The immunobiology of tumor-infiltrating dendritic cells (DCs) can be strongly influenced by the cytokine environment present in the malignant tissue. We have previously identified discrete melanoma lines, inducing E-cadherin expression on monocyte-derived DCs in vitro. We demonstrate here that this effect, independent of cell contact, is not inducible in the presence of tumor lysates and requires the constitutive expression of IFN stimulated gene 15 (ISG15) by malignant cells.

High-density oligonucleotide arrays were used to investigate the expression pattern of 7000 genes in RNA from two melanoma cell clones competent for E-cadherin induction and two clones devoid of DC-modulating capacity. A total of 13 genes encoding soluble proteins were expressed at higher magnitude in melanomas able to induce E-cadherin expression on DCs. Combining those data with quantitative protein assays, we could narrow our investigation down to three factors: the chemokine CCL5 and the cytokines ISG15 and type I IFNs. Strikingly, >7 ng/ml of ISG15 could be detected in the corresponding melanoma-conditioned medium and induction of E-cadherin on DCs failed in the presence of antibodies neutralizing ISG15 protein. Most importantly, strong cytoplasmic expression of ISG15 was detected by immunohistochemistry in the original tumor specimen from which the melanoma cell lines under investigation were derived.

These data describe a novel property of ISG15 targeting induction of E-cadherin on DCs and possibly influencing their migratory behavior.

DCs3 are potent inducers of immunity, the properties of which can be strongly influenced by the nature of the microenvironment they infiltrate (1). Experimental data indicate that the migratory behavior of mature DCs can be profoundly different, even when their apparent antigen-presenting capacity is similar (2). In particular, homing of mature DCs to secondary lymphoid organs, following the induction of specific chemokine receptors (3, 4), allows priming of naïve T cells. On the other hand, induction of chemokines specifically attracting antigen-specific T cells (5, 6) will enhance effector functions in the periphery and recruitment of memory lymphocytes (7). Derangements of these migratory patterns could crucially influence the outcome of immune responses, including those that are tumor specific (8, 9).

The presence of DCs in malignant tissues has been extensively studied, with conflicting results. For some types of cancers, such as oral (10), ovarian (11), colorectal cancers (12) and renal carcinoma (13), DC infiltration has been associated with prolonged patients survival and reduced metastatic disease, specially if associated with T-cell infiltration (10, 12).

On the other hand, factors secreted by neoplastic cells can compromise the antigen-presenting cell functions of infiltrating DCs, thereby favoring tumor immune escape. In a consistent number of reports, discrete tumor-derived factors have been shown to prevent DC differentiation and maturation and hamper the induction of antitumor immunity (14, 15, 16, 17). Along with these observations, we have previously identified discrete melanoma cell lines inducing E-cadherin expression on monocyte-derived DCs in vitro, potentially impairing their migratory behavior (9).

In the present study, melanoma cell clones endowed with different functional capacities were subjected to gene profiling to identify differentially expressed genes. We provide evidence that defined soluble factors are responsible for the melanoma-induced modulation of DC phenotype.

Media and Reagents for Cell Culture.

Cells were cultured in RPMI 1640 supplemented with 1% Ciproxin (Bayer, Zürich, CH), 2 mml-glutamine, 1% nonessential amino acids, 1% sodium pyruvate, 5 × 10−5m 2-mercaptoethanol and 10% FCS (Life Technologies, Inc., Paisley, Great Britain), thereafter referred to as complete medium. LPS content in serum was tested by Limulus LAL assay, and only LPS-free batches were used. Human recombinant IL-4 was produced in our laboratory. Granulocyte/macrophage-colony stimulating factor was provided by Novartis (Basel, CH).

Cell Cultures.

The original cell line Me67 was generated in our laboratory from a metastatic melanoma. Lines Me67.3, Me67.5, Me67.9, and Me67.10 were derived by culturing the parental cell line in limiting dilution at 0.3 cells/well in 96-well plates. All cell lines were grown in complete medium and were free from Mycoplasma infection, as monitored by specific reverse transcription-PCR. Cell lysates were prepared by three cycles of freeze and thawing.

Generation of DCs from Peripheral Blood Monocytes.

Immature DCs were generated from human peripheral blood mononuclear cells according to published methods (18). Briefly, monocytes were purified by positive sorting using anti-CD14 conjugated microbeads (Milteny I., Bergisch-Gladbach, D). The sorted cells were cultured for 6–7 days in complete medium supplemented with 50 ng/ml granulocyte/macrophage-colony stimulating factor and 1000 units/ml IL-4.

Induction of DC Phenotypic Modulation.

Immature DCs were cocultured with either tumor cells at a 5:1 ratio, in the presence of tumor cell culture supernatant or their lysates (1:2 dilution). In some experiments, purified rabbit IgG anti-ISG15 (19), goat anti-IFNARI, mouse anti-CCR5 IgG1 antibodies, or their isotype matched controls (R&D Systems, Oxon, United Kingdom and BD Biosciences, Heidelberg, Germany), were added to the cultures at a 50 μg/ml final concentration. Cell cultures were analyzed, after 24-h incubation time, by FACS staining.

FACS Analysis.

