Skip to main content
Proceedings of the National Academy of Sciences of the United States of America logoLink to Proceedings of the National Academy of Sciences of the United States of America
. 2010 Mar 22;107(14):6471–6476. doi: 10.1073/pnas.0908396107

Targeting the protein prenyltransferases efficiently reduces tumor development in mice with K-RAS-induced lung cancer

Meng Liu a,b,1, Anna-Karin M Sjogren a,1, Christin Karlsson a, Mohamed X Ibrahim a, Karin M E Andersson a, Frida J Olofsson a, Annika M Wahlstrom a, Martin Dalin a, Huiming Yu b, Zhenggang Chen a,b, Shao H Yang c, Stephen G Young c, Martin O Bergo a,2
PMCID: PMC2852017  PMID: 20308544

Abstract

RAS and RHO proteins, which contribute to tumorigenesis and metastasis, undergo posttranslational modification with an isoprenyl lipid by protein farnesyltransferase (FTase) or protein geranylgeranyltransferase-I (GGTase-I). Inhibitors of FTase and GGTase-I were developed to block RAS-induced malignancies, but their utility has been difficult to evaluate because of off-target effects, drug resistance, and toxicity. Moreover, the impact of FTase deficiency and combined FTase/GGTase-I deficiency has not been evaluated with genetic approaches. We found that inactivation of FTase eliminated farnesylation of HDJ2 and H-RAS, prevented H-RAS targeting to the plasma membrane, and blocked proliferation of primary and K-RASG12D-expressing fibroblasts. FTase inactivation in mice with K-RAS-induced lung cancer reduced tumor growth and improved survival, similar to results obtained previously with inactivation of GGTase-I. Simultaneous inactivation of FTase and GGTase-I markedly reduced lung tumors and improved survival without apparent pulmonary toxicity. These data shed light on the biochemical and therapeutic importance of FTase and suggest that simultaneous inhibition of FTase and GGTase-I could be useful in cancer therapeutics.

Keywords: mouse models, non-small-cell lung cancer, protein farnesyltransferase, protein geranylgeranyltransferase type I


Many intracellular proteins, such as the RAS and RHO family proteins, are posttranslationally lipidated at a carboxyl-terminal CAAX motif. This process is called isoprenylation and is carried out by a pair of cytosolic enzymes, protein farnesyltransferase (FTase) and protein geranylgeranyltransferase type I (GGTase-I) (1). FTase and GGTase-I share a common α-subunit but have unique β-subunits that determine substrate specificity (2, 3). Some CAAX proteins, such as H-RAS, HDJ2, and prelamin A, are substrates for FTase, whereas others, such as RAP1A and RHOA, are substrates for GGTase-I (3). Protein isoprenylation facilitates membrane interactions, promotes protein–protein interactions, and can affect protein turnover (4).

The RAS proteins, by far the most thoroughly studied CAAX proteins, are involved in the pathogenesis of many forms of cancer (5). Because isoprenylation is essential for the plasma membrane targeting of the RAS proteins and their ability to transform cells (4), FTase inhibitors (FTIs) have been developed and tested as anticancer agents (6). FTIs showed efficacy in preclinical studies of malignancies, including those without RAS mutations (79). However, clinical trials of FTIs in humans have not been particularly successful (6). The mechanism by which FTIs inhibit cell growth is not entirely clear, but it likely involves interfering with the farnesylation of several CAAX proteins, in addition to RAS (6, 10, 11). Also, different FTIs have different properties (1215), complicating efforts to define compound- versus mechanism-related effects.

A few years ago, Mijimolle et al. (16) attempted to address the functional relevance of FTase by generating mice with a conditional knockout allele for the gene encoding the β-subunit of FTase (Fntb). Cre-mediated recombination appeared to inhibit the farnesylation of HDJ2 and H-RAS, but only partially, and, most remarkably, H-RAS remained in the membrane fraction of cells. They also reported that Fntb-deficient fibroblasts grew in culture and that the development of K-RAS-induced tumors was unaffected by Fntb deficiency. These findings were surprising for several reasons. First, FTI treatment studies had suggested that the membrane association of H-RAS is utterly dependent on protein farnesylation (17). Second, a nonprenylated mutant of H-RAS (C186S) is found exclusively in the soluble, cytosolic fraction of cells (18). Third, FTI treatment of cells typically results in cell-cycle arrest (19, 20). Fourth, in mouse models, FTIs are efficacious against many tumors, including those without RAS mutations (9, 21).

A potential explanation for the differences between the genetic and pharmacologic studies is that FTIs might affect other proteins aside from FTase. Another is that the Fntb knockout allele generated by Mijimolle et al. (16) yielded a transcript with an in-frame deletion (22), and it is conceivable that this mutant transcript yielded a protein with some residual enzymatic activity.

In FTI-treated cells, K-RAS and N-RAS are alternately prenylated by GGTase-I (2325). That finding prompted both pharmaceutical companies and academic laboratories to develop GGTase-I inhibitors (GGTIs) (26), which have shown promise in preclinical studies (2731). The rationale for inhibiting GGTase-I is supported by genetic studies in mice: Inactivating the gene for the β-subunit of GGTase-I (Pggt1b) reduced tumor formation and prolonged survival in mice with K-RAS-induced lung cancer (32).

Because neither an FTI alone nor a GGTI alone inhibits the prenylation of K- and N-RAS, FTI/GGTI combinations and dual-prenylation inhibitors (DPIs) were developed (33, 34). DPIs and FTI/GGTI combinations block K-RAS prenylation in vivo, but only at high doses that are toxic in mice. However, some studies with FTI/GGTI combinations did not report significant toxicity (27, 35). Thus far, no one has used genetic approaches to study dual inhibition of FTase and GGTase-I.

