.1073/pnas.092576699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Mestas J, Hughes CC. Endothelial cell costimulation of T cell activation through CD58-CD2 interactions involves lipid raft aggregation. J Immunol. 2001;167(8):4378–4385. doi: 10.4049/jimmunol.167.8.4378. [DOI] [PubMed] [Google Scholar]
  • 32.Reymond N, Imbert AM, Devilard E, Fabre S, Chabannon C, Xerri L, Farnarier C, Cantoni C, Bottino C, Moretta A, et al. DNAM-1 and PVR regulate monocyte migration through endothelial junctions. J Exp Med. 2004;199(10):1331–1341. doi: 10.1084/jem.20032206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Bianchi C, Sellke FW, Del Vecchio RL, Tonks NK, Neel BG. Receptor-type protein-tyrosine phosphatase mu is expressed in specific vascular endothelial beds in vivo. Exp Cell Res. 1999;248(1):329–338. doi: 10.1006/excr.1999.4428. [DOI] [PubMed] [Google Scholar]
  • 34.Koop EA, Lopes SM, Feiken E, Bluyssen HA, van der Valk M, Voest EE, Mummery CL, Moolenaar WH, Gebbink MF. Receptor protein tyrosine phosphatase mu expression as a marker for endothelial cell heterogeneity; analysis of RPTPmu gene expression using LacZ knock-in mice. Int J Dev Biol. 2003;47(5):345–354. [PubMed] [Google Scholar]
  • 35.Miyazaki Y, Satoh T, Nishioka K, Yokozeki H. STAT-6-mediated control of P-selectin by substance P and interleukin-4 in human dermal endothelial cells. Am J Pathol. 2006;169(2):697–707. doi: 10.2353/ajpath.2006.051211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Sheikh AM, Ochi H, Manabe A, Masuda J. Lysophosphatidylcholine posttranscriptionally inhibits interferon-gamma-induced IP-10, Mig and I-Tac expression in endothelial cells. Cardiovasc Res. 2005;65(1):263–271. doi: 10.1016/j.cardiores.2004.09.017. [DOI] [PubMed] [Google Scholar]
  • 37.Helbig KJ, Ruszkiewicz A, Semendric L, Harley HA, McColl SR, Beard MR. Expression of the CXCR3 ligand I-TAC by hepatocytes in chronic hepatitis C and its correlation with hepatic inflammation. Hepatology. 2004;39(5):1220–1229. doi: 10.1002/hep.20167. [DOI] [PubMed] [Google Scholar]
  • 38.Siekmann AF, Lawson ND. Notch signalling limits angiogenic cell behaviour in developing zebrafish arteries. Nature. 2007;445(7129):781–784. doi: 10.1038/nature05577. [DOI] [PubMed] [Google Scholar]
  • 39.Fischer A, Schumacher N, Maier M, Sendtner M, Gessler M. The Notch target genes Hey1 and Hey2 are required for embryonic vascular development. Genes Dev. 2004;18(8):901–911. doi: 10.1101/gad.291004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Abuazza G, Becker A, Williams SS, Chakravarty S, Truong HT, Lin F, Baum M. Claudins 6, 9, and 13 are developmentally expressed renal tight junction proteins. Am J Physiol Renal Physiol. 2006;291(6):F1132–F1141. doi: 10.1152/ajprenal.00063.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Carmeliet P, Tessier-Lavigne M. Common mechanisms of nerve and blood vessel wiring. Nature. 2005;436(7048):193–200. doi: 10.1038/nature03875. [DOI] [PubMed] [Google Scholar]
  • 42.Eichmann A, Makinen T, Alitalo K. Neural guidance molecules regulate vascular remodeling and vessel navigation. Genes Dev. 2005;19(9):1013–1021. doi: 10.1101/gad.1305405. [DOI] [PubMed] [Google Scholar]
  • 43.Zhang Y, Chung Y, Bishop C, Daugherty B, Chute H, Holst P, Kurahara C, Lott F, Sun N, Welcher AA. Regulation of T cell activation and tolerance by PDL2. Proc Natl Acad Sci USA. 2006;103(31):11695–11700. doi: 10.1073/pnas.0601347103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Lopez M, Aoubala M, Jordier F, Isnardon D, Gomez S, Dubreuil P. The human poliovirus receptor related 2 protein is a new hematopoietic/endothelial homophilic adhesion molecule. Blood. 1998;92(12):4602–4611. [PubMed] [Google Scholar]
  • 45.Hwang J, Kim CW, Son KN, Han KY, Lee KH, Kleinman HK, Ko J, Na DS, Kwon BS, Gho YS, et al. Angiogenic activity of human CC chemokine CCL15 in vitro and in vivo. FEBS Lett. 2004;570(1-3):47–51. doi: 10.1016/j.febslet.2004.06.023. [DOI] [PubMed] [Google Scholar]
  • 46.Pietila TE, Veckman V, Lehtonen A, Lin R, Hiscott J, Julkunen I. Multiple NF-kappaB and IFN regulatory factor family transcription factors regulate CCL19 gene expression in human monocyte-derived dendritic cells. J Immunol. 2007;178(1):253–261. doi: 10.4049/jimmunol.178.1.253. [DOI] [PubMed] [Google Scholar]
  • 47.Romagnani P, Lasagni L, Annunziato F, Serio M, Romagnani S. CXC chemokines: the regulatory link between inflammation and angiogenesis. Trends Immunol. 2004;25(4):201–209. doi: 10.1016/j.it.2004.02.006. [DOI] [PubMed] [Google Scholar]
  • 48.Sauty A, Colvin RA, Wagner L, Rochat S, Spertini F, Luster AD. CXCR3 internalization following T cell-endothelial cell contact: preferential role of IFN-inducible T cell alpha chemoattractant (CXCL11) J Immunol. 2001;167(12):7084–7093. doi: 10.4049/jimmunol.167.12.7084. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplementary Figures and Tables
neo0912_1038SD1.pdf (530KB, pdf)

Articles from Neoplasia (New York, N.Y.) are provided here courtesy of Neoplasia Press

RESOURCES