Ewing's sarcoma (EWS) is an aggressive tumor of children and young adults that requires intensive treatment. The search for new prognostic factors is very important to choose the most appropriate therapy and to better understand the biology of the disease for the development of new therapeutic tools. We found that Xg, a thus far poorly described molecule and member of the CD99 family, is expressed in EWS cell lines and EWS primary tumors. Immunohistochemical analysis confirmed the expression of Xg in 24% of patients. We found that Xg expression in EWS defines a subgroup of patients with worse prognosis compared with those with Xg-negative localized tumors, indicating a clinical relevance of Xg expression in EWS. Forced expression of Xg in an EWS cell line upregulated cell migration and invasion in vitro. Furthermore, knockdown of Xg expression with specific short hairpin RNA significantly reduced migration and invasion of EWS cells. Consistent with these data, in vivo xenotransplant studies in nude mice revealed that Xg expression increased the incidence and the number of metastases of EWS cells. Thus, Xg expression is associated with lower overall survival in EWS patients with localized tumors and is implicated in metastasis. Cancer Res; 70(9); 3730–8. ©2010 AACR.

Ewing's sarcoma (EWS) is a highly aggressive malignant tumor that develops in bones and soft tissues and occurs preferentially in children and young adults. Survival rates reach approximately 60% to 70% for patients with localized tumors; however, the presence of metastases at diagnosis defines worse prognosis (survival rates approximately 20–40%). Bone metastases confer a poorer outcome than lung/pleura metastases (<20% compared with 20–40% 5-year survival; ref. 1). Two biological features characterize EWS and help in diagnosis: chromosomal translocation between the 5′ portion of the EWS gene with the 3′ end of the ETS family genes (FLI1, ERG, ETV1, E1AF, or FEV; refs. 2, 3). The most frequent aberrant transcription factor generated is the EWS-FLI1 chimeric protein (85% of cases), which is the result of the t(11;22)(q14;q12) translocation. The oncogenic role of this aberrant transcription factor has been described in fibroblasts, murine primary bone-derived cells, and primary bone marrow–derived mesenchymal progenitor cells (46). It has also been shown recently that EWS can originate from mesenchymal stem cells (7). The other feature is the large amount of CD99 molecules in almost all EWS cases (8, 9). CD99 is also displayed at high density on T-cell acute lymphoblastic leukemia (10). CD99 is a 32-kDa transmembrane, highly glycosylated and sialylated protein, which is ubiquitously expressed on hematopoëitic cells. CD99 has two isoforms a long form of 32 kDa and a short one of 28 kDa that possess adverse functional properties (1113). From a functional viewpoint, we recently described CD99 as a regulator of HLA class I expression on the cell surface (14). Moreover, CD99 is involved in cell adhesion and motility (11), including intercellular adhesion of lymphocyte endothelial cells (15) and transendothelial migration of monocytes (16), neutrophils (17), and CD34+ cells (18). In addition, it can induce immediate lymphocyte arrest on an inflammatory endothelium (15). In a pathologic context, CD99 can exert an oncosuppressor effect on osteosarcoma (19) and, in contrast, contributes to the malignant properties of the EWS family of tumors (13, 20).

CD99 exhibits 48% and 47% of sequence homology with two other proteins, Xg and mouse CD99L2, respectively, whose genes are located on the X chromosome (21, 22). Xg is a 26-kDa molecule, whose gene which is located on/at the pseudoautosomal boundary of the short arm of the X chromosome (Xp22), in the vicinity of the CD99 gene (21, 23). The gene of CD99L2 (40 kDa) is located at the end of the long arm of the X chromosome (Xq28; ref. 22). From a structural viewpoint, CD99, Xg, and CD99L2 present four highly conserved regions including the FXLXDAL motif (in which X is any amino acid) and a glycine-rich region on the extracytoplasmic domain, the transmembrane domain. The fourth domain is the juxtamembrane region that is composed of positively charged lysine and arginine residues (22). CD99 and Xg also present a supplemental highly conserved domain that is a proline-rich region. From a functional viewpoint, CD99L2 has also been characterized as an adhesion molecule required for neutrophil extravasation in mice (23).

The Xg protein was initially characterized on RBC and defines a blood group (24). Studies into families revealed a difference in the Xg(+) frequency according to gender. In fact, 89% of females and 62% of males are Xg(a+) (ref. 24). The CD99 and Xg antigens present a quantitative polymorphism on RBC (25, 26). However, the exact tissue distribution of the Xg protein, initially described as limited to RBC, and its biological function(s) remain to be characterized.

Here, we found the Xg protein to be expressed on cancer cells in 24% of EWS cases. Moreover, we found that the expression of Xg is associated with worse prognosis of EWS patients. To study the role of the Xg protein in the malignancy of EWS cells, we chose two approaches based on forced and silenced expression of Xg in two EWS cell lines. Furthermore, we show that Xg contributes to the metastatic properties of EWS cells by regulating migration in vitro and in vivo.

Cell lines and transfection

EWS and osteosarcoma cell lines (IOR/BRZ, IOR/CAR/, IOR/BER, IOR/CLB, LAP35, U-2OS, OS9, SARG, and MOS) were generated at the Istituto Ortopedici Rizzoli. EWS cell lines SK-ES-1, SK-N-MC, and RDES were obtained from the American Type Culture Collection. TC-71 and 6647 were a generous gift from TJ Triche (Childrens Hospital, Los Angeles, CA). WE-68, NT-68, TC-83, and VH-64 were a generous gift of Dr. Frans van Valen (Laboratory for Experimental Orthopaedic Research, University Hospital of Munster, Germany) (27). EWS cells were cultured in DMEM (Life Technologies; Invitrogen) supplemented with 100 U/mL penicillin, 100 μg/mL streptomycin, 1 mmol/L sodium pyruvate, and 10% inactivated fetal bovine serum (FBS; Perbio). Cells were maintained at 37°C in a humidified 5% CO2 atmosphere.