In this study, we created a conditional knockout allele for Fntb and reevaluated the impact of Fntb deficiency on protein isoprenylation, cell proliferation, and the growth of K-RAS-induced tumors. We also bred mice homozygous for conditional knockout alleles in both Fntb and Pggt1b and assessed the effect of combined FTase/GGTase-I deficiency on the development of K-RAS-induced lung cancer.

Results

Generation and Validation of a Conditional FTase Knockout Allele.

To create a conditional knockout allele (Fntbfl) for the β-subunit of FTase, we introduced loxP sites 1 kb upstream and 1 kb downstream of exon 1 (Fig. 1A). Mice homozygous for the conditional allele (Fntbfl/fl) were healthy and fertile. Fntbfl/fl mice were bred first with EIIa-Cre mice (36) to remove a floxed neo cassette and then with deleter-Cre mice (37) to produce mice harboring one conditional knockout allele and one knockout allele (Fntbfl/Δ). Primary fibroblasts were cultured from E13.5 Fntbfl/fl embryos. When these cells were incubated with Cre-adenovirus (adCre), they were converted to FntbΔ/Δ derivatives that had no detectable Fntb expression (Fig. 1B).

Fig. 1.

Fig. 1.

A conditional knockout allele for the β-subunit of FTase (Fntbfl). (A) Schematic of the Fntb gene-targeting vector. LoxP sites were inserted 1 kb upstream and downstream of exon 1. Arrows show the locations of primers for genotyping. neo, neomycin-resistance cassette; tk, thymidine kinase cassette. (B) PCR and RT-PCR analyses demonstrating the inactivation of Fntb by treatment with adCre. Fntbfl/+ and Fntbfl/fl fibroblasts were incubated with adβgal or adCre, and genomic DNA and total RNA were isolated 4 days later. (C) Western blots of extracts from Fntbfl/+ and Fntbfl/Δ fibroblasts incubated with adβgal or adCre. An antibody against actin was used as a loading control. (D) Western blots showing the distribution of proteins in the membrane (mem) and cytosolic (cyt) fractions of adβgal- or adCre-treated fibroblasts. (E) (Upper) K-RAS western blot of extracts from Fntbfl/flPggt1bfl/fl fibroblasts that had been incubated with adβgal or adCre. (Lower) Western blot showing the distribution of K-RAS in the membrane and cytosolic fractions of Fntbfl/flPggt1bfl/fl fibroblasts incubated with adβgal or adCre. (F) N-RAS western blot of Fntbfl/Δ fibroblasts treated with adCre and 10 μM GGTI for 4 days.

To assess the impact of Fntb deficiency on the isoprenylation of FTase substrates, we performed western blots of lysates from β-gal adenovirus- (adβgal) and adCre-treated Fntbfl/Δ fibroblasts and control fibroblasts (Fntbfl/+). HDJ2 and H-RAS in extracts of adCre-treated Fntbfl/Δ cells exhibited reduced electrophoretic mobilities (Fig. 1C), characteristic of the nonfarnesylated forms of these proteins. The absolute levels of HDJ2 did not change, but the levels of H-RAS in adCre-treated Fntbfl/Δ cells were 2- to 4-fold higher than in control cells, as judged by densitometry (Fig. 1C). The electrophoretic mobilities of K-RAS and N-RAS were unchanged, likely because these proteins are isoprenylated by GGTase-I (2325). HDJ2 and H-RAS accumulated in the cytosolic fraction of FntbΔ/Δ cells, whereas K- and N-RAS remained associated with the membrane fraction (Fig. 1D). Inactivating a single Fntb allele in Fntbfl/+ fibroblasts with adCre did not affect the electrophoretic mobilities or the membrane/cytosolic partitioning of HDJ2 and H-RAS (Fig. 1 C and D).

To determine whether K-RAS and N-RAS are geranylgeranylated in FntbΔ/Δ cells, we isolated Fntbfl/flPggt1bfl/fl fibroblasts and treated them with adCre to inactivate both Fntb and Pggt1b (FntbΔ/ΔPggt1bΔ/Δ). In FntbΔ/ΔPggt1bΔ/Δ cells, a substantial proportion of K-RAS accumulated in the cytosolic fraction and exhibited a reduced electrophoretic mobility (Fig. 1E). A similar shift in the mobility of N-RAS was observed in FntbΔ/Δ cells treated with a GGTI (Fig. 1F). FntbΔ/ΔPggt1bΔ/Δ cells remained viable for a few days, but they underwent apoptosis and died within 4 days (Fig. S1 A and B). Inactivating Fntb and Pggt1b in cells expressing oncogenic H-RAS targeted to the plasma membrane by an amino-terminal myristoylation sequence also underwent apoptosis (Fig. S1 C and D).

Fntb Deficiency Blocks Proliferation of Primary and K-RASG12D-Expressing Fibroblasts.

Inactivating a single Fntb allele in Fntbfl/+ fibroblasts with adCre did not affect cell proliferation (Fig. 2A). However, inactivating both alleles by treating primary Fntbfl/Δ fibroblasts with adCre dramatically reduced proliferation (Fig. 2A). Similar results were found with primary and immortalized Fntbfl/fl cells. Genotypically confirmed FntbΔ/Δ fibroblasts were large and flat and accumulated in the G2M phase of the cell cycle (Fig. 2 B and C), but the number of apoptotic cells was low (Fig. S1A). Inactivating Fntb in primary fibroblasts increased p21CIP1 levels and delayed serum-stimulated phosphorylation of AKT but did not affect levels of phosphorylated MEK and ERK1/2 (Fig. S2).

