Cell Growth Inhibition by Farnesyltransferase Inhibitors Is Mediated by ...

6 downloads 157 Views 3MB Size Report
Hundley, S. K. Koester, D. A. Troyer, D. J. Bearss, M. W. Conner, J. B. Gibbs, .... Graham, G. D. Hartman, J. Gibbs, and A. Oliff. 1994. ... Leonard, D. M. 1997.
MOLECULAR AND CELLULAR BIOLOGY, Mar. 1999, p. 1831–1840 0270-7306/99/$04.0010 Copyright © 1999, American Society for Microbiology. All Rights Reserved.

Vol. 19, No. 3

Cell Growth Inhibition by Farnesyltransferase Inhibitors Is Mediated by Gain of Geranylgeranylated RhoB WEI DU, PETER F. LEBOWITZ,

AND

GEORGE C. PRENDERGAST*

The Wistar Institute, Philadelphia, Pennsylvania 19104 Received 16 July 1998/Returned for modification 8 September 1998/Accepted 19 November 1998

Recent results have shown that the ability of farnesyltransferase inhibitors (FTIs) to inhibit malignant cell transformation and Ras prenylation can be separated. We proposed previously that farnesylated Rho proteins are important targets for alternation by FTIs, based on studies of RhoB (the FTI-Rho hypothesis). Cells treated with FTIs exhibit a loss of farnesylated RhoB but a gain of geranylgeranylated RhoB (RhoB-GG), which is associated with loss of growth-promoting activity. In this study, we tested whether the gain of RhoB-GG elicited by FTI treatment was sufficient to mediate FTI-induced cell growth inhibition. In support of this hypothesis, when expressed in Ras-transformed cells RhoB-GG induced phenotypic reversion, cell growth inhibition, and activation of the cell cycle kinase inhibitor p21WAF1. RhoB-GG did not affect the phenotype or growth of normal cells. These effects were similar to FTI treatment insofar as they were all induced in transformed cells but not in normal cells. RhoB-GG did not promote anoikis of Ras-transformed cells, implying that this response to FTIs involves loss-of-function effects. Our findings corroborate the FTI-Rho hypothesis and demonstrate that gain-of-function effects on Rho are part of the drug mechanism. Gain of RhoB-GG may explain how FTIs inhibit the growth of human tumor cells that lack Ras mutations. models have offered dramatic examples of tumor regression in the absence of detectable toxic side effects, indicating that FTIs pinpoint a specific feature of neoplastic pathophysiology (4, 31, 43). Notably, recent investigations into the biological mechanisms that underlie FTI treatment have raised questions about their exact mode of action (reviewed in reference 38). While it is quite clear that FTIs act by specifically inhibiting FT activity, it is much less clear that inhibiting Ras farnesylation is essential for the drugs’ antitransforming effects. First, the kinetics of phenotypic reversion of Ras transformation are too rapid to be explained by loss of the function of Ras through inhibition of its farnesylation. Reversion is largely complete within 24 h of cell treatment (49), even though Ras has a half-life of ;24 h (60) and is only partially depleted by the time reversion is complete (49). Second, soluble forms of oncogenic Ras generated in drug-treated cells do not accumulate to steady-state levels which are sufficient to interfere with prenylated Ras (49) and, in any case, only the Ras L61 but not the Ras V12 mutant allele used in published experimental models can exert dominant negative effects (19). Third, FTIs can inhibit the anchorage-independent growth of cells transformed with oncogenic Ras proteins engineered to function independently of farnesylation, due to N-myristylation or geranylgeranylation (10, 37, 47a). Similarly, K-Ras-transformed cells are susceptible to growth inhibition, despite the fact that FTIs do not inhibit K-Ras prenylation due to geranylgeranylation of K-Ras by GGT-I when FT activity is blocked in cells (41, 53, 67). Lastly, there is no correlation between the susceptibility of human tumor cell lines to growth inhibition by FTIs and their Ras status (57). Thus, the biological susceptibility to FTIs can be separated to a significant degree from Ras inhibition. These investigations have stimulated efforts to identify farnesylated proteins other than Ras whose functional alteration is germane to the drugs’ antitransforming mechanism (9, 38). Our previous work in this area led us to suggest an alternate model for drug action termed the FTI-Rho hypothesis (38). The FTI-Rho hypothesis proposes that the antitransforming effects of FTIs are mediated at least in part through altering

Farnesyltransferase inhibitors (FTIs) are a novel class of antitumor agents whose development was based upon the discovery that posttranslational prenylation is required for the oncogenic properties of Ras (reviewed in references 17, 18, 40, and 56). Protein prenylation involves C-terminal addition of C15 (farnesyl) or C20 (geranylgeranyl) isoprenoids, each of them intermediates in cholesterol biosynthesis. Protein prenylation reactions are carried out by one of three enzymes in the cell: farnesyltransferase (FT), geranylgeranyltransferase type I (GGT-I), or geranylgeranyltransferase type II (GGT-II; Rab GGT). FT and GGT-I are related heterodimeric enzymes that share a common subunit. They mediate prenylation of members of the Ras and Rho subfamilies of the Ras superfamily of small GTPases that include C-terminal CAAX prenylation motifs. GGT-II is an enzyme that is unrelated to the FT and GGT-I. It mediates geranylgeranylation of members of the Rab subfamily of Ras superfamily small GTPases through a reaction that is mechanistically distinct from the reactions catalyzed by FT or GGT-I (7, 71). Prenylation facilitates association with cellular membranes and mediates protein-protein interactions (71). Geranylgeranylation is the predominant type of prenylation in cells. It is unclear why two types of prenylation occur, but there are examples in which protein function can be altered by switching prenylation type (11, 34). Farnesylation of Ras proteins is the crucial modification for oncogenicity (28). Therefore, compounds which specifically inhibit FT were sought as a strategy to block Ras function and suppress the growth of Ras-dependent tumors while leaving cellular geranylgeranylation intact (17). While the ultimate clinical potential of this strategy has yet to be assessed, proofof-principle cell culture and animal experiments have firmly established the ability of FTIs to effectively reverse Ras-dependent cell transformation and to impede tumorigenesis (27, 30, 32, 44, 46, 49, 57, 62). In particular, studies with Ras oncomice

* Corresponding author. Mailing address: The Wistar Institute, 3601 Spruce St., Philadelphia, PA 19104. Phone: (215) 898-3792. Fax: (215) 898-2205. E-mail: [email protected]. 1831

1832

DU ET AL.