The Xg expression vector (pcDNA3.1) was a generous gift from JP Cartron [Institut National de la Sante et de la Recherche Medicale (INSERM) U76, Biologie des groupes sanguins humains, Paris, France; ref. 28]. Stably transfected IOR/BRZ cells expressing Xg were obtained using a calcium phosphate transfection (Invitrogen). Cells transfected with empty pcDNA3.1 (+) were used as a negative control. Transfectants were selected with 1 mg/mL neomycin and analyzed by phycoerythrin (PE)-conjugated Xg staining using FACScan (Becton Dickinson). The NBL-1 hybridoma (Anti-Xg) was generated and validated by Ellis and colleagues (24). This clone has also been validated by Fouchet and colleagues (28, 29). Purified NBL-1 was coupled to PE (Proteogenix).

Short hairpin RNA constructs

Using the calcium phosphate reagent (Invitrogen), the TC-71 EWS cell line was stably transfected with a mix of two short hairpin RNA (shRNA) targeting Xg expression plasmids (pSilencer4.1-CMV puro, Ambion). The target sequences used were 5′-GCCGAGGTCA AAGAGACTTTGATTTCAAGAGAATCAAAGTCTCTTTGACCTCGGTTTTTT-3′ and 5′-GGGAGGTTACTTCAATGATGTGGATCAAGAGTCCACATCATTGAAGTAACCTC CTTTTTT-3′. The empty vector was used as a negative control. Xg-silenced cells were selected with 0.2 μg/mL puromycin.

Quantitative real-time reverse transcription-PCR

Messenger RNA was extracted using an Oligotex direct mRNA Micro kit (Qiagen) and reverse transcribed to cDNA using the High Capacity cDNA Reverse Transcription kit (Applied Biosystems). Taq platinium (Invitrogen) was used for PCRs performed using an ABI PRISM 7900 Sequence Detection System (Applied Biosystems). The relative mRNA expression of Xg was determined by the comparative cycle threshold method using the mean of four housekeeping genes: Human Actin, GAPDH, HPRT, and ubiquitin. The Xg primers used for Xg PCR amplification were 5′-GATGTGGACCGTGATGACGGAC-3′ (forward) and 3′-TACCATATTGCCTTCTGGATTG CC-5′ (reverse).

Patient population

Xg expression was analyzed on the primary tumors of 97 patients suffering from EWS collected from the tissue bank of the Laboratory of Experimental Oncology, Rizzoli Institute. Seventy-five patients presented with a primary localized tumor at diagnosis, whereas 22 showed metastases at diagnosis (median follow-up, 60 mo; range, 13–60 mo). Chemotherapy was administered according to consecutive protocols sequentially activated EW-REN1-3 and ISG/SSGIII-IV (3032). A panel of pathologists reviewed histology. A standard follow-up surveillance protocol was adopted for up to 5 years and clinical data were updated. Adverse events were defined as metastasis at any site or death during remission and event-free survival was calculated from the date of initial diagnosis. Clinical pathologic features of the different series of EWS patients included in the study are summarized in Table 1. Sample sets were handled in a coded fashion. The ethical committee of the Rizzoli Institute approved the studies and informed consent was obtained from all subjects involved. Kaplan-Meier and log-rank methods were used, respectively, to draw and evaluate the significance of survival curves in EWS patients. Cox's proportional hazards regression analysis was used for multivariate analysis.

Table 1.

Clinicopathologic features of EWS patients evaluated for Xg expression

CharacteristicsAll patients (n = 97)Patients with localized tumors (n = 75)
No.%Association with prognosis (EFS)*No.%Association with prognosis (EFS)*
Gender 
    Female 46 47 0.57 36 48 0.69 
    Male 51 53  39 52  
Age 
    ≤14 y 41 58 0.45 43 57 0.93 
    >14 y 56 42  32 43  
Location 
    Extremity 57 58 0.35 50 67 0.11 
    Pelvis 20 21  15 20  
    Other 20 21  10 13  
Diagnosis 
    EWS 71 73 0.55 58 77 0.5 
    PNET 12 15   
    Atypical 14 12  10 13  
Xg 
    Positive 23 24 0.2 19 25 0.1 
    Negative 74 76  56 75  
Surgery 
    Yes 74 76 0.53 61 81 0.83 
    No 23 24  14 19  
Chemotherapy protocol 
    Yes 95 95 Nd 75 100 Nd 
    No   
Chemotherapy protocol 
    EW-REN1-3 32 34 0.11 27 36 0.07 
    ISG/SSGIII-IV 63 66  48 64  
Necrosis 
    Total 16 24 0.02 15 27 0.01 
    Nontotal 51 76  40 73  
CharacteristicsAll patients (n = 97)Patients with localized tumors (n = 75)
No.%Association with prognosis (EFS)*No.%Association with prognosis (EFS)*
Gender 
    Female 46 47 0.57 36 48 0.69 
    Male 51 53  39 52  
Age 
    ≤14 y 41 58 0.45 43 57 0.93 
    >14 y 56 42  32 43  
Location 
    Extremity 57 58 0.35 50 67 0.11 
    Pelvis 20 21  15 20  
    Other 20 21  10 13  
Diagnosis 
    EWS 71 73 0.55 58 77 0.5 
    PNET 12 15   
    Atypical 14 12  10 13  
Xg 
    Positive 23 24 0.2 19 25 0.1 
    Negative 74 76  56 75  
Surgery 
    Yes 74 76 0.53 61 81 0.83 
    No 23 24  14 19  
Chemotherapy protocol 
    Yes 95 95 Nd 75 100 Nd 
    No   
Chemotherapy protocol 
    EW-REN1-3 32 34 0.11 27 36 0.07 
    ISG/SSGIII-IV 63 66  48 64  
Necrosis 
    Total 16 24 0.02 15 27 0.01 
    Nontotal 51 76  40 73  

Abbreviations: EFS, event-free survival; PNET, primitive neuroectodermal tumors; Nd, not determined.