Fig. 2.

Fig. 2.

Inactivating Fntb stops fibroblast proliferation. (A) Proliferation of Fntbfl/+ and Fntbfl/Δ primary mouse fibroblasts incubated with adβgal or adCre. Data show a single cell line assayed in triplicate. Similar results were obtained with two different cell lines of each genotype. **P < 0.01; ***P < 0.001. (B) Photomicrographs of Fntbfl/Δ cells incubated with adβgal or adCre. (C) Cell-cycle analysis of Fntbfl/Δ fibroblasts incubated with adβgal or adCre. PI, propidium iodide. (D) PCR genotyping of genomic DNA from Fntbfl/fl fibroblasts at various times after incubation with adβgal or adCre. (E) PCR genotyping of genomic DNA from individual clones of adβgal- or adCre-treated Fntbfl/+ and Fntbfl/fl fibroblasts. (F) Proliferation of primary Fntbfl/+KLSL and Fntbfl/ΔKLSL fibroblasts incubated with adβgal or adCre. Data represent the mean of a single cell line/genotype assayed in triplicate. Similar results were obtained in two independent experiments with two cell lines/genotypes. Inset shows PCR genotyping of genomic DNA demonstrating the activation of the KG12D allele in cells incubated with adCre (ii and iv) but not in cells incubated with adβgal (i and iii). (G) Cell-cycle analysis of adβgal- and adCre-treated Fntbfl/ΔKLSL fibroblasts. The experiment was repeated three times with similar results.

When adCre-treated Fntbfl/fl cells were left on the culture plates for more than a week, cell growth gradually resumed. However, this growth was due to overgrowth by Fntbfl/Δ cells (rather than FntbΔ/Δ cells) (Fig. 2D). Despite extensive efforts, we were unable to clone FntbΔ/Δ fibroblasts from adCre-treated Fntbfl/fl cells. In more than 12 independent experiments, cell growth late after adCre treatment was invariably due to overgrowth by Fntbfl/Δ cells (Fig. 2E). In contrast, we had no difficulty in obtaining stable FntbΔ/+ cell lines after treating Fntbfl/+ cells with adCre (Fig. 2E).

To assess the impact of Fntb deficiency on the proliferation of cells expressing K-RASG12D, we isolated primary fibroblasts from Fntbfl/Δ and Fntbfl/+ embryos harboring an inducible oncogenic K-RAS allele (KLSL) (38). The KLSL allele is normally silent but K-RASG12D expression can be induced with Cre. Incubation of Fntbfl/+KLSL fibroblasts with adCre yielded FntbΔ/+KG12D cells that proliferated more rapidly (Fig. 2F). In contrast, adCre treatment of Fntbfl/ΔKLSL cells (producing FntbΔ/ΔKG12D cells) resulted in G2M cell-cycle arrest (Fig. 2 F and G).

Inactivating Fntb Increases Survival of Mice with K-RAS-Induced Lung Cancer.

To determine the effect of an Fntb knockout on the development of a K-RAS-induced malignancy, we bred mice car-rying the KLSL allele (K) and a lysozyme M-Cre allele (L) on Fntbfl/Δ and Fntbfl/+ backgrounds (mice harboring both alleles were designated KL). Littermate Fntbfl/ΔL mice were monitored to assess the impact of Fntb deficiency in the absence of K-RAS-induced tumors; Fntbfl/+L, Fntbfl/+K, and Fntbfl/ΔK mice were used as healthy controls (Ctr mice).

We previously showed that KL mice express K-RASG12D in most or all type II pneumocytes and develop lung cancer that is fatal by 25 days of age. Lung weight is increased ∼10-fold and alveoli are obliterated by diffuse hyperplasia and adenocarcinoma (32). In keeping with those findings, the maximum survival of Fntbfl/+KL mice was 24 days (Fig. 3A). The survival of Fntbfl/ΔKL mice was significantly longer, >100 days in some cases (P < 0.0001; Fig. 3A). The lung weights in 3-week-old Fntbfl/ΔKL mice were lower than in Fntbfl/+KL mice (P < 0.0001) but higher than in Fntbfl/ΔL mice or Ctr mice (Fig. 3B). Moreover, histologic analyses revealed nearly complete obliteration of alveoli in Fntbfl/+KL lungs, whereas Fntbfl/ΔKL lungs retained areas of normal histology (Fig. 3C). Survival, lung weight, and lung histology in Fntbfl/ΔL mice were indistinguishable from those of Ctr mice (Fig. 3 AC).

Fig. 3.

Fig. 3.

Inactivation of Fntb reduces tumor development and prolongs survival of mice with K-RAS-induced lung cancer. (A) Kaplan–Meier curve showing increased survival in Fntbfl/ΔKL mice (n = 16) compared with Fntbfl/+KL mice (n = 10). All Fntbfl/ΔL mice (n = 9) were alive at the end of the experiment (150 days). (B) Lung weight in 3-week-old Fntbfl/+KL (n = 8), Fntbfl/ΔKL (n = 7), Fntbfl/ΔL (n = 6), and Ctr (n = 5) mice. (C) Hematoxylin/eosin-stained sections of lungs from 3-week-old mice. (Scale bars, 100 μm.) (D) Genotyping of wild-type and activated KG12D alleles and the recombined FntbΔ allele by PCR amplification of genomic DNA from lung tissue. (E) Western blot showing the distribution of H-RAS and K-RAS in membrane and cytosolic fractions of lung lysates. (F) Western blots of protein lysates from the lungs of 3-week-old mice. Actin was used as a loading control.

