Expression of human NDRG2 by myeloid dendritic cells inhibits down ...

40 downloads 217 Views 994KB Size Report
of Pathology, College of Medicine, Chungnam National University, Daejeon, Korea; ... Research Center for Women's Diseases, Sookmyung Women's University, ...
Expression of human NDRG2 by myeloid dendritic cells inhibits down-regulation of activated leukocyte cell adhesion molecule (ALCAM) and contributes to maintenance of T cell stimulatory activity Seung-Chul Choi,*,† Kwang Dong Kim,* Jong-Tae Kim,* Jae Wha Kim,* Hee Gu Lee,* Jin-Man Kim,‡ Yong-Suk Jang,† Do-Young Yoon,§ Keun Il Kim,ⱍⱍ Young Yang,ⱍⱍ Dae Ho Cho,ⱍⱍ and Jong-Seok Limⱍⱍ,1 *Laboratory of Cell Biology, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea; ‡Department of Pathology, College of Medicine, Chungnam National University, Daejeon, Korea; †Division of Biological Sciences and the Institute for Molecular Biology and Genetics, Chonbuk National University, Chonju, Korea; §Department of Molecular Biotechnology, Konkuk University, Seoul, Korea; and ⱍⱍDepartment of Biological Sciences and the Research Center for Women’s Diseases, Sookmyung Women’s University, Seoul, Korea

Abstract: We reported previously that N-myc downstream-regulated gene 2 (NDRG2), a member of a new family of differentiation-related genes, is expressed specifically in dendritic cells (DC) differentiated from monocytes, CD34ⴙ progenitor cells, and the myelomonocytic leukemic cell line. In this study, we demonstrate that NDRG2 protein expression is detected, not only in in vitro-differentiated DC but also in primary DC from lymph nodes, thymus, and skin when anti-NDRG2 antibodies are used. As predicted from previous studies investigating the mRNA expression pattern of several types of cell lines, progenitor cells, and DC, NDRG2 protein was expressed strongly in DC. Its expression was detected at significant levels after differentiation from progenitor cells. RNA interference of NDRG2 demonstrated that activated leukocyte cell adhesion molecule (ALCAM) expression is down-regulated specifically in DC differentiated from NDRG2 small interfering RNA (siRNA)-transfected monocytes. This was consistent with our observation that U937 cells transfected with NDRG2 became resistant to the GM-CSF/IL-4-induced ALCAM reduction. Furthermore, DC, which had differentiated from NDRG2 siRNA-transfected monocytes, showed a reduced ability to induce T cell proliferation. Taken together, our results indicate that NDRG2 is able to preserve ALCAM expression during DC differentiation from monocytes under cytokine culture conditions and that its expression helps DC maintain costimulatory signals necessary for T cell stimulation. J. Leukoc. Biol. 83: 89 –98; 2008.

T lymphocytes and to generate primary T cell responses. Naive T cells achieve stimulation by forming synapses with antigenpresenting DC in the T cell areas of secondary lymphoid organs [2]. Proliferating T cells are continuously poised to form synapses with DC [3], thus allowing the signaling process to be sustained in dividing cells for as long as they are retained in T cell areas in contact with antigen-carrying DC. It has been well established that optimal T cell activation requires two stimulatory signals. One of these signals is the TCR, which recognizes antigen in the form of peptide fragments bound to MHC class molecules on APC. The second signal is generally referred to as the costimulatory signal, which does not, by itself, induce any proliferation in resting T cells but significantly lowers the number of TCR complexes, which need to be triggered to induce the activation and acceleration of T cell responses [4, 5]. A variety of costimulatory molecules has been described, which may alter the overall functional avidity of selection by affecting TCR-dependent signals or by inducing signals independently of TCR. CD5 and CD6 antigens are examples of similar costimulatory proteins, which are important during thymocyte selection [6 –9]. In addition, CD6 is capable of providing the costimulatory signals needed to synergize with signals mediated through the TCR to enhance T cell proliferation [10, 11]. Activated leukocyte cell adhesion molecule (ALCAM; CD166), also known as KG-CAM, neurolin, and BEN/DMGRASP/SC1, is a novel member of the Ig superfamily, which is involved in homophilic adhesion and in binding to CD6 [12, 13]. It is expressed on activated leukocytes [14], monocytes [15], hematopoietic progenitor cells [16], bone marrow stromal cells [17], and hematopoiesis-supporting osteoblastic cells

Key Words: dendritic cells (DC) 䡠 siRNA

INTRODUCTION Dendritic cells (DC) are the most potent APC found in lymphoid organs [1]. They have the unique ability to activate naive 0741-5400/08/0083-0089 © Society for Leukocyte Biology

1 Correspondence: Department of Biological Sciences and the Research Center for Women’s Diseases, Sookmyung Women’s University, ChungpaDong, Yongsan-Gu, Seoul 140-742, Korea. E-mail: [email protected] Received May 14, 2007; revised September 2, 2007; accepted September 8, 2007. doi: 10.1189/jlb.0507300

Journal of Leukocyte Biology Volume 83, January 2008 89

[18]. ALCAM-mediated interactions are important during neural development [19], maturation of hematopoietic stem cells in blood-forming tissues [16, 17, 20], immune responses [21], and tumor progression [22]. Although ALCAM was shown to mediate homotypic and heterotypic cell-cell clustering through homophilic (ALCAM-ALCAM) and heterophilic (ALCAMCD6) interaction [14], Hassan et al. [23] demonstrated recently that homophilic interaction was approximately 100 times weaker than heterophilic interaction. It was also observed that ALCAM/CD6 engagement made a positive contribution during specific immune responses. It has also been shown that a fraction of CD6 is physically associated with the TCR/CD3 complex and that CD6 and its ligand ALCAM colocalize with the TCR/CD3 complex at the central supramolecular activation clusters [24]. It is interesting that it has been shown in a more recently published work that ALCAM is another cell surface protein expressed by DC upon differentiation from monocytes and mediated adhesion to CD6, and ALCAM and CD6 interactions contribute to stabilization of the immunological synapse [25]. In this study, we examined N-myc downstream-regulated gene 2 (NDRG2) expression in monocyte-derived DC as well as primary DC from lymph nodes, thymus, and skin using antiNDRG2 polyantibodies and mAb and found a DC-specific expression of NDRG2 protein. To assess whether NDRG2 expression affects APC function of DC, U937 and NDRG2transfected U937 cells were evaluated for their surface expression of costimulatory or adhesion molecules. When U937 cells showing NDRG2 overexpression were stimulated with GM-CSF and IL-4, NDRG2 specifically inhibited the cytokine-induced reduction of ALCAM. Last, DC differentiated from NDRG2 small interfering RNA (siRNA)-transfected monocytes exhibited a decreased level of ALCAM expression and showed a decreased ability to stimulate allogeneic T cells. Thus, our data indicate that expression of NDRG2 during the differentiation of DC may contribute to their sustained ability to activate T lymphocytes by preserving the expression of costimulatory molecules.

