• No results found

Biology of monocyte interactions with the endothelium : the platelet factor - Chapter 6 "DC-SIGN mediates adhesion and rolling of dendritic cells on primary human umbilical vein endothelial cells through LewisY antig

N/A
N/A
Protected

Academic year: 2021

Share "Biology of monocyte interactions with the endothelium : the platelet factor - Chapter 6 "DC-SIGN mediates adhesion and rolling of dendritic cells on primary human umbilical vein endothelial cells through LewisY antig"

Copied!
21
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

UvA-DARE (Digital Academic Repository)

Biology of monocyte interactions with the endothelium : the platelet factor

da Costa Martins, P.A.

Publication date

2005

Link to publication

Citation for published version (APA):

da Costa Martins, P. A. (2005). Biology of monocyte interactions with the endothelium : the

platelet factor.

General rights

It is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), other than for strictly personal, individual use, unless the work is under an open content license (like Creative Commons).

Disclaimer/Complaints regulations

If you believe that digital publication of certain material infringes any of your rights or (privacy) interests, please let the Library know, stating your reasons. In case of a legitimate complaint, the Library will make the material inaccessible and/or remove it from the website. Please Ask the Library: https://uba.uva.nl/en/contact, or a letter to: Library of the University of Amsterdam, Secretariat, Singel 425, 1012 WP Amsterdam, The Netherlands. You will be contacted as soon as possible.

(2)

Chapterr 6

"DC-SIGNN mediates adhesion and rolling

off dendritic cells on primary human

umbilicall vein endothelial cells through

Lewis

YY

antigen expressed on ICAM-2"

JuanJuan J, Garcia-Vallejo

1

, Ellis van Liempt

1

, Paula da Costa Martins

2

, Cora

BeckersBeckers

33

,, Bert van het Hof

1

, Sonja I. Gringhuis

1

, Jaap-Jan Zwaginga

2,4

,

WillemWillem van Dijk

1

, Teunis B. H. Geijtenbeek

1

, Yvette van Kooyk

1

, and

IrmaIrma van Die

1

1

Dept.. of Molecular Cell Biology & Immunology, VU University Medical Centre 2Dept.. of Experimental Immunohematology, Sanquin Research, Location CLB laboratoryy for Physiology, VU University Medical Centre

4Dept.. of Hematology, Academical Medical Center; Amsterdam,, The Netherlands Manuscriptt in preparation

(3)

Abstract t

Immaturee dendritic ceils (DCs) are recruited from blood into tissues to patrol for foreignn antigens. After antigen uptake and processing, DCs mature and migrate to thee secondary lymphoid organs where they initiate immune responses. DC-SIGN is a DC-specificc C-type lectin that acts both as a pattern recognition receptor and as an adhesionn molecule. As an adhesion molecule, DC-SIGN is able to mediate rolling andd adhesion over endothelial cells under shear flow. The binding partner of DC-SIGNN in endothelial cells is the carbohydrate epitope LewisY (LeY), expressed on ICAM-2.. ICAM-2 expressed on CHO cells only served as a ligand for DC-SIGN when properlyy glycosylated, underscoring its function as a scaffolding protein. The expressionn of LeY in endothelial cells is directed by the enzyme FUT1. Silencing of FUT11 results in an inhibition of the rolling and adhesion of immature DCs over endotheliall cells. The identification of LeY as the carbohydrate ligand of DC-SIGN in endotheliall cells opens new possibilities for the manipulation of DC migration.

(4)

Introduction n

Dendriticc cells (DC) have a key role in the control of immunity by surveying peripherall tissues in the search for self or non-self antigens \ In order to create a networkk of tissue-resident DCs, precursor DCs continuously migrate from the blood intoo peripheral tissues, where they are highly efficient in capturing and processing

antigenss as immature DCs 23. Once activated, immature DCs mature and migrate

fromm the peripheral tissues to secondary lymphoid organs in order to interact with specificc T-cells and initiate an immune response 1. The molecular basis for the migratoryy capacity of DCs is starting to be unraveled 4 5, and several molecules have beenn described to be involved, such as DC-SIGN 6, MR 7 8, and selectins 4. DC-SIGN (CD209)) is a C-type lectin expressed by precursor and immature DCs that was primarilyy identified through its high affinity interaction with ICAM-3 9. In addition, DC-SIGNN also functions as an HIV-1 frans-receptor important in the dissemination of HIV-11 10. Importantly, DC-SIGN mediates rolling and adhesion of precursor DC over thee endothelium, which is suggested to be mediated through interactions with ICAM-26. .

Thuss DC-SIGN appears as a molecule with a dual role, acting as a pattern recognitionn receptor and as an adhesion molecule. As a pattern recognition receptor, thee carbohydrate specificity of DC-SIGN has been carefully evaluated. It is now clear thatt DC-SIGN is able to recognize high-mannose type N-glycans, as well as glycoconjugatess carrying non-sialylated, non-sulfated Lewis antigens 11"16. This relativelyy large recognition profile converts DC-SIGN into a sort of broad-spectrum patternn recognition receptor. Many pathogens have been found to be recognized and

internalizedd by DC-SIGN 4 and, although this mechanism is meant to allow the

developmentt of an immune response, often is used by the pathogen to escape

immunee surveillance 17. As an adhesion molecule, however, the identity of the

endogenouss carbohydrate ligand(s) of DC-SIGN, especially in endothelial cells, still remainss inconclusive.

Thee present study was undertaken to identify the DC-SIGN ligands that are crucial forr the adhesion and rolling of dendritic cells on endothelial cells. We show here that ICAM-22 expressed on endothelial cells constitutes the major scaffold protein ligand forr DC-SIGN. Importantly, the interaction of DC-SIGN with ICAM-2 is carbohydrate-dependent,, and we provide evidence that LeY antigens within ICAM-2 are of crucial importancee for the binding of DC-SIGN to endothelial cells, as well as for the rolling andd adhesion of dendritic cells over endothelial cells.

(5)

Materialss and Methods

CellCell lines and primary cells. Human umbilical vein endothelial cells (HUVEC)

weree isolated from 5 healthy donors by a modification of the method of Jaffe et al1 8, ass previously described 19. The cells were resuspended in M199 (Biowhittaker, USA) supplementedd with 100 U/mL Penicilin-Streptomycin (Biowhittaker, USA), 10 % humann serum (Biowhittaker, USA), 10 % new born calf serum (Biowhittaker, USA), 5 U/mll heparine (Leo Pharmaceutical Products, The Netherlands), and 150 ug/mL bFGFF (Sigma, The Netherlands) and plated in gelatin-coated plates. The cells were

culturedd to confluency in the mentioned media in a 5 % C02 atmosphere at 37 .

