Oncogene (2011) 30, 3661–3671 & 2011 Macmillan Publishers Limited All rights reserved 0950-9232/11 www.nature.com/onc ORIGINAL ARTICLE The human Rgr oncogene is overexpressed in T-cell malignancies and induces transformation by acting as a GEF for Ras and Ral

K Osei-Sarfo1, L Martello1,3, S Ibrahim2 and A Pellicer1

1Department of Pathology, New York University Langone Medical Center, New York, NY, USA and 2Department of Hematopathology, New York University Langone Medical Center, New York, NY, USA

The Ras superfamily of GTPases is involved in the Introduction modification of many cellular processes including cellular motility, proliferation and differentiation. Our laboratory Small GTPase are molecular switches that has previously identified the RalGDS-related (Rgr) transduce extra- and intracellular signals by cycling oncogene in a DMBA (7,12-dimethylbenz[a]anthracene)- between inactive GDP-bound and active GTP-bound induced rabbit squamous cell carcinoma and its human states. The cycling between inactive and active forms is orthologue, hRgr. In this study, we analyzed the expres- regulated by guanine nucleotide exchange factors sion levels of the human hRgr transcript in a panel (GEFs) and GTPase-activating proteins (GAPs). GEFs of human hematopoietic malignancies and found that activate these GTPases by inducing the dissociation of a truncated form (diseased-truncated (Dtr-hrgr)) was GDP and the binding of GTP, which is present at higher significantly overexpressed in many T-cell-derived concentration in cells. GAPs inactivate GTPases by neoplasms. Although the Rgr proto-oncogene belongs to hydrolyzing the g-phosphate group, resulting in a GDP- the RalGDS family of guanine nucleotide exchange bound molecule (Takai et al., 2001). The superfamily of factors (GEFs), we show that upon the introduction of GTPases can be subdivided into five distinct classes hRgr into fibroblast cell lines, it is able to elicit the (Ras, Rho, Rab, Sar1/Arf and Ran) based on sequence activation of both Ral and Ras GTPases. Moreover, homology (Takai et al., 2001). Ras , the more in vitro guanine nucleotide exchange assays confirm that extensively studied members, typically are mutated and/ hRgr promotes Ral and Ras activation through GDP or activated in approximately 30% of human cancers dissociation, which is a critical characteristic of GEF and are frequently mutated in several animal proteins. hRgr has guanine nucleotide exchange activity tumor models (Bos, 1989). Mutations involving one for both small GTPases and this activity was reduced of the three Ras isoforms have been described in when a point mutation within the catalytic domain several malignancies, including, but not limited to, (CDC25) of the , (cd) Dtr-hRgr, was utilized. squamous head and neck tumors, pancreatic adenocar- These observations prompted the analysis of the biological cinoma, colorectal malignancies, lung adenocarcinomas, effects of hRgr and (cd) hRgr expression in cultured cells. acute leukemias and myelodysplastic syndromes Here, we show that hRgr increases proliferation in low (Corominas et al., 1989; Rodenhuis, 1992). serum, increases invasion, reduces anchorage dependence Shortly upon the identification of Ras GTPases, Ral and promotes the progression into the S phase of the cell GTPases (Ral A and Ral B) were isolated and found to cycle; properties that are abolished or severely reduced in be downstream members of the Ras signaling pathway. the presence of the catalytic dead mutant. We conclude (Chardin and Tavitian, 1986). More specifically, Ral that the ability of hRgr to activate both Ral and Ras is GTPases were found to be activated by Ras GTPases responsible for its transformation-inducing phenotype and through the activation of a GEF for these GTPases, it could be an important contributor in the development of namely Ral guanine nucleotide dissociation stimulator some T-cell malignancies. (RalGDS) (Albright et al., 1993). Previous studies, Oncogene (2011) 30, 3661–3671; doi:10.1038/onc.2011.93; through inhibiting the expression of Ral GTPases, have published online 28 March 2011 shown that the activation of Ral GTPases cooperates with Ras-induced cellular transformation (Urano et al., Keywords: Rgr; GEFs; Ras GTPases; Ral GTPases; 1996; Lim et al., 2005). For many years, it was assumed T-cell lymphoma that Ral GTPases merely facilitated the cascade of Ras GTPase signaling events. Recent studies have deter- mined that the effectors of Ral GTPases have been Correspondence: Professor A Pellicer, Department of Pathology, implicated in vesicle transport (Camonis and White, New York University Langone Medical Center, 550 First Avenue, 2005; Rosse et al., 2006), endocytosis (Jullien-Flores MSB 594, New York, NY 10016, USA. et al., 2000), transcription (Frankel et al., 2005), E-mail: [email protected] development of filopodia (Ohta et al., 1999) and cellular 3Current address: Division of Gastroenterology and Hepatology, SUNY Downstate Medical Center, New York, NY, USA proliferation (Smith et al., 2006). In addition to these Received 13 August 2010; revised 7 January 2011; accepted 16 February effectors, recent research has determined that improper 2011; published online 28 March 2011 activation of Ral GTPases has been implicated in hRgr expression induces cellular transformation K Osei-Sarfo et al 3662 oncogenesis through their role in metastasis (Oxford In this study, we investigated the importance of the and Theodorescu, 2003; Lim et al., 2005, 2006), hRgr-truncated form in human leukemias and in the resistance to anoikis (Zahir et al., 2003), activation biological