MATERIALS AND METHODS Reagents for cell culture, cytokines, and antibodies All cultures were performed in RPMI-1640 medium (Sigma Chemical Co., St. Louis, MO, USA), supplemented with L-glutamine (2 mM), penicillin (100 U/ml), streptomycin (100 ␮g/ml), HEPES (10 mM), and 10% FBS (Gibco-BRL, Grand Island, NY, USA). The growth factors used in the primary cultures of DC precursors were recombinant human (rh)GM-CSF (kindly provided by LG LS, Iksan, Republic of Korea) and rhIL-4 (Endogen, Woburn, MA, USA). FITC- or PE-conjugated mAb, specific for CD1a, CD3, CD11a, CD14, CD54, CD80, CD83, CD86, CD166, CD209 [DC-specific ICAM-grabbing nonintegrin (SIGN)], HLA-A,B,C, and HLA-DR, were purchased from BD Biosciences (San Jose, CA, USA).

Generation of DC in vitro Human PBMC of healthy donors (Red Cross Blood Center, Daejeon, Republic of Korea) were isolated by density centrifugation on a Histopaque 1077 (Sigma Chemical Co.). The Red Cross Blood Center Committee (Seoul, Republic of Korea) approved use of these cells. Monocytes were purified by negatively

90

Journal of Leukocyte Biology Volume 83, January 2008

depleting T, B, and NK cells, erythrocytes, and granulocytes using mouse antibody-reactive immunomagnetic beads (Dynal, Oslo, Norway). In brief, anti-CD2-, -CD7-, -CD16-, -CD19-, -CD56-, and -CD235a-labeled PBMC were incubated with the immunomagnetic beads for 30 min at 4ºC with gentle rotation, and positive cells were removed using a Dynal magnet. Purified monocytes were found to be ⬎90% positive for the CD14 marker. Immature DC were generated by culturing monocytes in human DC medium containing GM-CSF (400 ng/ml) and IL-4 (20 ng/ml) in a 12-well plate at a concentration of 5 ⫻ 105 cells per well.

Preparation of NDRG2 antibody Mouse polyclonal antibodies and NDRG2 mAb were generated using recombinant protein consisting of amino acids 150 –357 of NDRG2 (NM_201541). In brief, NDRG2 C terminus cDNA (corresponding to 150 –357 aa) was cloned into the pET28a vector by PCR and restriction enzyme digestion methods. The cloned pET28a-NDRG2-C plasmid was transformed into BL21 DE3 to produce recombinant protein production. Recombinant NDRG2-C protein expression was induced by the addition of isopropylthiogalactoside to a final concentration of 1 mM at 37°C for 4 h. The recombinant NDRG2-C protein extraction and purification were performed according to the protocol for the QIAexpressionist system (Qiagen, Hilden, Germany). The eluted recombinant NDRG2-C protein was mixed with Freund’s adjuvant, and the protein/adjuvant complex was injected into BALB/c mice. During boosting, anti-NDRG2 antibody production was examined by ELISA. After the fifth boosting, antisera were collected for polyclonal antibody and transferred to microtubes for subsequent experiments. NDRG2 mAb was produced using BALB/c mice and a myeloma cell line, P3/NS/1-Ag4-1. Spleen cells of mice immunized with recombinant NDRG2-C protein were harvested, and roughly 50 – 60% of all cells were placed in culture after fusion. Cell culture supernatants from selected cell lines were tested by ELISA. Among the growing clones, which tested positive, the 19C-4 clone (isotype IgG1) was used for further study. mAb was produced in ascites from BALB/c mice injected with 19C-4 cells.

Immunohistochemistry For immunohistochemistry, tissues were fixed routinely in 10% buffered formalin and embedded in paraffin. Sections (3 ␮m) were cut from the paraffin blocks and mounted on 3-amino-propyltriethoxysilane-coated slides. After deparaffinization and antigen retrieval using a pressure cooker with 10 mM sodium citrate buffer (pH 6.0) at full power for 4 min, primary antibody raised against NDRG2 was diluted at 1/3200 with background-reducing diluent (Dako, Carpinteria, CA, USA). Immunostaining using antibodies against NDRG2, S-100 protein (Dako, 1/400 dilution), and CD1a (Dako, 1/50 dilution) was performed using the mouse EnVision kit (Dako). After preincubation with blocking serum for 15 min, the primary antibody was applied and incubated for 30 min in a humid chamber and then washed with PBS. Slides were incubated for 30 min with the EnVision peroxidase reagent (Dako) and were then incubated sequentially with 3,3-diaminobenzidine chromogen for 5 min. The slides were counterstained with Meyer’s hematoxylin and mounted. Careful rinses with several changes of PBS were performed between each stage of the procedure. Negative controls were used by excluding the primary antibody or by using preimmune IgG1.

Western blot analysis Cells were homogenized on ice in Pro-PREPTM protein extraction solution (iNtRon Biotechnology, Seongnam, Republic of Korea) containing protease inhibitor, 1 mM PMSF, 1 mM EDTA, 1 ␮M pepstatin A, 1 ␮M leupeptin, and 0.1 ␮M aprotinin. After 30 min on ice, the extracts were centrifuged (13,000 g) for 10 min at 4ºC, and the supernatants were recovered. The protein concentration was measured (Bio-Rad, Hercules, CA, USA), and ⬃30 ␮g of each extract was loaded for 12% SDS-PAGE gel and was then transferred from the gel onto a polyvinylidene difluoride membrane (Amersham Biosciences, Little Chalfont, UK). The nonspecific antibody-binding sites on the membrane were blocked by incubation of the membrane in PBS (pH 7.0) containing 0.1% Tween 20 and 4% nonfat dry milk for 1 h at room temperature. The membrane was washed in PBS containing 0.1% Tween 20 and incubated overnight at 4ºC with NDRG2 antibody. After washing for 1 h, the membrane was incubated with HRP-conjugated secondary antibody (Sigma Chemical Co.) for 1 h at room

http://www.jleukbio.org

temperature. The resulting blot was visualized by ECL-Plus Western detection reagents (Amersham Biosciences).