Whenn confluency was reached, cells were trypsinized (0.18 % trypsin, 10 mM EDTA) andd plated again to 1/3 of their density. All endothelial cells used displayed the presencee of Von Willebrand factor, platelet endothelial cell adhesion molecule-1 (CD31),, and VE-Cadherin 20. No immunoreactivity to the anti-cytokeratin 20 antibody orr the anti-a-smooth muscle actin antibody was observed.

Chinesee Hamster Ovary (CHO) cells were cultured in RPMI-1640 (Gibco BRL, USA) supplementedd with 10% FCS and 100 U/ml Penicillin-Streptomycin. Where indicated, cellss were cultured during 5 days in the presence of kifunensine (Kitasatosporia

kifunense,kifunense, 2 ug/ml; Calbiochem, USA). Efficiency of treatment was assessed by flow

cytometryy analysis using Con A as described under Flow Cytometry.

Immaturee DCs were obtained from a buffycoat as previously described 21. In short, humann peripheral blood mononuclear cells (PBMCs) were isolated from a buffycoat byy a Ficoll gradient and followed by a CD14 magnetic microbeads isolation (MACS; Miltenyibiotec,, USA). The obtained CD14+ monocytes were differentiated into immaturee DCs in the presence of interleukine-4 and granulocyte-macrophage colony stimulatingg factor (500 and 800 U/ml, respectively; Schering-Plough, Belgium). At day 6,, the phenotype of the cultured DCs was confirmed by flow cytometric analysis. The immaturee DCs expressed high levels of major histocompatibility complex class I and II,, CD11 b, CD11c, and ICAM-1; and low levels of CD80 and CD86.

WesternWestern blotting. SIGN-Fc consists of the extracellular portion of

DC-SIGNN (amino acid residues 64-404) fused at the C-terminus to a human lgG1-Fc

fragmentt into the Sig-plgG1-Fc vector 22. DC-SIGN-Fc was produced in Chinese

hamsterr ovary K1 cells by cotransfection of DC-SIGN-Sig-plgG1 Fc (20 ug) and pEE144 (5 ug) vector. DC-SIGN-Fc concentrations in the supernatant were determinedd by an anti-lgG1 Fc ELISA. Cells were grown to confluency in a T175 flaskk (Corning, USA), washed and resuspended in TSM, and lysed in 0.1 M Tris-HCI (pHH 7.4), 0.05 M CHAPS. Lysate was centrifuged and the supernatant incubated with DC-SIGN/Fcc (0.5 mg/ml) at 4 C in a rotating device (18 h). Subsequently, 20 ul protA/G-agarosee beads (Santa Cruz, USA) were added and incubated at 4 C in a

(6)

rotatingg device (4 h). The beads were washed twice in TSM, resuspended in Laemmli

samplee buffer and incubated at C for 5 minutes prior to resolving in 10 %

SDS-PAGE,, according to Laemmli23.

Afterr electrophoresis, the gel was blotted on to a PVDF (Millipore, The Netherlands) membranee and stained with DC-SIGN/Fc or mouse anti-human-ICAM-2 (12A2) using peroxidase-labeledd goat anti-human (Jackson, USA) and rabbit anti-mouse immunoglobulinss (DakoCytomation, Denmark). The membrane was developed using SuperSignall WestPico Chemiluminescence substrate (Pierce, USA) and the chemiluminescencee detected in an Epi Chemi II Darkroom (UVP, USA) using the Labworkss (UVP, USA) software.

TransientTransient transfection of CHO cells. Cells were incubated until 50-80 %

confluent.. The transfection was performed according to the manufacturer's protocol. Inn short, both DNA (5 ug pcDM8-ICAM2 or pcDNA1-FUT4) and LipofectAMINE (Gibcoo BRL, USA) were diluted in serum free medium and combined. After 30 min thee cells were washed with serum free medium and the complex solution was added too the cells. After 5 h serum-enriched medium (20 %) was added. The medium was replacedd with fresh, complete medium 24 h after transfection. 24 h later the cells

(5104)) were resuspended, washed with TSM and analysed by flow cytometry as

indicatedd under Flow cytometry. Alternatively, cells (106) were lysed in 0.1 M Tris-HCI (pHH 7.4), 0.05 M CHAPS and analyzed by ELISA as indicated under ELISA.

CaptureCapture ELISA. Goat anti-mouse-Fc was coated on ELISA plates (Nunc,

USA;; 2 ug/ml, 100 pl/well), followed by mouse anti-human ICAM2 (12A2) antibodies (11 ug/ml, 50 Ml/well). Plates were blocked with 1 % ELISA grade BSA (Fraction V, Fattyy acid free; Calbiochem, USA), cell lysates were added, and incubated overnight att 4 . After washing, the wells were incubated with DC-SIGN-Fc (5 ug/ml), anti-LewisYY (5 ug/ml , clone F3, Calbiochem, USA) and digoxin-labeled Con A (5 ug/ml, Roche,, Switzerland), in the presence or absence of 50 mM a-D-CH3-Mannose/a-D-Chh-Glucosee (both Sigma, The Netherlands). Binding was detected using a peroxidasee labelled anti-human IgG-Fc, goat anti-mouse IgM (both Jackson, West Grove,, PA) or sheep anti-digoxin (Roche, Switzerland), respectively. Color developmentt after adding POD substrate (Roche, Switzerland) was measured in a spectrophotometerr (BioRad, USA) at 410 nm.

FlowFlow Cytometry. Cells were washed twice in cold TSM (20 mM Tris, pH 7.4,

1500 mM NaCI, 1 mM CaCI2 and 2 mM MgCI2), resuspended in 1 % BSA/TSM and incubatedd 30 minutes at room temperature with 25 uL of 1 % BSA-TSM diluted

(7)

primaryy antibody/lectin (10 ug/ml), washed twice with TSM and incubated 30 minutes att room temperature with secondary antibody according to manufacturers instructions.. After the second incubation, cells were washed twice with PBS and resuspendedd in a final volume of 100 uL 1 % BSA/TSM for analysis in the FACS-Caliburr (Becton-Dickinson, USA). The primary monoclonal antibodies (mouse IgM)

usedd were specific for Lewisx (DakoCytomation, Denmark), LewisY (clone F3,

Calbiochem,, USA), Lewis3 (clone T174, Calbiochem, USA), Lewisb (clone T128,

Calbiochem,, USA). The lectins used were Con A (concanavalin A, digoxin-labeled, Roche,, Switzerland), UEA-I (Ulex europaeus agglutinin, biotin-labeled, EY Labs, USA),, and AAL (Aleuria aurantia, digoxin-labeled, Roche, Switzerland). The anti-carbohydratee antibodies, were counter-stained with Alexa 488-labeled goat-anti-mousee secondary antibody (Molecular Probes, The Netherlands). For the secondary stainingg of the biotin-labeled lectins Alexa 488-streptavidin (Vector Laboratories, USA)) and FITC-labeled anti-digoxin (Sigma, USA) were used.