and biochemical aspects of cellular transfor- of transcription factors (Frankel et al., 2005) and mation. In addition, we show that the abnormal inappropriate cell survival signals (Chien et al., 2006; transcript of hRgr is overexpressed in a wide range of Smith et al., 2006). human hematological malignancies. Our results reveal Like Ras GTPases, Ral GTPases have their own that the expression of the truncated protein produced by cadre of activating proteins or GEFs. On the basis of the the abnormal transcript of hRgr can induce cellular elucidation of the first RalGEF, RalGDS (Albright transformation through the activation of Ras and Ral et al., 1993; Hofer et al., 1994), several other GEFs for GTPases. Ral GTPases have been uncovered, including Rlf (Wolthuis et al., 1996), Rgl 1-3 (Shao and Andres, 2000; Sood et al., 2000; Takaya et al., 2007), RalGPS 1-2 Results (Ceriani et al., 2007; Tazmini et al., 2009) and AND-34 (Gotoh et al., 2000). The majority of these GEFs are The abnormal hRgr transcript is overexpressed in T-cell activated by Ras GTPases through a Ras-binding malignancies domain (RBDs), whereas RalGPS 1-2 and AND-34 do To determine the expression patterns of the abnormal not have detectable RBDs. As several RalGEFs do not hRgr transcript in hematological malignancies, a quan- have RBDs, studies have confirmed that Ral activation titative real-time (QRT)–PCR screen of T- and B-cell can be achieved through Ras-independent mechanisms neoplasms and normal tissues was conducted. To detect (Linnemann et al., 2002). Several groups have observed the presence of the abnormal transcript, we utilized Ras-independent activation of Ral GTPases through primers from the intronic region that becomes exonic at 0 signals propagated by the Ca2 þ ionophore ionomycin the 5 -end of exon 9 and from a portion of the normal (Hofer et al., 1998), b-arrestins and the formyl-Met-Leu- exon 9 sequence (Figure 1a). As shown in Table 1, Phe receptor (Bhattacharya et al., 2002), nitric oxide the abnormal hRgr transcript was only observed in (Mittar et al., 2004) and JAK/STAT 3 activation (Senga T-cell neoplasms, including T-ALL, peripheral T-cell et al., 2001). lymphomas, mycosis fungoides, anaplastic large T-cell The RalGDS-related (Rgr) oncogene was isolated in lymphoma, adult T-cell lymphoma leukemia and our laboratory from a DMBA (7,12-dimethylbenz[a]an- angioimmunoblastic T-cell lymphoma. Figure 1b, which thracene)-induced squamous cell carcinoma and was quantifies the expression of the abnormal transcript found to induce tumorigenesis in the nude mouse assay from selected samples, shows that the abnormal (D’Adamo et al., 1997). Protein sequencing analysis transcript is highly overexpressed in T-cell neoplasms. determined that Rgr had 40% sequence identity to In addition, the amplicon produced by the QRT–PCR RalGDS and up to 72% identity within the CDC25 of a T-ALL sample was sequenced and was found to be domain (D’Adamo et al., 1997). The mechanism of located on 22 at the expected location of enhanced guanine nucleotide dissociation caused by Rgr hRgr (data not shown). was due to overexpression through a loss of transla- For a comparative analysis, we sought to observe the tional regulation by the elimination of 50-inhibitory putative expression of the normal hRgr transcript in the sequences (Hernandez-Munoz et al., 2003). The human samples mentioned previously. Thus, QRT–PCR analy- orthologue of Rgr (hRgr) was also identified in our sis was used to characterize the expression pattern of the laboratory by screening a human testis cDNA library normal transcript by using primers directed to exon 5 and was found to have 13 exons, of which the first three (Figure 1a), which is not present in the abnormal hRgr exons contained a 50-untranslated region (UTR) (Leo- transcript. Here, we find that the normal transcript has a nardi et al., 2002). Further analysis of the hRgr sequence low expression pattern in both T- and B- cell neoplasms found a significant nucleotide similarity to an expressed as well as in normal tissues (Figure 1c). sequence tag from Jurkat cells, which is a T-cell acute These data confirm that both a truncation and the lymphoblastic leukemia (T-ALL)-derived cell line exonic expression of a portion of an intronic sequence (Leonardi et al., 2002). Northern blotting and lead to the overexpression of the abnormal hRgr reverse transcription-PCR (RT–PCR) of Jurkat cells transcript only in neoplasms derived from the T-cell showed a severely truncated transcript of hRgr lineage. In addition, these data suggest that a genomic (Leonardi et al., 2002). In addition, the truncated alteration, possible due to a rearrangement on chromo- transcript-initiated protein translation by using a start some 22, may initiate the abnormal expression of hRgr codon within an intronic region that allows the open- culminating in the development or maintenance of T-cell reading frame to be maintained through exons 9–13. malignancies. Interestingly, Rgr does not contain an RBD or a Ras exchange motif typically seen in RalGEFs (D’Adamo Rgr activates Ras and Ral GTPases in vivo et al., 1997; Leonardi et al., 2002). However, the lack of As previous studies from our laboratory have showed the RBD and Ras exchange motifs has been observed in that the ability of rabbit Rgr to transform cells is other RalGEFs, including RalGPS1 (Tazmini et al., dependent on Ras (Hernandez-Munoz et al., 2000), we 2009), RalGPS2 (Ceriani et al., 2007) and AND-34 wanted to confirm this ability for hRgr. To determine (Gotoh et al., 2000). this property, we used effectors for activated Ras (RBD