Confocal laser scan microscopy (CLSM) Monocyte-derived DC were grown on 12-mm diameter glass coverslips coated with poly-L-lysin in 12-well plates. Cells were fixed with 4% paraformaldehyde for 20 min at room temperature. After washing with PBS, blocking was completed with 1% BSA (Sigma Chemical Co.) in PBS for 30 min at room temperature. Cells were incubated with mouse anti-NDRG2 polyclonal antibody, followed by incubation with tetramethyl rhodamine isothiocyanate-conjugated secondary antibody and FITC-conjugated HLA-DR antibody. Cells were then treated with Hoechst 33342 (Molecular Probes, Eugene, OR, USA) in PBS for 10 min and washed with 0.3% Triton X-100 in PBS. Coverslips, which held cells, were mounted onto cover glasses with Vectashield mounting medium, and the slides were kept in darkness prior to examination. Confocal images were obtained using a Zeiss LSM 510 META laser scanning microscope (Carl Zeiss, Jena, Germany).

Construction and transfection of NDRG2 in U937 cells The NDRG2 cDNA region containing the entire open reading frame was amplified by PCR with primers (5⬘-ATGGCGGAGCTGCAGG-3⬘ and 3⬘TCAACAGGAGACCTCCAT-5⬘). The PCR products were cloned into topoisomerase plasmid (Invitrogen, Carlsbad, CA, USA), and the nucleotide sequence was determined by automatic sequencing. After digestion with XhoI and BamHI, NDRG2 was subcloned into pcDNA3.1 (Invitrogen). For stable expression of NDRG2, U937 cells were transfected with pcDNA3.1-NDRG2 by electroporation with a Gene Pulser (Bio-Rad) at 960 ␮F and 300 V, resulting in U937-NDRG2. After stable transfection, U937 cells were maintained in culture medium containing 1 mg/ml Geneticin disulfate salt (G418, Sigma Chemical Co.). Two weeks after the transfection, single cells were plated in 96-well plates (0.5 cell/well) for growth. Expression of NDRG2 in cell clones was confirmed by Northern and Western blotting.

Transfection of siRNA in monocytes and DC A mixture of four siRNAs (SMARTPool) specific for NDRG2 or ALCAM and control SMARTPool siRNA were purchased from Dharmacon (Lafayette, CO, USA). siRNAs were introduced into monocytes and DC using a monocyte or DC nucleofection kit (Amaxa, Cologne, Germany), according to the instructions of the manufacturer. In brief, 1 ␮g SMARTPool siRNA was mixed with 0.1 ml cell suspension, transferred to a 2-mm electroporation cuvette, and nucleofected with a NucleofectorTM apparatus (Amaxa), as described. After electroporation, cells were transferred immediately to 3 ml-supplemented medium or DC medium and were then cultured in six-well plates. Using pmaxGFP (Amaxa), transfection efficiency was evaluated in monocytes or DC after 24 h of transfection. Using cytometric analysis, GFP expression was detected in 70 –75% of the monocytes and in 25–29% of DC (data not shown).

RT-PCR and Northern blot analysis Total RNA was extracted from the cultured cells using the acid guanidinium thiocyanate-phenol-chloroform extraction method [26]. First-strand cDNA was synthesized from total RNA under RNase-free conditions. The reaction was performed with 10 ␮g total RNA using a ProSTARTM first-strand RT-PCR kit (Stratagene, La Jolla, CA, USA), according to the instructions of the manufacturer. PCR was performed in a GeneAmp PCR system 2700 (Perkin-Elmer/ Centus, Norwalk, CT, USA) using the following first-strand cDNA and Taq polymerase (TaKaRa Shuzo, Kyoto, Japan) for each primer: 5⬘-GCCATGTACGTTGCTATCCAGGCTG-3⬘ and 3⬘-AGCCGTGGCCATCTCTTGCTCGAAG-5⬘ for ␤-actin, 5⬘-AATGGCCCTTGTTGCCCT-3⬘ and 3⬘-TCCTTCCCCACACTCGTT-5⬘ for NDRG2, and 5⬘-AGTTCCTGCCGTCTGCTCTT-3⬘ and 3⬘-TTTTTCCTTTACCTGGGTCA-5⬘ for ALCAM. PCR-amplified products were separated on 1.5% agarose gels containing 0.1 ␮g/ml ethidium bromide and visualized under UV light. Northern blot analysis was performed according to a method described previously [27]. In brief, total RNA (3–5 ␮g) was separated in a 1% denaturing formaldehyde-agarose gel and transferred to a nylon membrane (Boehringer Mannheim, Mannheim, Germany). After UV fixation, a random primer DNA labeling kit (Roche, Mannheim, Germany) was used to hybridize the membrane at 68ºC in ExpressHybTM solution (Clontech,

Palo Alto, CA, USA) with the cDNA probes labeled with [32p]dCTP (PerkinElmer/Centus). In the case of NDRG2, a cDNA probe was prepared from the RT-PCR product using a specific primer set, 5⬘-ATGGCGGAGCTGCAGG-3⬘ and 3⬘-TCAACAGGAGACCTCCAT-5⬘. The probed membrane was then washed in 2⫻ SSC and 0.1% SDS at room temperature and exposed to autoradiographic film using an intensifying screen for 2–3 days at –70ºC.

Proliferation assay of allogeneic PBL and measurement of IL-2 production Monocyte-depleted PBL (1⫻105 cells), isolated from buffy coats of donor blood, were incubated with graded numbers of irradiated (3000 rad) DC transfected with control or NDRG2- or ALCAM-specific SMARTPool siRNA. Experiments were performed in each well of 96-well round-bottom plates. Cell proliferation was quantified by measuring the thymidine uptake of cells after they had been incubated with 1 ␮Ci [methyl-3H]thymidine (Amersham Pharmacia, Piscataway, NJ, USA) during the last 18 h of the 4-day culture. Cells were harvested onto glass fibers, and radioactivity was measured using a scintillation counter. Results were presented as mean cpm of cultures performed in triplicate. For determining IL-2 production, DC (5⫻105) were incubated with allogeneic PBL (1⫻106), and IL-2 production on days 1–3 was measured from supernatants using an ELISA kit (Endogen).