the function of farnesylated Rho proteins, including RhoB (34, 37, 39, 49, 50). Rho proteins are a family of small GTPases that are required for Ras transformation (29, 50, 51) and that regulate cytoskeletal actin, focal adhesion formation, cell adhesion signaling, and transcription (reviewed in references 63, 64, and 66). RhoB is closely related to RhoA, but it is differently localized, regulated, and prenylated. RhoB is localized in endosomes where it could conceivably participate in receptormediated endocytosis events where Rho has been implicated (33, 55). RhoB is short-lived and part of the immediate-early genetic response to v-Src and epidermal growth factor that may contribute to regulating cell cycle progression (25, 37, 70). However, RhoB also has cell cycle inhibitory roles suggested by its upregulation by UV irradiation and the stress responseassociated kinases p38 and JNK (15, 16) and by its ability to govern transforming growth factor b (TGF-b)-regulated transcription (14). RhoB is unique among prenylated proteins in that it exists normally in vivo in two populations that are either farnesylated or geranylgeranylated (RhoB-F or RhoB-GG) (1). FT is responsible for the generation of RhoB-F, whereas GGT-I is responsible for the generation of RhoB-GG (34). RhoB has a half-life of only ;2 h (37), so the steady-state levels of RhoB-F decrease rapidly in FTI-treated cells. However, there is a simultaneous elevation in the steady-state levels of RhoB-GG, and a great increase in the ratio of RhoB-GG to RhoB-F in cells, because newly synthesized RhoB still serves as a substrate for GGT-I. The increased levels of RhoB-GG therefore represent a biochemical gain-of-function effect of FTI treatment. Interestingly, this shift in RhoB prenylation was associated with a loss of RhoB’s ability to promote cell proliferation (34). This observation implied that the functions of RhoB-F and RhoB-GG might be dissimilar and that cell proliferation may be influenced by altering the ratio of RhoB-F to RhoB-GG. In this study, we tested the hypothesis that elevation of RhoB-GG could mimic the ability of FTIs to specifically inhibit the growth of Ras-transformed cells. In support of this hypothesis, we found that RhoB-GG was not only inhibitory but also caused a phenotypic reversion which was similar in its features to that produced by FTI treatment. The conclusions of this study indicate that FTIs act in part through gain-of-function effects. Our findings suggest that Rho regulates oncogenic Ras biology and offers a Ras-independent mechanism for how FTIs are able to broadly inhibit the growth of human tumor cells. MATERIALS AND METHODS Plasmid constructions. An epitope-tagged RhoB polypeptide (HA-RhoBGG) that is preferentially geranylgeranylated in cells was constructed in the following manner. A HindIII-BstXI restriction fragment was excised from cytomegalovirus (CMV) HA-RhoB-WT (34) to provide the RhoB N terminus, including an influenza hemagglutinin (HA) epitope. A BstXI-EcoRI restriction fragment was excised from RhoB-ACT (2) to provide the remainder of RhoB and a modified C terminus which substituted the 13 C-terminal residues of RhoA in place of the 16 C-terminal residues of RhoB. The 13 residues derived from the RhoA C terminus include the CAAX box sequences responsible for prenyltransferase specificity (7), and they are sufficient for directing geranylgeranylation of RhoB-ACT in cells (2). The recombinant product, designated HA-RhoB-GG, was generated by ligation of the restriction fragments into the HindIII-EcoRI site of pcDNA3.1zeo (Invitrogen) to generate the plasmid zeoCMV-HA-RhoB-GG. Similar expression vectors for HA-RhoB-S, an epitope-tagged polypeptide that is unprenylated due to a C193S mutation in the RhoB CAAX box, HA-RhoBV14-S, an activated version that includes a V14 mutation, and HA-RhoA have been described (34). Cell culture. The cell lines used in this study were derived from Rat1/ras, a clonal Rat1 fibroblast line generated by transformation with v-H-ras (30, 49). Rat1/ras cells were cultured in Dulbecco modified Eagle medium (DMEM; Gibco) containing 10% fetal bovine serum (Atlanta Biological) and 10 U of penicillin-streptomycin (Mediatech) per ml. Where indicated, the FT-specific inhibitor L-744,832 (31) was added to cultures to a final concentration of 10 mM. Rat1/Ras cell lines stably expressing HA-RhoB-GG were obtained by modified

MOL. CELL. BIOL. calcium phosphate transfection of zeoCMV-HA-RhoB-GG, selection in 200 mg of zeosin (Invitrogen) per ml, ring cloning of zeosin-resistant colonies, and expansion into mass culture. Expression was verified by Western analysis with the anti-HA antibody 12CA5 (BABCO, Inc.). Control cell lines harboring empty vector or expressing the unprenylated HA-RhoB-S or HA-RhoBV14-S mutants (34) were derived similarly with zeocin resistance vectors (Invitrogen). MTT assay. Cell growth was measured by MTT [3-(4,5-diethylthiazoly-2-yl)2,5-diphenyltetrazolium bromide] assay (6). Briefly, cells were seeded at 500 cells per well in 96-well culture plates in quadruplicate. At various points, medium was removed, and cells were incubated with 180 ml of RPMI 1640 containing 5% fetal calf serum (FCS), 0.25 mg of MTT (Sigma) per ml at 37°C for 4 h, followed by solubilization with 20% sodium dodecyl sulfate–50% dimethyl formamide in water for another 4 h or overnight at 37°C. The absorbance of each well was measured with a microplate reader (Rainbow reader) at 595- and 655-nm dual wavelengths. The viable cell number is proportional to the absorbance. Soft agar assay. One milliliter of 0.5% NuSieve agarose (FMC Biochemicals) in DMEM containing 10% FCS was used to coat the bottom of each well in 6-well culture dishes. After hardening, 104 cells were suspended in 1 ml of 0.3% NuSieve agarose, DMEM, and 10% FCS solution and plated onto the bottom layer. The cell solution was allowed to set 30 min at room temperature before moving it to 37°C. Where indicated, L-744,832 was added to top and bottom agar mixtures to a final concentration of 10 mM. Colonies formed in the soft agarose culture were photographed 10 to 16 days later with an Olympus microscope with a 35-mm camera attachment. Actin immunofluorescence. Cells were seeded onto coverslips in six-well dishes and treated the next day for 48 h with 10 mM L-744,832 or carrier. Cells were fixed and stained with fluorescein-phalloidin (Molecular Probes) as described previously (49). Photographs of stained cells were generated on a Leitz immunofluorescence microscope. Apoptosis analysis. For apoptosis analysis, 1 3 106 to 2 3 106 cells were seeded onto polyHEMA-coated dishes. After 12 to 14 h of plating, 10 mM FTI was added as described earlier (39). After 48 h of FTI treatment, cells were collected, trypsinized, washed with phosphate-buffered saline (PBS), and fixed in 70% ethanol. The cells were then stained in PBS containing 5 mg of propidium iodide per ml, 10 mg of RNase A per ml, and 0.1% glucose. Flow cytometry was performed by using an EPIC/XL cell analyzer (Coulter). Western analysis. At appropriate times, cells were washed in cold PBS and harvested in Nonidet P-40 lysis buffer containing phenylmethylsulfonyl fluoride, pepstatin, and leupeptin (20a). Cellular protein was quantitated by Bradford assay, and 40 mg was fractionated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis and electrophoretically transferred onto nitrocellulose membranes (Amersham). Blots was probed with 2.5 mg of anti-HA antibody 12CA5 (BABCO, Inc.) per ml followed by an anti-mouse immunoglobulin G horseradish peroxidase-conjugated secondary antibody (Boehringer Mannheim) at a 1:10,000 dilution by standard protocols. A chemiluminescence kit (Pierce) was used to detect the antibody complex according to the protocol recommended by the vendor. Western analysis of p21WAF1 was performed similarly under the same conditions with the C19 anti-p21 antibody as suggested by the vendor (Santa Cruz Biotechnology). Northern analysis. Total cytoplasmic RNA was isolated and subjected to Northern blotting and hybridization as described previously (48). Probes were generated by random-primed labeling of RhoB and RhoA cDNAs (34) with [a-32P]dCTP (NEN). Transactivation assay. NIH 3T3 cells were maintained in DMEM containing 10% calf serum, 1 mM of sodium pyruvate, and 10 U of penicillin-streptomycin (Mediatech) per ml. Cells were transfected for 20 h in six-well dishes by modified calcium phosphate transfection (8) with 3 mg of total DNA including 0.2 mg of CMV-RasV12 (42), 0.4 mg of the p21 promoter reporter WWP-luc (13), 0.2 mg of CMV-bgal (to normalize for transfection efficiency), and 2.2 mg of pBS1 as plasmid filler. Where indicated, 0.2 mg of CMV-HA-RhoB-GG or HA-RhoA and 2 mg of pBS1 was included. Cells were washed and refed with standard growth medium and 10 mM FTI L-744,832 was added where indicated (31). After an additional 24-h incubation, cells were harvested and extracts were prepared and assayed for b-galactosidase and luciferase activities with a commercial kit under conditions recommended by the vendor (Promega). Relative luciferase activity was normalized to b-galactosidase activity in each trial before the data were plotted.