Oncogene hRgr expression induces cellular transformation K Osei-Sarfo et al 3663

Figure 1 Expression of the abnormal and normal hRgr transcripts. The abnormal hRgr transcript is overexpressed only in T-cell neoplasms, whereas the normal hRgr transcript has low levels of expression in T- and B-cell neoplasms and in normal tissues. (a) The genomic organization of the normal (Fl-hRgr; top), full-length truncated (Tr-hRgr; middle) and abnormal (Dtr-hRgr; bottom) hRgr transcripts. The untranslated region (UTR) is represented by the white box; exons by red vertical boxes; and introns by green horizontal boxes. The green vertical box designates the coding sequence that is present in the abnormal transcript, which is intronic in the normal transcript. The black arrows represent primer sets designed against specific regions of hRgr. (b, c) QRT–PCR analysis of selected T- and B-cell neoplasms and normal tissues to determine the expression of the abnormal (b) and normal (c) hRgr transcripts, respectively. Total RNA was harvested from cyropreserved patient samples, reverse transcribed and amplified using specific hRgr primers for the abnormal and normal transcripts. Data in this QRT–PCR analysis were normalized by using primers specific for human glyceraldehyde 3-phosphate dehydrogenase. Results represent at least four independent experiments. of Raf-1) and Ral A (Ral-binding domain of Ral mant-GDP exchange assay was utilized. This assay binding protein 1) to measure the amount of active or measures the ability of a GEF to dissociate GDP from a GTP-bound Ras and Ral in cells stably expressing Rgr GTPase by observing the amount of mant-GDP released (rabbit Rgr (R-Rgr) and Dtr-hRgr). In addition, we over time. For this analysis, we used the full-length used a catalytic dead mutant of Dtr-hRgr ((cd) Dtr- truncated hRgr (Tr-hRgr), which is a derivative of hRgr hRgr), which has a point mutation within the catalytic that lacks the 50-UTR of the full-length hRgr or CDC25 domain. We found that the expression of (Figure 1a). Tr-hRgr was used to determine if only the Dtr-hRgr in NIH3T3 cells increases the activation of complete coding sequence of hRgr could induce a higher Ras GTPases compared with non-transfected NIH3T3 amount of GDP dissociation on Ras and Ral GTPases. cells (Figures 2a and c). Consistent with these results, It is important to note that Tr-hRgr is a purely synthetic NIH3T3 cells that stably express Dtr-hRgr and R-Rgr form that has not been naturally observed. Here 6X- increased the activation of Ral A compared with non- histidine-tagged Rgr fusion proteins (Tr-hRgr, Dtr- transfected NIH3T3 cells and NIH3T3 cells expressing hRgr and R-Rgr) have the ability to dissociate GDP an ‘empty’ pCGN vector (Figures 2b and d). Although from Ral A (Figure 3a and Supplementary Figure 1), slight, Ras and Ral GTPase activation was reduced in Ral B (Figure 3b and Supplementary Figure 1) and cells expressing (cd) Dtr-hRgr. Interestingly, cells that N-Ras (Figure 3c and Supplementary Figure 2). expressed R-Rgr had a higher amount of Ral A Between the hRgr constructs (Tr-hRgr and Dtr-hRgr), activation compared with those expressing constitutively we did not observe any significant differences in mant- active N-Ras (N-Ras V12), which served as a positive GDP dissociation. Also, we found that the catalytic control (Figure 2b). This observation is appropriate dead mutants of Dtr-hRgr, (cd) Dtr-hRgr, and Tr-hRgr, because of the dual nature of Rgr-induced activation of (cd) Tr-hRgr, had comparable dissociation properties to Ras and Ral GTPases. Rgr could activate Ral A by Ras- the negative control experiment samples (the GTPase dependent events (as a GEF for Ras) and also directly alone and the GTPase incubated with GST, a non-GEF activate Ral A (as a GEF for Ral). The activation of Ras molecule; Figure 3). and Ral A GTPases by hRgr could account for the In this study, we found that the more characterized induction or maintenance of T-cell malignancies. GEFs for Ral A and Ral B (RalGDS, Rlf and RalGEF 2) and N-Ras (Sos and RasGRP 1) had a higher rate of Rgr enhances GDP dissociation from N-Ras, Ral A mant-GDP dissociation within the first 200 s compared and Ral B with the Rgr constructs (Supplementary Figures 1 and To gain a better perspective concerning the mechanism 2). Even though the Rgr constructs had less dissociation of Ras and Ral GTPases activation by Rgr, the activity than the more characterized GEFs for N-Ras

Oncogene hRgr expression induces cellular transformation K Osei-Sarfo et al 3664 Table 1 The presence of the abnormal hRgr transcript in patient those expressing Dtr-hRgr. In addition to increased samples cellular proliferation, Dtr-hRgr expression in NIH3T3 Expression of Number of cells promoted increased colony formation in soft abnormal hRgr samples agar compared with non-transfected NIH3T3 cells (Figure 4b). T-cell neoplasms As Figure 5 shows, both Dtr-hRgr and R-Rgr can T-ALL 11 11 PTCL 2 2 also alter invasive properties in vivo. Stably expressing MF 1 1 Rgr cells were subjected to the modified Boyden T-ALCL 2 2 chamber assay, which measures the ability of trans- T-ATLL 1 1 formed cells to invade and migrate through an extra- T-AILT 2 2 Total 19 19 cellular matrix. Consistent with the biological results observed above, we found that Dtr-hRgr-expressing B-cell neoplasms cells display enhanced invasive properties. We found B-ALL 0 2 that there was close to three times the amount of cellular CLL 0 2 invasion in cells expressing Dtr-hRgr, relative to non- MCL 0 3 FL 0 3 transfected NIH3T3 cells. In addition, cells expressing HL 0 2 (cd) Dtr-hRgr had invasive properties that were DLBCL 0 2 comparable to non-transfected NIH3T3 cells (Figure 5). MZL 0 2 Taken together, the biological assays described herein Total 0 16 confirm that the expression of the abnormal hRgr Normal tissue protein can alter biological properties, which were Thymus 0 1 consistent with the results observed in the in vitro and Spleen 0 2 in vivo Ras and Ral GTPases activation analyses. Tonsil 0 1 To determine a biochemical connection to the Liver 0 1 Adenoid 0 1 biological processes altered by Rgr, cell cycle progres- Blood 0 1 sion analysis was conducted by flow cytometry. Analysis Lymph node 0 2 of cell cycle progression showed that Dtr-hRgr and Total 0 9 R-Rgr caused a moderate, but significant, increase in the S-phase events (Figure 6). Consistent with this observa- Abbreviations: ALCL, anaplastic large T-cell lymphoma; B-ALL, B-cell acute lymphoblastic leukemia; CLL, chronic lymphocytic tion, we found that there was a reduction of G0/G1 leukemia; DLBCL, diffuse large B-cell lymphoma; FL, follicular events in cells expressing Dtr-hRgr or R-Rgr. These lymphoma; HL, Hodgkin’s lymphoma; MCL, mantle cell lymphoma; results suggest that Rgr can drive cell cycle events, which MF, mycosis fungoides; MZL, marginal zone lymphoma; PTCL, could be a contributing factor in the induction of a peripheral T-cell lymphoma; T-AILT, angioimmunoblastic T-cell malignant phenotype. lymphoma; T-ALL, T-cell acute lymphoblastic leukemia; T-ATLL, Adult T-cell leukemia/lymphoma.