*Log-rank P values.

Tissue array construction and immunohistochemistry

Duplicate 1-mm cores were taken from representative areas of tumors and inserted into a recipient paraffin block to create a tissue microarray containing a total of 120 cores, including six control cores of nontumor tissue (normal kidney or liver). In all cases, CD99 positivity was confirmed. Immunohistochemistry was performed on tumor cores using the NBL-1 monoclonal antibody (mAb; Anti-Xg) as the primary antibody (1:100 dilution) followed by a horseradish peroxidase secondary antibody (1:300 dilution; VECTASTAIN ABC KIT; ref. 24). Analysis of Xg expression in normal tumors was made on tissue microarrays purchased from Ambion, Inc., Cambridgeshire UK. Each tissue sample was obtained in duplicate from four individual donors, except endometrium samples that came from two individual donors.

Metastatic ability in athymic mice

Six-week old female athymic Nude-Foxn1 nu/nu mice were purchased from Harlan. Metastasis was determined after i.v. injection of 1 × 2.106 cells. The number of pulmonary metastases was assessed 2 months after cell inoculation by counting after staining with black India ink. Twelve mice were used for each experimental group. All mice were cared for in accordance with the INSERM guidelines. Experimental procedures were approved by the local ethical committee.

Flow cytometry

Cells (1 × 5.106) were stained with a PE-conjugated NBL-1 mAb (Anti-Xg) in PBS for 20 minutes at 4°C (24). After washing with PBS, cells were fixed with 1% paraformaldehyde and analyzed using a FACScan (Becton Dickinson).

Western blotting

Cells were lysed in radioimmunoprecipitation assay buffer containing Tris-HCl (pH 7.4), 1 mmol/L NaCl, 1% SDS, 1% Triton X-100, 1% deoxycholate and protease inhibitors mmol/L phenylmethylsulfonyl fluoride, 10 μg/mL aprotinin, and 10 μg/mL leupeptin. The protein concentration of the samples was determined by the Bio-Rad protein assay (Bio-Rad Laboratories). Fifty micrograms of total cell lysates were separated by 10% SDS-PAGE and transferred onto a nitrocellulose membrane (Amersham). The membrane was blocked with PBS containing 0.4% Tween and 5% dried nonfat milk for 1 h at room temperature and probed with the NBL-1 mAb (Anti-Xg) overnight at 4°C with gentle rocking (24). Proteins were revealed using a horseradish peroxydase–linked anti-mouse antibody (1:3000; CST) and visualized with enhanced chemiluminescence reagents (Pierce). Erk2 (TEBU) was used as a loading control.

Cell migration and invasion assays

Motility assays were performed using Transwell Permeable Supports chambers (Corning Incorporated). Cells (1 × 1.106) in serum-free culture medium were seeded in the upper compartment, whereas the lower compartment contained 600 μL of medium with 10% FBS used as a source of chemoattractant. Cells were allowed to migrate for 20 h at 37°C. EWS cells were harvested with 2 mmol/L EDTA PBS. Migrated cells were counted using 25 μL of Flow-Count Fluorospheres (Beckman Coulter) and were analyzed by flow cytometry. For the invasion assay, 1 mg/mL growth factor–reduced BD Matrigel matrix (BD Biosciences) was added onto the insert and incubated for 30 minutes at 37°C to allow polymerization. Experiments were performed in triplicate.

Xg expression on EWS cell lines and tissues

As CD99 and Xg belong to the same family of proteins, we looked for Xg expression in EWS, in which almost all cases possess the salient feature of CD99 expression (8, 9). Quantitative real-time PCR analysis revealed that Xg transcripts were always detected in EWS cell lines, in contrast to osteosarcoma cell lines that were Xg negative (Fig. 1A). Figure 1B shows that the expression level of the Xg protein is variable in EWS cell lines, whereas it was constantly negative in osteosarcoma cell lines (Supplementary Fig. S1). In accordance with the Western blotting data, the presence of Xg on the cell surface was detected in 13 of 15 cell lines of the EWS family with widespread median cytofluorimetric fluorescence intensity (Fig. 1C). Although it was reported that Xg positivity and the CD99 level of expression are coregulated on erythrocytes (28), we found no significant correlation (P = 0.8439) for Xg and CD99 expression when evaluated by immunohistochemistry on tissues from EWS patients (data not shown). Moreover, Xg expression in EWS tumors was independent of the gender of the patients, despite the fact that the Xg molecule has X-linked expression on RBC (24). It must be noted that we examined a hundred samples of leukemia and hematosarcoma cells for which we could not find a detectable level of the Xg molecule (data not shown).

Figure 1.

Xg expression on EWS cell lines. A, quantification of Xg transcripts from osteosarcoma and EWS cell lines by real-time PCR. The 10 osteosarcoma cell lines investigated were U2OS, SAOS, MG83, MOS, SARG, OS9, OS10, OS14, OS15, and OS17 (□); the 14 EWS cell lines investigated were IOR/CLB, IOR/RCH, H1474-P2, H825, WE68, MM83, TC83AH, NT68, VH64, RM82, STAET2.1, STAET2.2, TC-71, and IOR/BRZ (▪). B, Xg protein levels in EWS cell lines. Western blotting was performed with an anti-Xg mAb NBL-1 antibody. Erk2 expression was used as a loading control. C, cytofluorimetric analysis of Xg expression on EWS cell lines. Solid line, cells stained using a PE-conjugated anti-Xg mAb NBL-1 antibody. Dotted line, cells stained with a PE-isotype control.

Figure 1.