Cellss were analyzed for immunofluorescence on a FACS-Calibur flow cytometer by collectingg data for 104 cells per histogram. Corresponding negative controls were performedd by omitting the antibody or lectin of interest.

mRNAmRNA isolation and cDNA synthesis. mRNA was isolated by capturing of

poly(A+)) RNA in streptavidin-coated tubes with an mRNA Capture kit (Roche, Switzerland)) and cDNA was synthesized with the Reverse Transcription System kit (Promega,, USA) following manufacturer's guidelines. Cells (2*105/well) were washed twicee with ice-cold PBS and harvested with 200 uL of lysis buffer. Lysates were incubatedd with biotin-labeled oligo(dT)2o for 5 min at 37 C and then 50 uL of the mix weree transferred to streptavidin-coated tubes and incubated for 5 min at 37 . After washingg 3 times with 250 uL of washing buffer, 30 uL of the reverse transcription mix (55 mM MgCb, 1x reverse transcription buffer, 1 mM dNTP, 0.4 U recombinant RNasin ribonucleasee inhibitor, 0.4 U AMV reverse transcriptase, 0.5 ug random hexamers in nuclease-freee water) were added to the tubes and incubated for 10 min at room

temperaturee followed by 45 min at 42 . To inactivate AMV reverse transcriptase

andd separate mRNA from the streptavidin-biotin complex, samples were heated at 99 CC for 5 min, transferred to microcentrifuge tubes and incubated in ice for 5 min, dilutedd 1:2 in nuclease-free water and stored at -20 C until analysis.

QuantitativeQuantitative real-time PCR. Oligonucleotides (Table I) have been designed

byy using the computer software Primer Express 2.0 (Applied Biosystems, USA). All oligonucleotidess were synthesized by Invitrogen (Invitrogen, Belgium). Oligonucleotidee specificity was computer tested (BLAST, NCBI) by homology search withh the human genome and specifically, with all the known galactosyltransferases

(8)

(CLUSTALW,, EMBL), and later confirmed by dissociation curve analysis and resolvingg the PCR products in agarose electrophoresis. In the case of FUT3, FUT5 andd FUT6, genes with a high homology, the specificity of the primers was tested usingg plasmids (kindly provided by Dr. JB Lowe) encoding for each of the fucosyltransferases.. The efficiency 24 of the oligonucleotides was determined using thee computer program LinReg 25 and resulted in an average of 90 %. PCR reactions weree performed with the SYBR Green method in an ABI 7900HT sequence detection systemm (Applied Biosystems, USA). The reactions were set on a 96 well-plate by mixingg 4 uL of the 2 times concentrated SYBR Green Master Mix (Applied Biosystems,, USA) with 2 uL of a oligonucleotide solution containing 5 nmol/uL of bothh oligonucleotides and 2pL of a cDNA solution corresponding to 1/60 of the cDNA

synthesiss product. The thermal profile for all the reactions was 2 min at 50 ,

followedd by 10 min at 95 C and then 40 cycles of 15 sec at 95 C and 1 min at 60 . Thee fluorescence monitoring occurred at the end of each cycle.

Tablee I. Oligonucleotides used in the present study.

Gene e Genee ID Oligonucleotides

GAPD D

H H 2597 7 Fwd:: aggtcatccctgagctgaacgg Rev: cgcctgcttcaccaccttcttg

FX X FUT1 1 FUT2 2 FUT3 3 FUT4 4 FUT5 5 FUT6 6 FUT7 7 FUT8 8 FUT9 9 7264 4 2523 3 2524 4 2525 5 2526 6 2527 7 2528 8 2529 9 2530 0 10690 0 Fwd:: agccatccagaaggtggtagc Fwd:: gcaggccatggactggtt Fwd:: ctcgctacagctccctcatctt Fwd:: ccagtgggtcctcccga Fwd:: gagctacgctgtccacatcacc Fwd:: gtcccgagacgatgccact Fwd:: atcccactgtgtaccctaatgg Fwd:: tccgcgtgcgactgttc Fwd:: tcttcatccccgtcctcca Fwd:: caaatcccatgcagttctgatc Rev:: gacgtgtgtgggttggacc Rev:: cctgggaggtgtcgatgttt Rev:: cgtgggaggtgtcaatgttct Rev:: gccatgtccatagcaggatca Rev:: cagctggccaagttccgtatg Rev:: ccggtgacaggttccactg Rev:: tgccaggcaccatctctgag Rev:: accctcaaggtcctcatagacttg Rev:: gagacacccaccacactgca Rev:: gtggcctagcttgctgaggta

Additionally,, dissociation curve analysis was performed at the end of every run by

increasingg the temperature of the block from 60 to 95 C at a rate of 1.75 n

whilee continuously monitoring fluorescence. The plot of the first derivative of the decreasee in fluorescence with respect to temperature showed in all cases one single

(9)

peakk at the Tm predicted by the Primer Express 2.0 software. The Ct value is defined ass the number of PCR cycles where the fluorescence signal exceeds the detection thresholdd value, which is fixed above 10 times the standard deviation of the fluorescencee during the first 15 cycles and typically corresponds to 0.2 relative fluorescencee units. This threshold is set constant throughout the study and correspondss to the log linear range of the amplification curve. The normalized amountt of target, or relative abundance 26, reflects the relative amount of target transcriptss with respect to the expression of the endogenous reference gene. Due to thee low expression of glycosyltransferases, the results are shown as 100-times the relativee abundance. In this study, the endogenous reference gene chosen was GAPDH,, based on previous results 19.

