Inhibition of the 3-Hydroxy-3-methylglutaryl-coenzyme A Reductase Pathway Induces p53-independent Transcriptional Regulation of p21WAF1/CIP1 in Human Prostate Carcinoma Cells*

Su Jae Lee, Mahn Joon Ha, Jeen Lee, PhuongMai Nguyen, Yung Hyun Choi, Farzaneh Pirnia, Won-Ki Kang, Xiao-Fan WangDagger , Seong-Jin Kim§, and Jane B. Trepel

From the Medicine Branch, Division of Clinical Sciences, NCI, National Institutes of Health, Bethesda, Maryland 20892, the Dagger  Department of Pharmacology, Duke University Medical Center, Durham, North Carolina 27710, and the § Laboratory of Chemoprevention, Division of Basic Sciences, NCI, National Institutes of Health, Bethesda, Maryland 20892

    ABSTRACT
Top
Abstract
Introduction
Materials & Methods
Results
Discussion
References

Progression through the cell cycle is controlled by the induction of cyclins and the activation of cognate cyclin-dependent kinases. The 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitor lovastatin induces growth arrest and cell death in certain cancer cell types. We have pursued the mechanism of growth arrest in PC-3-M cells, a p53-null human prostate carcinoma cell line. Lovastatin treatment increased protein and mRNA levels of the cyclin-dependent kinase inhibitor p21WAF1/CIP1, increased binding of p21 with Cdk2, markedly inhibited cyclin E- and Cdk2-associated phosphorylation of histone H1 or GST-retinoblastoma protein, enhanced binding of the retinoblastoma protein to the transcription factor E2F-1 in vivo, and induced the activation of a p21 promoter reporter construct. By using p21 promoter deletion constructs, the lovastatin-responsive element was mapped to a region between -93 and -64 relative to the transcription start site. Promoter mutation analysis indicated that the lovastatin-responsive site coincided with the previously identified transforming growth factor-beta -responsive element. These data indicate that in human prostate carcinoma cells an inhibitor of the HMG-CoA reductase pathway can circumvent the loss of wild-type p53 function and induce critical downstream regulatory events leading to transcriptional activation of p21.

    INTRODUCTION
Top
Abstract
Introduction
Materials & Methods
Results
Discussion
References

Cell cycle progression is controlled by the orderly activation of cyclin-dependent kinases (Cdks)1 (1-4). Cdk activation is regulated at multiple levels including the induction and degradation of cyclin protein, Cdk phosphorylation by CAK, the cyclin-activating kinase, and the synthesis of Cdk-inhibitory proteins (5). Cdk inhibitors can negatively regulate cell cycle progression in response to a variety of antiproliferative signals including DNA damage, differentiation, contact inhibition, and senescence (6-8). p21WAF1/CIP1, the first Cdk inhibitor to be identified, is required for p53-induced growth arrest (9). The p21 protein negatively regulates cell cycle progression by inhibiting Cdk catalytic activity and by inhibiting proliferating cell nuclear antigen, which is required for DNA synthesis (10). By inhibiting the activity of G1 cyclin-Cdk complexes, p21 blocks phosphorylation of the retinoblastoma protein (pRB). This leaves pRB in its active, hypophosphorylated form, in which it is able to induce G1 arrest through negative regulation of the heterodimeric transcription factor E2F/DP (11-13).

We have found that human prostate carcinoma cells are very sensitive to the induction of growth arrest and cell death by the HMG-CoA reductase inhibitor lovastatin (14).2 Lovastatin blocks the rate-limiting step in the cholesterol-synthesis pathway, the formation of mevalonic acid from HMG-CoA (15-17). The study presented here investigates the mechanism by which lovastatin induces cell cycle arrest in the androgen-independent prostate carcinoma line PC-3-M, which is lacking both p53 alleles (18). The data show that lovastatin treatment caused a marked shift of pRB to its underphosphorylated form and enhanced the in vivo binding of pRB and E2F-1. In addition, lovastatin inhibited Cdk activity, and this inhibition was associated with elevated protein and RNA expression of the Cdk inhibitor p21, similar to the induction of p21 by lovastatin in human breast cells (19). We investigated the mechanism of lovastatin-induced p21 expression using p21 promoter-reporter assays. Lovastatin treatment induced transcriptional activation of the p21 promoter as indicated by increased activity of a p21 promoter-luciferase reporter construct, and this transcriptional response did not require the p53 response element. All effects of lovastatin were completely reversed by addition of mevalonic acid. These data demonstrate a link between the mevalonate pathway and activation of the promoter of the critical cell cycle-regulatory protein p21. To begin to understand the mechanism of lovastatin-induced transcription, promoter deletion and mutation studies were performed. This analysis demonstrated that lovastatin responsiveness is mediated by a promoter-proximal region that coincides with the recently identified transforming growth factor-beta (TGF-beta )-responsive element.