Xg expression on EWS cell lines. A, quantification of Xg transcripts from osteosarcoma and EWS cell lines by real-time PCR. The 10 osteosarcoma cell lines investigated were U2OS, SAOS, MG83, MOS, SARG, OS9, OS10, OS14, OS15, and OS17 (□); the 14 EWS cell lines investigated were IOR/CLB, IOR/RCH, H1474-P2, H825, WE68, MM83, TC83AH, NT68, VH64, RM82, STAET2.1, STAET2.2, TC-71, and IOR/BRZ (▪). B, Xg protein levels in EWS cell lines. Western blotting was performed with an anti-Xg mAb NBL-1 antibody. Erk2 expression was used as a loading control. C, cytofluorimetric analysis of Xg expression on EWS cell lines. Solid line, cells stained using a PE-conjugated anti-Xg mAb NBL-1 antibody. Dotted line, cells stained with a PE-isotype control.

Close modal

Xg expression is correlated with a lower overall survival in EWS patients with localized tumors

To obtain a better appraisal of Xg expression in primary EWS cells, we performed immunohistochemical analysis of Xg on paraffin-embedded tissue sections from 97 primary EWS tumors (Table 1). Patients ages ≤40 years with biopsy-proven histologic diagnosis of Ewing family of tumors (EFT) were eligible for the study. Histology was reviewed by a panel of pathologists. The patients included in this study represent ∼40% of all patients treated at the Rizzoli Institute in the period between 1995 and 2005. We selected the cases for which representative tissue material was available for the construction of tissue array and the clinical data and the follow up could be completed. This cohort included 75 patients with localized tumors and 22 patients presenting with detectable metastasis at diagnosis. Therefore, the series of patients investigated was representative of a typical EWS patient population (33). Figure 2A displays typical positive staining of EWS tissue sections, which can be compared with the Xg negativity observed in our series of EWS patients. The percentages of positivity were 24% (18 of 75) in localized tumors and 23% (5 of 22) in disseminated tumors. We evaluated the prognostic relevance of Xg expression either considering all the patients or considering only those with localized tumors. Taking into account clinical parameters, patients who showed total necrosis after preoperative chemotherapy had a favorable prognosis (Table 1). Xg expression was instead associated with adverse prognosis. This association is clearer when considering only the localized tumors and seemed to be significant when overall survival (P = 0.047) but not event-free survival (P = 0.1) was determined (Fig. 2B). Considering that the final patient outcome depends on both tumor intrinsic malignancy and response to treatment, which very likely determines overall and event-free survival, respectively, this information may indicate that Xg expression correlates with EWS aggressiveness rather than chemosensitivity. Multivariate analyses confirmed that Xg expression and the percentage of histologic tumor response to neoadiuvant chemotherapy are independent variables and confirmed a significant association with prognosis for both the percentage of necrosis and Xg expression. Cox proportional hazards regression analysis for overall survival in 75 EWS patients details were as follows: adjusted risk rate ratio of 5.67 and 2.9 for necrosis and Xg expression, respectively, 95% confidence interval of 1.27 to 25.32 and 1.12 to 7.52 for necrosis and Xg expression, respectively, P value of 0.02 and 0.03 for necrosis and Xg expression, respectively.

Figure 2.

Expression of Xg in EWS primary tumors. A, immunohistochemical analysis of Xg expression on a tissue array of 97 EWS patients. The anti-Xg mAb NBL-1 was used for specific staining. Pictures of Xg-positive and Xg-negative staining observed in EWS tissue. B, survival of EWS patients according to Xg expression. Top and bottom, show event-free survival and OVS for all patients (n = 97) and localized EWS patients (n = 75), respectively. Comparison of survival curves was performed by the log-rank test. Time scale refers to months from diagnosis. Thick lines, Xg-positive patients.

Figure 2.

Expression of Xg in EWS primary tumors. A, immunohistochemical analysis of Xg expression on a tissue array of 97 EWS patients. The anti-Xg mAb NBL-1 was used for specific staining. Pictures of Xg-positive and Xg-negative staining observed in EWS tissue. B, survival of EWS patients according to Xg expression. Top and bottom, show event-free survival and OVS for all patients (n = 97) and localized EWS patients (n = 75), respectively. Comparison of survival curves was performed by the log-rank test. Time scale refers to months from diagnosis. Thick lines, Xg-positive patients.

Close modal

Xg expression enhances the invasive capacity of EWS cells

To investigate the effect of Xg positivity in Ewing's tumors, we transfected the naturally Xg-negative IOR/BRZ EWS cell line with an expression vector carrying Xg or with an empty vector. Using flow cytometry, two stable transfectants (Xg1 and Xg2) were selected for Xg expression at a similar level to that of EWS cell lines presented in Fig. 1C (Fig. 3A).

Figure 3.

Effect of Xg expression on the invasive capacity of EWS cells. A, analysis of Xg expression on IOR/BRZ cells: IOR/BRZ cells were transfected with either the Xg cDNA or with the empty vector; the level of expression of Xg was evaluated by flow cytometry. Solid line, cells stained using a PE-conjugated anti-Xg mAb NBL-1; dotted line, cells stained with a PE-isotype control. B, for migration assays, cells were seeded in the upper compartment of a Transwell chamber. For the invasion assays, Matrigel was added to the upper chamber to mimic the extracellular matrix. In the lower compartment, medium was supplemented (▪) or not (□) with 10% FBS as a source of chemoattractant. After 20 h, migrated cells were counted using Flow-Count Fluorospheres. Columns, mean of two independent experiments performed in triplicate; bars, SEM. Each histogram represents the mean ± SEM of two independent experiments performed in triplicate. *, P < 0.05, Dunnett's Method. C, analysis of Xg expression on TC-71 cells: TC-71 cells were transfected with a mix of two plasmids containing a shRNA targeting Xg or the empty vector; the level of expression of Xg was evaluated by flow cytometry. Solid line, cells stained using the PE-conjugated anti-Xg mAb NBL-1; dotted line, cells stained with a PE-isotype control. D, experiments were performed in the same conditions as in B. Columns, mean of two independent experiments performed in triplicate; bars, SEM. *, P < 0.05, Dunnett's Method.