RESULTS RhoB-GG induces morphological reversion and actin stress fiber formation in transformed cells. Previous work suggested a role for alteration of RhoB in the mechanism by which FTIs induce phenotypic reversion, loss of anchorage-independent growth capacity, and apoptosis (34, 37, 39, 49, 50). Since FTI treatment leads to an elevation of RhoB-GG in cells and a concomitant loss of growth-promoting activity (34), we investigated the possibility that the gain of RhoB-GG may be sufficient to mediate cell growth inhibition or other drug responses. This

VOL. 19, 1999

CELL GROWTH INHIBITION BY FTIs MEDIATED BY Rho

FIG. 1. HA-RhoB-GG expression. (A) Western blot analysis. Extracts from different stable Rat1 and Rat1/ras transfectants were immunoblotted with the anti-HA antibody 12CA5. A total of 40 mg of cellular protein was loaded into each lane of the gels. Expression and morphology of the lines examined in each set were highly similar. (B) Northern blot analysis. Total cytoplasmic RNA for randomly selected cell lines from each set generated was fractionated by Northern analysis and hybridized to RhoB and RhoA cDNA probes.

was performed by expressing in cells a RhoB-RhoA chimera in which the 16 C-terminal residues of RhoB, which direct both farnesylation and geranylgeranylation, were replaced with the 13 C-terminal residues of RhoA, which directs only geranylgeranylation (1, 34). The localization of this construction, termed RhoB-GG, is more similar to RhoB in FTI-treated cells or to the strictly geranylgeranylated RhoA protein (2, 37). An epitope-tagged version of this chimera, HA-RhoB-GG, was stably expressed in normal Rat1 fibroblasts or in Rat1/ras, an H-Ras-transformed Rat1 derivative (30, 49). Cell lines expressing either only vector sequences or HA-RhoB-S or HARhoBV14-S, two unprenylated CAAX box mutants, were also generated as controls. Four sets of cell lines each for Rat1 and Rat1/ras were each derived by transfection of these vectors and are referred to as Rat1/B-GG, Rat1/B-S, Rat1/BV-S, and Rat1/vect and as Ras/B-GG, Ras/B-S, Ras/BV-S, and Ras/vect, respectively. Subsequent analyses demonstrated consistent phenotypes among the clones in each set, arguing against significant clonal variation. In addition, cell clones expressing the unprenylated RhoB-S or RhoB-VS proteins were identical in all tests. Therefore, a subset of cell lines generated in this study which are representative is presented. Exogenous expression of various RhoB mutants was confirmed in Rat1 or Rat1/ras derivatives by Western analysis with an anti-HA epitope antibody (Fig. 1A). Northern analysis indicated that the expression of the exogenous RhoB gene in the cell lines was a fewfold higher than the endogenous gene but not in gross excess (Fig. 1B).

1833

While Rat1/vect and Rat1/B-GG clones were similar in morphology, Ras/B-GG clones all exhibited a strikingly flattened morphology relative to the Ras/vect control lines that was similar to the reverted phenotype induced in such lines by FTI treatment (Fig. 2). This effect appeared to be dose dependent, paralleling that seen with FTIs (49), insofar as two cell lines with reduced levels of RhoB-GG exhibited a slightly less flattened morphology (data not shown). Geranylgeranylation of RhoB was necessary to produce the flattened phenotype in Rat1/ras cells because the unprenylated RhoB-S or RhoBV14-S mutants had no effect on the morphology of Rat1/ras cells (Fig. 2 and data not shown). Rat1/ras cells expressing either mutant also reverted after exposure to FTIs in the same manner as the Ras/vect cells, arguing against a dominant inhibitory effect of accumulation of unprenylated RhoB in the cellular response to FTI. RhoBV14-GG which included an activating mutation that abolished GTPase activity produced the same reversion and growth inhibition as did RhoB-GG (data not shown), a finding consistent with the notion that RhoB is already in the GTPbound state in Ras-transformed cells (50). The trivial possibility that RhoB-GG acted by suppressing Ras expression was ruled out by demonstrating that Ras levels were similar in Ras/B-GG and Ras/vect cells (data not shown). To examine the structure of cytoskeletal actin in each cell type, fixed cells were stained with fluorescein isothiocyanate-conjugated phalloidin and examined by immunofluorescence microscopy. Phenotypic reversion caused by FTI treatment was associated with a shift from membrane ruffles in transformed cells to stress fibers in reverted cells (Fig. 3). Stress fibers predominated in Ras/B-GG cells and in FTI-treated Ras/vect cells, whereas membrane ruffles predominated in untreated Ras/vect cells. The shift in actin structure was specific insofar as RhoB-GG had no detectable effect on the cytoskeletal actin structure of Rat1 cells. Geranylgeranylation was also required to produce the shift in actin structure because no changes were seen in Ras/B-S or Ras/BV-S cell lines (data not shown). The requirement for geranylgeranylation in shifting the morphology and actin cytoskeleton in Ras-transformed cells was specific, because unprenylated RhoB is capable of activating serum response factor (35). We concluded that the gain of RhoB-GG elicited by FTI treatment was sufficient to induce morphological reversion and to stimulate stress fiber formation in Rastransformed cells. RhoB-GG selectively inhibits the growth of transformed cells. The first indication that RhoB-GG could inhibit Ras transformation was that fewer colonies emerged after transfection of Rat1/ras and selection for stably transfected cells. The efficiency of colony formation by HA-RhoB-GG vector was decreased ;2-fold in Rat1/ras cells relative to vector, but no similar decrease was observed in Rat1 cells (data not shown). To determine explicitly whether RhoB-GG inhibited Rat1/ras growth, we compared the anchorage-dependent growth and the anchorage-independent growth of the cell lines obtained. We also compared the growth of cells in the presence or absence of FTI to determine whether RhoB-GG might mimic the growth-inhibitory effects of FTI. MTT assay was used to monitor proliferation in monolayer culture of replicate cultures of Ras/B-GG and Ras/vect lines (see Materials and Methods). We observed that Ras/B-GG cells grew ;50% more slowly than Ras/vect cells, with an average doubling time of ;24 h compared to ;16 h for Ras/vect cells (Fig. 4A). A similar experiment comparing the flat Rat1/vect and Rat1/ B-GG cell lines indicated that RhoB-GG had only limited effects on normal fibroblast growth (Fig. 4B). Thus, RhoB-GG inhibited the growth of transformed cells relatively specifically. This resembled the effects of FTI treatment, which reduces the

1834

DU ET AL.

MOL. CELL. BIOL.

FIG. 2. RhoB-GG reverts the Ras-transformed phenotype. Photomicrographs of various Rat1/ras and Rat1 derivatives in monolayer culture in the presence or absence of FTI are shown (see text for nomenclature). Ras/BV-S cells exhibited the same morphology and reversion response as Ras/B-S cells (not shown). Where indicated, cells were treated for 40 h with FTI before photography.

growth rate of Rat1/ras cells in monolayer culture substantially but has only limited effects on Rat1 cells cultured under the same conditions (49). We tested whether RhoB-GG and FTI treatment produced a similar reduction in monolayer growth rate. Consistent with previous observations (49), FTIs reduced the growth of Ras/vect cells in a way similar to that displayed by untreated Ras/B-GG cells (Fig. 5A). FTI treatment slightly potentiated the inhibition of cell growth by RhoB-GG but this effect was minimal. FTIs also slightly potentiated the effects of RhoB-GG on the growth of Rat1 cells (Fig. 5B). The slight effects of FTIs observed in Ras/B-GG or Rat1/B-GG cells could be explained by effects on the endogenous RhoB genes in those cells. In addition, the effects of RhoB-GG and FTI on normal Rat1 cells were similar in degree to those observed previously (49). Taken together, the results indicated that gain of RhoB-GG by FTI was sufficient to mediate selective inhibition of anchorage-dependent growth of Ras-transformed cells. To determine whether RhoB-GG could mimic FTI in its ability to inhibit the anchorage-independent growth of Rastransformed cells, we compared the colony formation efficiency of Ras/vect and Ras/B-GG cells in soft agar culture. Cells were seeded in triplicate cultures and colony formation was monitored 2 to 3 weeks later. As expected, the Ras/vect cells formed colonies with an efficiency comparable to that of the parental Rat1/ras cells (data not shown). In contrast, Ras/ B-GG cells exhibited a dramatically reduced ability to prolif-