The DC phenotype was monitored by cell surface staining using FITC-conjugated mouse antibodies from BD Biosciences (Heidelberg, Germany) to human CD86 (clone IT2.2) and CD15 (clone MMA). The mouse antihuman E-cadherin (clone SHE78–7; R&D Systems) antibody was used in combination with a goat antimouse IgG2a FITC-conjugated (Southern Biotechnology Associates, Birmingham, AL) antibody. Samples were analyzed on a FACSCalibur (Becton Dickinson, Mountain View, CA) using propidium iodide to exclude dead cells.

Oligonucleotide Array Analysis.

Cultured melanoma cells were harvested by scraping, and total cellular RNA was extracted (20). Ten μg from each sample were reverse transcribed, labeled, and processed by using a commercial kit (Affymetrix, Santa Clara, CA) according to the supplier’s instructions. Upon alkaline heat fragmentation, cDNA were hybridized to the arrays following standard procedures as supplied with the microchips (Affymetrix). Raw data were collected with a confocal laser scanner (Hewlett Packard, Palo Alto, CA), and pixel levels were analyzed using a commercial software (GeneChip v3.1; Affymetrix). Three repeats for each array were performed. Expression levels for each gene were calculated as normalized average difference of fluorescence intensity as compared with hybridization to mismatched oligonucleotides, expressed in arbitrary units. On average, >25% of the genes under investigation were positive in the cell lines tested. A threshold of 20 normalized average difference units was assigned to any gene with a calculated expression level <20, because mRNA levels in this low range could not be reliably assessed.

Chemokines and Cytokines Detection.

CXCL1, CCL5, IL-1β, IL-6, and IGF-II production was determined by quantitative ELISA assays (sensitivity, ≥10 pg/ml) using cell supernatants of confluent cultures. Antibody pairs and standards were provided by BD Biosciences or R&D Systems. ISG15 was detected by ELISA assay (19). Release of type I IFNs was quantified on HeLa cells as described elsewhere (21). Assays were performed on coded samples.

Immunohistochemistry.

The original tumor specimen from which Me67 melanoma lines were derived, conserved as paraffin-embedded material, was retrieved and analyzed as follows. Serial sections were incubated overnight at 4°C with anti-ISG15 or isotype-matched control antibodies, followed by avidin-biotin-peroxidase complex (ABC; Vectastain, Burlingame, CA).

E-Cadherin Induction on Melanoma-conditioned DCs Is Independent from Cell Contact.

We have shown previously that melanoma cell lines can activate monocyte-derived DCs in vitro(9). To obtain an insight into the molecular mechanisms underlining these modifications, we first analyzed the effects of coculturing Me67 melanoma cells, their conditioned culture medium, or their lysates with human DCs. Induction of E-cadherin, as well as the increased expression of CD15 and CD86 on DCs was observed in the presence of tumor cells or their culture supernatant, whereas cell lysates were unable to induce these effects (Fig. 1 A). Thus, phenotypic DC modulation induced by Me67 was cell contact independent and relied on the production of active soluble factors present in the cell culture medium.

To identify the molecular entity(s) responsible for these effects, specific cell clones were derived from Me67 parental cell line. Supernatants from clones Me67.3 and Me67.9 induced E-cadherin expression, whereas clones Me67.5 and Me67.10 were devoid of any modulatory effect on DC phenotype (Fig. 1 B). These clonal cell populations, all expressing the same genetic background, were subjected to gene profiling to identify differentially expressed gene sequences.

Pattern of Soluble Factors Expressed in Me67 Melanoma Clones.

Total cellular RNA extracted from Me67.3, Me67.9, Me67.5, and Me67.10 melanoma clones was processed for hybridization to oligonucleotide arrays containing probe sets from ∼7000 full-length human genes. A total of 52 expressed genes were selected based on a >3-fold changes between E-cadherin inducing (Me67.3 + Me67.9) and noninducing (Me67.5 + Me67.10) clones and grouped according to their putative physiological function (Table 1). Most of the genes identified were specific for membrane-associated, intracellular, or nuclear proteins and therefore excluded from further analysis.

A total of 11 genes encoding secreted proteins were found to be expressed at higher levels in clones Me67.3 and Me67.9 than clones Me67.5 and Me67.10 (Table 1). Among these, 5 were specific for proteins present in the extracellular matrix such as peptidoglycan, collagen, fibronectin, and mucin (Table 1). Because these proteins are involved in the formation of three-dimensional structures and poorly detectable in solution, they were excluded from further investigations.

The other genes were analyzed in detail. Two genes for chemotactic factors (GRO-α/CXCL1 and RANTES/CCL5) and 4 encoding cytokines (IL-1β, IL-6, IGF-II, and ISG15) were strongly expressed in clones Me67.3 and Me67.9 but barely detectable in clones Me67.5 and Me67.10 (Table 1).

When the corresponding conditioned melanoma cell culture media were assessed for proteins detection, the chemokine CXCL1 and the cytokine IL-1β were found to be produced to different extents by three of the melanoma lines (Fig. 2, A and C, respectively). No quantitative differences existed for IL-6 and IGF-II secretion among the different melanoma clones (Fig. 2, D and E). Considering these patterns, the involvement of these factors in the modulation of DC phenotype was unlikely. On the contrary, production of the chemokine CCL5 and the cytokine ISG15 was consistently different among the two groups of melanoma clones. More than 4000 pg/ml of CCL5 and >7000 pg/ml of ISG15 were measured in the supernatants of clones Me67.3 and Me67.9 but were undetectable in supernatants from clones Me67.5 and Me67.10 that were devoid of any DC-conditioning capacity.