RESULTS Monocyte-derived DC express the NDRG2 protein We showed previously that NDRG2 mRNA is expressed specifically in DC differentiated from monocytes, CD34⫹ progenitor cells, and MUTZ-3 cells [28]. To confirm the expression of NDRG2 protein in monocyte-derived DC, Western blot analysis was performed. As expected, DC, but not PBMC or monocytes, expressed a 45-kDa NDRG2 protein (Fig. 1A). Moreover, monocytes, which did not show NDRG2 expression at the beginning of the cultures, showed progressively higher NDRG2 expression during the differentiation of DC (Fig. 1B). When the myelomonocytic leukemia cell line MUTZ-3 cells were differentiated to DC by culturing with GM-CSF, IL-4, and TNF-␣, NDRG2 expression increased gradually during differentiation (Fig. 1C). In addition, NDRG2 protein was not detected at all in leukemia, lymphoma, B-lymphocyte, or NK cell lines (Fig. 1D). Taken together, these results indicate that NDRG2 protein is expressed specifically during the differentiation of DC, as shown previously in NDRG2 mRNA expression in DC. To investigate the subcellular localization of NDRG2 in monocyte-derived DC, CLSM analysis was performed. Qu et al. [29] demonstrated that NDRG2, NDRG3, and NDRG4 were cytosolic proteins and that NDRG4 even accumulated in certain organelles of the cell. Our data also showed that NDRG2 protein expression was localized mostly in the cytoplasm, rather than in the nuclei of monocyte-derived DC (Fig. 1E). The plasma membrane and nucleus were stained with the HLA-DR antibody and Hoechst, respectively.

NDRG2 is expressed specifically in primary DC in human lymph node, thymus, and skin To investigate the expression pattern of NDRG2 in human tissues, we performed immunohistochemistry against NDRG2 using normal human thymus, reactive lymph nodes, and skin. In addition, several pathologic lesions were studied for the expression of this

Choi et al. Specific expression and function of NDRG2 in DC

91

Fig. 1. NDRG2 protein is expressed specifically in human DC. (A–D) Cell lysate was prepared from human cell lines, MUTZ-3 DC, PBMC, primary monocytes, and monocyte-derived DC. Using antiNDRG2 mAb, NDRG2 protein expression was determined by Western blot analysis. (E) Analysis of the cellular distribution of NDRG2 in monocyte-derived DC by CLSM. DC were stained with Hoechst 33342, FITC-conjugated HLA-DR mAb, and NDRG2 polyclonal antibody, respectively.

molecule. In a reactive lymph node, NDRG2 was expressed characteristically in interdigitating DC (IDC), and lymphocytes and follicular DC were found to be negative for NDRG2 expression (Fig. 2A). In the thymus, the expression of NDRG2 was

noted in DC and thymic epithelial cells, including Hassall’s corpuscles (Fig. 2B). Its expression was also demonstrated in the epidermal Langerhans cells and dermal DC in the skin (dendrocytes; Fig. 3A, upper). Expression of NDRG2 in

Fig. 2. Immunohistochemistry for NDRG2 expression in the human lymph node and thymus. (A) In lymph nodes, NDRG2 was expressed specifically in IDC, and all lymphocytes and follicular DC were found to be nonreactive. Upper left and right (100⫻ original magnification), lower left (200⫻ original magnification), lower right (400⫻ original magnification). Arrows indicate typical positive staining in DC. (B) In the normal thymus, the expression of NDRG2 was noted in DC and thymic epithelial cells including Hassall’s corpuscles. M, Medullar; C, Cortex. Upper left (200⫻ original magnification), upper right, lower left, and lower right (400⫻ original magnification). Arrows indicate typical positive staining in DC.

92

Journal of Leukocyte Biology Volume 83, January 2008

http://www.jleukbio.org

pathologic lesions such as dermatopathic lymphadenitis was confirmed additionally but not in tuberculosis (Fig. 3A, lower). When we prepared 3 ␮m serial sections to confirm the expression of NDRG2, IDC of the lymph node showed coexpression of NDRG2 and S-100 protein (Fig. 3B, top). Epidermal Langerhans cells and LCH showed coexpression of NDRG2 and CD1a in staining of sequential sections (Fig. 3B, middle and bottom). In LCH, most of the tumor cells were strongly reactive for our NDRG2 antibody (Fig. 3B, bottom). When the immunostaining was performed for DCSIGN, it was distributed mainly in the sinusoidal DC of reactive lymph nodes (data not shown). These results confirm that NDRG2 protein is expressed specifically in IDC of lymph nodes and Langerhans cells.

NDRG2 prevents GM-CSF and IL-4-induced down-regulation of ALCAM in U937 cells To address NDRG2 function in cell differentiation, U937 cells were transfected with full-length, wild-type NDRG2 cDNA to generate the stable U937-NDRG2 transfectant. Northern blot hybridization revealed expression of the NDRG2 transcript with an mRNA size smaller than the endogenous NDRG2 transcript in DC (Fig. 4A). The smaller size of NDRG2 mRNA represents that non-coding region of NDRG2 plasmid’s probably smaller than that of endogenous NDRG2 transcript. The expression level of NDRG2 protein in several U937-NDRG2 clones was compared with that of the U937-mock transfectant, and three clones (numbers 45, 61, and 91), which strongly expressed NDRG2 protein, were selected (Fig. 4B). Although no significant difference in morphology was

observed between the U937-NDRG2 transfectant and U937-mock transfectant, the U937-NDRG2 transfectant showed a slightly reduced cell proliferation rate (data not shown), as reported previously [30]. To examine the response for cytokines, U937NDRG2 transfectants were cultured under the same conditions as those used for DC generation from monocytes. It is interesting that as shown in Figure 4C, ALCAM expression was inhibited completely, particularly in the U937-mock transfectant, but not in U937-NDRG2 transfectants. ALCAM reduction at the mRNA level in the U937-mock transfectant was also observed 2 days after culture in GM-CSF and IL-4 (Fig. 4E). In a previous report, it was shown that GM-CSF was able to increase expression of CD11b, a myeloid-specific adhesion molecule, in U937 cells [31]. When U937-mock and U937-NDRG2 transfectants were stimulated with GM-CSF and IL-4, we found no difference in CD11b expression between the two cell types (data not shown). We next examined the time courses of ALCAM reduction in the U937mock transfectant by GM-CSF and IL-4. A significant reduction in ALCAM expression was obtained after 2 days, and the expression was inhibited completely after 4 days (Fig. 4D). These results, therefore, provide evidence that NDRG2 expression might prevent ALCAM reduction through cultivation with cytokines, GM-CSF and IL-4.