PCR genotyping of genomic DNA from Fntbfl/ΔKL lungs revealed activation of the K-RASG12D allele and a recombined Fntb allele (Fig. 3D). We harvested protein lysates from lung tissue from 3-week-old Fntbfl/+KL and Fntbfl/ΔKL mice and performed western blots with antibodies against the CAAX proteins, H-RAS, HDJ2, and prelamin A, which serve as markers of cellular FTase activity (39). In lung extracts of Fntbfl/ΔKL mice, a substantial proportion of H-RAS accumulated in the cytosolic fraction, whereas K-RAS remained in the membrane fraction (Fig. 3E). Approximately 50% of the HDJ2 in Fntbfl/ΔKL lungs exhibited a reduced electrophoretic mobility (Fig. 3F). Also, reduced FTase activity resulted in an accumulation of nonfarnesylated prelamin A (Fig. 3F). There was no difference in levels of phosphorylated ERK1/2 in lung extracts from Fntbfl/+KL and Fntbfl/ΔKL lungs (Fig. 3F).

Simultaneous Inactivation of Fntb and Pggt1b Reduces Tumor Load.

To assess the effect of combined Fntb and Pggt1b deficiency on K-RAS-induced tumors, we bred Fntbfl/ΔPggt1bfl/ΔKL mice and control KL mice in which neither prenyltransferase was inactivated. Littermate Fntbfl/ΔPggt1bfl/ΔL mice were analyzed to determine the impact of Fntb/Pggt1b deficiency in the absence of K-RAS-induced tumors. Fntbfl/+Pggt1bfl/+L, Fntbfl/ΔPggt1bfl/+L, and Fntbfl/+Pggt1bfl/ΔL mice (collectively designated Ctr) were used as healthy controls.

Lung weight and histology were indistinguishable in 3-week-old Fntbfl/ΔPggt1bfl/ΔKL and Ctr mice (Fig. 4 A and B). But in 3-week-old KL mice, lung weight was 10-fold higher than in Ctr mice (as a result of tumor burden), and the maximum survival was 24 days (Fig. 4 AC). The levels of phosphorylated ERK1/2 in lung lysates were lower in Fntbfl/ΔPggt1bfl/ΔKL mice than in KL mice (Fig. 4D), as were Ki-67 levels (as judged by immunofluorescence confocal microscopy) (Fig. 4E). Fntbfl/ΔPggt1bfl/ΔKL mice survived far longer than KL mice (median, 170 vs. 22 days; P < 0.0001) (Fig. 4C), but all eventually developed tumors and were euthanized. Lung histology and survival were indistinguishable in Fntbfl/ΔPggt1bfl/ΔL and Ctr mice (Fig. 4 B and C).

Fig. 4.

Fig. 4.

Simultaneous inactivation of Fntb and Pggt1b in mice with K-RASG12D-induced lung cancer. (A) Lung weight of 3-week-old KL (n = 4), Fntbfl/ΔPggt1bfl/ΔKL (n = 6), Fntbfl/ΔPggt1bfl/ΔL (n = 5), and Ctr (n = 9) mice. BW, body weight. (B) Representative hematoxylin/eosin-stained lung sections of mice from the experiment shown in A. (Scale bars, 100 μm.) (C) Kaplan–Meier curve showing survival of KL (n = 12), Fntbfl/ΔPggt1bfl/ΔKL (n = 10), and Fntbfl/ΔPggt1bfl/ΔL (n = 9) mice. Black tick marks indicate healthy Fntbfl/ΔPggt1bfl/ΔL mice that were euthanized for tissue analyses. (D) Western blots of protein extracts from the lungs of 3-week-old KL and Fntbfl/ΔPggt1bfl/ΔKL mice showing levels of phosphorylated ERK1/2. Total ERK1/2 was used as a loading control. (E) Confocal immunofluorescence micrographs showing expression of Ki-67 (pink) in cells from KL, Fntbfl/ΔPggt1bfl/ΔKL, and Ctr mice. Nuclei were visualized with DAPI (blue).

To determine whether farnesylation and geranylgeranylation were inhibited in lungs of Fntbfl/ΔPggt1bfl/ΔKL mice, we performed western blots on lung lysates from 3-week-old mice. Approximately 25% of HDJ2 exhibited a reduced electrophoretic mobility, characteristic of the nonfarnesylated protein. Protein geranylgeranylation was also inhibited, as the nonprenylated (np) form of RAP1A accumulated in lung lysates (Fig. 5A). Moreover, a large proportion of K-RAS accumulated in the cytosolic fraction of lung lysates from Fntbfl/ΔPggt1bfl/ΔKL mice, suggesting that many cells lacked both FTase and GGTase-I activity (Fig. 5B). Immunofluorescence and confocal microscopy revealed prelamin A staining in lung sections from Fntbfl/ΔKL and Fntbfl/ΔPggt1bfl/ΔKL mice, whereas np-RAP1A staining was detected only in the lungs of Fntbfl/ΔPggt1bfl/ΔKL mice (Fig. 5C). Some cells in lungs from Fntbfl/ΔPggt1bfl/ΔKL mice exhibited strong staining for both prelamin A and np-RAP1A (Fig. 5C and Fig. S3A). These double-positive cells were also identified in lungs of Fntbfl/ΔPggt1bfl/ΔL mice, and some of those cells were type II pneumocytes because they were also positive for SP-C (Fig. S3B). Moreover, K-RAS was identified in the cytosolic fraction of lung lysates from Fntbfl/ΔPggt1bfl/ΔL mice (Fig. S3C). Despite the presence of cells that apparently lacked both FTase and GGTase-I activity, we found no evidence of apoptosis in lung sections of Fntbfl/ΔPggt1bfl/ΔL mice (Fig. S4).