erate under these conditions (Fig. 6). This reduction in colony formation did not reflect clonal variation because it was observed in all of the Ras/B-GG cell lines tested. The ability of RhoB-GG to elicit this effect suggested a role in the mechanism by which FTIs inhibit the anchorage-dependent and anchorage-independent proliferation of transformed and human tumor cell lines (30, 46, 57), the outgrowth of tumors in certain H-Ras and N-Ras oncomice (4, 43), and the outgrowth of human tumors in xenograft models (46, 62). We concluded that gain of RhoB-GG by FTI was sufficient to mediate cytostatic effects in Ras-transformed cells. RhoB-GG does not promote apoptosis of Ras-transformed cells denied substratum adhesion. FTIs are normally cytostatic to Ras-transformed cells but are cytotoxic if cells are cultured on polyHEMA, a nonadherent substrate that sensitizes them to anoikis (39). To test whether RhoB-GG could mimic the ability of FTIs to promote anoikis, we compared the sensitivity of Ras/vect and Ras/B-GG cells to undergo apoptosis when cultured on polyHEMA. Apoptosis was measured by the detection of sub-G1 phase DNA by flow cytometry, an assay which has been validated previously in this cell system (39). Interestingly, both Ras/vect and Ras/B-GG cells survived in polyHEMA culture, indicating that oncogenic Ras could promote survival in both cases and that FTI addition to either type of cell caused apoptosis within 24 h in a way similar to that observed with parental Rat1/ras cells (Fig. 7). Since it has been demonstrated previously that farnesyl-independent RhoB

VOL. 19, 1999

CELL GROWTH INHIBITION BY FTIs MEDIATED BY Rho

FIG. 3. RhoB-GG activates actin stress fibers in Ras-transformed cells. Cells were treated with FTI or carrier, and actin was visualized by phalloidin staining and fluorescence microscopy as described in Materials and Methods.

(myristylated RhoB) can inhibit FTI-induced death in Rat1/ras cells (39), this result suggested that FTI-induced apoptosis occurred through a distinct mechanism that was separable from the growth inhibitory and morphological effects mediated by RhoB-GG and where loss of RhoB-F was implicated. We concluded that gain of RhoB-GG by FTI was insufficient to sensitize Ras-transformed cells to anoikis. RhoB-GG mimics the ability of FTIs to induce expression of the CKI p21WAF1. The cell cycle kinase inhibitors (CKIs) are key focal points for negative regulation of cell cycle progression. Since gain of RhoB-GG was sufficient to inhibit the proliferation of Ras-transformed cells, one might predict that one or more CKIs might be elevated in Ras/B-GG cells. Some human tumor cell lines which are susceptible to growth inhibition by FTIs exhibit induction of the CKI p21WAF1 (58). Therefore, we investigated whether FTIs could increase p21WAF1 expression in Rat1/ras cells and whether RhoB-GG might act similarly. Western analysis of cell extracts was performed to monitor steady-state levels of p21WAF1. We found that p21WAF1 was undetectable in all Ras/vect cell lines examined but was significantly elevated in all Ras/B-GG cell lines examined (Fig. 8A). The levels of induction were robust, being similar in degree to the levels stimulated by the activation of wild-type p53 in a control E1A-immortalized BRK cell line

1835

FIG. 4. RhoB-GG inhibits the proliferation of Ras-transformed cells. (A) Growth curve of Ras/vect and Ras/B-GG cells. Cell growth was measured with the MTT assay. Three representative cell lines containing vector (solid symbols) or RhoB-GG (open symbols) were tested. Each point represents the average of four individual measurements. (B) Growth curve of Rat1/vect and Rat1/B-GG cells. Two representative cell lines containing vector (open symbols) or RhoB-GG (solid symbols) were tested as described above.

(12). A similar but transient elevation of p21WAF1 occurred in Rat1/ras or Ras/vect cells treated with FTI. After drug addition, p21WAF1 was first detected at 12 to 24 h (Fig. 8B), a period that is consistent with the kinetics of RhoB alteration, growth inhibition, and morphological reversion in this system (49). The relative increase elicited by FTI treatment was similar to that detected in Ras/B-GG cells, arguing that induction by the latter was not due to an overexpression artifact. Since p53 can activate expression of p21WAF1 and since it was conceivable that RhoB-GG gain stressed cells such that p53 was stabilized, we examined the same extracts by immunoblotting with a p53 antibody. p53 was not detected, however, arguing against the possibility that p21WAF1 was elevated by p53 activation. Notably, p21WAF1 was not elevated in Rat1/B-GG cells (Fig. 8C). This argued that p21WAF1 elevation was a synthetic effect produced by the combination of Ras activation and RhoB-GG elevation and that the induction of p21WAF1 was associated with growth inhibition. Interestingly, Ras/B-S and Ras/BV-S cells expressing unprenylated wild-type or activated RhoB also

1836

DU ET AL.

MOL. CELL. BIOL.

FIG. 6. RhoB-GG inhibits anchorage-independent growth. 104 Cells from two Ras/vect and Ras/B-GG lines each were seeded at 104 cells per well in six-well dishes in soft agar culture. A control trial confirming FTI suppression of Rat1/ras parental cells was also performed as described elsewhere (30). Colonies were scored after 2 to 3 weeks. A photograph of representative colony formation in wells for each cell line tested is shown.

FIG. 5. RhoB-GG mimics the selective inhibitory effects of FTI treatment. (A) FTI treatment versus RhoB-GG expression in Ras-transformed cells. Growth curves were determined as before. FTI was added where indicated and replenished every 48 h. A representative comparison between Ras/vect and Ras/B-GG cells is shown; similar results were obtained with other cell lines. (B) FTI treatment versus RhoB-GG expression in normal fibroblasts. Growth curves were determined as described above, and FTI was added as before where indicated. A representative comparison between Rat1/vect and Rat1/B-GG cells is shown; similar results were obtained with another cell line.

exhibited elevation of p21WAF1 (Fig. 8C). This implied that in the presence of oncogenic Ras RhoB does not have to be prenylated to activate p21WAF1, a result reminiscent of previous demonstrations that prenylation is dispensible for RhoB to activate SRF (35). It also implied that induction of p21WAF1 by FTIs may be a necessary but perhaps not a sufficient cause of phenotypic reversion, a conclusion consistent with the results of a recent study of human tumor cells in which p21WAF1 induction is not consistently correlated with FTI-induced growth inhibition (58). To determine whether FTIs and RhoB-GG upregulated p21WAF1 expression at the level of transcription, we performed a set of transient promoter activation experiments in NIH 3T3 cells with a luciferase reporter plasmid. Consistent with a transcriptional mechanism of activation, FTI and RhoB-GG each activated the p21WAF1 reporter when introduced into cells with activated Ras (Fig. 8D). The activa-

tion by FTI was slightly more robust in these experiments, but RhoB-GG clearly mimicked the action of the drug. In support of the possibility that RhoB-GG had properties similar to the geranylgeranylated RhoA protein, p21WAF1 transcription was activated by RhoA when it was cotransfected with activated Ras (data not shown). We concluded that RhoB-GG was sufficient to mediate transcriptional activation of p21WAF1 by FTI in the presence of activated Ras. DISCUSSION In this study, we demonstrated that gain-of-function effects on RhoB elicited by FTIs are sufficient to mediate phenotypic reversion and cell cycle inhibition, two major effects of FTIs on transformed cells. Unlike FTIs, RhoB-GG did not sensitize Ras-transformed cells to anoikis, suggesting that this effect is mechanistically distinct and may instead involve loss of function of farnesylated RhoB or other proteins. These observations are important because they provide the first evidence that FTIs may act not only by causing loss of function but also by inducing gain of function of proteins that are normally farnesylated in cells. A caveat to this study is that the engineered RhoB-GG molecule is not exactly identical to RhoB-GG found in FTI-treated cells. We believe this caveat is small, however, because of the brevity of the C-terminal replacement (only ;10 amino acids are different) and because of other studies which have also implicated functional alteration of RhoB in the drug mechanism (34, 37, 39, 50). The extent of the pheno-