Inhibition of NDRG2 decreases ALCAM expression during DC differentiation As reported previously that ALCAM (CD166) was expressed in activated leukocytes [21] and monocytes [15], ALCAM expression was detected in leukocytes, including monocytes, macrophages,

Fig. 3. Expression of NDRG2 in the skin DC and lymphadenitis (A) and comparison of NDRG2 expression with other DC marker expression by staining of sequential sections (B). (A) In the skin, NDRG2 expression was demonstrated in the epidermal Langerhans cells (upper left, 200⫻ original magnification) and dermal DC (upper right, 200⫻ original magnification). Arrows indicate typical positive staining in Langerhans cells and dermal DC. The actively proliferating IDC in dermatopathic lymphadenitis (lower right, 100⫻ original magnification) were also positive, and epithelioid histiocytes of tuberculosis were negative (lower left, 200⫻ original magnification). (B) In parallel with NDRG2 expression, paracortex of lymph node in 3 ␮m serial sections was stained with anti-S-100 antibody (top, 400⫻ original magnification). Expression of NDRG2 and CD1a in serial sections was compared in epidermal Langerhans cells (middle, 400⫻ original magnification) and Langerhans cell histiocytosis (LCH; bottom, 400⫻ original magnification).

Choi et al. Specific expression and function of NDRG2 in DC

93

Fig. 4. NDRG2 inhibits the GM-CSF/IL-4-induced ALCAM reduction in U937 cells. (A) U937 cells were transfected with pcDNA3.1 or pcDNA3.1-NDRG2, and NDRG2 expression was detected by Northern blotting. (B) Several clones overexpressing NDRG2 were selected by G418 (1 mg/ml), and NDRG2 protein expression in the clones and DC was detected by immunoblotting with anti-NDRG2 mouse mAb. (C) U937-mock and three U937-NDRG2 clones were cultured with or without GM-CSF (400 ng/ml) and IL-4 (20 ng/ml) for 4 days, and ALCAM expression was analyzed by flow cytometry. (D) U937-mock and U937-NDRG2 (clone 61) were cultured in the presence of GM-CSF and IL-4 for the times indicated and stained with PE-conjugated ALCAM mAb (BD Biosciences) for flow cytometric analysis. (E) ALCAM mRNA expression was detected 2 days after culture in the absence or presence of GMCSF and IL-4 by RT-PCR. M, Mock; N, NDRG2.

DC, and B cells, but it was not expressed in T lymphocytes (data not shown). We next examined the specificity of NDRG2 siRNAinduced gene silencing in monocytes, which were subsequently induced to differentiate into DC. They showed a significant decrease in the induction of NDRG2 transcript and protein (Fig. 5, A and B). In addition, although the transfection efficiency using NDRG2 siRNA was low, a decrease in NDRG2 transcript and protein levels was also observed after direct transfection of DC with NDRG2 siRNA (data not shown), indicating that NDRG2specific SMARTPool siRNA-mediated NDRG2 silencing was specific for NDRG2-expressing DC. To investigate ALCAM expression in NDRG2 siRNA-transfected, monocyte-derived DC and NDRG2 siRNA-transfected DC, an RT-PCR specific for ALCAM was performed. As expected, siRNA transfection decreased the expression of NDRG2 mRNA in siRNA-transfected, monocyte-derived DC and siRNA-transfected DC. It is interesting that ALCAM mRNA expression was also suppressed in siRNA-transfected, monocyte-derived DC (Fig. 5C), whereas it was not decreased in siRNA-transfected DC (data 94

Journal of Leukocyte Biology Volume 83, January 2008

not shown). We next addressed whether NDRG2 siRNA affects DC differentiation directly. Monocytes were transfected with NDRG2 siRNA, and differentiation was then induced with GMCSF and IL-4. DC differentiation was assessed by analyzing expression levels of CD1a, CD14, CD80, CD83, CD86, HLAABC, and HLA-DR. Neither control siRNA- nor NDRG2 siRNAtransfected, monocyte-derived DC showed changes in surface expression of most DC-related antigens. They expressed high levels of CD1a, CD80, HLA-ABC, and HLA-DR (Fig. 5D) but did not express CD14 and CD83 antigens (data not shown). These results, therefore, provide evidence that NDRG2 expression may prevent the reduction of ALCAM expression or may be able to prolong ALCAM expression in an early stage of DC differentiation from monocytes.

NDRG2 down-regulation affects the T cell stimulatory ability of DC ALCAM has been demonstrated to interact in a homophilic manner in cis or in trans [18, 32] or in an heterophilic manner http://www.jleukbio.org

did not change the expression level of DC-SIGN (CD209) that may be involved in interactions between T cells and DC. NDRG2 expression, however, was not affected in DC differentiated from ALCAM siRNA-transfected monocytes (Fig. 6B). Furthermore, ALCAM siRNA transfection in monocytes did not affect DC differentiation (data not shown). While the interference of NDRG2 expression with siRNA during DC differentiation did not affect CD54 expression on DC1a-positive DC or CD11a on CD3-positive T cells in the mixed lymphocyte reaction, it clearly decreased allogeneic T cell proliferation (Fig. 6, C and D). Interestingly, NDRG2 siRNA transfection induced more effective reduction of allogeneic T cell proliferation than ALCAM siRNA transfection, indicating the possibility that NDRG2 inhibition may induce diverse effects on DC function other than ALCAM reduction. However, as previously reported that ALCAM is essential for T cell proliferation [25], DC transfected with ALCAM siRNA also showed a decreased induction in the T cell proliferation. Similarly, these findings were also confirmed in IL-2 level produced by allogeneic T cells (Fig. 6E). No significant difference in the cytokine production such as IL-12 and IL-10 between NDRG2 or ALCAM siRNA- and control siRNA-transfected DC was observed under these coculture conditions (data not shown). These results therefore suggest that although the involvement of soluble factors in the low level of stimulation may not be excluded completely, inhibition of NDRG2 during DC differentiation at least down-regulates ALCAM expression, which influences the ability of DC to stimulate allogeneic T cell proliferation.