Fig. 5.

Fig. 5.

Reduced FTase and GGTase-I activities in lungs of Fntbfl/ΔPggt1bfl/ΔKL mice. (A) Western blots of protein extracts from lungs of 3-week-old KL and Fntbfl/ΔPggt1bfl/ΔKL mice. Actin was used as a loading control. (B) Western blot showing the distribution of K-RAS in the membrane and cytosolic fractions of KL and Fntbfl/ΔPggt1bfl/ΔKL mice. (C) Confocal immunofluorescence microscopy showing prelamin A (red) and np-RAP1A (green) expression in lung sections from 3-week-old KL, Fntbfl/ΔKL, Fntbfl/ΔPggt1bfl/ΔKL, and Ctr mice. Nuclei were stained with DAPI (blue). The specificity of prelamin A staining was established with lung sections of Zmpste24−/− mice (where prelamin A accumulates due to a defect in the conversion of farnesyl-prelamin A to mature lamin A). Arrows indicate cells positive for both prelamin A and np-RAP1A. Asterisk, alveolus. (Scale bars, 25 μm.) (D) Recombination efficiency of the “floxed” KLSL, Fntbfl, and Pggt1bfl alleles determined by quantitative PCR of genomic DNA from lung biopsies of 3-week-old Fntbfl/ΔPggt1bfl/ΔKL mice (n = 5) and from lung tumors of 28-week-old Fntbfl/ΔPggt1bfl/ΔKL mice (n = 2).

In the immunohistochemistry analyses, we detected cells in lungs of Fntbfl/ΔPggt1bfl/ΔKL mice that were positive for np-RAP1A but not prelamin A (Fig. S3A) and cells that were negative for both. We suspect that incomplete recombination in the Fntb and Pggt1b alleles underlies the development of tumors in older Fntbfl/ΔPggt1bfl/ΔKL mice. Indeed, the percentage of genomic DNA with an inactivated Fntb allele fell from 18% in 3-week-old mice to 5% in the tumors of 198-day-old mice. Simultaneously, the percent inactivation of the Pggt1b gene fell from 20% in 3-week-old mice to 10% in tumors of 198 day-old mice (Fig. 5D).

Simultaneous Inactivation of Fntb and Pggt1b Inhibits Tumorigenesis in a Second K-RAS-Induced Lung Tumor Model.

To further investigate how inactivating Fntb and Pggt1b affects tumor development, we administered adCre to Fntbfl/ΔPggt1bfl/ΔK mice, K mice heterozygous for one or both of the conditional alleles, and healthy Ctr mice (38). AdCre results in KG12D expression in a limited number of cells, leading to a limited number of tumors that can be characterized in number, grade, and lesion area. Eight weeks after administration of 5 × 107 pfu adCre, K mice had large tumors that were easily visible on the surface of the lungs (Fig. 6 AC). However, the lung surface of adCre-treated Fntbfl/ΔPggt1bfl/ΔK mice was nearly indistinguishable from that of Ctr mice, and Fntbfl/ΔPggt1bfl/ΔK mice had 76% fewer tumors and 79% smaller lesion area than K mice (Fig. 6 AC). The lung lesions in K mice ranged from atypical adenomatous hyperplasia and epithelial hyperplasia to adenocarcinoma, the most common lesion (identified in 7 of 8 mice; Fig. 6D). The lung histology of Fntbfl/ΔPggt1bfl/ΔK mice ranged from entirely normal to the presence of epithelial hyperplasia (Fig. 6D) and small adenomas; adenocarcinoma was observed in only 1 of 11 mice.

Fig. 6.

Fig. 6.

Inactivation of Fntb and Pggt1b reduces tumor burden in a second K-RASG12D-induced lung cancer model. (A) Photographs of lungs 8 weeks after inhalation of adCre. Note the extensive lesions (white areas) on the surface of lungs from K mice and the relatively normal appearance of the lungs from Fntbfl/ΔPggt1bfl/ΔK mice. (B and C) Tumor number (B) and surface area (C) in lungs from K (n = 8) and Fntbfl/ΔPggt1bfl/ΔK (n = 11) mice 8 weeks after inhalation of adCre. (D) Hematoxylin/eosin-stained sections of typical lesions in lungs from K and Fntbfl/ΔPggt1bfl/ΔK mice. (Scale bars, 100 μm.)

Discussion

In this study, we produced mice with a conditional knockout allele for Fntb and showed that inactivation of Fntb eliminated farnesylation of HDJ2 and H-RAS, prevented membrane targeting of H-RAS, and blocked the proliferation of primary, immortalized, and K-RASG12D-expressing fibroblasts in vitro. Moreover, inactivating Fntb in mice with K-RAS-induced lung cancer reduced tumor growth and improved survival. In addition, simultaneous inactivation of Fntb and Pggt1b had a strong antitumor effect.

Our findings differ significantly from those of Mijimolle et al. (16). In the latter study, the Fntb knockout did not affect H-RAS membrane association or stop fibroblast proliferation, nor did it affect the development of K-RAS-induced tumors in mice. When their report was published, it was provocative because it challenged both the widely accepted notion that H-RAS association with membranes depends on protein farnesylation and the view that HDJ2 prenylation depends on FTase. The explanation for the differences between our studies and theirs is unknown; however, we suspect that the differences might relate to the fact that Mijimolle et al.’s allele yielded an unexpected splicing event. Rather than generating a transcript with a nonsense mutation, the mutation yielded a short in-frame deletion in the Fntb transcript (22). In contrast, the recombination event in our Fntb allele deleted the promoter and exon 1 and created a bona fide null allele.