and Ral GTPases, these results confirm that Rgr has a The abnormal hRgr transcript contributes to the role in the activating properties for Ras and Ral malignant phenotype of T-cell malignancies GTPases through the dissociation of GDP. In this study as well as in previous studies (Leonardi et al., 2002), it has been established that the abnormal hRgr transcript is overexpressed in T-ALL-derived cell hRgr alters biological and biochemical processes and lines and neoplasms. To fully understand the influence initiates cellular transformation of transcript expression in the maintenance of a To determine the importance of Rgr-mediated cellular malignant phenotype in these cell lines, RNA interfering transformation, we characterized the alteration of oligonucleotides directed against hRgr were synthesized biological activities of Rgr stably expressing NIH3T3 to inhibit the expression of the abnormal hRgr cell lines. Previous results confirmed that R-Rgr could transcript. RNA interfering oligonucleotides directed induce cellular transformation (D’Adamo et al., 1997; against two regions of hRgr were introduced into Hernandez-Munoz et al., 2000, 2003). As we have T-ALL 1 and Jurkat cells, which were cultured in low shown that the abnormal hRgr transcript can modulate serum conditions, and cell cycle events were measured. the activation of Ras and Ral GTPases, it was necessary Here, we found that Jurkat cells (Figure 7a) and T-ALL to confirm that hRgr-induced activation of these 1 cells (Figure 7b) transfected with hRgr RNA interfer- GTPases could contribute to cellular transformation. ing oligonucleotides (hRgr Oligo 1) had a significant NIH3T3 cells expressing Dtr-hRgr and R-Rgr promoted reduction in the S phase and an increase in the G0/G1 cellular proliferation in low serum (1%) conditions phase in comparison to non-transfected cells or those (Figure 4a). These results were significant by day 9 as transfected with the non-targeting oligonucleotides. As shown by the Kruskal–Wallis rank-sum test and an experimental control, we confirmed using QRT–PCR analysis of variance when compared with non-trans- analysis that hRgr Oligo 1 decreased hRgr expression by fected NIH-3T3 cells. Supporting the role of hRgr- at least 65% (Supplementary Figure 3). These data induced transformation, cells expressing (cd) Dtr-hRgr indicate that proper translation of the abnormal hRgr had reduced proliferation when compared with transcript stimulates cellular proliferation by promoting

Oncogene hRgr expression induces cellular transformation K Osei-Sarfo et al 3665

Figure 2 hRgr increases the levels of GTP-bound forms of Ras and Ral A in vivo.(a, b) Cumulative expression of activated Ras (a) and Ral A (b) GTPases relative to non-transfected NIH3T3 cells in three independent experiments; *Po0.05 and ***Po0.001. (c, d) A representative experiment for Ras (c) and Ral A (d) activation, in which protein lysates (1 mg) extracted from non-transfected NIH3T3 cells and stably expressing Rgr and N-RasV12 cells were used to recover the GTP-bound forms of Ras (with GST-Raf 1-RBD fusion protein) and Ral A (with GST-Ral BP1-Ral binding domain fusion protein); upper panels. Total Ras and Ral A present in 20 mg of the lysates used for the activation assays; lower panels. (e) SDS–PAGE analysis to determine the expression of (cd) Dtr-hRgr, Dtr-hRgr, R-Rgr and N-Ras V12 with anti-HA antibody. the entrance of these cells into the S phase. The results transcript initiated translation using a portion of the from these experiments are consistent with the postu- 50-intronic sequence preceding exon 9 that becomes lated role in hRgr-induced transformation for T-cell exonic in the abnormal form (Leonardi et al., 2002). The neoplasms. aim of this study was to further characterize the biological and biochemical roles that hRgr have in cellular transformation. Discussion

Previous studies in our laboratory have isolated Rgr as a Abnormal hRgr is an oncogene expressed in T-cell GEF with significant homology to RalGDS with the malignancies ability to dissociate guanine nucleotides from Ral In this study, we performed an extensive screening of GTPases (D’Adamo et al., 1997; Hernandez-Munoz various hematological malignances and normal tissues et al., 2000). In addition to its dissociation activity, it to determine the expression patterns of hRgr’s tran- was determined that Rgr could induce cellular transfor- scripts, both normal and abnormal. Consistent with mation through the loss of translational regulation by previous results (Leonardi et al., 2002), we detected the the elimination of 50-inhibitory sequences (Hernandez- abnormal hRgr transcript in a wide range of T-cell Munoz et al., 2003). The human version of Rgr (hRgr) neoplasms and its levels of expression were significantly was identified in our laboratory through the screening of elevated. In the same hematological neoplasms a human testis cDNA library (Leonardi et al., 2002). and normal tissue samples, we established that the Through additional examination, a severely truncated normal transcript was very lowly expressed. This is transcript was observed in an mRNA sequence derived consistent with the low levels of expression observed for from an expressed sequence tag of the Jurkat cell line, the wild-type Rgr transcript in most rabbit tissues which is a T-ALL-derived cell line. Also, this truncated (D’Adamo et al., 1997). RNA interfering knockdown

Oncogene hRgr expression induces cellular transformation K Osei-Sarfo et al 3666

Figure 3 hRgr has GDP dissociation activity on Ral A, Ral B and N-Ras. In vitro fluorescence-based guanine nucleotide exchange assays were performed to determine the amount of GDP dissociation from Ral A, Ral B and N-Ras induced by Rgr and more characterized GEFs for Ras and Ral GTPase. Purified 6X His recombinant fusion GTPase proteins, preloaded with mant-GDP were incubated with GTP and purified (GST or a GEF) and GDP dissociation was measured as the decrease in fluorescence for 600 s. Tr-hRgr, Dtr-hRgr and R-Rgr had the ability to dissociate mant-GDP from Ral A (a), Ral B (b) and N-Ras (c). However, this dissociation was typically less than the more characterized GEFs for Ral A and B (RalGDS, Rlf and RalGEF 2) and N-Ras (Sos and RasGRP 1). In addition, the addition of a point mutation within the catalytic region of hRgr, (cd) Tr-hRgr and (cd) Dtr-hRgr, severely perturbed this dissociation activity. Data shown are representative of at least three experiments, except for (cd) Tr-hRgr and (cd) Dtr-hRgr (n ¼ 2); ***Po0.001.