RNARNA interference. The mammalian expression vector, pSUPER.retro.puro

27288

(a kind gift of Dr. R. Agami, Netherlands Cancer Institute, Amsterdam, The Netherlands)) was used for expression of siRNA in HUVEC. The gene-specific insert identifiess a 19-nucleotide sequence corresponding to nucleotides 272-291 (tcagatgggacagtatgcc)) of FUT-1 (NM_000148), nucleotides 362-381 (gacgacctacccgatagat)) of FX (NM_003313), or the sequence 5'-ctgaatgaatcgtgacacg withh no significant similarity to any human gene sequence, therefore used as a silencingg control. The gene-specific insert was separated by a 9-nucteotide non-complementaryy spacer (ttcaagaga) from the reverse complement of the same 19-nucleotidee sequence, and flanked by restriction sites for the enzymes Bgl II and Hind III,, producing a final insert of 60 nucleotides. These sequences were inserted into the pSUPER.retro.puroo backbone. The different vectors were referred to as pSUPER/FUT-1,, pSUPER/FX, and pSUPER/Scrambled, respectively. Plasmids were transfectedd into HUVEC using the Basic Nucleofector Kit for Primary Mammalian Endotheliall Cells (Amaxa, Germany) in an Amaxa Nucleofector (Amaxa, Germany), accordingg to manufacturer's instructions. Immediately after transfection, cells were seededd in glass coverslips coated with crosslinked gelatin (1 %) and fibronectin (5 mg/ml).mg/ml). Transfection efficiency was higher than 90 % as evaluated by flowcytometry analysiss of HUVEC co-transfected pmax/GFP (Amaxa, Germany) and the different pSUPERR constructs (data not shown). To test the efficiency of RNA interference, cellss were lysed after 48 h, mRNA isolated (mRNA Capture Kit, Roche, Switzerland) andd retrotranscribed into cDNA (Reverse Transcription System, Promega, USA), accordingg to manufacturer's instructions. Gene expression of FUT-1, FX, and ICAM-2 wass assessed by means of quantitative real-time PCR in an ABI 7900HT platform (Appliedd Biosystems, USA) using the SYBR Green I chemistry (Applied Biosystems,

USA),, as previously described 19, using the primers described in Table I. The

silencingg resulted in a decrease in gene expression higher than 80 % (data not shown). .

(10)

ImmatureImmature DC perfusion and evaluation of adhesion and rolling velocity.

Immaturee DCs in suspension (2x106 cells/ml in incubation buffer) were aspirated fromm a reservoir through plastic tubing and perfused through a chamber with a Harvardd syringe pump (Harvard Apparatus, South Natic, MA). The flow rate through thee chamber was precisely controlled and the immature DCs were perfused over endotheliall cells at 0.8 dyn/cm2. During perfusions the flow chamber was mounted on aa microscope stage (Axiovert 25, Zeiss, Germany), equipped with a B/W CCD video

cameraa (Sanyo, Osaka, Japan), and coupled to a VHS video recorder 29,3 . Video

imagess were evaluated for the number of adherent monocytes and the rolling velocity perr cell, with dedicated routines made in the image analysis software Optimas 6.1 (Mediaa Cybernetics Systems, Silverspring, MD, USA). The immature dendritic cells thatt were in contact with the surface appeared as bright white-centered cells after properr adjustment of the microscope during recording. The number of surface-adherentt immature dendritic cells was measured after 5 min of perfusion at a minimumm of 25 randomized high-power fields. To automatically determine the velocity off rolling cells, custom-made software was developed in Optimas 6.1. A sequence of 500 frames representing an adjustable time interval (8t, with a minimal interval of 80 milliseconds)) was digitally captured. The position of every cell was detected in each frame,frame, and for all subsequent frames the distance traveled by each cell and the numberr of images in which a cell appears in focus was measured. The cut-off value too distinguish between rolling and static adherent cells was set at 1 (im/s. With this method,, static adherent, rolling and free flowing cells (which were not in focus) could bee clearly distinguished.

Results s

ICAM-2ICAM-2 is the major DC-SIGN ligand on endothelial cells. Precursor DCs

continuouslyy traffic to peripheral tissues. The migration process is highly dependent onn the interaction of DC-SIGN with its ligand 1. In earlier studies, ICAM-2 was identifiedd as a ligand for DC-SIGN that supports binding under shear stress

conditionss using a chimeric construct produced in CHO cells (ICAM-2/Fc) 6. To

identifyy counter receptors for SIGN on primary endothelial cells, chimeric DC-SIGN/Fcc protein was used to immunoprecipitate ligands from a HUVEC cell lysate of whichh the surface proteins had been labeled by biotin. Subsequently, the immunoprecipitatee and the supernatant were subjected to SDS-PAGE under reducingg conditions, and the isolated proteins analyzed by western blot. As shown in Figuress 1A and 1C, the major surface-labeled protein that was selectively precipitatedd by DC-SIGN-Fc has an apparent molecular weight of 55-60 KDa, which coincidess with the molecular weight of ICAM-2 31,32. The selective precipitation of ICAM-22 could be confirmed after western blotting analysis using an anti-ICAM-2 antibodyy for immunodetection (Figure 1B).

(11)

4'JJ 1 mm W W 50 40 0

Figuree 1. DC-SIGN binds to ICAM-2 in endothelial cells. HUVECs were labeled with biotin

priorr to lysis. SIGN ligands were immunoprecipitated in a HUVEC lysate by incubation with DC-SIGN/Fcc and Prot-A/G agarose beads as described in Materials and Methods. A. 10 % SDS-PAGE. Onee major band of approximatedly 55-60 KDa was detected in the immunoprecipitated fraction. 6. Westernn Blot, immunodetection with anti-ICAM-2 antibody (12A2). The main immunoprecipitated band correspondss to ICAM-2. C. Western Blot, immunodetection with Streptavidin. The immunoprecipitated bandd identified as ICAM-2, is present in the extracellular membrane of HUVECs. Results are representativee of 3 experiments.

DC-SIGN-ICAM-2DC-SIGN-ICAM-2 interaction is carbohydrate-dependent. ICAM-2 is also a

ligandd for LFA-1 and this interaction is carbohydrate-independent 33. To investigate whetherr the DC-SIGN-ICAM-2 interaction is carbohydrate dependent, CHO cells weree transfected with a cDNA coding for ICAM-2, and either grown in the presence of kifunensinee or co-transfected with a cDNA coding for FUT4. Kifunensine is an a-mannosidasee I inhibitor that stops the processing of N-glycans at the

Man9-GlcNAc2-Asnn stage 34. Cells grown in the presence of kifunensine produce mainly

high-mannosee type N-glycans. FUT4 is an a1,3-fucosyltransferase implicated in the synthesiss of Lex 35. Both Lex and high-mannose N-glycans are recognized by DC-SIGNN 11'13.