VOL. 19, 1999

CELL GROWTH INHIBITION BY FTIs MEDIATED BY Rho

1837

FIG. 7. RhoB-GG does not induce apoptosis on polyHEMA. Cells were seeded into 10-cm dishes coated with polyHEMA at 106 cells per well. Where indicated FTI was added, and 48 h later the cell suspensions were collected. Cells were trypsinized briefly to disperse clumps, pelleted, fixed in ice-cold ethanol, and processed for flow cytometry as described previously (54).

types induced by RhoB-GG suggests that the antitransforming effects of FTIs that are mediated through gain of function may be rather broad. Our findings also provide a mechanism to explain how FTIs can inhibit the growth of neoplastically transformed cells that lack Ras mutations or that are dependent on K-Ras or N-Ras, which each remain active in the presence of FTIs due to their ability to be alternately geranylgeranylated in FTI-treated cells (26, 41, 43, 53, 67). K-Ras is the predominant Ras oncogene in human cancer but unlike RhoB and H-Ras (11) geranylgeranylation of either is normal or oncogenic KRas does not change its function (8a). We previously proposed an alternate model for FTI action, termed the FTI-Rho hypothesis, to explain the biological effects of the drugs which cannot be attributed directly to functional inhibition of Ras (34). The FTI-Rho hypothesis, which emerged from studies of a role for RhoB alteration in the drug mechanism (34, 37, 39), proposes that inhibition or alteration of farnesylated Rho functions are crucial steps for the antitransforming properties of FTIs. This model was developed to address anomalies evident during the earliest phases of FTI research which argued that Ras inhibition could not be the sole basis for FTI action (49). Additional anomalies and difficulties with the original Ras-based model for FTI action emerged subsequently with evidence that the growth of neoplastically transformed cells can be blocked even if FTIs are ineffectual at blocking K-Ras farnesylation and function (37, 43, 57, 61). Animal experiments which further illustrate Ras-independent effects include xenograft experiments employing K-Ras- and N-Ras-transformed cells (32) or human tumor cells (46, 61), N-Ras oncomouse experiments (43), and oncomouse experiments where cell cycle inhibition represents the major mechanism for drug action (4). When taken together, the existing results indicate that Ras inhibition may be sufficient but is not necessary for FTIs to reverse or inhibit malignant cell growth. The FTI-Rho hypothesis offers a new viewpoint to interpret

cellular responses where loss of function in oncogenic Ras cannot explain the response. The loss-of-function and gain-offunction arms of the mechanism are distinguished in this model (Fig. 9). Gain of function through RhoB-GG elevation is proposed to mediate phenotypic effects and cell cycle inhibition. RhoB-GG did not potentiate anoikis (adhesion deprivalinduced apoptosis), so loss of function through loss of RhoB-F, H-Ras, or other farnesylated proteins would be implicated in this response, a conclusion in support of the results of previous observations (39). Elevation of RhoB-GG in Ras-transformed cells caused dramatic morphological reversion to a flat phenotype characteristic of normal fibroblasts. The reverted phenotype closely resembled that produced by FTI treatment of Ras-transformed cells. Reversion was associated with a shift in cytoskeletal actin structure, such that there was a loss of membrane ruffles associated with transformation to an increase in actin stress fibers characteristic of normal cells. RhoB-GG did not affect the morphology of normal cells but slightly increased stress fiber formation in those cells, also mimicking FTI treatment (49). While the exact mechanism(s) underlying this phenomenon is not yet clear, the ability of RhoB-GG to induce stress fiber formation addresses an apparent anomaly in the FTI-Rho hypothesis. Rho proteins induce stress fiber formation, so if FTIs act in part by altering RhoB, as the model proposed, one might have been expected FTIs to disperse rather than induce stress fibers in Ras-transformed cells. FTI treatment produces the opposite effect, however, such that stress fiber formation increases (44, 49). The observation that RhoB-GG can induce stress fibers addresses this anomaly. RhoB-GG has certain RhoA-like features (being not only structurally related but rendered more similar to RhoA by geranylgeranylation), so one consequence of FTI treatment is to shift RhoB to a RhoA-like localization and therefore elevate a RhoA-like function in cells. Based on accepted definitions of RhoA function, this

1838

DU ET AL.

MOL. CELL. BIOL.

FIG. 8. RhoB-GG is sufficient to mediate activation of p21WAF1 by FTIs. (A) Constitutive elevation of p21WAF1 in Ras/B-GG cells. Cell extracts were prepared from the cell lines indicated and analyzed by immunoblotting with a p21WAF1 antibody. BRK An1 is a control cell line that contains a temperature-sensitive p53 gene that is mutant at 38°C. When cells are shifted to 32°C, the mutant assumes a wild-type configuration and function that leads to p21 induction. A single Ras/vect line lacking expression is representative of all lines examined. (B) Transient induction of p21WAF1 in Ras-transformed cells after FTI treatment. Cell extracts were prepared at the times indicated after addition of FTI to Rat1/ras cells and analyzed as described above. The cell extract from a Ras/B-GG cell line was included to illustrate the similar levels of expression in FTI-treated Rat1/ras cells and Ras/B-GG cells. (C) p21WAF1 is not induced in Rat1 cells expressing RhoB-GG but is induced in Rat1/ras cells expressing unprenylated RhoB-S or RhoBV-S. Cell extracts were prepared and analyzed as described above. RhoBV-S is the same as RhoB-S except that the former also includes an activating mutation (V14). An extract prepared from a Ras/B-GG cell line provides a positive control (right lane). (D) FTIs activate p21WAF1 at the level of transcription. NIH 3T3 cells were transiently transfected with an oncogenic Ras vector (42) and the p21WAF1 reporter plasmid WWP-luc (13), along with vectors expressing no insert or HA-RhoB-GG. Where indicated, cells were treated for 24 h with FTI before being processed for relative luciferase activity as described in Materials and Methods. The standard error was computed from three trials.

elevation would be predicted to increase stress fiber formation (51, 52). This interpretation is simple and consistent with the effects of FTIs on RhoB function, cellular morphology, and cytoskeletal actin (34, 37, 49, 51). Cell growth inhibition by RhoB-GG or by FTIs correlated with increased expression of p21WAF1. Transformed cells but not normal cells were susceptible to RhoB-GG, similar to the effects of FTI treatment (49). Increased expression of p21WAF1 in Ras/B-GG cells may be part of the mechanism through which RhoB-GG may inhibit cell cycle progression because of the role of p21WAF1 in inhibiting cell cycle progression in many cells (21). Notably, p21WAF1 was elevated in Ras/B-S cells which did not exhibit evidence of phenotypic reversion or growth inhibition, implying that although p21WAF1 may be necessary it was not sufficient to mediate these responses to FTI. This finding is consistent with observations in human tumor cells, where p21WAF1 elevation by FTIs has been seen but not correlated with FTI growth inhibition (58), as well as with previous observations that RhoB-S can stimulate gene expression by activating SRF (35). The mechanism of p21WAF1 activation was p53 independent insofar as RhoB-GG did not stabilize p53 and elevate its level; moreover, RhoB-GG was still able to activate a p21WAF1 promoter in which the p53 binding site was mutated. Identification of RhoB-GG as a potential mediator of the inhibitory effects of FTIs on cell cycle progression does not immediately shed light on why normal and transformed cells should respond so differently. It is tempting to speculate that morphological and/or cytoskeletal effects of RhoB-GG related to cell adhesion are important, because Rho proteins have been implicated in integrin-dependent cell growth control (3, 5, 22, 23, 65) and because reversion indirectly affects cell cycle regulation by returning integrin-mediated adhesion dependence. Reengagement of an appropriate adhesion signaling program could underlie the response of transformed cells, since normal cells already have such a program in place—as an adhesion checkpoint control—and do not