DISCUSSION

Fig. 5. Inhibition of NDRG2 in monocytes decreases ALCAM expression during DC differentiation. Monocytes were transfected with control or NDRG2 siRNA using a monocyte nucleofection kit (Amaxa). After differentiation into DC with GM-CSF and IL-4, inhibition of NDRG2 was analyzed by Northern blotting (A) on day 6 and immunoblotting (B) on days 3 and 6. C, Control; N, NDRG2. (C) ALCAM expression on day 6 was detected in DC, which were differentiated from control (Cont) or NDRG2 siRNA (NDRG2)-transfected monocytes using RT-PCR. The relative values of PCR products were calculated as follows: (mean intensity⫻band area) of NDRG2 or ALCAM/(mean intensity⫻band area) of ␤-actin. (D) After DC had differentiated from monocytes transfected with control or NDRG2 siRNA, cells were analyzed for surface antigen expression by immunostaining with their respective antibodies by flow cytometry on days 3 and 6 (thin line, control SMARTPool siRNA; bold line, NDRG2-specific siRNA).

with CD6 mediated through CD6d3 and the N-terminal domain of ALCAM [14]. We hypothesized that NDRG2 siRNA transfection affects the allostimulatory activity of DC, as the inhibition of NDRG2 during DC differentiation decreases ALCAM expression. To compare with DC, which have a decreased ALCAM expression, reduction of ALCAM expression was induced directly in monocytes using ALCAM-specific siRNA transfection. As predicted, ALCAM expression was decreased during DC differentiation of monocytes by a transfection with not only NDRG2-specific siRNA, but also ALCAM-specific siRNA (Fig. 6A). NDRG2 or ALCAM silencing in monocytes

In the present study, we identified that the NDRG2 protein is strongly expressed in DC, including monocyte- and myelomonocytic leukemia cell-derived DC. It is also strongly expressed in lymph node, thymus, and skin DC in vivo. It was observed that NDRG2 protein localized mostly to the cytoplasm. The NDRG2 isoform, which has an insertion sequence of 14 amino acids, was also detected in the cytoplasm (data not shown). In contrast to the positive staining of DC in lymphoid organs and skin, plasmacytoid monocytes associated with Kikuchi’s lymphadenitis were not stained when treated with our NDRG2 antibody, suggesting the possibility for specific expression of NDRG2 in type 1 DC (data not shown). Therefore, we found that our antibody specific for NDRG2 is a useful additional marker for the identification of IDC, dermal DC, Langerhans cells, and their pathologic lesions. NDRG2 belongs to the NDRG family, a new family of differentiation-related genes. This family is comprised of four members, NDRG1–NDRG4 [33]. NDRG2 is highly expressed in the adult brain, salivary gland, and skeletal muscle [29, 34]. NDRG2 cDNAs, corresponding to four isoforms (Ndrg2a1, Ndrg2a2, Ndrg2b1, and Ndrg2b2), which differ in terms of their 5⬘-untranslated and N-terminal coding regions, have been reported in rat renal tissue [34]. NDRG2 has been implicated in cell growth [30], differentiation [28], and apoptosis [35] and is rapidly responsive to mineralcorticoid stimuli in the kidney [34]. Most recently, it has been shown that its inactivation is involved in the development of aggressive forms of cancer [36].

Choi et al. Specific expression and function of NDRG2 in DC

95

Fig. 6. DC derived from NDRG2 siRNA-transfected monocytes show a decreased ability to induce allogeneic T cell proliferation. (A) Down-regulation of ALCAM 3 days after induction of DC differentiation from control siRNA- (filled histogram) and NDRG2 siRNA- or ALCAM siRNA-transfected (open histogram) monocytes. Cells were analyzed for surface antigen expression by immunostaining with their respective antibodies by flow cytometry. Numbers indicate mean fluorescence intensity (upper; filled histogram, lower; open histogram). (B) ALCAM siRNA transfection into monocytes does not affect the NDRG2 expression level of DC. After induction of DC differentiation from control, NDRG2, or ALCAM siRNA-transfected monocytes, NDRG2 expression in DC was detected by immunoblotting. (C) After induction of DC differentiation from control (filled histogram) or NDRG2 siRNA-transfected monocytes (open histogram), DC-SIGN expression was detected by flow cytometry. Expression of CD54 and CD11a is not affected by NDRG2 or ALCAM siRNA transfection. After DC differentiation from control, NDRG2, or ALCAM siRNA-transfected monocytes, DC (5⫻105) were incubated with allogeneic PBL (1⫻106) for 1 day. ICAM (CD54) expression was measured on CD1a-positive cells (R1), whereas LFA-1 (CD11a) expression was measured on CD3-positive cells (R2) by using flow cytometer (filled histogram, control SMARTPool siRNA; open histogram, NDRG2- or ALCAM-specific SMARTPool siRNA). (D) Different numbers of DC derived from monocytes transfected with control, NDRG2, or ALCAM siRNA were incubated with allogeneic PBL (1⫻105) for 4 days. Cell proliferation was measured by the incorporation of radioactive thymidine using a scintillation counter. Data are representative of at least two independent experiments. Mean ⫾ SEM of triplicates is shown (Student’s t-test; *, P⬍0.05, compared with control siRNA). (E) DC (5⫻105) were incubated with allogeneic PBL (1⫻106), and IL-2 production on days 1, 2, and 3 was measured from supernatants using the ELISA kit. Results are means ⫾ SD of two independent experiments (Student’s t-test; *, P⬍0.05; **P⬍0.5, compared with control siRNA).

96

Journal of Leukocyte Biology Volume 83, January 2008

http://www.jleukbio.org

In this study, we demonstrated that interference of NDRG2 expression induced a down-regulation of ALCAM expression, and overexpression of NDRG2 strongly prevented cytokineinduced ALCAM reduction in monocytic cell lines. In parallel with this observation, NDRG2 siRNA transfection decreased the ability of DC to stimulate allogenic T cell proliferation. Unfortunately, it was not possible to compare the effect of siRNA directly with the neutralizing antibody capable of binding to ALCAM, as the commercially available form of the anti-ALCAM antibody was not effective for interfering with the ALCAM-CD6 interaction. However, it is worth noting that the inhibition of NDRG2 expression does not affect the DC differentiation phase and the morphological changes, which occur during their differentiation, although it seems to induce minimal changes in the growth rate of the NDRG2-overexpressing U937 cell line (data not shown). Originally, based on reports that NDRG2 is implicated in cell differentiation and growth, we expected that the inhibition of its expression could effectively result in the inhibition of differentiation. This was not the case, however, although RNA interference could not induce complete inhibition of its expression. Thus, our observations indicate that NDRG2 expression in DC may be involved in DC-related functions. ALCAM is a member of a small subgroup of transmembrane glycoproteins, which belong to the Ig superfamily. ALCAM is expressed on a wide variety of cells and mediates homotypic and heterotypic cell-cell clustering through homophilic (ALCAM-ALCAM) and heterophilic (ALCAM-CD6) interaction [14, 22, 37]. However, it has been shown that the homophilic interaction has 100 times lower affinity than does the heterophilic interaction [23]. Gimferrer et al. [24] reported that in image analysis of antigen-specific T-APC conjugates, CD6 and its ligand (ALCAM) colocalize with TCR/CD3 at the center of the immunological synapse and that a soluble recombinant CD6 form reduces the number of mature antigen-specific TAPC conjugates significantly. Therefore, these results demonstrate the important role played by the interaction of ALCAM with CD6 during T cell activation and proliferation processes. In a more recently published work, it has been demonstrated that ALCAM-CD6 interactions are involved in the stabilization of DC-T cell contact and the proliferative phase of the T cell response [25]. Moreover, it shows that ALCAM and CD6 are actively recruited at the DC-T cell interface and localize all along the contact site. These findings indicate that the downregulation of ALCAM expression in DC may profoundly affect the ability of DC to stimulate T cells. Therefore, it will be interesting to examine whether NDRG2 expression in APC is involved in the activation of CD6. It is interesting that NDRG2 inhibition in monocyte-derived DC was more effective in inhibiting allogenic T cell proliferation than ALCAM inhibition, whereas the transfection of NDRG2 siRNA directly into DC did not result in the reduction of ALCAM or the inhibition of T cell proliferation (data not shown), although ALCAM inhibition still induced significant down-regulation of proliferation. It is therefore worth considering that the inhibition of NDRG2 during DC differentiation might modulate other DC functions such as cytokine production. In fact, although we were not able to detect any significant difference of IL-12 or IL-10 production in DC-T cell cocultures, NDRG2-transfected U937 cells, when