Ass shown in Figure 2A, the DC-SIGN/Fc chimera does not bind to ICAM-2 expressed inn untreated cells, whereas it binds with high affinity to ICAM-2 expressing CHO cells thatt were grown in the presence of kifunensine. Remarkably, DC-SIGN/Fc also recognizedd glycoproteins other than ICAM-2 on the CHO cells that were grown in the presencee of kifunensine, as can be observed in the mock transfected CHO cells. In ann ICAM-2-immobilizing ELISA (Figure 2B), we could show that the kifunensine treatmentt resulted in an ICAM-2 population that showed increased binding of the lectinn Con A, as well as an increased DC-SIGN/Fc binding (Figure 2B).

(12)

Thee co-transfection with FUT4 also resulted in an increase in the binding of DC-SIGN/Fc,, which correlated with an increase in the expression of Lex-containing ICAM-22 (Figure 2A and B). Together, these data demonstrate that binding of DC-SIGN/Fcc to ICAM-2 is strictly carbohydrate dependent, and that the glycosylate potentiall of the cells expressing ICAM-2 determines whether or not ICAM-2 can functionn as a counter receptor for DC-SIGN.

' H ) I A ( 5 « M | | Untreated d

L L

U U

kifunensmc c

M M

iJ iJ

Untreated d

L L

U U

KK t Aniens me

!'A !'A

U U

-- FUT-4

LJ J

aj j

B B S S ê ê

n n

_L L j . . M * * J L L X X U

--HSH H

JL L

11 I

Figuree 2. The DC-SIGN-ICAM-2 interaction is carbohydrate dependent. CHO cells were

transfectedd with the plasmid pcDM8-ICAM-2 and either left untreated, treated with kifunensine or co-transfectedd with the plasmid pcDNA1-FUT4. Untreated or kifunensine-treated mock transfectant were usedd as controls. A Flow cytometry analysis using an anti-ICAM-2 antibody (12A2) or the DC-SIGN/Fcc chimeric protein (+ EDTA as a negative control). Gray lines denote the isotype control, while solidd areas represent the staining with the above mentioned antibody or chimeric molecule. B. The bindingg of DC-SIGN/Fc, Con A and anti-Lex to ICAM-2 captured from the CHO cells transfected with thee plasmid pcDM8-ICAM-2 was analyzed by ELISA in plates coated with the antibody 12A2. Results aree representative of 3 experiments.

ICAM-2ICAM-2 carries LeY in endothelial cells. In order to explore the glycosylate

off ICAM-2 in endothelial cells, HUVECs were analyzed by flow cytometry, ELISA, andd western blotting. Using antibodies specific for the Lewis antigens Lex, LeY, Lea,

(13)

andd Leb, it was demonstrated that HUVEC expressed significant amounts of LeY (Figuree 3A). Presence of the other Lewis antigens could not be detected on HUVECs,, whereas all antibodies showed specific binding to control neoglycoconjugatess expressing the respective Lewis antigens (data not shown). Con AA was also able to bind to endothelial cells (Figure 3A). Con A recognizes unsubstitutedd hydroxyl groups in mannose, as those present in high-mannose structures,, hybrid-type N-glycans and diantennary N-glycans (with this order of affinity)) 36'37. Using increasing concentrations of methyl-mannoside it is possible to discriminatee whether Con A binds to the high affinity ligand (high-mannose glycans) orr to the low affinity ligands (diantennary N-glycans). A priori, as indicated by previouss in vitro studies 11,13, both LeY and mannose-rich structures are potential ligandss for DC-SIGN.

i i

i i

AAL L

Le' '

A A

UEA-l l

k k

Conn A

jt" "

1CAM-2 2 B B 55 «1 FL-l l

XX

JL

n n

OO 01 < <

Figuree 3. Endothelial cells express LeY and Con A-reactive epitopes. A. HUVEC were

grownn to confluency, mechanically detached, incubated with the corresponding lectin or antibody, and analyzedd by flow cytometry. Gray lines denote the isotype control, while solid areas represent the stainingg with the above mentioned lectin or antibody. B. Alternatively, HUVEC were lysed and the bindingg of DC-SIGN/Fc (+ EDTA as a negative control), anti-LeY, Con A, and Con A + 50 mM methyl-mannosidee to captured ICAM-2 was analyzed by ELISA in plates coated with anti-ICAM-2 (12A2). Resultss are representative of 3 experiments.

(14)

Too identify the carbohydrates present in ICAM-2, immobilized ICAM-2 from an endotheliall cell extract was tested for binding with anti-LeY antibodies and Con A in ELISA.. The data in Fig. 3B show that both anti-LeY antibodies and Con A bound to thee captured ICAM-2. Binding of Con A, however, could be completely abolished by additioinn of low concentrations of methyl-mannoside (Figure 3B), indicating that the loww affinity Con A-reactive glycans on ICAM-2 most likely correspond to diantennary N-glycans,, rather than high-mannose type N-glycans. This indicates that LeY most likelyy represents the ligand for DC-SIGN on ICAM-2.

HUVECHUVEC express FUT1 and FUT4 as the main a2- and

a3-fucosyltransferases,fucosyltransferases, respectively. The expression of fucosylated carbohydrates

dependss upon the expression of fucosyltransferases 38. To identify the

fucosyltransferasess that are involved in the synthesis of LeY structures in HUVEC, a highlyy sensitive and specific real-time PCR assay was designed. Special care was takenn to design oligonucleotides able to discriminate FUT3, FUT5 and FUT6, which havee a large sequence identity. Plasmids encoding for FUT3, FUT5 and FUT6 were usedd as positive control (data not shown). The results showed significant mRNA levelss for only three fucosyltransferase genes in HUVEC. Amongst them, FUT4, whichh encodes an a3-fucosyltransferase involved in the synthesis of Lewis-type

structures,, as well as the VIM-2 antigen 35, and FUT1 that encodes an

a2-fucosyltransferasee 39, can contribute to the biosynthesis of LeY The third

fucosyltransferase,, FUT8, encodes for an a6-fucosyltransferase that catalyzes the transferr of fucose to the first acetylglucosamine of the chitobiose core of an N-glycann 40.