Moreover, genes encoding IFN-inducible proteins (i.e., rig-g, the IFN-induced Mr 56,000 protein encoding sequence, ISG20, trip14, 27-sep, and 16-jun) were consistently expressed in clones Me67.3 and Me67.9 but poorly amplified in clones Me67.5 and Me67.10. Indeed, the two groups of clones differed for the capacity to secrete type I IFNs; clones Me67.9 and Me67.3 produced 64 and 128 IU/ml of type I IFNs, respectively, whereas Me67.5 and Me67.10 clone supernatants were completely negative (Fig. 2 G). Considering the immunomodulatory properties of type I IFNs, these data might be of relevance also in the experimental system described here.

Altogether, by combining the gene chip technology with quantitative protein detection assays, we were able to identify three soluble factors (i.e., RANTES, ISG15, and type I IFNs most likely involved in the DC phenotypic modulation induced by Me 67.3 and Me67.9 conditioned media.

Anti-ISG15 Antibodies Inhibit the Expression of E-Cadherin and Hamper the Up-Regulation of CD15 and CD86.

To define to what extent RANTES, ISG15, and/or type I IFNs induced DC phenotypic modulation, the cellular in vitro assays were repeated in the presence of specific neutralizing antibodies. The following experiments crucially depended on the use of reagents unable to induce any modification of DC phenotype per se, e.g., devoid of endotoxin contamination. Commercially available anti-CCL5 antibodies were excluded from our tools because they were found to induce up-regulation of CD83 and CD86 on immature DCs. Anti-ISG15, anti-IFNARI, and anti-CCR5 antibodies were suitable for our assays.

Neutralization of ISG15 markedly suppressed the melanoma-conditioned modulation of DC phenotype (Table 2). The results of three different experiments, independently performed, confirmed that ISG15 played a crucial role in inducing expression of E-cadherin and strongly influenced up-regulation of CD15 induced by both Me67.3 and Me67.9 conditioned media (Table 2, Exp. I, II, and III). In contrast, the up-regulation of CD86 expression on melanoma-conditioned DCs was not reproducibly dependent on ISG15. Antibodies to IFN receptors did not inhibit E-cadherin induction and CD15 expression but slightly affected the up-regulation of CD86 induced by both Me67.3 and Me67.9 conditioned media (Table 2, Exp. IV). Finally, because anti-CCR5 antibodies did not interfere with the induction of E-cadherin induced by Me67.3 conditioned medium (Table 2, Exp. V), involvement of CCL5 in conditioned media DC phenotypic modulation can be unlikely.

Detection of ISG15 in Tumor Specimens.

In previous work, tumor-infiltrating CD15+ cells with a typical DC morphology were identified within malignant melanoma tissues. The potential in vivo relevance of our in vitro findings relied on the expression of ISG15 in original tumor specimens. Indeed, immunohistochemical staining of Me67 metastatic melanoma tissue on paraffin sections revealed that tumor cells were strongly positive for ISG15, in contrast to tumor-infiltrating lymphocytes that were completely negative (Fig. 3,A). In malignant cells, ISG15 protein was localized within the cytoplasm (Fig. 3,B). WM9 melanoma human cells grown in nude mice as xenografts were examined, in the same settings, as specific control. These cells were strongly positive in immunocytochemistry only upon treatment with IFN-β, a potent inducer of ISG15 (Fig. 3, C and D, respectively). Altogether, eight tumor specimens were analyzed in immunohistochemistry. Among these, four samples were found strongly positive, three showed a moderate positive staining, and one specimen was negative (data not shown). These findings suggest that constitutive expression of ISG15 by malignant cells might frequently occur in vivo, at least in melanomas.

In this study, we explored the capacity of melanoma cell-derived proteins to induce phenotypic modulation of human monocyte-derived DCs (9). We performed microchip analysis on cellular reagents, all from the same genetic background, combined with standard ELISA assays, and this narrowed our search from 52 genes most likely involved in the DC phenotypic modulation induced by melanoma-conditioned media down to three soluble products, ISG15, type I IFNs, and CCL5.

ISG15 cytokine is strongly induced by IFN-α/β stimulation in different types of cells, including epithelial tumor cell lines in vitro(19, 22), and requires a functional proteasome (23). Its expression in malignant tissues was, thus far, never investigated. ISG15 is synthesized as a Mr 17,000 precursor protein and processed to a mature Mr 15,000 product by cleavage of the COOH-terminal amino acidic tail (24). Mature ISG15 may be released as monomer or in the form of high molecular weight conjugates (25). At present, the nature and the biological functions of these conjugates are not fully understood (26), although the immunoregulatory properties of ISG15 have been demonstrated on T lymphocytes and natural killer cells. In particular, exposure to ISG15 in vitro induces IFN-γ production by T lymphocytes and proliferation of natural killer cells (27).