Figure 4 hRgr expression increases proliferative growth and anchorage independence in NIH3T3 cells. (a) Cells stably expressing Dtr-hRgr and R-Rgr increase cellular proliferation over 15 days when compared with those expressing (cd) Dtr-hRgr, an empty vector and non-transfected NIH3T3 cells. Cell proliferation was determined by the MTT assay, in which 500 cells were cultured in low serum (1%) conditions. Every three days for 15 days, cellular proliferation was measured by the absorbance due to the conversion of yellow MTT to purple formazin. Data are represented by six independent experiments performed in triplicate. Statistical relevance was determined by the Kruskal–Wallis test (using each plate as a testing unit) and analysis of variance (using each well as a testing unit). (b) Fibroblasts that express Dtr-hRgr and R-Rgr reduce anchorage dependence, relative to non-transfected NIH3T3 cells. The soft agar assay for anchorage independence was conducted by plating 1000 cells (expressing empty pCGN, Dtr-hRgr, R-Rgr and N-Ras V12) resuspended in 0.25% onto 0.5% solidified agar. The formation of colonies was scored 4 weeks post-plating. These data are representative of three independent experiments, performed in triplicate; **Po0.01 and ***Po0.001.

showed that blocking translation of Dtr-hRgr These observations advance the hypothesis that a could lead to reduced cell cycle progression. These genomic modification or rearrangement may cause the data suggest that hRgr may have a role in main- overexpression of the abnormal transcript, which con- taining the malignant phenotype of T-ALL-derived tains an intronic sequence that becomes exonic. The cell lines. long arm of chromosome 22 is highly recognized as a

Oncogene hRgr expression induces cellular transformation K Osei-Sarfo et al 3667

Figure 6 hRgr expression in NIH3T3 increases the proportion of cells in the S phase and reduce those in the G0/G1 phase. Stably (cd) Dtr-hRgr-, R-Rgr- and N-Ras V12-expressing NIH3T3 cells (10 000) were subjected to propidium iodide staining to determine hRgr’s role in cell cycling events. Data are representative of at least three independent experiments; *Po0.05. Figure 5 NIH3T3 cells that express Dtr-hRgr have increased invasion properties and this phenotype can be perturbed in cells expressing (cd) Dtr-hRgr. (a) Invasion assay using non-transfected chromosome 22 in adult ALL (Paulsson et al., 2008) and NIH3T3 cells or those stably expressing an empty vector, (cd) Dtr- mesotheliomas (Sekido et al., 1995). hRgr, Dtr-hRgr, R-Rgr and N-Ras V12 were carried out in 0.125 mg/ml Matrigel-coated Boyden chamber stimulated with hRgr activates Ras and Ral GTPases conditioned medium, containing 10% bovine serum. Data are representative of at least four independent experiments; Here, the abnormal version of hRgr (Dtr-hRgr) can ***Po0.001. (b) Pictographs of an individual representative activate Ras and Ral GTPases in vivo and in vitro. experiment, in which data were acquired from three randomly Previous studies have shown that Rgr can activate selected fields. extracellular signal-regulated kinase 1/2, Jun N-terminal kinase 1/2 and p38 through activating the Ras GTPase signaling pathway (Hernandez-Munoz et al., 2000). These data suggest that hRgr would activate these participant of the Philadelphia translocation, which kinases in vivo, as they are downstream participants of involves reciprocal translocation of a portion of the Ras GTPase signaling pathway. In addition, we chromosome 22 with (Rowley, 1973). found that the introduction of a point mutation within More specifically, this chromosomal translocation, the catalytic domain of hRgr can severely impair its t(9;22)(q34;q11), causes the juxtaposition of BCR and guanine nucleotide dissociation activity in vitro. This ABL genes forming an oncogenic fusion protein (Muller result is consistent with other studies in which a similar et al., 1991). It was further determined that genomic point mutation was introduced into the CDC25 domain translocations or rearrangements on chromosome 22 of Sos (Hall et al., 2001). were identified in 90–95% of chronic myelogenous Although both human and rabbit Rgr constructs were leukemias, 20% of adult ALLs, 5% of pediatric ALLs able to activate Ras and Ral GTPases in vitro, the and 2% of acute myeloid leukemias (Kurzrock et al., amount of GDP dissociation was considerably less than 2003). Owing to the proximity of hRgr to the region GDP dissociation initiated by the better characterized involving the translocation of BCR on chromosome 22, Ras and Ral GEFs (that is, Sos, RasGRP, RalGDS, it is highly probable that hRgr may participate in the RalGEF 2, and so on). This weaker dissociation activity Philadelphia translocation. It has been shown that the could suggest that Rgr may require a cooperating BCR gene translocation can extend over a 200 kb region molecule to stimulate more robust dissociation activity. (Chissoe et al., 1995), which is well within the reach of For example, Sos facilities the GDP dissociation from hRgr on chromosome 22. At this point, we are not in a Ras and can couple this activity to Rac GTPases position to suggest that hRgr have a role in this disease, through its Dbl and pleckstrin homology domains but this would be an intriguing possibility to explore in (Sondermann et al., 2004). This phenomenon suggests the future. Another possible mechanism for the genera- the possibility that hRgr may interact with other tion of the abnormal hRgr transcript is through genomic proteins resulting in the activation of these GTPases. rearrangements caused by microdeletions. Studies Nevertheless, as evident by the in vivo and in vitro have confirmed the involvement of microdeletions of activation of Ras and Ral GTPases, hRgr displays its

Oncogene hRgr expression induces cellular transformation K Osei-Sarfo et al 3668 domain of hRgr reduced the malignant phenotype in these fibroblasts, confirming that hRgr initiates trans- formation by guanine nucleotide dissociation. These results are consistent with studies that have shown that both Ral A and Ral B GTPases can cooperate with other oncogenes, resulting in cellular transformation (Tchevkina et al., 2005; Rosse et al., 2006). Although hRgr can activate both Ras and Ral A GTPases in vitro, perhaps it may have a stronger dissociation activity on Ral in vivo. Although Ral A and Ral B have 85% similarity to each other, the interactions of these isoforms with specific RalGEFs may vary. For instance, specific RalGEFs can modify downstream activities of Ral A and Ral B in cytokinesis (Ceriani et al., 2007). Ral A can be activated by RalGDS and RalGPS 2 through Ras-dependent and -indepen- dent mechanisms (Cascone et al., 2008). Similar GEF specificity has been observed for the activation of Ral B by RGL and RalGPS 1 in a Ras-dependent and -independent manner, respectively (Cascone et al., 2008). Perhaps, in line with these studies, hRgr may have a specific role in vivo with a particular GTPase.