    MATERIALS AND METHODS
Top
Abstract
Introduction
Materials & Methods
Results
Discussion
References

Cells-- The human prostate carcinoma cell line PC-3-M was a gift of Dr. James Kozlowski, Department of Urology, Northwestern University Medical School (20). Cells were cultured as described previously (21).

Antibodies-- Polyclonal antibodies to cyclin E, Cdk2, Cdk4, and monoclonal anti-E2F-1 (clone 20) were obtained from Santa Cruz Biotechnology. Monoclonal anti-pRB antibody (clone G3-245) and monoclonal anti-p21 antibody were purchased from PharMingen.

Cell Cycle Analysis-- Cells were fixed in 50% ethanol and incubated with RNase A and the DNA intercalating dye propidium iodide, and cell cycle phase analysis was performed by flow cytometry using a Becton Dickinson FACStar flow cytometer and Becton Dickinson Cell Fit software.

Immunoprecipitation and Western Blot Analysis-- Immunoprecipitations were performed as described (22). Cell lysates were incubated with anti-Cdk2 polyclonal antibody, anti-E2F-1, or normal rabbit serum. The immunocomplex was collected on protein A-Sepharose (Sigma) and analyzed by SDS-polyacrylamide gel electrophoresis using enhanced chemiluminescence detection (Amersham Pharmacia Biotech). Western blot analysis was performed as described (23).

Northern Blot Analysis-- Total RNA was isolated from cells lysed in guanidinium thiocyanate solution, and Northern blot analysis was carried out as described previously (24) using 32P-labeled (ICN) cDNA probes and 30 µg of total RNA per lane. The human p21 DNA probe was a gift of Dr. David Givol.

Immune Complex Kinase Assay-- Immune complex kinase assays were performed as described (22). Briefly, cell lysates were incubated with primary antibody, and immune complexes were collected on protein A-Sepharose beads and resuspended in kinase assay mixture containing [gamma -32P]ATP (ICN) and GST-RB or histone H1 (Life Technologies, Inc.) as substrate. The GST-RB fusion protein containing amino acids 379-928 of human pRB (25) was a gift of Dr. Frederic Kaye, NCI, National Institutes of Health. The proteins were separated on 10% SDS-polyacrylamide gels, and bands were detected by autoradiography.

p21 Promoter-Luciferase Constructs-- Drs. Bert Vogelstein and Todd Waldman, The Johns Hopkins Oncology Center, provided the full-length 2.4-kb human p21 promoter-chloramphenicol acetyltransferase constructs as well as p21 promoter deletion constructs, in which 300 base pairs were progressively removed from the 3' terminus. Three constructs were tested as follows: a 2.4-kb p21 promoter fragment containing the p21 transcription initiation start site at its 3' end and a p53 response element at the 5' end, a 2.1-kb fragment consisting of the 2.4-kb fragment from which 300 base pairs, including the p53 response element, had been deleted from the 5' end, and a 0.3-kb fragment consisting of the 300 base pairs most proximal to the transcription start site (9). The original chloramphenicol acetyltransferase constructs were digested with HindIII and XhoI and subcloned into the HindIII-XhoI sites of the luciferase reporter vector pGL2-basic (Promega Corp.), to create p21 promoter-luciferase constructs. The deletion constructs p21P 93-S (0 to -93), p21P Sma Delta 1 (0 to -63) p21P Sma Delta 2 (full-length p21 promoter from which the region -64 to -110 was deleted) and p21P (0 to -100), and the p21P 93-S mutants were created as described previously (26).

Luciferase Assay-- PC-3-M cells were co-transfected with the p21 promoter-luciferase reporter constructs and with the beta -galactosidase reporter vector pCMVbeta (Promega) using LipofectAMINE (Life Technologies, Inc.), as recommended by the manufacturer. Following transfection the cells were incubated for 6 h; the medium was exchanged, and the cells were incubated for an additional 36 h in the presence or absence of 10 µM lovastatin. The cells were then lysed, and luciferase activity in the lysates was assayed using a Dynatech ML1000 luminometer (Dynatech Laboratories). Luciferase activity was normalized to beta -galactosidase activity which was assayed using the beta -galactosidase Enzyme Assay System (Promega).

    RESULTS
Top
Abstract
Introduction
Materials & Methods
Results
Discussion
References

Lovastatin Induces G1 Arrest and Cell Death-- 24 h after 10 µM lovastatin addition there was an increase in the percent of PC-3-M cells in G1. By 48 h following lovastatin addition greater than 90% of the PC-3-M cells were arrested in G1 (Table I). Cell viability was consistently greater than 75% in cells treated for 48 h, after which there was a marked loss of viability, culminating in cell death (data not shown).