DC-SIGNDC-SIGN interacts with the LeY structure on ICAM-2 expressed by endothelialendothelial cells. Based on the fucosyltransferase gene expression profile, FUT1

wass the a priori candidate to direct the synthesis of the DC-SIGN ligand LeY in HUVEC.. To further investigate this point, a silencing approach was followed 27,28 to knockk down the expression of the enzymes that are expected to be crucial for the

synthesiss of LeY. HUVECs were transfected with either pSUPER/Scrambled

(non-silencingg control), pSUPER/FX or pSUPER/FUT-1. The plasmid pSUPER/FX targets thee expression of the gene FX, which encodes GDP-4-keto-6-deoxymannose 3,5-epimerase-4-reductase,, one of the enzymes necessary for the synthesis GDP-fucose fromm GDP-mannose 41,42. This pathway accounts for the vast majority of cellular

GDP-fucosee production 43. GDP-fucose is the sugar donor used in the reactions

catalyzedd by fucosyltransferases. In the absence of this enzyme, the pool of GDP-Fucosee can be rescued by adding fucose to the culture media, which is then

metabolyzedd to GDP-Fucose via the salvage pathway 44. The efficiency of the

(15)

ann anti-LeY antibody (data not shown). As shown in Figure 5, the rolling velocity of immaturee DCs on a cell-layer of primary HUVEC is increased when either FX or FUT11 were silenced. Simultaneously, the tethering and adhesion (B) of the immature DCss to HUVEC is decreased. The degree of increase in rolling velocity and decrease inn tethering and adhesion was to the same extent as was achieved using the monoclonall antibody AZN-D1, which is a DC-SIGN blocking antibody. Furthermore, thee effects obtained with pSUPER/FX could be rescued by adding fucose to the HUVECs. . 0.400 ££ 0.35 Q Q

<< °

M

'

*Z*Z 0.10 Ü Ü II 0.08 c c II 0.06 aa 0 04 s s && 0.02 0 0

A A

—— <N f i - t "~. \ £ r- 3C 5-D D D D D D D D D D U . U . U . L L . U . U . U - U . U . .

Figuree 4. Fucosyltransferase gene expression profile. HUVEC were grown to confluency

andd assayed for the expression of a panel of fucosyltransferases (FUT1-9) using real-time PCR. GAPDHH was used as an endogenous reference. Results are shown as the average SE of 5 experiments. .

Discussion n

Immaturee DCs use DC-SIGN as an adhesion molecule to recognize a counter-receptorr in endothelial cells. As a result of this interaction, immature DCs tether and adheree to endothelial cells, starting the migration to the underlying tissue. In this studyy we have shown that ICAM-2 expressed in endothelial cells constitutes the majorr scaffold protein ligand for DC-SIGN. Furthermore, we have demonstrated that

LeYY present on ICAM-2 acts as the major carbohydrate ligand that mediates

DC-SIGNN adhesion and tethering to HUVEC.

ICAM-22 was identified as the major endothelial ligand for SIGN emplying in a DC-SIGN/Fcc immuneprecipitation from HUVEC lysates (Figure 1). Although the main visiblee band is situated around 55-60 KDa (Figure 1A), coinciding with ICAM-2 as

(16)

detectedd by Western-Blot (Figure 1B), two other minor bands (molecular weight > 1000 KDa) are visible that are surface located (Figure 1C), indicating that other proteinss expressed by HUVEC may contribute to the binding. It was also demonstratedd that the ICAM-2-DC-SIGN interaction is carbohydrate-dependent, as ICAM-22 expressed in CHO cells is only able to be recognized by DC-SIGN when properlyy glycosylated, excluding an integrin-like interaction (Figure 2). This system illustratess a complex model in which both interaction partners perform dual functions, DC-SIGNN as a pattern-recognition receptor and an adhesion molecule, and ICAM-2 ass an integrin ligand and a scaffold molecule for a lectin-ligand

Inn this study, we have identified LeY as the major carbohydrate ligand for DC-SIGN onn endothelial cell-expressed ICAM-2, as is suggested by the flow cytometry analysis off endothelial cells, and further demonstrated by an ICAM-2 specific capture ELISA (Figuree 3), and the FUT-1 silencing experiments (Figure 5). Recently, it was publishedd that ICAM-2 presents high-mannose N-glycans, which serve as carbohydratee ligands for DC-SIGN 45.

Figuree 5. The binding of DC-SIGN to ICAM-2 in endothelial cells is blocked by silencing

thee expression of LeY using RNA interference. The expression of FX and FUT-1 was silenced in

HUVEC.. The rolling velocity (A) and adhesion (B) of monocyte-derived DCs over the transfected HUVECC was measured as indicated in Materials and Methods. Results are shown as the average SEE of 3 experiments.

However,, the work of Jimenez et al. has a serious technical disadvantage, the conclusionss are based on ICAM-2 produced in large amounts in COS cells. It is very welll known that glycosylation is a species and cell-type specific property 46"48. This

(17)

hass also been evidenced in this study (Figure 2). Additionally, our rolling/adhesion assayss demonstrate that fucosylation is essential for the rolling and adhesion of monocyte-derivedd DCs (Figure 5). Additionally, the silencing of FUT-1 results in a reductionn in rolling and adhesion analogue to the inhibition obtained with the anti-DC-SIGNN antibody AZN-D1 (Figure 5). In our opinion, this unequivocally proves the identityy of LeY as the carbohydrate ligand of DC-SIGN in HUVEC.

Interestingly,, LeY is expressed by many endothelial cell glycoproteins (data not shown),, however only ICAM-2, and perhaps other high-molecular weight minor ligands,, can support the binding of DC-SIGN. This may be explained by spatial considerations,, since there is no evidence so far in other post-translational modificationss being necessary for the DC-SIGN-carbohydrate ligand interaction 11,12,155 a s j s t n e c a s e for p_selectin 49. The discovery of FUT-1 as a key enzyme in thee synthesis of the endothelial DC-SIGN-ligand opens new possibilities in the manipulationn of DC migration.

Acknowledgements s

Wee thank Dr J. B. Lowe for the kind gift of the plasmids pCDM7-FUT3, pcDNA1-FUT5,, and pcDNA1-FUT6, and Dr. R. Agami for the plasmid pSUPER.retro.puro.. We also thank K. van Gisbergen for developing the ELISA assay,, Dr. R. Beelen for his help in providing umbilical cords, and Dr. G. Kraal for helpfull discussions and critical reading of the manuscript.

(18)

References s

1 .. Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature 1998; 392: 245-52. .

2.. Inaba K, Inaba M, Naito M, Steinman RM. Dendritic cells progenitors phagocytose particulates, includingg bacillus Calmette-Guerin organisms, and sensitize mice to micobacterial antigens in vivo.

J.Exp.Med.J.Exp.Med. 1993; 178: 479-88.

3.. Svensson M, Stockinger B, Wick MJ. Bone marrow-derived dendritic cells can process bacteriafor MHC-II and MHC-II presentation to T cells. J.lmmunol. 1997; 158: 4229-36.