Cooperation of different GEFs could enhance transformation Blocking the translation of Dtr-hRgr using small interfering RNA reduced, but did not completely abolish, cell cycle progression in T-ALL-derived cell lines. This observation suggests that hRgr is not the only oncogene driving transformation in these cell lines. Recently, interesting studies characterizing Ras GTPase Figure 7 Inhibiting the abnormal hRgr transcript reduces the activation in lymphocytes showed a cooperation among proliferation of T-ALL-derived cell lines. (a, b) Abnormal hRgr Ras GEFs, Sos and RasGRP 1. Structural analysis of transcript silencing with hRgr-specific siRNA (hRgr oligo 1) or Sos confirms that Sos has two GTP-bound Ras binding negative control siRNA (non-targeting oligo) oligonucleotides in Jurkat (a) and T-ALL 1 (b) cell lines cultured in low serum pockets (Margarit et al., 2003). Loading one of these conditions, respectively. The effects of hRgr silencing were assessed pockets with an active form of Ras enhances GEF by propidium iodide (PI) staining; the reduction of hRgr expression activity of Sos, suggesting a positive feedback mechan- caused an increase in cells in the G0/G1 phases and a reduction in ism for the activation of Ras by Sos (Boykevisch et al., those in the S phase. Data are representative of four independent 2006). In lymphocytes, Ras can be activated through experiments; *P 0.05. o Sos- or RasGRP-mediated guanine nucleotide dissocia- tion mechanisms. Small interfering RNA studies con- oncogenic mechanism through its dissociation activity. firmed that RasGRP 1, a RasGEF that is specifically The dual activation capabilities reported here for expressed in T lymphocytes, and Sos could act in hRgr provide the molecular bases for its transforming concert to activate Ras GTPases in T lymphocytes activity, which allows a single molecule to activate both (Roose et al., 2007). As the catalyst of Sos-induced Ras Ras and Ral. activation is its own product (that is, GTP-bound Ras), it is necessary for T lymphocytes to have an additional hRgr can modulate biological and biochemical activities molecule capable of creating a GTP-bound Ras important for cellular transformation molecule (Roose et al., 2007; Das et al., 2009). In this As confirmed through biological assays, Dtr-hRgr can study, we found that abnormal hRgr is overexpressed in modify several biological functions, including cellular malignant T lymphocytes and has the ability to proliferation, migration and invasion, and anchorage dissociate GDP, which suggests that hRgr could provide independence. Interestingly, of all the biological assays activated Ras molecules (albeit in a lower amount performed in this study, Dtr-hRgr had a stronger effect compared with RasGRP 1) to initiate Sos’ positive on the migratory and invasive properties of murine feedback loop. fibroblasts. Several studies have implicated Ral GTPases In this report, we have shown that the abnormal hRgr in the mobilization of Sec5 and Exo84 of the exocyst, is an oncogene that augments cellular transformation driving cellular migration and facilitating cellular through the activation of Ras and Ral GTPases. As the transformation (Oxford et al., 2005; Rosse et al., abnormal hRgr transcript was observed in several 2006). In addition, a mutation within the CDC25 subsets of T-cell neoplasms, this study suggests that

Oncogene hRgr expression induces cellular transformation K Osei-Sarfo et al 3669 hRgr should be investigated as a diagnostic tool for Odyssey Blocking Buffer (LI-COR Biosciences, Lincoln, NE, determining subsets of T-cell malignancies, as a poten- USA), filters were incubated with the appropriate antibodies tial prognostic marker and/or as a target for the for Cullin 1 (Invitrogen, Carlsbad, CA, USA) or hybridoma development of therapeutic compounds against these clone (12CA5) HA epitope (gift from Dr D Levy, NYU malignancies. Langone Medical Center, New York, NY, USA). Then, the filters were incubated with IRDye 800 anti-goat immunoglo- bulin G (LI-COR Biosciences) and scanned on the Odyssey Materials and methods imager (LI-COR Biosciences). When necessary, the filters were subjected to densitometry by analysis with Odyssey Applica- Further details and additional methods are provided as tion software, version 3.0 (LI-COR). Supplementary Information. Ras and Ral A pull-down assays Cell culture and patient materials Ras and Ral A activation detection assays were performed Detailed descriptions of cell culture and patient samples are using Active Ras and Ral Pull-Down and Detection Kits provided in the Supplementary Experimental procedures. (Thermo Fisher Scientific, Rockford, IL, USA). See Supple- mentary Experimental procedures for more details. Plasmid construction and generation of (cd) hRgr mutants Description is provided in Supplementary Experimental Fusion protein expression and purification procedures. For the detailed description for the purification of 6X His fusion proteins of Tr-hRgr, Dtr-hRgr, (cd) Dtr-hRgr, R-Rgr, Sos, RasGRP 1, RalGEF 2, Rlf, RalGDS, N-Ras, Ral A or Transfections Ral B expressed in ArticExpress Escherichia coli (Stratagene, NIH3T3 cells were transfected by the calcium phosphate Santa Clara, CA, USA), see the Supplementary Experimental precipitation method, as described previously (Wigler et al., procedures. 1978). Descriptions are provided in Supplementary Experi- mental procedures. Guanine nucleotide dissociation assays Guanine nucleotide dissociation assays were conducted on N- RNA preparation and quantitative real-time (QRT–PCR) Ras, Ral A or Ral B loaded with mant-GDP. See Supplemen- QRT–PCR was performed using iCYCLER iQ5 with a SYBR tary Experimental procedures for more details. green master mix (Bio-Rad, Hercules, CA, USA). Gene- specific oligonucleotides were used for abnormal hRgr, normal hRgr and human glyceraldehyde 3-phosphate dehydrogenase; Statistical analysis see details in Supplementary Experimental procedures. Analysis of variance was used to analyze differences between a control group and experimental groups, with *Po0.05, **P 0.01 and ***P 0.001. For the MTT assay for cellular Cell cycle analysis o o proliferation, analysis of variance was used to treat each well At least 10 000 events of ethanol-fixed, RNase A-treated and as a testing unit and the Kruskal–Wallis test was used to treat propidium iodide-stained cells were acquired using a BD LSR each plate as the testing unit. II flow cytometer (BD Biosciences, San Jose, CA, USA). Resolution of specific cell cycle stages was determined by ModFit LT software (Verity Software House, Topsham, ME, Conflict of interest USA). See Supplementary Experimental procedures for more details. The authors declare no conflict of interest. Biological assays to determine cellular invasion, cellular proliferation and cellular anchorage independence Acknowledgements Cellular invasion, cellular proliferation and cellular anchorage independence were measured by the modified Boyden chamber We are grateful to Drs M Philips, D Bar-Sagi and J Bos assay, MTT assay for cellular proliferation and soft agar assay for the gift of expression plasmids. We thank Kamlesh for anchorage independence, respectively. See Supplementary Yadav and Pamela Sung for their assistance with the mant- Experimental procedures for more details. guanine nucleotide exchange and cellular proliferation assays, respectively, and Dr M Liu for her assistance with the Western blot analysis statistical analysis for the cellular proliferation assay. These Western blots were performed as described previously (Garcia- studies were supported by NIH Grants CA50434 and Espana et al., 2005). Membranes were blocked for 1 h with CA36327 to AP.