Our experimental data support the hypothesis that ISG15 is crucially involved in the modulation of DC phenotype induced by melanoma cell conditioned medium: (a) the ISG15 gene was expressed at higher magnitude in tumor cells that induced DC phenotypic modulation than in cells devoid of this capacity; (b) high amounts of ISG15 protein were detected in the corresponding conditioned media and, remarkably, in original tumor specimens; and (c) most importantly, de novo expression of E-cadherin on monocyte-derived DCs in vitro was not inducible in the presence of anti-ISG15 antibodies, and up-regulation of CD15 and CD86 were strongly hampered in these conditions. In contrast, neutralization of type I IFN receptors partially inhibited CD86 up-regulation, confirming previous results (6), but did not affect E-cadherin induction; anti-CCR5 antibodies did not have any effect. Finally, in vitro exposure to recombinant CCL5 (used in the range 10–1000 ng/ml) and IFN-α2a (from 10 to 1000 IU/ml) never resulted in E-cadherin expression on immature DCs (data not shown). Thus, the most straightforward explanation of these results is that E-cadherin induction required ISG15 expression, whereas CD86 up-regulation relied on the contribution of both ISG15 and tumor-derived type I IFNs.

Impaired DC mobility, eventually caused by E-cadherin expression, and sequestration of DCs into malignant tissues have been described previously as potential mechanisms of tumor immune escape not affecting antigen-presenting capacities (8, 9). These data could have important implications for cancer immunotherapy. As interventional strategy, neutralization of tumor-derived ISG15 might be of difficult application. On the contrary, targeting the migratory pattern of immunocompetent cells might represent a possibility of immune intervention. Regarding DC mobility, it has been shown that cytokines promoting mobilization of circulating DCs (i.e., Flt3 ligand) could increase infiltration of DCs in the peritumoral region and enhance the responses to recall antigen in cancer patients (28). However, the newly recruited DCs might also become the target of tumor-derived factors and be sequestered into the malignant tissue. In this context, the use of cytokines promoting recruitment and expansion of effector T cells could be suggested to counteract DC immobilization (6, 29). The aim will be the concomitant intratumoral infiltration of DC and T lymphocytes that has been related to more favorable prognosis in different types of malignancies (10, 12).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1

Supported by the Basel Regional Cancer League Grant 6/00 (to E. P.) and National Cancer Institute CA90914 (to E. C. B.).

3

The abbreviations used are: DC, dendritic cell; IL, interleukin; FACS, fluorescence-activated cell sorter; ISG, interferon stimulated gene.

Fig. 1.

DC phenotypic modulation induced by melanoma cells is cell contact independent. A, phenotypic maturation of DCs upon 24-h coculture with Me67 melanoma cells, their conditioned medium or lysates was monitored by measuring the expression of E-cadherin, CD15, and CD86, as indicated. In each plot, dotted lines represent the background staining with isotype-matched control antibodies; thin lines show the staining profile of immature DC; overlayed black lines indicate the phenotype of mature DCs. B, modulatory capacity of clones derived from Me67 parental cell line. In each plot, data are expressed as median fluorescence intensity (MFI).

Fig. 1.

DC phenotypic modulation induced by melanoma cells is cell contact independent. A, phenotypic maturation of DCs upon 24-h coculture with Me67 melanoma cells, their conditioned medium or lysates was monitored by measuring the expression of E-cadherin, CD15, and CD86, as indicated. In each plot, dotted lines represent the background staining with isotype-matched control antibodies; thin lines show the staining profile of immature DC; overlayed black lines indicate the phenotype of mature DCs. B, modulatory capacity of clones derived from Me67 parental cell line. In each plot, data are expressed as median fluorescence intensity (MFI).

Close modal
Fig. 2.

Pattern of soluble factors expressed by Me67 melanoma clones. Concentration of CXCL1, CCL5, IL-1β, IL-6, IGF-II, and ISG15 proteins in conditioned media from Me67.3, Me67.9, Me67.5, and Me67.10 clones was determined by specific ELISAs and amounts expressed in pg/ml. Type I IFNs were quantified with a biological assay using HeLa cells, and concentrations were expressed in IU/ml. Mean values of triplicates are shown; bars, SD.

Fig. 2.

Pattern of soluble factors expressed by Me67 melanoma clones. Concentration of CXCL1, CCL5, IL-1β, IL-6, IGF-II, and ISG15 proteins in conditioned media from Me67.3, Me67.9, Me67.5, and Me67.10 clones was determined by specific ELISAs and amounts expressed in pg/ml. Type I IFNs were quantified with a biological assay using HeLa cells, and concentrations were expressed in IU/ml. Mean values of triplicates are shown; bars, SD.

Close modal
Fig. 3.

Immunohistochemical detection of ISG15 expressing cells on surgically excised Me67 metastatic melanoma tissue. Sections derived from paraffin-embedded Me67 specimen were stained with polyclonal rabbit anti-ISG15 antibodies. Two different magnifications of a representative field are shown in A and B, respectively. Results obtained in the same settings with WM9 melanoma cells grown in nude mice, untreated or treated with IFN-β, are shown in C and D, respectively.

Fig. 3.