4.. Geijtenbeek TB, van Vliet SJ, Engering A, 't Hart BA, van Kooyk Y. Self- and nonself-recognition by C-typee lectins on dendritic cells. Annu.Rev.lmmunol. 2004; 22: 33-54.

5.. van Kooyk Y, Geijtenbeek TB. A novel adhesion pathway that regulates dendritic cell trafficking and TT cell interactions. Immunol.Rev. 2002; 186: 47-56.

6.. Geijtenbeek TB, Krooshoop DJ, Bleijs DA, van Vliet SJ, van Duijnhoven GC, Grabovsky V et al. DC-SIGN-ICAM-22 interaction mediates dendritic cell trafficking. Natlmmunol. 2000; 1 (4): 353-7.

7.. Leteux C, Chai W, Loveless RW, Yuen CT, Uhlin-Hansen L, Combamous Y et al. The cysteine-rich domainn of the macrophage mannose receptor is a multispecific lectin that recognizes chondroitin sulfatess A and B and sulfated oligosaccharides of blood group Lewis(a) and Lewis(x) types in addition too the sulfated N-glycans of lutropin. J.Exp.Med. 2000; 191 (7): 1117-26.

8.. Irjala H, Johansson EL, Grenman R, Alanen K, Salmi M, Jalkanen S. Mannose receptor is a novel ligandd for L-selectin and mediates lymphocyte binding to lymphatic endothelium. J.Exp.Med. 2001; 194:: 1033-42.

9.. Geijtenbeek TB, Torensma R, van Vliet SJ, van Duijnhoven GC, Adema GJ, van Kooyk Y et al. Identificationn of DC-SIGN, a novel dendritic cell-specific ICAM-3 receptor that supports primary immunee responses. Ce//2000; 100 (5): 575-85.

10.. Geijtenbeek TB, Kwon DS, Torensma R, van Vliet SJ, van Duijnhoven GC, Middel J et al. DC-SIGN,, a dendritic cell-specific HIV-1-binding protein that enhances trans-infection of T cells. Cell 2000; 1000 (5): 587-97.

H.Feinbergg H, Mitchell DA, Drickamer K, Weiss Wl. Structural basis for selective recognition of oligosaccharidess by DC-SIGN and DC-SIGNR. Science 2001; 294: 2163-6.

12.. Guo Y, Feinberg H, Conroy E, Mitchell DA, Alvarez R, Blixt O et al. Structural basis for distinct ligand-bindingg and targeting properties of the receptors DC-SIGN and DC-SIGNR. Nat.Stnict.Mol.Biol. 2004;; 11 (7): 591-8.

13.. Van Die I, van Vliet SJ, Nyame AK, Cummings RD, Bank CM, Appelmelk B et al. The dendritic cell-specificc C-type lectin DC-SIGN is a receptor for Schistosoma mansoni egg antigens and recognizess the glycan antigen Lewis x. Glycobiology 2003; 13 (6): 471-8.

14.. Appelmelk BJ, Van Die I, van Vliet SJ, Vandenbroucke-Grauls CM, Geijtenbeek TB, van Kooyk Y. Cuttingg edge: carbohydrate profiling identifies new pathogens that interact with dendritic cell-specific ICAM-3-grabbingg nonintegrin on dendritic cells. J.Immunol. 2003; 170 (4): 1635-9.

15.. Van Liempt E, Imberty A, Bank CM, van Vliet SJ, van Kooyk Y, Geijtenbeek TB et al. Molecular basiss of the differences in binding properties of the highly related C-type lectins DC-SIGN and L-SIGN too Lewis X trisaccharide and Schistosoma mansoni egg antigens. J.Biol.Chem. 2004; 279 (32): 33161-7. .

(19)

16.. Blixt O, Head S, Mondala T, Scanlan C, Huflejt ME, Alvarez R et al. Printed covalent glycan array forr ligand profiling of diverse glycan binding proteins. Proc.Natl.Acad.Sci.U.S.A. 2004; 101 (49): 17033-8. .

17.. van Kooyk Y, Geijtenbeek TB. DC-SIGN: escape mechanism for pathogens. Nat Rev.Immunol. 2003;; 3 (9): 697-709.

18.. Jaffe EA, Nachman RL, Becker CG, Minick CR. Culture of human endothelial cells derived from umbilicall veins. Identification by morphologic and immunologic criteria. J.Clin.Invest 1973; 52 (11): 2745-56. .

19.. Garcia-Vallejo JJ, Van het Hof B, Robben J, Van Wijk JAE, Van Die I, Joziasse DH et al. Approach forr defining endogenous reference genes in gene expression experiments. Anal.Biochem. 2004; 329: 293-9. .

20.. van Hinsbergh VW, Sprengers ED, Kooistra T. Effect of thrombin on the production of plasminogenn activators and PA inhibitor-1 by human foreskin microvascular endothelial cells.

Thromb.Haemost.Thromb.Haemost. 1987; 57 (2): 148-53.

21.. Sallusto F, Lanzavecchia A. J.Exp.Med. 1994; 179: 1109-18.

22.. Geijtenbeek TB, van Duijnhoven GC, van Vliet SJ, Krieger E, Vriend G, Figdor CG et al. Identificationn of different binding sites in the dendritic cell-specific receptor DC-SIGN for intercellular adhesionn molecule 3 and HIV-1. J.Biol.Chem. 2002; 277 (13): 11314-20.

23.. Laemmli UK. Cleavage of structural proteins during the assembly of the head of bacteriophage T4.

NatureNature 1970; 227: 680-5.

24.. Pfaffl MW. A new mathematical model for relative quantification in real-time RT-PCR. Nucleic

AcidsAcids Res. 2001; 29 (9): e45.

25.. Ramakers C, Ruijter JM, Lekanne Deprez RH, Moorman AFM. Assumption-free analysis of quantitativee real-time polymerase chain reaction (PCR) data. Neuroscience Letters 2003; 339 (1): 62-6. .

26.. Livak KJ, Schmittgen TD. Analysis of Relative Gene Expression Data Using Real-Time Quantitativee PCR and the 2"CT Method. Methods 2001; 25: 402-8.

27.. Brummelkamp TR, Bernards R, Agami R. A system for stable expression of short interfering RNAs inn mammalian cells. Science 2002; 296: 550-3.

28.. Brummelkamp TR, Bernards R, Agami R. Stable suppression of tumorigenicity by virus-mediated RNAA interference. Cancer Cell 2002; 2 (3): 243-7.