Figure 3.

Effect of Xg expression on the invasive capacity of EWS cells. A, analysis of Xg expression on IOR/BRZ cells: IOR/BRZ cells were transfected with either the Xg cDNA or with the empty vector; the level of expression of Xg was evaluated by flow cytometry. Solid line, cells stained using a PE-conjugated anti-Xg mAb NBL-1; dotted line, cells stained with a PE-isotype control. B, for migration assays, cells were seeded in the upper compartment of a Transwell chamber. For the invasion assays, Matrigel was added to the upper chamber to mimic the extracellular matrix. In the lower compartment, medium was supplemented (▪) or not (□) with 10% FBS as a source of chemoattractant. After 20 h, migrated cells were counted using Flow-Count Fluorospheres. Columns, mean of two independent experiments performed in triplicate; bars, SEM. Each histogram represents the mean ± SEM of two independent experiments performed in triplicate. *, P < 0.05, Dunnett's Method. C, analysis of Xg expression on TC-71 cells: TC-71 cells were transfected with a mix of two plasmids containing a shRNA targeting Xg or the empty vector; the level of expression of Xg was evaluated by flow cytometry. Solid line, cells stained using the PE-conjugated anti-Xg mAb NBL-1; dotted line, cells stained with a PE-isotype control. D, experiments were performed in the same conditions as in B. Columns, mean of two independent experiments performed in triplicate; bars, SEM. *, P < 0.05, Dunnett's Method.

Close modal

The data described above indicate that Xg expression correlated with the survival of EWS patients. As metastatic dissemination is a determining factor of prognosis of EWS patients, we examined the functional role for Xg in the occurrence of metastases. We assessed the in vitro migration and invasion of two Xg transfectants (Xg1 and Xg2) compared with empty vector–transfected and wild-type (WT) IOR/BRZ cells. Experiments were performed over 20 hours using a Boyden chamber in the presence or absence of FBS, which was used as a chemoattractant. Forced expression of Xg significantly increased the number of migrating cells compared with that of empty vector–transfected and WT EWS cells (P < 0.05; Fig. 3B, top). In addition, Xg expression induced a significant increase in the capacity of EWS cells to invade Matrigel, which was used to mimic the extracellular matrix (P < 0.05; Fig. 3B, bottom). These data suggest a role for Xg in the invasiveness of EWS sarcoma.

To confirm the functional role(s) of Xg in EWS cells, Xg expression was silenced in the naturally Xg-positive TC-71 EWS cell line. As described in Materials and Methods section, two distinct transfectants obtained with a mix of two anti-Xg shRNA were selected for Xg negativity (Fig. 3C). Figure 3D shows that the silencing of Xg expression significantly decreased (P < 0.05) the migratory and invasive ability of TC-71 EWS cells. Thus, Xg regulates the invasive properties of EWS cells in vitro, suggesting an important role for Xg in metastatic progression of EWS.

Xg expression increases the metastatic ability of EWS cells in vivo

To investigate the importance of Xg in EWS tumor progression in vivo, WT, empty vector– and Xg-transfected IOR/BRZ cells were subsequently injected into nude mice, as indicated in the Materials and Methods section. The incidence of lung metastasis was increased in mice injected with Xg-transfected cells compared with WT and empty vector–transfected cells (Fig. 4A). In the same way, the cumulative number of lung metastases was three times higher in the Xg-injected groups of mice (Fig. 4B). To confirm the function of Xg on metastatic formation, we performed xenotransplantation in immunodeficient mice with WT, vector-, shXg1-, and shXg2-transfected TC-71 EWS cells. Consistent with these data, the shRNA-mediated knockdown of Xg in TC-71 cells decreased the incidence and the absolute number of lung metastases (Fig. 4C and D). Yet, the differences we observed between all the groups we tested did not reach significance; this could be due to the heterogeneity in the number and occurrence of metastases between all groups. Together, our results indicate an important role for Xg in EWS metastatic formation. On the basis that the occurrence of metastasis is linked to progression of EWS and so to the prognosis of EWS patients (33), our data suggest that Xg expression could be linked to the survival of patients through its involvement in metastatic formation.

Figure 4.

Xg expression increases the formation of metastases of EWS cells after grafting into immunocompromised mice. A, incidence of lung metastases in mice injected i.v. with IOR/BRZ, vector-, Xg1- or Xg2-transfected cells (n = 12 per group). B, incidence of lung metastases in mice injected i.v. with TC-71, vector-, shXg1-, or shXg2-transfected cells (n = 12 per group). C and D, cumulative number of lung metastases observed for each group of mice.

Figure 4.

Xg expression increases the formation of metastases of EWS cells after grafting into immunocompromised mice. A, incidence of lung metastases in mice injected i.v. with IOR/BRZ, vector-, Xg1- or Xg2-transfected cells (n = 12 per group). B, incidence of lung metastases in mice injected i.v. with TC-71, vector-, shXg1-, or shXg2-transfected cells (n = 12 per group). C and D, cumulative number of lung metastases observed for each group of mice.

Close modal

We report here the first characterization of a biological function of the Xg protein that is of clinical relevance to EWS patients. Xg, initially described as a RBC protein, was expressed on nearly all the EWS cell lines tested. Among the EWS primary tumors, we found that 24% of EWS cases expressed the Xg molecule, which was significantly associated with a shorter overall survival of EWS patients with localized tumors. Xg expression was indicative of strong tumor aggressiveness. As the invasiveness of cancer cells is dependent on migratory and invasive properties, we investigated the role of Xg in the motility of EWS cells. We observed a contribution of the Xg protein to tumor invasiveness and metastatic formation.