Inactivating Fntb in fibroblasts induced a G2M arrest associated with large flattened cells, up-regulated p21CIP1, and reduced serum-stimulated phosphorylation of AKT. Previously, we showed that inactivation of Pggt1b induces a G1 arrest associated with cell rounding and up-regulation of p21CIP1. The cell rounding and cell-cycle arrest could be overcome, at least temporarily, by expressing farnesylated mutants of RHOA and CDC42, suggesting that a limited number of geranylgeranylated proteins are important for those phenotypes. The creation of a bona fide knockout allele for the Fntb allele opens the door to performing similar experiments to determine whether geranylgeranylated versions of FTase substrates reverse the phenotypes of Fntb-deficient cells.

Fntb deficiency reduced tumor growth and improved survival in mice with K-RAS-induced lung cancer, similar to the effects of inactivating Pggt1b (32). Because K-RAS remains prenylated and associated with membranes in both Fntb- and Pggt1b-deficient cells, these studies support the notion (40) that the therapeutic effects of FTIs and GGTIs are independent of the RAS proteins.

We hypothesized that simultaneous inactivation of Fntb and Pggt1b would limit tumor growth more effectively than inactivation of either gene alone—in part because this approach would be predicted to inhibit K-RAS prenylation and membrane association. The simultaneous inactivation strategy was effective, at least to an extent, because a substantial proportion of K-RAS in lysates from Fntb/Pggt1b-deficient fibroblasts, lung tissue, and lung tumors accumulated in the cytosolic fraction. As we predicted, the simultaneous inactivation of Fntb and Pggt1b had a far greater inhibitory effect on K-RAS-induced tumors than inactivation of either gene alone. However, the main effect of inhibiting FTase and GGTase-I is likely independent of the RAS proteins because both control and myristoylated H-RAS-transfected fibroblasts underwent apoptosis after inactivation of both Fntb and Pggt1b.

The main concern surrounding the combined inhibition of FTase and GGTase-I has been toxicity (33, 34). Whereas the simultaneous inactivation of Fntb and Pggt1b clearly induced cell death in fibroblasts, the inactivation of both genes in type II pneumocytes in Fntbfl/ΔPggt1bfl/ΔL mice did not produce pulmonary disease phenotypes and did not induce apoptosis. Moreover, Fntbfl/ΔPggt1bfl/ΔKL mice appeared healthy for several months despite widespread expression of K-RASG12D in the lung. Some cells in Fntbfl/ΔPggt1bfl/ΔKL lungs clearly lacked both enzymes, as a large proportion of K-RAS in lung lysates was found in the cytosolic fraction. Also, immunochemical studies revealed that some cells were positive for both prelamin A and np-RAP1A. It is intriguing that some cells in the lung were apparently viable despite the absence of both FTase and GGTase-I. However, our genetic approach does not allow us to address the impact of FTase and GGTase-I deficiency in every lung cell (such as lung stem cells), and we cannot rule out the possibility that a more widespread inactivation of these enzymes would be toxic.

In summary, our findings support the idea that farnesylation is essential for H-RAS membrane association, HDJ2 prenylation, and fibroblast proliferation in vitro. In mice harboring a mutationally activated form of K-RAS in the lung, blocking protein farnesylation retarded tumor growth and improved survival. Our results also support the idea that simultaneous inhibition of FTase and GGTase-I could be therapeutically useful. Finally, the experimental approach described here should be useful for dissecting the in vivo importance of protein farnesylation and geranylgeranylation in other cell types in a variety of diseases.

Materials and Methods

A Conditional Knockout Allele for Fntb.

A 2.2-kb fragment spanning promoter sequences, exon 1, and parts of intron 1 was amplified by PCR from the genomic DNA of strain 129/OlaHsd embryonic stem (ES) cells. The fragment was cloned into pNB1, which contains a polylinker flanked by loxP sites. The floxed fragment was excised and cloned into pKSloxPNTmod (41) upstream of a floxed neo cassette. Finally, 5′- and 3′-flanking arms were amplified by PCR and cloned upstream and downstream, respectively, of the floxed exon 1 fragment and neo cassette. The gene-targeting vector was electroporated into 129/OlaHsd ES cells, and targeted clones (identified by Southern blotting with flanking probes) were used to produce germline-transmitting chimeric mice. A detailed description of all other methods appears in SI Materials and Methods.

Supplementary Material

Supporting Information

Acknowledgments

We thank Drs. Hans Nordlinder and Aziz Hussein for assistance with lung histopathology. This study was supported by a Starting Investigator Grant from the European Research Council; by grants from the Swedish Cancer Society, the Swedish Medical Research Council, the Swedish Children's Cancer Fund, and Västra Götalandsregionen (to M.O.B.); by the Medical Society of Gothenburg and the Foundations of Assar Gabrielsson, Serena Ehrenströms, and Konrad and Helfrid Johansson (to M.L.); and by National Institutes of Health Grants AR050200 and HL76839 and an Ellison Medical Foundation Senior Scholar Award (to S.G.Y.). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Footnotes

The authors declare no conflict of interest.

This article is a PNAS Direct Submission.

This article contains supporting information online at www.pnas.org/cgi/content/full/0908396107/DCSupplemental.