Immunohistochemical detection of ISG15 expressing cells on surgically excised Me67 metastatic melanoma tissue. Sections derived from paraffin-embedded Me67 specimen were stained with polyclonal rabbit anti-ISG15 antibodies. Two different magnifications of a representative field are shown in A and B, respectively. Results obtained in the same settings with WM9 melanoma cells grown in nude mice, untreated or treated with IFN-β, are shown in C and D, respectively.

Close modal
Table 1

Genes differentially regulated in melanoma clones endowed with (Me67.3 and 67.9) or devoid of (Me67.5 and 67.10) DC phenotypic modulation capacity.

Sequences are grouped based on putative function and localization of specific gene products. Genes are listed by accession number and description. Data are expressed as average difference as compared with mismatched oligonucleotide controls; only genes showing a >3-fold change factor between competent (Me67.3 + Me67.9) and incompetent (Me67.5 + Me67.10) clones were considered.

Accession no.ProductAverage difference
Me67.3Me67.9Me67.5Me67.10
Secreted protein       
Chemokines       
 X54489 Melanoma growth stimulatory activity (mgsa; GRO-α/CXCL1) 961 579 20 61 
 M21121 T cell-specific protein (RANTES/CCL5) mrna, complete cds 2673 245 38 20 
Cytokines       
 X04500 Prointerleukin 1β (IL-1β) 8958 6126 1791 1123 
 X04602 IL bsf-2 (IL-6) 1865 749 119 51 
 J03242 Insulin-like growth factor 2 (IGF-II) 487 378 109 83 
 M13755 Human interferon-induced 17-KDa/15-KDa protein (ISG17/15) 7204 5916 750 286 
Extracellular matrix       
 J04599 hpgi mrna encoding bone small proteoglycan i (biglycan) 417 655 20 20 
 X02761 Fibronectin (fn precursor) 14710 15016 2378 2942 
 M55998 α-1 collagen type I gene, 3′ end 1687 1873 272 192 
 HG3044-HT3742 Fibronectin, alt. splice 1 6318 6711 827 1082 
 HG880-HT880 Mucin 6 157 567 20 20 
IFN-induced       
Unknown function       
 U52513 rig-g mrna 395 200 98 42 
 M24594 IFN-inducible 56 KD protein 912 1058 47 20 
Nuclear bodies       
 U88964 hem45 (ISG20) mrna 718 742 165 163 
Membrane protein       
 L40387 Thyroid receptor interactor (trip14) gene, 3′ end of cds. 1285 596 203 21 
 J04164 IFN-inducible protein 27-sep 4817 6161 20 20 
 U22970 16-jun gene (IFN-inducible peptide precursor) 4785 4284 22 20 
Membrane proteins       
 U96094 Sarcolipin (sln) 20 20 464 303 
 D16593 bdr-2 mrna for hippocalcin 63 124 201 379 
 Z68228 Plakoglobin 171 314 52 20 
 U59302 Steroid receptor coactivator-1 f-src-1 261 372 20 20 
 M24439 Liver/bone/kidney-type alkaline phosphatase (alpl) 306 336 20 20 
Type I       
 U48705 Receptor tyrosine kinase 80 96 426 344 
 M24283 Major group rhinovirus receptor (hrv) 314 386 52 73 
 S76475 trkc 431 551 65 88 
 Z48481 Matrix metalloproteinase 1 244 306 20 97 
Type II       
 X90846 Mixed lineage kinase 2 20 20 314 228 
 X94612 Type II cgmp-dependent protein kinase 20 20 208 547 
 L16862 G protein-coupled receptor kinase (grk6) 478 534 88 148 
Type III       
 M36284 Glycophorin C 341 455 68 129 
Integral membrane proteins       
 U40223 Uridine nucleotide receptor (unr) 227 374 28 20 
 X16662 Vascular anticoagulant-beta (Annexin 8) 252 304 20 20 
 U50136 Leukotriene c4 synthase (ltc4s) 500 365 20 77 
Intracellular proteins       
 17793 kiaa0119 53 20 347 253 
 U14417 ral guanine nucleotide dissociation stimulator 173 301 54 34 
 X53414 Peroxisomal 1-alanine:glyoxylate aminotransferase 20 20 328 237 
 AF006041 fas-binding protein (daxx) 299 663 78 103 
 X15331 Phosphoribosylpyrophosphate synthetase subunit one 514 546 129 105 
 X68277 Protein tyrosine phosphatase 586 526 144 124 
 U80226 γ-Aminobutyric acid transaminase 336 266 81 79 
 M19267 Tropomyosin 1428 2158 332 303 
 Z24727 Tropomyosin isoform 3037 2631 687 735 