Thus far, Xg expression has been observed only on the RBC lineage. During erythropoïesis, Xg expression appears after band 3, i.e., from the orthochromatic normoblast stage up to mature erythrocytes. However, Northern blotting and reverse transcription-PCR showed that Xg could be expressed on other cell lineages (24, 29). The Xg mRNA has also been detected in fetal tissues (liver, spleen, and thymus). Using immunohistochemical staining on an array of normal tissues, we observed that Xg was particularly well expressed in the myometrium, kidney, lung, and skeletal muscle (summarized in the Table 2). Interestingly, the distribution of the Xg protein, although rather limited, is not restricted to the RBC lineage.

Table 2.

Xg distribution in normal tissues

TissuesXg expression
Myometrium +++ 
Lung +++; +/++ 
Kidney ++/+++ 
Seminal vesicle ++/+++ 
Skeletal muscle +/++ 
Thyroid 
Adrenal gland 
Heart 
Testis 
Endometrium 
Placenta (third trimester) 
Prostate (muscle) 
Liver +/−; + 
Spleen +/−; + 
Ovary (stromal) +/− 
Tonsil −; +/− 
Lymph node − 
Pancreas − 
Cerebellum − 
Cerebrum − 
Prostate − 
Breast − 
TissuesXg expression
Myometrium +++ 
Lung +++; +/++ 
Kidney ++/+++ 
Seminal vesicle ++/+++ 
Skeletal muscle +/++ 
Thyroid 
Adrenal gland 
Heart 
Testis 
Endometrium 
Placenta (third trimester) 
Prostate (muscle) 
Liver +/−; + 
Spleen +/−; + 
Ovary (stromal) +/− 
Tonsil −; +/− 
Lymph node − 
Pancreas − 
Cerebellum − 
Cerebrum − 
Prostate − 
Breast − 

NOTE: The staining intensity of healthy tissue samples labelled with an anti-Xg mAb was classified from high expression to negative expression. Strong positivity (+++; ++); positive (+); weak positivity (+/−); negative (−).

Because EWS behave as very aggressive tumors, they require intensive therapy including surgery, radiotherapy, and chemotherapy to abolish micrometastasis. EWS occurs in children and young adults with a long life expectancy; it seems particularly useful to identify high-risk patients to adapt the therapy regimen and avoid toxicity. Indeed, a series of prognostic laboratory markers have been described in EWS including lactate dehydrogenase, S-100 protein, CCN3, p53, and EWS FLI fusion transcripts (3443). Given the almost constant and the particularly high level of CD99 expression in EWS, we looked for Xg expression in these tumors in comparison with the most frequent bone tumors in children, namely osteosarcoma. Remarkably, we detected Xg at the mRNA level in EWS but not in osteosarcoma cell lines. The expression of Xg at the cell surface was confirmed by flow cytometry on 13 EWS cell lines, whereas it could not be detected at the osteosarcoma cell surface. As for primary tumors, immunohistochemistry analysis revealed, strikingly, that although 100% of EWS carry CD99, only 24% of patients displayed Xg. The expression of Xg in primary tumors of EWS patients was less frequent, possibly reflecting selection events. Xg expression in EWS patients has two major features: (a) it is independent of the gender of the patients and (b) it is not significantly linked to the level of CD99 expression. As pointed out in the introduction, CD99 is present in two isoforms due to alternative splicing (11). This is not the case for Xg, which was always expressed as a short form. Thus far we could not detect a long Xg isoform. Comparison of Xg and CD99 sequences shows that Xg shares the best homology with the short isoform of CD99 (data not shown). It is noteworthy that the forced expression of the short isoform of CD99 reduces cell-cell adhesion and enhances the cell migratory capacity (11, 13). Thus it is tempting to speculate that Xg acts in a similar fashion to the CD99 short form. Although we could not detect physical links between both molecular species, we could not exclude functional interactions between CD99 and Xg, which would reinforce the migratory capacity of the carrying tumor cells.

Therefore, we investigated the prognostic relevance of Xg expression and performed retrospective analysis on a cohort of 97 patients: 75 patients with localized tumors and 22 presenting with metastases at diagnosis. We found that in localized patients, ectopic expression of Xg is significantly linked to a shorter overall survival at 5 years (P = 0.047), indicating a putative role of this molecule in regulating the aggressiveness and metastatic ability of EWS cells.

To assess the direct effect of Xg on EWS progression, we injected immunodeficient mice with an IOR/BRZ cell line displaying forced Xg expression. We observed that forced expression of Xg in EWS cells lead to the upregulation of cellular migration and invasion in vitro and metastatic formation in vivo. Accordingly, our results, obtained with Xg-silenced EWS cell lines, confirmed the biological function of Xg. The function exerted by Xg is in line with the function already described for the other members of the CD99 family. Each of these proteins has a potent global effect on cellular adhesion and migration. Indeed, CD99 was described as being involved in the migration of different cell types (1618).

Taken together, our results show that Xg, a member of the CD99 family of molecules, is expressed in aggressive EWS tumors. This view, however, would not exclude the possibility that CD99 and Xg could act synergistically in EWS progression and that therapeutic strategies would benefit from a combined approach targeting both molecules.

No potential conflicts of interest were disclosed.

We thank J.P. Cartron for providing us with the Nbl1 hybridoma and the Xg expression vector.

Grant Support: INSERM, European project PROgnosis and THerapeutic targets in the Ewing family of TumorS, Ministère de la Recherche et des Technologies, Institut National du Cancer Cancéropôle PACA, Association pour la Recherche sur le Cancer, La Ligue contre le cancer, Fondation de France, Fondation de la Recherche Médicale, Agence Nationale de Biomédecine, Italian Association for Cancer Research, Associazione Italiana per la Ricerca sul Cancro (K. Scotlandi), and the Italian Ministry of Health (Strategico Oncologia RFPS-2006-3-340280; Alliance against Cancer).