References

  • 1.Lane KT, Beese LS. Lipid posttranslational modifications. Structural biology of protein farnesyltransferase and geranylgeranyltransferase type I. J Lipid Res. 2006;47:681–699. doi: 10.1194/jlr.R600002-JLR200. [DOI] [PubMed] [Google Scholar]
  • 2.Seabra MC, Reiss Y, Casey PJ, Brown MS, Goldstein JL. Protein farnesyltransferase and geranylgeranyltransferase share a common α subunit. Cell. 1991;65:429–434. doi: 10.1016/0092-8674(91)90460-g. [DOI] [PubMed] [Google Scholar]
  • 3.Casey PJ, Seabra MC. Protein prenyltransferases. J Biol Chem. 1996;271:5289–5292. doi: 10.1074/jbc.271.10.5289. [DOI] [PubMed] [Google Scholar]
  • 4.Kato K, et al. Isoprenoid addition to Ras protein is the critical modification for its membrane association and transforming activity. Proc Natl Acad Sci USA. 1992;89:6403–6407. doi: 10.1073/pnas.89.14.6403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Bos JL. ras oncogenes in human cancer: A review. Cancer Res. 1989;49:4682–4689. [PubMed] [Google Scholar]
  • 6.Basso AD, Kirschmeier P, Bishop WR. Lipid posttranslational modifications. Farnesyl transferase inhibitors. J Lipid Res. 2006;47:15–31. doi: 10.1194/jlr.R500012-JLR200. [DOI] [PubMed] [Google Scholar]
  • 7.Kohl NE, et al. Inhibition of farnesyltransferase induces regression of mammary and salivary carcinomas in ras transgenic mice. Nat Med. 1995;1:792–797. doi: 10.1038/nm0895-792. [DOI] [PubMed] [Google Scholar]
  • 8.Omer CA, et al. Mouse mammary tumor virus-Ki-rasB transgenic mice develop mammary carcinomas that can be growth-inhibited by a farnesyl:protein transferase inhibitor. Cancer Res. 2000;60:2680–2688. [PubMed] [Google Scholar]
  • 9.Sepp-Lorenzino L, et al. A peptidomimetic inhibitor of farnesyl:protein transferase blocks the anchorage-dependent and -independent growth of human tumor cell lines. Cancer Res. 1995;55:5302–5309. [PubMed] [Google Scholar]
  • 10.Sebti SM, Hamilton AD. Farnesyltransferase and geranylgeranyltransferase I inhibitors and cancer therapy: Lessons from mechanism and bench-to-bedside translational studies. Oncogene. 2000;19:6584–6593. doi: 10.1038/sj.onc.1204146. [DOI] [PubMed] [Google Scholar]
  • 11.Sebti SM, Der CJ. Searching for the elusive targets of farnesyltransferase inhibitors. Nat Rev Cancer. 2003;3:945–951. doi: 10.1038/nrc1234. [DOI] [PubMed] [Google Scholar]
  • 12.End DW, et al. Characterization of the antitumor effects of the selective farnesyl protein transferase inhibitor R115777 in vivo and in vitro. Cancer Res. 2001;61:131–137. [PubMed] [Google Scholar]
  • 13.Liu M, et al. Antitumor activity of SCH 66336, an orally bioavailable tricyclic inhibitor of farnesyl protein transferase, in human tumor xenograft models and wap-ras transgenic mice. Cancer Res. 1998;58:4947–4956. [PubMed] [Google Scholar]
  • 14.Rose WC, et al. Preclinical antitumor activity of BMS-214662, a highly apoptotic and novel farnesyltransferase inhibitor. Cancer Res. 2001;61:7507–7517. [PubMed] [Google Scholar]
  • 15.Hunt JT, et al. Discovery of (R)-7-cyano-2,3,4,5-tetrahydro-1-(1H-imidazol-4-ylmethyl)-3-(phenylmethyl)-4-(2-thienylsulfonyl)-1H-1,4-benzodiazepine (BMS-214662), a farnesyltransferase inhibitor with potent preclinical antitumor activity. J Med Chem. 2000;43:3587–3595. doi: 10.1021/jm000248z. [DOI] [PubMed] [Google Scholar]
  • 16.Mijimolle N, et al. Protein farnesyltransferase in embryogenesis, adult homeostasis, and tumor development. Cancer Cell. 2005;7:313–324. doi: 10.1016/j.ccr.2005.03.004. [DOI] [PubMed] [Google Scholar]
  • 17.Lerner EC, et al. Ras CAAX peptidomimetic FTI-277 selectively blocks oncogenic Ras signaling by inducing cytoplasmic accumulation of inactive Ras-Raf complexes. J Biol Chem. 1995;270:26802–26806. doi: 10.1074/jbc.270.45.26802. [DOI] [PubMed] [Google Scholar]
  • 18.Willumsen BM, Christensen A, Hubbert NL, Papageorge AG, Lowy DR. The p21 ras C-terminus is required for transformation and membrane association. Nature. 1984;310:583–586. doi: 10.1038/310583a0. [DOI] [PubMed] [Google Scholar]
  • 19.Ashar HR, et al. Farnesyl transferase inhibitors block the farnesylation of CENP-E and CENP-F and alter the association of CENP-E with the microtubules. J Biol Chem. 2000;275:30451–30457. doi: 10.1074/jbc.M003469200. [DOI] [PubMed] [Google Scholar]
  • 20.Crespo NC, Ohkanda J, Yen TJ, Hamilton AD, Sebti SM. The farnesyltransferase inhibitor, FTI-2153, blocks bipolar spindle formation and chromosome alignment and causes prometaphase accumulation during mitosis of human lung cancer cells. J Biol Chem. 2001;276:16161–16167. doi: 10.1074/jbc.M006213200. [DOI] [PubMed] [Google Scholar]
  • 21.Nagasu T, Yoshimatsu K, Rowell C, Lewis MD, Garcia AM. Inhibition of human tumor xenograft growth by treatment with the farnesyl transferase inhibitor B956. Cancer Res. 1995;55:5310–5314. [PubMed] [Google Scholar]