29.. Sakariassen KS, Aarts PA, de Groot PG, Houdijk WP, Sixma JJ. A perfusion chamber developed too investigate platelet interaction in flowing blood with human vessel wall cells, their extracellular matrix,, and purified components. J.Lab Clin.Med. 1983; 102 (4): 522-35.

30.. van Zanten H, Saelman EU, Schut-Hese KM, Wu YP, Slootweg PJ, Nieuwenhuis HK et al. Platelet adhesionn to collagen type IV under flow conditions. Blood 1996; 88 (10): 3862-71.

31.. Gahmberg CG, Nortamo P, Zimmermann D, Ruoslahti E. The human leukocyte-adhesion ligand, intercellular-adhesionn molecule 2. Expression and characterization of the protein. Eur.J.Biochem. 1991;; 195(1): 177-82.

32.. Diacovo TG, deFougerolles AR, Bainton DF, Springer TA. A functional integrin ligand on the surfacee of platelets: intercellular adhesion molecule-2. J.Clin.Invest 1994; 94 (3): 1243-51.

33.. Bleijs DA, Geijtenbeek TB, Figdor CG, van Kooyk Y. DC-SIGN and LFA-1: a battle for ligand.

(20)

34.. Elbein AD, Tropea JE, Mitchell M, Kaushal GP. Kifunensine, a potent inhibitor of the glycoprotein processingg mannosidase I. J.Biol.Chem. 1990; 265 (26): 15599-605.

35.. Lowe JB, Kukowska-Latallo JF, Nair RP, Larsen RD, Marks RM, Macher BA et al. Molecular cloningg of a human fucosyltransferase gene that determines expression of the Lewis x and VIM-2 epitopess but not ELAM-1-dependent cell adhesion. J.Biol.Chem. 1991; 266 (26): 17467-77.

36.. Goldstein I, Hollerman C, Smith E. Protein-carbohydrate interaction. II. Inhibition studies on the interactionn of concanavalin A with polysaccharides. Biochemistry 1965; 41: 876-83.

37.. Goldstein I, Hollerman C, Merrick J. Protein-carbohydrate interaction. I. The interaction of polysaccharidess with concanavalin A. Biochim.Biophys.Acta 1965; 97: 68-76.

38.. Lowe JB. Glycosylation in the control of selectin counter-receptor structure and function.

Immunol.Rev.Immunol.Rev. 2002; 186:19-36.

39.. Larsen RD, Ernst LK, Nair RP, Lowe JB. Molecular cloning, sequence, and expression of a human GDP-L-fucose:beta-D-galactosidee 2-alpha-L-fucosyltransferase cDNA that can form the H blood group antigen.. Proc.Natl.Acad.Sci.U.S.A. 1990; 87: 6674-8.

40.. Yanagidani S, Uozumi N, lhara Y, Miyoshi E, Yamaguchi N, Taniguchi N. Purification and cDNA cloningg of GDP-L-Fuc:N-acetyl-beta-D-glucosaminide:alpha1-6 fucosyltransferase (alpha1-6 FucT) fromm human gastric cancer MKN45 cells. J.Biochem.(Tokyo) 1997; 121: 626-32.

41.. Camardella L, Carratore V, Ciardello MA, Damonte G, Benatti U, De Flora A. Primary structure of humann erythrocyte nicotinamide adenine dinucleotide phosphate (NADP[H])-binding protein FX: Identificationn with the mouse turn-transplantation antigen P35B. Blood 1995; 85: 264-7.

42.. Tonetti M, Sturla L, Bisso A, Benatti U, De Flora A. Synthesis of GDP-L-Fucose by the human FX protein.. J.Biol.Chem. 1996; 271: 27274-9.

43.. Yurchenko PD, Atkinson PH. Equilibration of fucosyl glycoprotein pools in HeLa cells.

BiochemistryBiochemistry 1977; 16: 944-53.

44.. Smith PL, Myers JT, Rogers CE, Zhou L, Petryniak B, Becker DJ et al. Conditional control of selectinn ligand expression and global fucosylation events in mice with a targeted mutation at the FX locus.. J.CellBiot, 2002; 158 (4): 801-15.

45.. Jimenez D, Roda P, Springer TA, Casasnovas JM. Contribution of N-linked glycans to the conformationn and function of intercellular adhesion molecules (ICAMs). J.Biol.Chem. 2005; in press. 46.. Wyss DF, Wagner G. The structural role of sugars in glycoproteins. Curr.Opin.Biotechnol. 1996; 7 (4):: 409-16.

47.. Elbein AD. The role of N-linked oligosaccharides in glycoprotein function. Trends Biotechnol. 1991; 99 (10): 346-52.

48.. Fussenegger M, Bailey JE, Hauser H, Mueller PP. Genetic optimization of recombinant glycoproteinn production by mammalian cells. Trends Biotechnol. 1999; 17 (1): 35-42.

49.. Leppanen A, Mehta P, Ouyang YB, Ju T, Helin J, Moore KL et al. A novel glycosulfopeptide binds too P-selectin and inhibits leukocyte adhesion to P-selectin. J.Biol.Chem. 1999; 274 (35): 24838-48.

(21)

Referenties

GERELATEERDE DOCUMENTEN

Other Rel-regulated prosur- vival genes are bcl-2 homolog Al, which is re- quired to prevent antigen receptor ligation- induced cell death in B cells (Grumont et al. 2000) and

In an attempt to identify new collaborating oncogenes in MoMLV-induced tumors in Eu.-myr transgenic mice, another common insertion site, pal-1 (proviral activation in lymphomas

As much as 2 5 % (10/40) of the lymphomas with proviral insertions near bmil also contain MoMLV integration in the eisl/gfi/pall/evi5 lo-.. Proviral integrations in bmil and

Gfil promotes thymocyte selection at different stages and inhibits death by neglect, during negative selec- tion and in common receptor y-chain deficient mice.. Blanca Scheijen 1

In the current study flow cytometric analysis was performed on spleen and bone marrow, to determine whether differentiation of the other major hematopoietic lineages

The zinc-finger protein Gfil and its close homologue GfilB have been implicated in regulating myeloid and T cell development (Schmidt et al. Here we report that enforced

To assess whether enforced gfil expres- sion could affect c-Myc-induced proliferation or inhibit apoptosis in pre-B cells, we analyzed bone marrow cells of wild type, single

The data described in this thesis indicate that the transcriptional regulators Bmil and Gfil control programmed cell death by different mechanisms in lymphoid cells, but each