reengage substratum after FTI treatment and RhoB-GG elevation. Several interpretations of our results can be considered in light of the inhibitory properties of oncogenic Ras in primary cells (59). Rho is necessary for Ras transformation (29, 50, 51), so one interpretation of our results is that RhoB-GG interferes with a Rho effector function that is required for Ras transformation. We believe that differences in the accessibility of RhoB-GG to transformation-associated Rho effectors could be important. Loss-of-function and gain-of-function scenarios can be imagined for how RhoB-GG might interfere with such functions. First, RhoB-GG may be localized away from certain RhoB effectors whose action is required for transformation. This concept can be illustrated by considering the RhoB effector PKN/PRK1, which is localized on endosomes (45) like RhoB and which is a likely effector for RhoB; since RhoB-GG is not on endosomes it may poorly interact with PKN/PRK1 in cells (2, 37). In this illustration RhoB-GG would represent a loss of function with regard to PKN/PRK1 signaling (whose role in transformation remains to be determined however). Second, since RhoB-GG localization overlaps with RhoA, RhoB-GG may acquire the ability to bind to certain RhoA effectors that are not normally accessible to RhoB. In this case, it is conceivable that RhoB-GG may competitively interfere with the operation of certain effectors of RhoA if RhoB-GG does not mimic RhoA function exactly. In this illustration RhoB-GG would represent a gain of function that dominantly inhibits certain functions of RhoA that may be associated with Ras transformation. This latter case offers an interpretation of our findings which are consistent with the findings of a recent report that suggests that Rho facilitates Ras transformation and that loss of Rho unleashes the growth-inhibitory properties of oncogenic Ras (47). If RhoB-GG disrupts or interferes with certain Rho effector functions, these functions themselves might even be nonphysiological in the sense that the effectors are themselves either mislocalized or dysfunc-

VOL. 19, 1999

CELL GROWTH INHIBITION BY FTIs MEDIATED BY Rho

1839

Merck and Co., Inc. G.C.P. is a Pew Scholar in the Biomedical Sciences.

FIG. 9. Gain of function and loss of function of RhoB mediate different antitransforming effects of FTI treatment. Cells treated with FTI undergo loss of RhoB-F and gain of RhoB-GG. Loss of RhoB-F is associated with anoikis in Ras-transformed cells, whereas gain of RhoB-GG is sufficient to mediate p21 activation and cell cycle inhibition. p21 activation may be required for RhoB-GG to direct phenotypic reversion and loss of anchorage independence. Other effector mechanisms are also required, since p21 elevation by itself may be necessary but not sufficient for the FTI response (see Results).

tional in transformed cells (a possibility since cell attachment and actin cytoskeletal regulation are subverted in cancer cells). It is possible that RhoB-GG has a gain of function directly related to growth inhibition. RhoB has been linked to some types of growth-inhibitory stimuli (14, 15, 16), so RhoB-GG may act through some physiological RhoB effectors. We favor the interpretation that RhoB-GG actions are based on altered cell localization, which is the simplest interpretation, but we cannot rule out the possibility that certain effectors distinguish RhoB-GG and RhoB-F such that differential biochemical specificities rather than localization changes underlie the effects of RhoB-GG (some RhoB-binding proteins have been identified which distinguish prenylation status [36, 69]). While implicated in inhibitory responses to UV irradiation and TGF-b (14, 15, 16), RhoB has not been linked previously to p21WAF1 regulation. In addition to connections between activated Ras and p21WAF1 (this study and reference 47), activated Raf has been reported to cause cell cycle arrest in normal 3T3 fibroblasts through a p21WAF1-dependent mechanism (68). Our findings would support a role for RhoB in mediating certain antiproliferative effects of oncogenic Ras but tend to suggest that p21WAF1 may be necessary but perhaps not sufficient to mediate FTI effects. The cell cycle inhibitory effects of RhoB-GG may be important to the FTI mechanism in human tumors because cytostatic rather than cytotoxic effects appear to predominate in xenograft models (20, 24, 46, 61, 62). At the pathophysiological level, clinical cancer can be defined by the ability of a cell to survive, proliferate, and invade in the absence of normal adhesion signals. Such signals normally act to stringently govern cell division so that anoikis occurs in their absence. Invasive and metastatic cancers are already substantially resistant to anoikis. If the apoptotic properties of FTIs are based on anoikis mechanisms (39), then one might predict that only premalignant or early-stage malignancies will exhibit apoptosis and show regression, whereas late-stage cancers of the type which are most often seen in the clinical setting will not. In support of this likelihood, while transformed cells are sensitive to anoikis when treated with FTIs or RhoB-GG, we have not seen a similar anoikis sensitivity in any human tumor cell lines tested (unpublished observations). Since these tumor cell lines retain cell cycle inhibitory responses to FTIs, human tumor xenograft models may actually be better models for predicting clinical responses than oncomouse models, which display an apoptotic response to FTIs. ACKNOWLEDGMENTS We are grateful to Allen Oliff and George Hartman for providing L-744,832 and to Wafik el-Deiry for a p21 reporter plasmid. Contributions by the Flow Cytometry Core Facility at The Wistar Institute are gratefully acknowledged. This study was supported by NIH grant CA65892. W.D. and P.F.L. were supported by fellowships from