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
Paulussen
M
,
Bielack
S
,
Jurgens
H
,
Casali
PG
,
On behalf of the EGWG
. 
Ewing's sarcoma of the bone: ESMO Clinical Recommendations for diagnosis, treatment and follow-up
.
Ann Oncol
2009
;
20
:
iv140
2
.
2
Arvand
A
,
Denny
CT
. 
Biology of EWS/ETS fusions in Ewing's family tumors
.
Oncogene
2001
;
20
:
5747
54
.
3
Burchill
SA
. 
Ewing's sarcoma: diagnostic, prognostic, and therapeutic implications of molecular abnormalities
.
J Clin Pathol
2003
;
56
:
96
102
.
4
Welford
SM
,
Hebert
SP
,
Deneen
B
,
Arvand
A
,
Denny
CT
. 
DNA binding domain-independent pathways are involved in EWS/FLI1-mediated oncogenesis
.
J Biol Chem
2001
;
276
:
41977
84
.
5
Castillero-Trejo
YES
,
Xiang
L
,
Richardson
JA
,
Ilaria
RL
. 
Expression of the EWS/FLI-1 oncogene in murine primary bone-derived cells Results in EWS/FLI-1-dependent, ewing sarcoma-like tumors
.
Cancer Res
2005
;
65
:
8698
705
.
6
Riggi
NCL
,
Provero
P
,
Suvà
ML
, et al
. 
Development of Ewing's sarcoma from primary bone marrow-derived mesenchymal progenitor cells
.
Cancer Res
2005
;
65
:
11459
68
.
7
Tirode
FL-DK
,
Prieur
A
,
Delorme
B
,
Charbord
P
,
Delattre
O
. 
Mesenchymal stem cell features of Ewing tumors
.
Cancer Cell
2007
;
11
:
421
9
.
8
Ambros
IMAP
,
Strehl
S
,
Kovar
H
,
Gadner
H
,
Salzer-Kuntschik
M
. 
MIC2 is a specific marker for Ewing's sarcoma and peripheral primitive neuroectodermal tumors. Evidence for a common histogenesis of Ewing's sarcoma and peripheral primitive neuroectodermal tumors from MIC2 expression and specific chromosome aberration
.
Cancer
1991
;
7
:
1886
93
.
9
LLombart-Bosch
A
,
Machado
I
,
Navarro
S
, et al
. 
Histological heterogeneity of Ewing's sarcoma/PNET: an immunohistochemical analysis of 415 genetically confirmed cases with clinical support
.
Virchows Arch
2009
;
5
:
397
411
.
10
Dworzak
MNFG
,
Printz
D
,
Zen
LD
, et al
. 
CD99 expression in T-lineage ALL: implications for flow cytometric detection of minimal residual disease
.
Leukemia
2004
;
18
:
703
8
.
11
Hahn
JHKM
,
Choi
EY
,
Kim
SH
, et al
. 
CD99 (MIC2) regulates the LFA-1/ICAM-1-mediated adhesion of lymphocytes, and its gene encodes both positive and negative regulators of cellular adhesion
.
J Immunol
1997
;
59
:
2250
8
.
12
Alberti
I
,
Bernard
G
,
Rouquette-Jazdanian
A
, et al
. 
CD99 isoforms expression dictates T cell Functional outcomes
.
FASEB J
2002
;
16
:
1946
8
.
13
Scotlandi
K
,
Zuntini
M
,
Manara
MC
, et al
. 
CD99 isoforms dictate opposite functions in tumour malignacy and metastases by activating or repressing c-Src kinase activity
.
Oncogene
2007
;
46
:
6604
18
.
14
Bremond
A
,
Meynet
O
,
Mahiddine
K
, et al
. 
Regulation of HLA class I surface expression requires CD99 and p230/golgin-245 interaction
.
Blood
2009
;
113
:
347
57
.
15
Bernard
G
,
Alberti
I
,
Pourtein
M
,
Widjenes
J
,
Ticchioni
M
,
Bernard
A
. 
CD99 (E2) up-regulates α4β1-dependent T cell adhesion to inflamed vascular endothelium under flow conditions
.
Eur J Immunol
2000
;
30
:
3061
5
.
16
Schenkel
ARMZ
,
Chen
X
,
Liebman
RM
,
Muller
WA
. 
CD99 plays a major role in the migration of monocytes through endothelial junctions
.
Nat Immunol
2002
;
3
:
143
50
.
17
Lou
O
,
Alcaide
P
,
Luscinskas
FW
,
Muller
WA
. 
CD99 is a key mediator of the transendothelial migration of neutrophils
.
J Immunol
2007
;
178
:
1136
43
.
18
Imbert
A-M
,
Belaaloui
G
,
Bardin
F
, et al
. 
CD99 expressed on human mobilized peripheral blood CD34+ cells is involved in transendothelial migration
.
Blood
2006
;
108
:
2578
86
.
19
Manara
MCBG
,
Lollini
PL
,
Nanni
P
, et al
. 
CD99 acts as an oncosuppressor in osteosarcoma
.
Mol Biol Cell
2006
;
17
:
1910
21
.
20
Kreppel
MAD
,
Schaefer
KL
,
Amann
G
,
Kofler
R
,
Poremba
C
,
Kovar
H
. 
Suppression of KCMF1 by constitutive high CD99 expression is involved in the migratory ability of Ewing's sarcoma cells
.
Oncogene
2006
;
25
:
2795
800
.
21
Ellis
NAYT
,
Patton
S
,
German
J
,
Goodfellow
PN
,
Weller
P
. 
Cloning of PBDX, an MIC2-related gene that spans the pseudoautosomal boundary on chromosome Xp
.
Nat Genet
1994
:
394
400
.
22
Suh
YH
,
Shin
YK
,
Kook
MC
, et al
. 