stimulated with GM-CSF and IL-4, could secrete IL-10 at a lower level than wild-type U937 cells (unpublished data). Therefore, we do not exclude the possibility that the effect of NDRG2 down-regulation on allogeneic T-cell stimulation may not be mediated solely by ALCAM reduction. In conclusion, this work confirms and expands earlier observations of the specific expression of NDRG2 in differentiated cell lineages on a protein level and suggests that NDRG2 expression in DC may play a role in maintaining their ability to activate T lymphocytes by preserving the expression of costimulatory molecules.

ACKNOWLEDGMENTS This work was supported by Grants R11-2005-017-03001 of the Research Center for Women’s Diseases of Korean Science and Engineering Foundation, KRF-2006-311-E00586 from the Korea Research Foundation, and 05092-CEL-326 from the Korean Food and Drug Administration, Republic of Korea.

REFERENCES 1. Banchereau, J., Briere, F., Caux, C., Davoust, J., Lebecque, S., Liu, Y. J., Pulendran, B., Palucka, K. (2000) Immunobiology of dendritic cells. Annu. Rev. Immunol. 18, 767– 811. 2. Bromley, S. K., Burack, W. R., Johnson, K. G., Somersalo, K., Sims, T. N., Sumen, C., Davis, M. M., Shaw, A. S., Allen, P. M., Dustin, M. L. (2001) The immunological synapse. Annu. Rev. Immunol. 19, 375–396. 3. Valitutti, S., Muller, S., Dessing, M., Lanzavecchia, A. (1996) Signal extinction and T cell repolarization in T helper cell-antigen-presenting cell conjugates. Eur. J. Immunol. 26, 2012–2016. 4. Janeway Jr., C. A., Bottomly, K. (1996) Responses of T cells to ligands for the T-cell receptor. Semin. Immunol. 8, 108 –115. 5. Sharpe, A. H. (1995) Analysis of lymphocyte costimulation in vivo using transgenic and ‘knockout’ mice. Curr. Opin. Immunol. 7, 389 –395. 6. Tarakhovsky, A., Kanner, S. B., Hombach, J., Ledbetter, J. A., Muller, W., Killeen, N., Rajewsky, K. (1995) A role for CD5 in TCR-mediated signal transduction and thymocyte selection. Science 269, 535–537. 7. Azzam, H. S., Grinberg, A., Lui, K., Shen, H., Shores, E. W., Love, P. E. (1998) CD5 expression is developmentally regulated by T cell receptor (TCR) signals and TCR avidity. J. Exp. Med. 188, 2301–2311. 8. Chan, S., Waltzinger, C., Tarakhovsky, A., Benoist, C., Mathis, D. (1999) An influence of CD5 on the selection of CD4-lineage T cells. Eur. J. Immunol. 29, 2916 –2922. 9. Singer, N. G., Fox, D. A., Haqqi, T. M., Beretta, L., Endres, J. S., Prohaska, S., Parnes, J. R., Bromberg, J., Sramkoski, R. M. (2002) CD6: expression during development, apoptosis and selection of human and mouse thymocytes. Int. Immunol. 14, 585–597. 10. Gangemi, R. M., Swack, J. A., Gaviria, D. M., Romain, P. L. (1989) Anti-T12, an anti-CD6 monoclonal antibody, can activate human T lymphocytes. J. Immunol. 143, 2439 –2447. 11. Bott, C. M., Doshi, J. B., Morimoto, C., Romain, P. L., Fox, D. A. (1993) Activation of human T cells through CD6: functional effects of a novel anti-CD6 monoclonal antibody and definition of four epitopes of the CD6 glycoprotein. Int. Immunol. 5, 783–792. 12. Burns, F. R., von Kannen, S., Guy, L., Raper, J. A., Kamholz, J., Chang, S. (1991) DM-GRASP, a novel immunoglobulin superfamily axonal surface protein that supports neurite extension. Neuron 7, 209 –220. 13. Peduzzi, J. D., Irwin, M. H., Geisert Jr., E. E. (1994) Distribution and characteristics of a 90 kDa protein, KG-CAM, in the rat CNS. Brain Res. 640, 296 –307. 14. Bowen, M. A., Aruffo, A. A., Bajorath, J. (2000) Cell surface receptors and their ligands: in vitro analysis of CD6-CD166 interactions. Proteins 40, 420 – 428. 15. Levesque, M. C., Heinly, C. S., Whichard, L. P., Patel, D. D. (1998) Cytokine-regulated expression of activated leukocyte cell adhesion molecule (CD166) on monocyte-lineage cells and in rheumatoid arthritis synovium. Arthritis Rheum. 41, 2221–2229.