References

Albright CF, Giddings BW, Liu J, Vito M, Weinberg RA. (1993). Bos JL. (1989). ras oncogenes in human cancer: a review. Cancer Res Characterization of a guanine nucleotide dissociation stimulator for 49: 4682–4689. a ras-related GTPase. EMBO J 12: 339–347. Boykevisch S, Zhao C, Sondermann H, Philippidou P, Halegoua S, Bhattacharya M, Anborgh PH, Babwah AV, Dale LB, Dobransky T, Kuriyan J et al. (2006). Regulation of ras signaling dynamics by Sos- Benovic JL et al. (2002). Beta-arrestins regulate a Ral-GDS Ral mediated positive feedback. Curr Biol 16: 2173–2179. effector pathway that mediates cytoskeletal reorganization. Nat Cell Camonis JH, White MA. (2005). Ral GTPases: corrupting the exocyst Biol 4: 547–555. in cancer cells. Trends Cell Biol 15: 327–332.

Oncogene hRgr expression induces cellular transformation K Osei-Sarfo et al 3670 Cascone I, Selimoglu R, Ozdemir C, Del Nery E, Yeaman C, White M Lim KH, O’Hayer K, Adam SJ, Kendall SD, Campbell PM, et al. (2008). Distinct roles of RalA and RalB in the progression Der CJ et al. (2006). Divergent roles for RalA and RalB in of cytokinesis are supported by distinct RalGEFs. EMBO J 27: malignant growth of human pancreatic carcinoma cells. Curr Biol 2375–2387. 16: 2385–2394. Ceriani M, Scandiuzzi C, Amigoni L, Tisi R, Berruti G, Martegani E. Linnemann T, Kiel C, Herter P, Herrmann C. (2002). The (2007). Functional analysis of RalGPS2, a murine guanine activation of RalGDS can be achieved independently of its Ras nucleotide exchange factor for RalA GTPase. Exp Cell Res 313: binding domain. Implications of an activation mechanism in 2293–2307. Ras effector specificity and signal distribution. J Biol Chem 277: Chardin P, Tavitian A. (1986). The ral gene: a new ras related gene 7831–7837. isolated by the use of a synthetic probe. EMBO J 5: 2203–2208. Margarit SM, Sondermann H, Hall BE, Nagar B, Hoelz A, Pirruccello Chien Y, Kim S, Bumeister R, Loo YM, Kwon SW, Johnson CL et al. M et al. (2003). Structural evidence for feedback activation by (2006). RalB GTPase-mediated activation of the IkappaB family Ras.GTP of the Ras-specific nucleotide exchange factor SOS. Cell kinase TBK1 couples innate immune signaling to tumor cell 112: 685–695. survival. Cell 127: 157–170. Mittar D, Sehajpal PK, Lander HM. (2004). Nitric oxide activates Chissoe SL, Bodenteich A, Wang YF, Wang YP, Burian D, Clifton Rap1 and Ral in a Ras-independent manner. Biochem Biophys Res SW et al. (1995). Sequence and analysis of the human ABL gene, the Commun 322: 203–209. BCR gene, and regions involved in the Philadelphia chromosomal Muller AJ, Young JC, Pendergast AM, Pondel M, Landau NR, translocation. Genomics 27: 67–82. Littman DR et al. (1991). BCR first exon sequences specifically Corominas M, Kamino H, Leon J, Pellicer A. (1989). Oncogene activate the BCR/ABL tyrosine kinase oncogene of Philadelphia activation in human benign tumors of the skin (keratoacanthomas): chromosome-positive human leukemias. Mol Cell Biol 11: is HRAS involved in differentiation as well as proliferation? Proc 1785–1792. Natl Acad Sci USA 86: 6372–6376. Ohta Y, Suzuki N, Nakamura S, Hartwig JH, Stossel TP. (1999). The D’Adamo DR, Novick S, Kahn JM, Leonardi P, Pellicer A. (1997). small GTPase RalA targets filamin to induce filopodia. Proc Natl Rsc: a novel oncogene with structural and functional homology Acad Sci USA 96: 2122–2128. with the gene family of exchange factors for Ral. Oncogene 14: Oxford G, Owens CR, Titus BJ, Foreman TL, Herlevsen MC, Smith 1295–1305. SC et al. (2005). RalA and RalB: antagonistic relatives in cancer cell Das J, Ho M, Zikherman J, Govern C, Yang M, Weiss A et al. (2009). migration. Cancer Res 65: 7111–7120. Digital signaling and hysteresis characterize ras activation in Oxford G, Theodorescu D. (2003). The role of Ras superfamily lymphoid cells. Cell 136: 337–351. proteins in bladder cancer progression. J Urol 170: 1987–1993. Frankel P, Aronheim A, Kavanagh E, Balda MS, Matter K, Bunney Paulsson K, Cazier JB, Macdougall F, Stevens J, Stasevich I, Vrcelj N TD et al. (2005). RalA interacts with ZONAB in a cell density- et al. (2008). Microdeletions are a general feature of adult and dependent manner and regulates its transcriptional activity. EMBO adolescent acute lymphoblastic leukemia: unexpected similarities J 24: 54–62. with pediatric disease. Proc Natl Acad Sci USA 105: 6708–6713. Garcia-Espana A, Salazar E, Sun TT, Wu XR, Pellicer A. (2005). Rodenhuis S. (1992). Ras and human tumors. Semin Cancer Biol 3: Differential expression of cell cycle regulators in phenotypic variants 241–247. of transgenically induced bladder tumors: implications for tumor Roose JP, Mollenauer M, Ho M, Kurosaki T, Weiss A. (2007). behavior. Cancer Res 65: 1150–1157. Unusual interplay of two types of Ras activators, RasGRP and Gotoh T, Cai D, Tian X, Feig LA, Lerner A. (2000). p130Cas SOS, establishes sensitive and robust Ras activation in lymphocytes. regulates