 L48546 Tuberin (tsc2) 187 303 37 20 
 U26266 Deoxyhypusine synthase 504 415 70 115 
 X81420 hhkb1 1308 791 82 181 
Nuclear proteins       
 X12517 u1 small nuclear rnp-specific c protein 20 20 441 430 
 Z49825 Hepatocyte nuclear factor 4 α 68 20 450 336 
 M96739 nscl-1 424 598 109 103 
 M76378 Cysteine-rich protein (crp) 2043 1656 483 365 
 X61755 Homeoprotein hox3d 155 444 20 20 
 M59465 Tumor necrosis factor α inducible protein a20 701 325 60 63 
 X68688 znf33b gene 410 482 20 20 
Accession no.ProductAverage difference
Me67.3Me67.9Me67.5Me67.10
Secreted protein       
Chemokines       
 X54489 Melanoma growth stimulatory activity (mgsa; GRO-α/CXCL1) 961 579 20 61 
 M21121 T cell-specific protein (RANTES/CCL5) mrna, complete cds 2673 245 38 20 
Cytokines       
 X04500 Prointerleukin 1β (IL-1β) 8958 6126 1791 1123 
 X04602 IL bsf-2 (IL-6) 1865 749 119 51 
 J03242 Insulin-like growth factor 2 (IGF-II) 487 378 109 83 
 M13755 Human interferon-induced 17-KDa/15-KDa protein (ISG17/15) 7204 5916 750 286 
Extracellular matrix       
 J04599 hpgi mrna encoding bone small proteoglycan i (biglycan) 417 655 20 20 
 X02761 Fibronectin (fn precursor) 14710 15016 2378 2942 
 M55998 α-1 collagen type I gene, 3′ end 1687 1873 272 192 
 HG3044-HT3742 Fibronectin, alt. splice 1 6318 6711 827 1082 
 HG880-HT880 Mucin 6 157 567 20 20 
IFN-induced       
Unknown function       
 U52513 rig-g mrna 395 200 98 42 
 M24594 IFN-inducible 56 KD protein 912 1058 47 20 
Nuclear bodies       
 U88964 hem45 (ISG20) mrna 718 742 165 163 
Membrane protein       
 L40387 Thyroid receptor interactor (trip14) gene, 3′ end of cds. 1285 596 203 21 
 J04164 IFN-inducible protein 27-sep 4817 6161 20 20 
 U22970 16-jun gene (IFN-inducible peptide precursor) 4785 4284 22 20 
Membrane proteins       
 U96094 Sarcolipin (sln) 20 20 464 303 
 D16593 bdr-2 mrna for hippocalcin 63 124 201 379 
 Z68228 Plakoglobin 171 314 52 20 
 U59302 Steroid receptor coactivator-1 f-src-1 261 372 20 20 
 M24439 Liver/bone/kidney-type alkaline phosphatase (alpl) 306 336 20 20 
Type I       
 U48705 Receptor tyrosine kinase 80 96 426 344 
 M24283 Major group rhinovirus receptor (hrv) 314 386 52 73 
 S76475 trkc 431 551 65 88 
 Z48481 Matrix metalloproteinase 1 244 306 20 97 
Type II       
 X90846 Mixed lineage kinase 2 20 20 314 228 
 X94612 Type II cgmp-dependent protein kinase 20 20 208 547 
 L16862 G protein-coupled receptor kinase (grk6) 478 534 88 148 
Type III       
 M36284 Glycophorin C 341 455 68 129 
Integral membrane proteins       
 U40223 Uridine nucleotide receptor (unr) 227 374 28 20 
 X16662 Vascular anticoagulant-beta (Annexin 8) 252 304 20 20 
 U50136 Leukotriene c4 synthase (ltc4s) 500 365 20 77 
Intracellular proteins       
 17793 kiaa0119 53 20 347 253 
 U14417 ral guanine nucleotide dissociation stimulator 173 301 54 34 
 X53414 Peroxisomal 1-alanine:glyoxylate aminotransferase 20 20 328 237 
 AF006041 fas-binding protein (daxx) 299 663 78 103 
 X15331 Phosphoribosylpyrophosphate synthetase subunit one 514 546 129 105 
 X68277 Protein tyrosine phosphatase 586 526 144 124 
 U80226 γ-Aminobutyric acid transaminase 336 266 81 79 
 M19267 Tropomyosin 1428 2158 332 303 
 Z24727 Tropomyosin isoform 3037 2631 687 735 
 L48546 Tuberin (tsc2) 187 303 37 20 
 U26266 Deoxyhypusine synthase 504 415 70 115 
 X81420 hhkb1 1308 791 82 181 
Nuclear proteins       
 X12517 u1 small nuclear rnp-specific c protein 20 20 441 430 
 Z49825 Hepatocyte nuclear factor 4 α 68 20 450 336 
 M96739 nscl-1 424 598 109 103 
 M76378 Cysteine-rich protein (crp) 2043 1656 483 365 
 X61755 Homeoprotein hox3d 155 444 20 20 
 M59465 Tumor necrosis factor α inducible protein a20 701 325 60 63 
 X68688 znf33b gene 410 482 20 20 
Table 2