  • 22.Yang SH, et al. Caution! Analyze transcripts from conditional knockout alleles. Transgenic Res. 2009;18:483–489. doi: 10.1007/s11248-008-9237-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.James G, Goldstein JL, Brown MS. Resistance of K-RasBV12 proteins to farnesyltransferase inhibitors in Rat1 cells. Proc Natl Acad Sci USA. 1996;93:4454–4458. doi: 10.1073/pnas.93.9.4454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Whyte DB, et al. K- and N-Ras are geranylgeranylated in cells treated with farnesyl protein transferase inhibitors. J Biol Chem. 1997;272:14459–14464. doi: 10.1074/jbc.272.22.14459. [DOI] [PubMed] [Google Scholar]
  • 25.Rowell CA, Kowalczyk JJ, Lewis MD, Garcia AM. Direct demonstration of geranylgeranylation and farnesylation of Ki-Ras in vivo. J Biol Chem. 1997;272:14093–14097. doi: 10.1074/jbc.272.22.14093. [DOI] [PubMed] [Google Scholar]
  • 26.El Oualid F, Cohen LH, van der Marel GA, Overhand M. Inhibitors of protein: Geranylgeranyl transferases. Curr Med Chem. 2006;13:2385–2427. doi: 10.2174/092986706777935078. [DOI] [PubMed] [Google Scholar]
  • 27.Sun J, Qian Y, Hamilton AD, Sebti SM. 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. 1998;16:1467–1473. doi: 10.1038/sj.onc.1201656. [DOI] [PubMed] [Google Scholar]
  • 28.Sun J, et al. Antitumor efficacy of a novel class of non-thiol-containing pepti-domimetic inhibitors of farnesyltransferase and geranylgeranyltransferase I: Combination therapy with the cytotoxic agents cisplatin, Taxol, and gemcitabine. Cancer Res. 1999;59:4919–4926. [PubMed] [Google Scholar]
  • 29.Sun J, et al. Geranylgeranyltransferase I inhibitor GGTI-2154 induces breast carcinoma apoptosis and tumor regression in H-Ras transgenic mice. Cancer Res. 2003;63:8922–8929. [PubMed] [Google Scholar]
  • 30.Peterson YK, Kelly P, Weinbaum CA, Casey PJ. A novel protein geranyl-geranyltransferase-I inhibitor with high potency, selectivity, and cellular activity. J Biol Chem. 2006;281:12445–12450. doi: 10.1074/jbc.M600168200. [DOI] [PubMed] [Google Scholar]
  • 31.Watanabe M, et al. Inhibitors of protein geranylgeranyltransferase I and Rab geranylgeranyltransferase identified from a library of allenoate-derived compounds. J Biol Chem. 2008;283:9571–9579. doi: 10.1074/jbc.M706229200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Sjogren A-KM, et al. GGTase-I deficiency reduces tumor formation and improves survival in mice with K-RAS-induced lung cancer. J Clin Invest. 2007;117:1294–1304. doi: 10.1172/JCI30868. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Lobell RB, et al. Evaluation of farnesyl:protein transferase and geranylgeranyl:protein transferase inhibitor combinations in preclinical models. Cancer Res. 2001;61:8758–8768. [PubMed] [Google Scholar]
  • 34.deSolms SJ, et al. Dual protein farnesyltransferase-geranylgeranyltransferase-I inhibitors as potential cancer chemotherapeutic agents. J Med Chem. 2003;46:2973–2984. doi: 10.1021/jm020587n. [DOI] [PubMed] [Google Scholar]
  • 35.Di Paolo A, et al. Inhibition of protein farnesylation enhances the chemotherapeutic efficacy of the novel geranylgeranyltransferase inhibitor BAL9611 in human colon cancer cells. Br J Cancer. 2001;84:1535–1543. doi: 10.1054/bjoc.2001.1820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Lakso M, et al. Efficient in vivo manipulation of mouse genomic sequences at the zygote stage. Proc Natl Acad Sci USA. 1996;93:5860–5865. doi: 10.1073/pnas.93.12.5860. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Schwenk F, Baron U, Rajewsky K. A cre-transgenic mouse strain for the ubiquitous deletion of loxP-flanked gene segments including deletion in germ cells. Nucleic Acids Res. 1995;23:5080–5081. doi: 10.1093/nar/23.24.5080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Jackson EL, et al. Analysis of lung tumor initiation and progression using conditional expression of oncogenic K-ras. Genes Dev. 2001;15:3243–3248. doi: 10.1101/gad.943001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Adjei AA, et al. A phase I trial of the farnesyl transferase inhibitor SCH66336: Evidence for biological and clinical activity. Cancer Res. 2000;60:1871–1877. [PubMed] [Google Scholar]
  • 40.Lebowitz PF, Prendergast GC. Non-Ras targets of farnesyltransferase inhibitors: Focus on Rho. Oncogene. 1998;17(11 Reviews):1439–1445. doi: 10.1038/sj.onc.1202175. [DOI] [PubMed] [Google Scholar]
  • 41.Hanks M, Wurst W, Anson-Cartwright L, Auerbach AB, Joyner AL. Rescue of the En-1 mutant phenotype by replacement of En-1 with En-2. Science. 1995;269:679–682. doi: 10.1126/science.7624797. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supporting Information

Articles from Proceedings of the National Academy of Sciences of the United States of America are provided here courtesy of National Academy of Sciences

RESOURCES