REFERENCES 1. Adamson, P., C. J. Marshall, A. Hall, and P. A. Tilbrook. 1992. Posttranslational modification of p21rho proteins. J. Biol. Chem. 267:20033–20038. 2. Adamson, P., H. F. Paterson, and A. Hall. 1992. Intracellular localization of the p21rho proteins. J. Cell Biol. 119:617–627. 3. Aepfelbacher, M. 1995. ADP-ribosylation of Rho enhances adhesion of U937 cells to fibronectin via the alpha 5 beta 1 integrin receptor. FEBS Lett. 363: 78–80. 4. Barrington, R. E., M. A. Subler, E. Rands, C. A. Omer, P. J. Miller, J. E. Hundley, S. K. Koester, D. A. Troyer, D. J. Bearss, M. W. Conner, J. B. Gibbs, K. Hamilton, K. S. Koblan, S. D. Mosser, T. J. O’Neill, M. D. Schaber, E. T. Senderak, J. J. Windle, A. Oliff, and N. E. Kohl. 1998. A farnesyltransferase inhibitor induces tumor regression in transgenic mice harboring multiple oncogenic mutations by mediating alterations in both cell cycle control and apoptosis. Mol. Cell. Biol. 18:85–92. 5. Burridge, K., and M. Chrzanowska-Wodnicka. 1996. Focal adhesions, contractility, and signaling. Annu. Rev. Cell Dev. Biol. 12:463–518. 6. Carmichael, J., W. G. DeGraff, A. F. Gazdar, J. D. Minna, and J. B. Mitchell. 1987. Evaluation of a tetrazolium-based semiautomated colorimetric assay: assessment of chemosensitivity testing. Cancer Res. 47:936–942. 7. Casey, P. J., and M. C. Seabra. 1996. Protein prenyltransferases. J. Biol. Chem. 271:5289–5292. 8. Chen, C., and H. Okayama. 1987. High-efficiency transformation of mammalian cells by plasmid DNA. Mol. Cell. Biol. 7:2745–2752. 8a.Cox, A. D. Personal communication. 9. Cox, A. D., and C. J. Der. 1997. Farnesyltransferase inhibitors and cancer treatment: targeting simply Ras? Biochim. Biophys. Acta 1333:F51–F71. 10. Cox, A. D., A. M. Garcia, J. K. Westwick, J. J. Kowalczyk, M. D. Lewis, D. A. Brenner, and C. J. Der. 1994. The CAAX peptidomimetic compound B581 specifically blocks farnesylated, but not geranylgeranylated or myristylated, oncogenic ras signaling and transformation. J. Biol. Chem. 269:1–4. 11. Cox, A. D., M. M. Hisaka, J. E. Buss, and C. J. Der. 1992. Specific isoprenoid modification is required for function of normal, but not oncogenic, Ras function. Mol. Cell. Biol. 12:2606–2615. 12. Debbas, M., and E. White. 1993. Wild-type p53 mediates apoptosis by E1A, which is inhibited by E1B. Genes Dev. 7:546–554. 13. el-Deiry, W. S., T. Tokino, V. E. Velculescu, D. B. Levy, R. Parsons, J. M. Trent, D. Lin, W. E. Mercer, K. W. Kinzler, and B. Vogelstein. 1993. WAF1, a potential mediator of p53 tumor suppression. Cell 75:817–825. 14. Engel, M. E., P. K. Datta, and H. L. Moses. 1998. RhoB is stabilized by transforming growth factor b and antagonizes transcriptional activation. J. Biol. Chem. 273:9921–9926. 15. Fritz, G., and B. Kaina. 1997. rhoB encoding a UV-inducible ras-related small GTP-binding protein is regulated by GTPases of the rho family and independent of JNK, ERK, and p38 MAP kinase. J. Biol. Chem. 272:30637– 30644. 16. Fritz, G., B. Kaina, and K. Aktories. 1995. The ras-related small GTPbinding protein RhoB is immediate-early inducible by DNA damaging treatments. J. Biol. Chem. 270:25172–25177. 17. Gibbs, J., A. Oliff, and N. E. Kohl. 1994. Farnesyltransferase inhibitors: Ras research yields a potential cancer therapeutic. Cell 77:175–178. 18. Gibbs, J. B., and A. Oliff. 1997. The potential of farnesyltransferase inhibitors as cancer chemotherapeutics. Annu. Rev. Pharmacol. Toxicol. 37:143– 166. 19. Gibbs, J. B., M. D. Schaber, T. L. Schofield, E. M. Scolnick, and I. S. Sigal. 1989. Xenopus oocyte germinal-vesicle breakdown induced by [Val12]Ras is inhibited by a cytosol-localized Ras mutant. Proc. Natl. Acad. Sci. USA 86: 6630–6634. 20. Hara, M., K. Akasaka, S. Akinaga, M. Okabe, H. Nakano, R. Gomez, D. Wood, M. Uh, and F. Tamanoi. 1993. Identification of Ras farnesyltransferase inhibitors by microbial screening. Proc. Natl. Acad. Sci. USA 90:2281– 2285. 20a.Harlow, E., and D. Lane (ed.). 1988. Antibodies: a laboratory manual. Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y. 21. Harper, J. W. 1997. Cyclin-dependent kinase inhibitors. Cancer Surv. 29:91– 107. 22. Hotchin, N. A., and A. Hall. 1995. The assembly of integrin adhesion complexes requires both extracellular matrix and intracellular rho/rac GTPases. J. Cell Biol. 131:1857–1865. 23. Hotchin, N. A., and A. Hall. 1996. Regulation of the actin cytoskeleton, integrins and cell growth by the Rho family of small GTPases. Cancer Surv. 27:311–322. 24. Ito, T., S. Kawata, S. Tamura, T. Igura, T. Nagase, J. I. Miyagawa, E. Yamazaki, H. Ishiguro, and Y. Matasuzawa. 1996. Suppression of human pancreatic cancer growth in BALB/c nude mice by manumycin, a farnesyl: protein transferase inhibitor. Jpn. J. Cancer Res. 87:113–116. 25. Jahner, D., and T. Hunter. 1991. The ras-related gene rhoB is an immediateearly gene inducible by v-Fps, epidermal growth factor, and platelet-derived growth factor in rat fibroblasts. Mol. Cell. Biol. 11:3682–3690.

1840

DU ET AL.

26. James, G. L., J. L. Goldstein, and M. S. Brown. 1995. Polylysine and CVIM sequences of K-RasB dictate specificity of prenylation and confer resistance to benzodiazepine peptidomimetic in vitro. J. Biol. Chem. 270:6221–6226. 27. James, G. L., J. L. Goldstein, M. S. Brown, T. E. Rawson, T. C. Somers, R. S. McDowell, C. W. Crowley, B. K. Lucas, A. D. Levinson, and J. C. Marsters. 1993. Benzodiazepine peptidomimetics: potent inhibitors of Ras farnesylation in animal cells. Science 260:1937–1942. 28. Kato, K., A. D. Cox, M. M. Hisaka, S. M. Graham, J. E. Buss, and C. J. Der. 1992. Isoprenoid addition to Ras protein is the critical modification for its membrane association and transforming activity. Proc. Natl. Acad. Sci. USA 89:6403–6407. 29. Khosravi-Far, R., P. A. Solski, G. J. Clark, M. S. Kinch, and C. J. Der. 1995. Activation of Rac1, RhoA, and mitogen-activated protein kinase are required for Ras transformation. Mol. Cell. Biol. 15:6443–6453. 30. Kohl, N. E., S. D. Mosser, S. J. deSolms, E. A. Giuliani, D. L. Pompliano, S. L. Graham, R. L. Smith, E. M. Scolnick, A. Oliff, and J. B. Gibbs. 1993. Selective inhibition of ras-dependent transformation by a farnesyltransferase inhibitor. Science 260:1934–1937. 31. Kohl, N. E., C. A. Omer, M. W. Conner, N. J. Anthony, J. P. Davide, S. J. deSolms, E. A. Giuliani, R. P. Gomez, S. L. Graham, K. Hamilton, L. K. Handt, G. D. Hartman, K. S. Koblan, A. M. Kral, P. J. Miller, S. D. Mosser, T. J. O’Neill, E. Rands, M. D. Schaber, J. B. Gibbs, and A. Oliff. 1995. Inhibition of farnesyltransferase induces regression of mammary and salivary carcinomas in ras transgenic mice. Nat. Med. 1:792–797. 32. Kohl, N. E., F. Redner, S. Mosser, E. A. Guiliani, S. J. deSolms, M. W. Conner, N. J. Anthony, W. J. Holtz, R. P. Gomez, T.-J. Lee, R. L. Smith, S. L. Graham, G. D. Hartman, J. Gibbs, and A. Oliff. 1994. Protein farnesyltransferase inhibitors block the growth of ras-dependent tumors in nude mice. Proc. Natl. Acad. Sci. USA 91:9141–9145. 33. Lamaze, C., T.-H. Chuang, L. J. Terlecky, G. M. Bokoch, and S. L. Schmid. 1996. Regulation of receptor-mediated endocytosis by Rho and Rac. Nature 382:177–179. 34. Lebowitz, P., P. J. Casey, G. C. Prendergast, and J. Thissen. 1997. Farnesyltransferase inhibitors alter the prenylation and growth-stimulating function of RhoB. J. Biol. Chem. 272:15591–15594. 35. Lebowitz, P., W. Du, and G. C. Prendergast. 1997. Prenylation of RhoB is required for its cell transforming functions but not its ability to activate SRE-dependent transcription. J. Biol. Chem. 272:16093–16096. 36. Lebowitz, P., and G. C. Prendergast. 1998. Functional interaction between RhoB and the transcription factor DB1. Cell Adhesion Commun. 4:1–11. 37. Lebowitz, P. F., J. P. Davide, and G. C. Prendergast. 1995. Evidence that farnesyl transferase inhibitors suppress Ras transformation by interfering with Rho activity. Mol. Cell. Biol. 15:6613–6622. 38. Lebowitz, P. F., and G. C. Prendergast. 1998. Non-Ras targets for farnesyltransferase inhibitors: focus on Rho. Oncogene 17:1439–1445. 39. Lebowitz, P. F., D. Sakamuro, and G. C. Prendergast. 1997. Farnesyltransferase inhibitors induce apoptosis in Ras-transformed cells denied substratum attachment. Cancer Res. 57:708–713. 40. Leonard, D. M. 1997. Ras farnesyltransferase: a new therapeutic target. J. Med. Chem. 40:2971–2990. 41. Lerner, E. C., T. T. Zhang, D. B. Knowles, Y. M. Qian, A. D. Hamilton, and S. M. Sebti. 1997. Inhibition of the prenylation of K-Ras, but not H- or N-Ras, is highly resistant to CAAX peptidomimetics and requires both a farnesyltransferase and a geranylgeranyltransferase-I inhibitor in human tumor cell lines. Oncogene 15:1283–1288. 42. Lin, H.-J., V. Eviner, G. C. Prendergast, and E. White. 1995. Activated H-ras rescues E1A-induced apoptosis and cooperates with E1A to overcome p53dependent growth arrest. Mol. Cell. Biol. 15:4536–4544. 43. Mangues, R., T. Corral, N. E. Kohl, W. F. Symmans, S. Lu, M. Malumbres, J. B. Gibbs, A. Oliff, and A. Pellicer. 1998. Antitumor effect of a farnesyl protein transferase inhibitor in mammary and lymphoid tumors overexpressing N-ras in transgenic mice. Cancer Res. 58:1253–1259. 44. Manne, V., N. Yan, J. M. Carboni, A. V. Tuomari, C. S. Ricca, J. G. Brown, M. L. Andahazy, R. J. Schmidt, D. Patel, R. Zahler, R. Weinmann, C. J. Der, A. D. Cox, J. T. Hunt, E. M. Gordon, M. Barbacid, and B. R. Seizinger. 1995. Bisubstrate inhibitors of farnesyltransferase: a novel class of specific inhibitors of ras transformed cells. Oncogene 10:1763–1779. 45. Mellor, J., P. Flynn, C. D. Nobes, A. Hall, and P. J. Parker. 1998. PRK1 is targeted to endosomes by the small GTPase, RhoB. J. Biol. Chem. 273:4811– 4814. 46. Nagasu, T., K. Yoshimatsu, C. Rowell, M. D. Lewis, and A. M. Garcia. 1995. Inhibition of human tumor xenograft growth by treatment with the farnesyltransferase inhibitor B956. Cancer Res. 55:5310–5314. 47. Olson, M. F., H. F. Paterson, and C. J. Marshall. 1998. Signals from Ras and Rho GTPases interact to regulate expression of p21Waf1/Cip1. Nature 394: 295–299.