Neutralization of ISG15 impairs E-cadherin induction on DCsa

Exp. IExp. IIExp. III
E-cadh.bCD15CD86E-cadh.CD15CD86E-cadh.CD15CD86
iDC+ RIg 3.3 20.1 7.5 5.0 22.2 23.2 4.9 24.1 24.1 
Me67.3+ RIg 18.1 643.5 33.6 34.2 609.7 55.4    
Me67.3+ anti-ISG15 3.5 342.8 13.8 3.8 387.1 69.1    
Me67.9+ RIg 13.5 577.7 21.2    29.1 729.9 62.8 
Me67.9+ anti-ISG15 3.4 321.9 9.9    3.7 425.5 80.5 
Exp. IExp. IIExp. III
E-cadh.bCD15CD86E-cadh.CD15CD86E-cadh.CD15CD86
iDC+ RIg 3.3 20.1 7.5 5.0 22.2 23.2 4.9 24.1 24.1 
Me67.3+ RIg 18.1 643.5 33.6 34.2 609.7 55.4    
Me67.3+ anti-ISG15 3.5 342.8 13.8 3.8 387.1 69.1    
Me67.9+ RIg 13.5 577.7 21.2    29.1 729.9 62.8 
Me67.9+ anti-ISG15 3.4 321.9 9.9    3.7 425.5 80.5 
Exp. IVExp. V
E-cadh.CD15CD86E-cadh.CD15CD86
iDC+ GIg 4.3 25.2 24.1  iDC+ IgG1 2.3 6.2 7.1 
Me67.3+ GIg 39.0 409.5 61.0 Me67.9+ IgG1 13.5 106.5 23.1 
Me67.3+ anti-IFNARI 34.0 549.0 55.1 Me67.9+ anti-CCR5 14.9 143.3 18.4 
Me67.9+ GIg 23.5 827.0 78.2     
Me67.9+ anti-IFNARI 25.0 743.1 63.0     
Exp. IVExp. V
E-cadh.CD15CD86E-cadh.CD15CD86
iDC+ GIg 4.3 25.2 24.1  iDC+ IgG1 2.3 6.2 7.1 
Me67.3+ GIg 39.0 409.5 61.0 Me67.9+ IgG1 13.5 106.5 23.1 
Me67.3+ anti-IFNARI 34.0 549.0 55.1 Me67.9+ anti-CCR5 14.9 143.3 18.4 
Me67.9+ GIg 23.5 827.0 78.2     
Me67.9+ anti-IFNARI 25.0 743.1 63.0     
a

Immature DCs were cultured for 24 h in the presence or absence of melanoma culture supernatants together with the indicated reagents. Cells were then collected and stained with specific mAbs.

b

Data on cell surphace expression of E-cadherin, CD15, CD86 are expressed as median of fluorescence intensity (MFI).

Thanks to M. Ferrantini, B. Hassel, and A. L. Haas for providing reagents and for helpful discussions and to E. Shultz and P. Zajac for technical help.