                              
View this table:
[in this window]
[in a new window]
 
Table I
Effect of lovastatin on cell cycle distribution in PC-3-M cells
Exponentially growing PC-3-M cells were treated at 0 time with lovastatin (10 µM), and flow cytometric cell cycle analysis was performed comparing untreated controls with cells treated with lovastatin for 24 and 48 h.

Loss of Phosphorylated pRB in Cells Exposed to Lovastatin-- In cells treated with lovastatin pRB appeared as a single band on Western blot analysis, migrating in the position characteristic of unphosphorylated or hypophosphorylated pRB (27) (Fig. 1A). The position of pRB after lovastatin coincided with the position of pRB detected with an antibody specific for the un- or hypophosphorylated form (Pharmingen clone G99-549, data not shown).


View larger version (36K):
[in this window]
[in a new window]
 
Fig. 1.   Hypophosphorylation of pRB and enhanced association of pRB and E2F-1 in PC-3-M cells after exposure to lovastatin. A, pRB was detected by Western blot analysis in cell lysates using 30 µg of protein from untreated cells (0 h) and in cells 12, 24, 36, 48, and 72 h after exposure to 10 µM lovastatin. B, total cell lysates (2 mg of protein) from control cells and cells incubated with 10 µM lovastatin for 36 h were immunoprecipitated with anti-E2F-1 antibody or normal rabbit serum (NRS). Immune complexes were separated by SDS-polyacrylamide gel electrophoresis, transferred to nitrocellulose, and probed with anti-pRB antibody. Proteins were detected by enhanced chemiluminescence.

Lovastatin Enhances Association of pRB and E2F-1-- Association of pRB with the transcription factor E2F-1 was almost undetectable by co-immunoprecipitation analysis of the untreated log phase cells (Fig. 1B). However, 36 h after addition of lovastatin there was a strong increase in the association of pRB and E2F-1.

Levels of Cdk2, Cdk4, and Cyclin E Protein Are Unchanged following Lovastatin Treatment-- The G1/S transition is regulated by D-type cyclins which bind to and activate Cdk4 and Cdk6 and cyclin E, which activates Cdk2. Because lovastatin arrests PC-3-M cells in G1, we tested whether lovastatin decreased protein levels of Cdk4, Cdk2, or cyclin E, and as demonstrated in Fig. 2A, there was little detectectable change in the level of these proteins, even after 48 h of exposure to lovastatin. It should be noted, however, that the actual effects of lovastatin on these proteins may be larger than is apparent in the Western blot because of the nonlinearity of the chemiluminescence detection system.


View larger version (36K):
[in this window]
[in a new window]
 
Fig. 2.   Effect of lovastatin on levels of Cdk2, Cdk4, and cyclin E protein and Cdk2-dependent and cyclin E-dependent kinase activity. A, PC-3-M cells were treated with 10 µM lovastatin for the times indicated. Total cell lysates were prepared, and Western blot analysis was performed using anti-Cdk2, anti-Cdk4, or anti-cyclin E antibody and enhanced chemiluminescence detection. B, effect of lovastatin on Cdk2 kinase and cyclin E-associated kinase activity. PC-3-M cells were treated with lovastatin, and cell lysates were immunoprecipitated (IP) with anti-Cdk2 or cyclin E antibody, and kinase activity was assayed using GST-RB or histone H1 as substrate.

Lovastatin Inhibits Cdk2 and Cyclin E-associated Kinase Activity-- Cdk2- and cyclin E-associated kinase activity were reduced to undetectable levels after 36 h treatment with lovastatin (Fig. 2B). These results indicated that the inhibition of Cdk2- and cyclin E-associated kinase activity might involve binding of a Cdk inhibitor protein to the Cdk2-cyclin E complex.

Lovastatin Induces p21 Protein and Association of p21 with Cdk2-- Western blot analysis showed that lovastatin induced a marked increase in the level of the Cdk inhibitor p21 (Fig. 3A). We next asked whether the p21 protein induced by lovastatin treatment was associated with Cdk2 in vivo. As shown in Fig. 3B, p21 was detected in anti-Cdk2 immunoprecipitates from lovastatin-treated cells and not in immunoprecipitates from untreated cells. Thus lovastatin induced the association of p21 with Cdk2, suggesting that lovastatin-induced p21 blocks Cdk2 kinase activity by direct binding to the cyclin-Cdk2 complex.


View larger version (24K):
[in this window]
[in a new window]
 
Fig. 3.   Lovastatin induces the cyclin-dependent kinase inhibitor p21 and association of p21 with Cdk2. A, cells were incubated with 10 µM lovastatin for the time indicated, and total cell lysates were subjected to Western blot analysis using anti-p21 antibody and enhanced chemiluminescence detection. B, cells were treated with 10 µM lovastatin for 36 h, and cell lysates (2 mg protein) were immunoprecipitated (IP) with anti-Cdk2 antibody, separated on SDS-polyacrylamide gels, transferred to nitrocellulose, and incubated with anti-p21 antibody followed by enhanced chemiluminescence detection.