the activity of AND-34, a novel Ral, Rap1, and Mol Cell Biol 27: 2732–2745. R-Ras guanine nucleotide exchange factor. J Biol Chem 275: Rosse C, Hatzoglou A, Parrini MC, White MA, Chavrier P, Camonis 30118–30123. J. (2006). RalB mobilizes the exocyst to drive cell migration. Mol Hall BE, Yang SS, Boriack-Sjodin PA, Kuriyan J, Bar-Sagi D. (2001). Cell Biol 26: 727–734. Structure-based mutagenesis reveals distinct functions for Ras Rowley JD. (1973). Letter: a new consistent chromosomal abnormality switch 1 and switch 2 in Sos-catalyzed guanine nucleotide exchange. in chronic myelogenous leukaemia identified by quinacrine fluores- J Biol Chem 276: 27629–27637. cence and Giemsa staining. Nature 243: 290–293. Hernandez-Munoz I, Benet M, Calero M, Jimenez M, Diaz R, Pellicer Sekido Y, Pass HI, Bader S, Mew DJ, Christman MF, Gazdar AF A. (2003). Rgr oncogene: activation by elimination of translational et al. (1995). Neurofibromatosis type 2 (NF2) gene is somatically controls and mislocalization. Cancer Res 63: 4188–4195. mutated in mesothelioma but not in lung cancer. Cancer Res 55: Hernandez-Munoz I, Malumbres M, Leonardi P, Pellicer A. (2000). 1227–1231. The Rgr oncogene (homologous to RalGDS) induces transforma- Senga T, Iwamoto T, Kitamura T, Miyake Y, Hamaguchi M. (2001). tion and gene expression by activating Ras, Ral and Rho mediated JAK/STAT3-dependent activation of the RalGDS/Ral pathway in pathways. Oncogene 19: 2745–2757. M1 mouse myeloid leukemia cells. J Biol Chem 276: 32678–32681. Hofer F, Berdeaux R, Martin GS. (1998). Ras-independent activation Shao H, Andres DA. (2000). A novel RalGEF-like protein, RGL3, as of Ral by a Ca(2+)-dependent pathway. Curr Biol 8: 839–842. a candidate effector for rit and Ras. J Biol Chem 275: 26914–26924. Hofer F, Fields S, Schneider C, Martin GS. (1994). Activated Ras Smith SC, Oxford G, Wu Z, Nitz MD, Conaway M, Frierson HF et al. interacts with the Ral guanine nucleotide dissociation stimulator. (2006). The metastasis-associated gene CD24 is regulated by Ral Proc Natl Acad Sci USA 91: 11089–11093. GTPase and is a mediator of cell proliferation and survival in Jullien-Flores V, Mahe Y, Mirey G, Leprince C, Meunier-Bisceuil B, human cancer. Cancer Res 66: 1917–1922. Sorkin A et al. (2000). RLIP76, an effector of the GTPase Ral, Sondermann H, Soisson SM, Boykevisch S, Yang SS, Bar-Sagi D, interacts with the AP2 complex: involvement of the Ral pathway in Kuriyan J. (2004). Structural analysis of autoinhibition in the Ras receptor endocytosis. J Cell Sci 113(Part 16): 2837–2844. activator Son of sevenless. Cell 119: 393–405. Kurzrock R, Kantarjian HM, Druker BJ, Talpaz M. (2003). Sood R, Makalowska I, Carpten JD, Robbins CM, Stephan DA, Philadelphia chromosome-positive leukemias: from basic mechan- Connors TD et al. (2000). The human RGL (RalGDS-like) gene: isms to molecular therapeutics. Ann Intern Med 138: 819–830. cloning, expression analysis and genomic organization. Biochim Leonardi P, Kassin E, Hernandez-Munoz I, Diaz R, Inghirami G, Biophys Acta 1491: 285–288. Pellicer A. (2002). Human rgr: transforming activity and alteration Takai Y, Sasaki T, Matozaki T. (2001). Small GTP-binding proteins. in T-cell malignancies. Oncogene 21: 5108–5116. Physiol Rev 81: 153–208. Lim KH, Baines AT, Fiordalisi JJ, Shipitsin M, Feig LA, Cox AD Takaya A, Kamio T, Masuda M, Mochizuki N, Sawa H, Sato M et al. et al. (2005). Activation of RalA is critical for Ras-induced (2007). R-Ras regulates exocytosis by Rgl2/Rlf-mediated activation tumorigenesis of human cells. Cancer Cell 7: 533–545. of RalA on endosomes. Mol Biol Cell 18: 1850–1860.

Oncogene hRgr expression induces cellular transformation K Osei-Sarfo et al 3671 Tazmini G, Beaulieu N, Woo A, Zahedi B, Goulding RE, Kay RJ. Wigler M, Pellicer A, Silverstein S, Axel R. (1978). Biochemical (2009). Membrane localization of RasGRP1 is controlled by transfer of single-copy eucaryotic genes using total cellular DNA as an EF-hand, and by the GEF domain. Biochim Biophys Acta donor. Cell 14: 725–731. 1793: 447–461. Wolthuis RM, Bauer B, vant’t Veer LJ, de Vries-Smits AM, Cool RH, Tchevkina E, Agapova L, Dyakova N, Martinjuk A, Komelkov A, Spaargarer M et al. (1996). RalGDS-like factor (Rlf) is a novel Ras Tatosyan A. (2005). The small G-protein RalA stimulates metastasis and Rap1A-associating protein. Oncogene 13: 353–362. of transformed cells. Oncogene 24: 329–335. Zahir N, Lakins JN, Russell A, Ming W, Chatterjee C, Rozenberg GI Urano T, Emkey R, Feig LA. (1996). Ral-GTPases mediate a distinct et al. (2003). Autocrine laminin-5 ligates alpha6beta4 integrin and downstream signaling pathway from Ras that facilitates cellular activates RAC and NFkappaB to mediate anchorage-independent transformation. EMBO J 15: 810–816. survival of mammary tumors. J Cell Biol 163: 1397–1407.

Supplementary Information accompanies the paper on the Oncogene website (http://www.nature.com/onc)

Oncogene