Cloning, genomic organization, alternative transcripts and expression analysis of CD99L2, a novel paralog of human CD99, and identification of evolutionary conserved motifs
.
Gene
2003
;
307
:
63
76
.
23
Bixel
MG
,
Petri
B
,
Khandoga
AG
, et al
. 
A CD99-related antigen on endothelial cells mediates neutrophil but not lymphocyte extravasation in vivo
.
Blood
2007
;
109
:
5327
36
.
24
Ellis
NATP
,
Petty
A
,
Reid
M
, et al
. 
PBDX is the XG blood group gene
.
Nat Genet
1994
;
8
:
285
90
.
25
Mann
JDCA
,
Gelb
AG
,
Fischer
N
, et al
. 
A sex-linked blood group
.
Lancet
1962
;
1
:
8
10
.
26
Goodfellow
PNTP
. 
A human quantitative polymorphism related to Xg blood groups
.
Nature
1981
;
289
:
404
5
.
27
Van Valen
F
. 
Ewing's sarcoma Family of tumors
. In:
Masters
JRW
,
Palsoon
B
, editors.
Human cell culture, Cancer cell lines
. 
1999
, p.
55
85
.
28
Fouchet
CGP
,
Cartron
JP
,
Lopez
C
. 
Quantitative analysis of XG blood group and CD99 antigens on human red cells
.
Immunogenetics
2000
;
51
:
688
94
.
29
Fouchet
CGP
,
Huet
M
,
Fellous
M
, et al
. 
A study of the coregulation and tissue specificity of XG and MIC2 gene expression in eukaryotic cells
.
Blood
2000
;
95
:
1819
26
.
30
Bacci
G
,
Longhi
A
,
Ferrari
S
,
Mercuri
M
,
Versari
M
,
Bertoni
F
. 
Prognostic factors in non-metastatic Ewing's sarcoma tumor of bone: an analysis of 579 patients treated at a single institution with adjuvant or neoadjuvant chemotherapy between 1972 and 1998
.
Acta Oncol
2006
;
45
:
469
75
.
31
Wiebe
T
,
Wiklund
T
,
Fossati Bellani
F
, et al
. 
Presentation of an Italian-Scandinavian treatment protocol for non-metastatic (ISG/SSG III) and high risk Ewing's family tumors (ISG/SSG IV)
.
Acta Orthop Scand
1999
;
70
:
28
9
.
32
Ferrari S, Alvegard T, Luksch R, et al. Non-metastatic Ewing's family tumors: High-dose chemotherapy with stem cell rescue in poor responder patients. Preliminary results of the Italian/Scandinavian ISG/SSG III protocol. J Clin Oncol 2007 Suppl. Vol. 25, 18S, p. 548, ASCO meeting, Chicago, US.
33
Cotterill
SJAS
,
Paulussen
M
,
Jürgens
HF
,
Voûte
PA
,
Gadner
H
,
Craft
AW
. 
Prognostic factors in Ewing's tumor of bone: analysis of 975 patients from the European Intergroup Cooperative Ewing's Sarcoma Study Group
.
J Clin Oncol
2000
;
18
:
3108
14
.
34
Llombart-Bosch
A
,
Contesso
G
,
Henry-Amar
M
, et al
. 
Histopathological predictive factors in Ewing's sarcoma of bone and clinicopathological correlations. A retrospective study of 261 cases
.
Virchows Archiv
1986
;
409
:
627
40
.
35
Picci
P
,
Rougraff
BT
,
Bacci
G
, et al
. 
Prognostic significance of histopathologic response to chemotherapy in nonmetastatic Ewing's sarcoma of the extremities
.
J Clin Oncol
1993
;
11
:
1763
9
.
36
Riley
RD
,
Burchill
SA
,
Abrams
KR
, et al
. 
A systematic review of molecular and biological markers in tumours of the Ewing's sarcoma family
.
Eur J Cancer
2003
;
39
:
19
30
.
37
Manara
MC
,
Perbal
B
,
Benini
S
, et al
. 
The Expression of ccn3(nov) Gene in Musculoskeletal Tumors
.
Am J Pathol
2002
;
160
:
849
59
.
38
De Alava
E
,
Kawai
A
,
Healey
JH
, et al
. 
EWS-FLI1 fusion transcript structure is an independent determinant of prognosis in Ewing's sarcoma [published erratum appears in J Clin Oncol 1998 Aug;16:2895]
.
J Clin Oncol
1998
;
16
:
1248
55
.
39
Zoubek
A
,
Dockhorn-Dworniczak
B
,
Delattre
O
, et al
. 
Does expression of different EWS chimeric transcripts define clinically distinct risk groups of Ewing tumor patients?
J Clin Oncol
1996
;
14
:
1245
51
.
40
De Alava
E
,
Antonescu
CR
,
Panizo
A
, et al
. 
Prognostic impact of P53 status in Ewing sarcoma
.
Cancer
2000
;
89
:
783
92
.
41
Matsunobu
T
,
Tanaka
K
,
Matsumoto
Y
, et al
. 
The prognostic and therapeutic relevance of p27kip1 in Ewing's family tumors
.
Clin Cancer Res
2004
;
10
:
1003
12
.
42
Ohali
A
,
Avigad
S
,
Zaizov
R
, et al
. 
Prediction of high risk Ewing's sarcoma by gene expression profiling
.
Oncogene
2004
;
23
:
8997
9006
.
43
Scotlandi
K
,
Remondini
D
,
Castellani
G
, et al
. 
Overcoming resistance to conventional drugs in Ewing sarcoma and identification of molecular predictors of outcome
.
J Clin Oncol
2009
;
27
:
2209
16
.

Supplementary data