1
Bell D., Young J. W., Banchereau J. Dendritic cells.
Adv. Immunol.
,
72
:
255
-324,  
1999
.
2
Sallusto F., Lanzavecchia A. Mobilizing dendritic cells for tolerance, priming, and chronic inflammation.
J. Exp. Med.
,
189
:
611
-614,  
1999
.
3
Sallusto F., Schaerli P., Loetscher P., Schaniel C., Lenig D., Mackay C. R., Qin S., Lanzavecchia A. Rapid and coordinated switch in chemokine receptor expression during dendritic cell maturation.
Eur. J. Immunol.
,
28
:
2760
-2769,  
1998
.
4
Hirao M., Onai N., Hiroishi K., Watkins S. C., Matsushima K., Robbins P. D., Lotze M. T., Tahara H. CC chemokine receptor-7 on dendritic cells is induced after interaction with apoptotic tumor cells: critical role in migration from the tumor site to draining lymph nodes.
Cancer Res.
,
60
:
2209
-2217,  
2000
.
5
Sallusto F., Lenig D., Mackay C. R., Lanzavecchia A. J. Flexible programs of chemokine receptor expression on human polarized T helper 1 and 2 lymphocytes.
J. Exp. Med.
,
187
:
875
-883,  
1998
.
6
Padovan E., Spagnoli G. C., Ferrantini M., Heberer M. IFN-α2a induces IP-10/CXCL10 and MIG/CXCL9 production in monocyte-derived dendritic cells and enhances their capacity to attract and stimulate CD8+ effector T cells.
J. Leuk. Biol.
,
71
:
669
-676,  
2002
.
7
Tang H. L., Cyster J. G. Chemokine up-regulation and activated T cell attraction by maturing dendritic cells.
Science (Wash. DC)
,
284
:
819
-822,  
1999
.
8
Lucas A. D., Halliday G. M. Progressor but not regressor skin tumors inhibit Langerhans’ cell migration from epidermis to local lymph nodes.
Immunology
,
97
:
130
-137,  
1999
.
9
Remmel E., Terracciano L., Noppen C., Zajac P., Heberer M., Spagnoli G. C., Padovan E. Modulation of dendritic cell phenotype and mobility by tumor cells in vitro.
Hum. Immunol.
,
62
:
39
-49,  
2001
.
10
Reichert T. E., Scheuer C., Day R., Wagner W., Whiteside T. L. The number of intratumoral dendritic cells and ζ-chain expression in T cells as prognostic and survival biomarkers in patients with oral carcinoma.
Cancer (Phila.)
,
91
:
2136
-2147,  
2001
.
11
Eisenthal A., Polyvkin N., Bramante-Schreiber L., Misonznik F., Hassner A., Lifschitz-Mercer B. Expression of dendritic cells in ovarian tumors correlates with clinical outcome in patients with ovarian cancer.
Hum. Pathol.
,
32
:
803
-807,  
2001
.
12
Schwaab T., Weiss J. E., Schned A. R., Barth R. J., Jr. Dendritic cell infiltration in colon cancer.
J. Immunother.
,
24
:
130
-137,  
2001
.
13
Schwaab T., Schned A. R., Heaney J. A., Cole B. F., Atzpodien J., Wittke F., Ernstoff M. S. In vivo description of dendritic cells in human renal cell carcinoma.
J. Urol.
,
162
:
567
-573,  
1999
.
14
Enk A. H., Jonuleit H., Saloga J., Knop J. Dendritic cells as mediators of tumor-induced tolerance in metastatic melanoma.
Int. J. Cancer
,
73
:
309
-316,  
1997
.
15
Gabrilovich D. I., Chen H. L., Girgis K. R., Cunningham H. T., Meny G. M., Nadaf S., Kavanaugh D., Carbone D. P. Production of vascular endothelial growth factor by human tumors inhibits the functional maturation of dendritic cells.
Nat. Med.
,
2
:
1096
-1103,  
1996
.
16
Menetrier-Caux C., Montmain G., Dieu M. C., Bain C., Favrot M. C., Caux C., Blay J. Y. Inhibition of dendritic cells from CD34+ progenitors by tumor cells: role of interleukin-6 and macrophage colony-stimulating factor.
Blood
,
92
:
4778
-4791,  
1998
.
17
Bronte V., Chappell D. B., Apolloni E., Cabrelle A., Wang M., Hwu P., Restifo N. P. Unopposed production of granulocyte-macrophage colony-stimulating factor by tumors inhibits CD8+ T cell responses by dysregulating antigen-presenting cell maturation.
J. Immunol.
,
162
:
5728
-5737,  
1999
.
18
Sallusto F., Lanzavecchia A. Efficient presentation of soluble antigen by cultured human dendritic cells is maintained by granulocyte/macrophage colony-stimulating factor plus interleukin 4 and downregulated by tumor necrosis factor α.
J. Exp. Med.
,
179
:
1109
-1118,  
1994
.
19
D’Cunha J., Ramanujam S., Wagner R. J., Witt P. L., Knight E., Jr., Borden E. C. In vitro and in vivo secretion of human ISG15, an IFN-induced immunomodulatory cytokine.
J. Immunol.
,
157
:
4100
-4108,  
1996
.
20
Certa U., Seiler M., Padovan E., Spagnoli G. C. High density oligonucleotide array analysis of interferon-α2a sensitivity and transcriptional response in melanoma cells.
Br. J. Cancer
,
85
:
107
-114,  
2001
.
21
Mecchia M., Matarrese P., Malorni W., D’Agostino G., Sestili P., Santini S. M., Gauzzi M. C., Venditti M., Mazzocchi A., Parmiani G., Belardelli F., Ferrantini M. Type I consensus interferon (CIFN) gene transfer into human melanoma cells up-regulates p53 and enhances cisplatin-induced apoptosis: implications for new therapeutic strategies with IFN-α.
Gene Ther.
,
7
:
167
-179,  
2000
.
22
Korant B. D., Blomstrom D. C., Jonak G. J., Knight E., Jr. Interferon-induced proteins. Purification and characterization of a 15,000-Dalton protein from human and bovine cells induced by interferon.
J. Biol. Chem.
,
259
:
14835
-14839,  
1984
.
23
Li X. L., Hassel B. A. Involvement of proteasomes in gene induction by interferon and double-stranded RNA.
Cytokine
,
14
:
247
-252,  
2001
.
24
Knight E., Jr., Fahey D., Cordova B., Hillman M., Kutny R., Reich N., Blomstrom D. A 15-kDa interferon-induced protein is derived by COOH-terminal processing of a 17-kDa precursor.
J. Biol. Chem.
,
263
:
4520
-4522,  
1988
.
25
Loeb K. R., Haas A. L. The interferon-inducible 15-kDa ubiquitin homolog conjugates to intracellular proteins.
J. Biol. Chem.
,
267
:
7806
-7813,  
1992
.
26
Yuan W., Krug R. M. Influenza B virus NS1 protein inhibits conjugation of the interferon (IFN)-induced ubiquitin-like ISG15 protein.
EMBO J.
,
20
:
362
-371,  
2001
.
27
D’Cunha J., Knight E., Jr., Haas A. L., Truitt R. L., Borden E. C. Immunoregulatory properties of ISG15, an interferon-induced cytokine.
Proc. Natl. Acad. Sci. USA
,
93
:
211
-215,  
1996
.
28
Morse M. A., Nair S., Fernandez-Casal M., Deng Y., St. Peter M., Williams R., Hobeika A., Mosca P., Clay T., Cumming R. I., Fisher E., Clavien P., Proia A. D., Niedzwiecki D., Caron D., Lyerly H. K. Preoperative mobilization of circulating dendritic cells by Flt3 ligand administration to patients with metastatic colon cancer.
J. Clin. Oncol.
,
18
:
3883
-3893,  
2000
.
29
Kirkwood J. M. Systemic adjuvant treatment of high-risk melanoma: the role of interferon α-2b and other immunotherapies.
Eur. J. Cancer
,
34S3
:
12
-17,  
1998
.