Choi et al. Specific expression and function of NDRG2 in DC

97

16. Uchida, N., Yang, Z., Combs, J., Pourquie, O., Nguyen, M., Ramanathan, R., Fu, J., Welply, A., Chen, S., Weddell, G., Sharma, A. K., Leiby, K. R., Karagogeos, D., Hill, B., Humeau, L., Stallcup, W. B., Hoffman, R., Tsukamoto, A. S., Gearing, D. P., Peault, B. (1997) The characterization, molecular cloning, and expression of a novel hematopoietic cell antigen from CD34⫹ human bone marrow cells. Blood 89, 2706 –2716. 17. Cortes, F., Deschaseaux, F., Uchida, N., Labastie, M. C., Friera, A. M., He, D., Charbord, P., Peault, B. (1999) HCA, an immunoglobulin-like adhesion molecule present on the earliest human hematopoietic precursor cells, is also expressed by stromal cells in blood-forming tissues. Blood 93, 826 – 837. 18. Nelissen, J. M., Torensma, R., Pluyter, M., Adema, G. J., Raymakers, R. A., van Kooyk, Y., Figdor, C. G. (2000) Molecular analysis of the hematopoiesis supporting osteoblastic cell line U2-OS. Exp. Hematol. 28, 422– 432. 19. Stephan, J. P., Bald, L., Roberts, P. E., Lee, J., Gu, Q., Mather, J. P. (1999) Distribution and function of the adhesion molecule BEN during rat development. Dev. Biol. 212, 264 –277. 20. Ohneda, O., Ohneda, K., Arai, F., Lee, J., Miyamoto, T., Fukushima, Y., Dowbenko, D., Lasky, L. A., Suda, T. (2001) ALCAM (CD166): its role in hematopoietic and endothelial development. Blood 98, 2134 –2142. 21. Bowen, M. A., Patel, D. D., Li, X., Modrell, B., Malacko, A. R., Wang, W. C., Marquardt, H., Neubauer, M., Pesando, J. M., Francke, U., et al. (1995) Cloning, mapping, and characterization of activated leukocyte-cell adhesion molecule (ALCAM), a CD6 ligand. J. Exp. Med. 181, 2213– 2220. 22. Degen, W. G., van Kempen, L. C., Gijzen, E. G., van Groningen, J. J., van Kooyk, Y., Bloemers, H. P., Swart, G. W. (1998) MEMD, a new cell adhesion molecule in metastasizing human melanoma cell lines, is identical to ALCAM (activated leukocyte cell adhesion molecule). Am. J. Pathol. 152, 805– 813. 23. Hassan, N. J., Barclay, A. N., Brown, M. H. (2004) Frontline: Optimal T cell activation requires the engagement of CD6 and CD166. Eur. J. Immunol. 34, 930 –940. 24. Gimferrer, I., Calvo, M., Mittelbrunn, M., Farnos, M., Sarrias, M. R., Enrich, C., Vives, J., Sanchez-Madrid, F., Lozano, F. (2004) Relevance of CD6-mediated interactions in T cell activation and proliferation. J. Immunol. 173, 2262–2270. 25. Zimmerman, A. W., Joosten, B., Torensma, R., Parnes, J. R., van Leeuwen, F. N., Figdor, C. G. (2006) Long-term engagement of CD6 and ALCAM is essential for T-cell proliferation induced by dendritic cells. Blood 107, 3212–3220. 26. Chomczynski, P., Mackey, K. (1995) Substitution of chloroform by bromochloropropane in the single-step method of RNA isolation. Anal. Biochem. 225, 163–164.

98

Journal of Leukocyte Biology Volume 83, January 2008

27. Choi, S., Kobayashi, M., Wang, J., Habelhah, H., Okada, F., Hamada, J., Moriuchi, T., Totsuka, Y., Hosokawa, M. (2000) Activated leukocyte cell adhesion molecule (ALCAM) and annexin II are involved in the metastatic progression of tumor cells after chemotherapy with Adriamycin. Clin. Exp. Metastasis 18, 45–50. 28. Choi, S. C., Kim, K. D., Kim, J. T., Kim, J. W., Yoon, D. Y., Choe, Y. K., Chang, Y. S., Paik, S. G., Lim, J. S. (2003) Expression and regulation of NDRG2 (N-myc downstream regulated gene 2) during the differentiation of dendritic cells. FEBS Lett. 553, 413– 418. 29. Qu, X., Zhai, Y., Wei, H., Zhang, C., Xing, G., Yu, Y., He, F. (2002) Characterization and expression of three novel differentiation-related genes belong to the human NDRG gene family. Mol. Cell. Biochem. 229, 35– 44. 30. Deng, Y., Yao, L., Chau, L., Ng, S. S., Peng, Y., Liu, X., Au, W. S., Wang, J., Li, F., Ji, S., Han, H., Nie, X., Li, Q., Kung, H. F., Leung, S. Y., Lin, M. C. (2003) N-Myc downstream-regulated gene 2 (NDRG2) inhibits glioblastoma cell proliferation. Int. J. Cancer 106, 342–347. 31. Okuma, E., Inazawa, Y., Saeki, K., Yuo, A. (2002) Potential roles of extracellular signal-regulated kinase but not p38 during myeloid differentiation of U937 cells stimulated by cytokines: augmentation of differentiation via prolonged activation of extracellular signal-regulated kinase. Exp. Hematol. 30, 571–581. 32. van Kempen, L. C., Nelissen, J. M., Degen, W. G., Torensma, R., Weidle, U. H., Bloemers, H. P., Figdor, C. G., Swart, G. W. (2001) Molecular basis for the homophilic activated leukocyte cell adhesion molecule (ALCAM)ALCAM interaction. J. Biol. Chem. 276, 25783–25790. 33. Zhou, R. H., Kokame, K., Tsukamoto, Y., Yutani, C., Kato, H., Miyata, T. (2001) Characterization of the human NDRG gene family: a newly identified member, NDRG4, is specifically expressed in brain and heart. Genomics 73, 86 –97. 34. Boulkroun, S., Fay, M., Zennaro, M. C., Escoubet, B., Jaisser, F., BlotChabaud, M., Farman, N., Courtois-Coutry, N. (2002) Characterization of rat NDRG2 (N-Myc downstream regulated gene 2), a novel early mineralocorticoid-specific induced gene. J. Biol. Chem. 277, 31506 –31515. 35. Wu, G. Q., Liu, X. P., Wang, L. F., Zhang, W. H., Zhang, J., Li, K. Z., Dou, K. F., Zhang, X. F., Yao, L. B. (2003) Induction of apoptosis of HepG2 cells by NDRG2. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi 19, 357–360. 36. Lusis, E. A., Watson, M. A., Chicoine, M. R., Lyman, M., Roerig, P., Reifenberger, G., Gutmann, D. H., Perry, A. (2005) Integrative genomic analysis identifies NDRG2 as a candidate tumor suppressor gene frequently inactivated in clinically aggressive meningioma. Cancer Res. 65, 7121–7126. 37. Swart, G. W. (2002) Activated leukocyte cell adhesion molecule (CD166/ ALCAM): developmental and mechanistic aspects of cell clustering and cell migration. Eur. J. Cell Biol. 81, 313–321.

http://www.jleukbio.org