MOL. CELL. BIOL. 47a.Prendergast, G. C. Unpublished observations. 48. Prendergast, G. C., and M. D. Cole. 1989. Posttranscriptional regulation of cellular gene expression by the c-myc oncogene. Mol. Cell. Biol. 9:124–134. 49. Prendergast, G. C., J. P. Davide, S. J. deSolms, E. Giuliani, S. Graham, J. B. Gibbs, A. Oliff, and N. E. Kohl. 1994. Farnesyltransferase inhibition causes morphological reversion of ras-transformed cells by a complex mechanism that involves regulation of the actin cytoskeleton. Mol. Cell. Biol. 14:4193– 4202. 50. Prendergast, G. C., R. Khosravi-Far, P. Solski, H. Kurzawa, P. Lebowitz, and C. J. Der. 1995. Critical role of Rho in cell transformation by oncogenic Ras. Oncogene 10:2289–2296. 51. Qiu, R. G., J. Chen, F. McCormick, and M. Symons. 1995. A role for Rho in Ras transformation. Proc. Natl. Acad. Sci. USA 92:11781–11785. 52. Ridley, A. J., and A. Hall. 1992. The small GTP-binding protein rho regulates the assembly of focal adhesions and actin stress fibers in response to growth factors. Cell 70:389–399. 53. Rowell, C. A., J. J. Kowalczyk, M. D. Lewis, and A. M. Garcia. 1997. Direct demonstration of geranylgeranylation and farnesylation of Ki-Ras in vivo. J. Biol. Chem. 272:14093–14097. 54. Sakamuro, D., P. Sabbatini, E. White, and G. C. Prendergast. 1997. The polyproline region of p53 is required to activate apoptosis but not growth arrest. Oncogene 15:887–898. 55. Schmalzing, G., H. P. Richter, A. Hansen, W. Schwarz, I. Just, and K. Aktories. 1995. Involvement of the GTP binding protein Rho in constitutive endocytosis in Xenopus laevis oocytes. J. Cell Biol. 130:1319–1332. 56. Sebti, S. M., and A. D. Hamilton. 1997. Inhibition of Ras prenylation: a novel approach to cancer chemotherapy. Pharmacol. Ther. 74:103–114. 57. Sepp-Lorenzino, L., Z. Ma, E. Rands, N. E. Kohl, J. B. Gibbs, A. Oliff, and N. Rosen. 1995. A peptidomimetic inhibitor of farnesyl:protein transferase blocks the anchorage-dependent and -independent growth of human tumor cell lines. Cancer Res. 55:5302–5309. 58. Sepp-Lorenzino, L., and N. Rosen. 1998. A farnesylprotein transferase inhibitor induces p21 expression and G1 block in p53 wild type tumor cells. J. Biol. Chem. 273:20243–20251. 59. Serrano, M., A. W. Lin, M. E. McCurrach, D. Beach, and S. W. Lowe. 1997. Oncogenic ras provokes premature cell senescence associated with accumulation of p53 and p16INK4a. Cell 88:593–602. 60. Shih, T. Y., P. E. Stokes, G. W. Smythers, R. Dhar, and S. Oroszlan. 1982. Characterization of the phosphorylation sites and the surrounding amino acid sequences of the p21 transforming proteins coded for by the Harvey and Kirsten strains of murine sarcoma viruses. J. Biol. Chem. 257:11767–11773. 61. Sun, J., Y. Qian, A. D. Hamilton, and S. M. Sebti. 1998. Both farnesyltransferase and geranylgeranyltransferase I inhibitors are required for inhibition of oncogenic K-Ras prenylation but each alone is sufficient to suppress human tumor growth in nude mouse xenografts. Oncogene 16:1467–1473. 62. Sun, J., Y. Qian, A. D. Hamilton, and S. M. Sebti. 1995. Ras CAAX peptidomimetic FTI 276 selectively blocks tumor growth in nude mice of a human lung carcinoma with K-Ras mutation and p53 deletion. Cancer Res. 55:4243–4247. 63. Symons, M. 1996. Rho family GTPases: the cytoskeleton and beyond. Trends Biochem. Sci. 21:178–181. 64. Tapon, N., and A. Hall. 1997. Rho, Rac and Cdc42 GTPases regulate the organization of the actin cytoskeleton. Curr. Opin. Cell Biol. 9:86–92. 65. Udagawa, T., and B. W. McIntyre. 1996. ADP-ribosylation of the G protein Rho inhibits integrin regulation of tumor cell growth. J. Biol. Chem. 271: 12542–12548. 66. Van Aelst, L., and C. D’Souza-Schorey. 1997. Rho GTPases and signaling networks. Genes Dev. 11:2295–2322. 67. Whyte, D. B., P. Kirschmeier, T. N. Hockenberry, I. Nunez-Olivia, L. James, J. J. Catino, W. R. Bishop, and J. K. Pai. 1997. K- and N-ras geranylgeranylated in cells treated with farnesyl protein transferase inhibitors. J. Biol. Chem. 272:14459–14464. 68. Woods, D., D. Parry, H. Cherwinski, E. Bosch, E. Lees, and M. McMahon. 1997. Raf-induced proliferation or cell cycle arrest is determined by the level of Raf activity with arrest mediated by p21Cip1. Mol. Cell. Biol. 17:5598–5611. 69. Zalcman, G., V. Closson, J. Camonis, N. Honore, M. F. Rousseau-Merck, A. Tavitian, and B. Olofsson. 1996. RhoGDI-3 is a new GDP dissociation inhibitor (GDI). Identification of a non-cytosolic GDI protein interacting with the small GTP-binding proteins RhoB and RhoG. J. Biol. Chem. 271: 30366–30374. 70. Zalcman, G., V. Closson, G. Linares-Cruz, F. Leregours, N. Honore, A. Tavitian, and B. Olofsson. 1995. Regulation of Ras-related RhoB protein expression during the cell cycle. Oncogene 10:1935–1945. 71. Zhang, F. L., and P. J. Casey. 1996. Protein prenylation: molecular mechanisms and functional consequences. Annu. Rev. Biochem. 65:241–269.