Lovastatin Induces p21 mRNA-- To examine the mechanism by which lovastatin induces p21 we performed Northern blot analysis of the effect of lovastatin on the level of p21 mRNA. The 2.4-kb p21 transcript was increased in PC-3-M cells as early as 12 h after lovastatin treatment (Fig. 4). The kinetics of induction were consistent with the time course of inhibition of Cdk activity. The increase in p21 mRNA induced by lovastatin was blocked by actinomycin D (data not shown), suggesting that lovastatin may be regulating p21 mRNA levels through an effect on p21 transcription.


View larger version (33K):
[in this window]
[in a new window]
 
Fig. 4.   Lovastatin induces expression of p21 mRNA. Cells were incubated with 10 µM lovastatin for the times indicated. Total RNA was prepared, and p21 mRNA was detected by Northern blot analysis. The same blot was probed with a cDNA to glyceraldehyde-3'-phosphate dehydrogenase (GAPDH) to demonstrate comparable levels of mRNA per lane. The fold increase in p21 mRNA level was determined by integrating the signal using NIH Image version 1.6 software.

p53 acts at the transcriptional level to increase p21 expression in response to DNA damage (9). In PC-3-M cells the p53 gene has been reported to be truncated and p53 protein absent (18). As a control we performed Western and Northern blot analysis for p53 and found no evidence of p53 expression in control or lovastatin-treated cells (data not shown). These data demonstrate that lovastatin is regulating the level of p21 mRNA through a p53-independent mechanism.

Transcriptional Activation of the p21 Promoter by Lovastatin-- To test for transcriptional regulation of p21 by lovastatin p21 promoter-luciferase reporter constructs were transfected into PC-3-M cells, and luciferase activity was measured in the presence and absence of lovastatin. The full-length 2.4-kb promoter construct containing a p53 response element was activated 3-4-fold by lovastatin (Fig. 5A). Activation was not seen with a control vector consisting of the pGL2-basic luciferase reporter vector alone, and activation of the p21 promoter by lovastatin was completely blocked by incubation of the cells with 5 mM mevalonic acid, demonstrating that the ability of lovastatin to activate the p21 promoter is mediated by a component of the HMG-CoA reductase pathway (data not shown).


View larger version (19K):
[in this window]
[in a new window]
 
Fig. 5.   Deletion analysis of the activation of the p21 promoter by lovastatin. PC-3-M cells were transiently transfected with p21 promoter luciferase constructs (10 µg) and incubated for 6 h; the medium was changed; the cells were incubated for an additional 36 h in the presence or absence of 10 µM lovastatin, and luciferase activity was measured. A, transfection of the full-length 2.4-kilobase promoter construct and three progressive deletion constructs. The results are expressed as the mean ± S.D. of data from three separate samples and are representative of five separate experiments. B, mapping of the lovastatin-responsive region using two transcription start site-proximal constructs and a full-length promoter construct with an interstitial deletion. RLU, relative luciferase units.

A 2.1-kb construct lacking the p53 response element was activated by lovastatin to the same extent as the full-length promoter (Fig. 5A). Constructs consisting of 300 base pairs and 100 base pairs proximal to the transcription start site were fully activated by lovastatin when compared with the 2.4-kb promoter. Thus, the lovastatin-responsive site is localized to the promoter proximal region between 0 and -100 base pairs. Three deletion constructs were employed to further map this region of the p21 promoter. A construct consisting of 0 to -93 base pairs was fully responsive to lovastatin (Fig. 5B). In contrast, a construct consisting of the region from 0 to -63 was non-responsive to lovastatin. A deletion construct consisting of the full-length promoter minus the region -64 to -110 was also non-responsive to lovastatin. These data localize the region responsive to lovastatin to the sequences between -64 and -93 base pairs.

The region between -64 and -93 contains two Sp1 binding sites. The second of these sites, between -78 and -84, has been identified as both the TGF-beta -responsive element (Tbeta RE) and the butyrate-responsive element (28). To test whether the lovastatin-responsive element coincides with this Sp1 binding site, we tested two constructs of the 0 to -93 region in which the Sp1 site was mutated. Mutant 1, containing 10 consecutive mutated base pairs, did not respond to lovastatin (Fig. 6). Mutation of bases -78 and -79 also resulted in loss of lovastatin responsiveness. These data localize the lovastatin-responsive region and suggest that Sp1 may play a critical role in the activation of p21 transcription in response to lovastatin.


View larger version (14K):
[in this window]
[in a new window]
 
Fig. 6.   Mutation analysis of the activation of the p21 promoter by lovastatin. PC-3-M cells were transfected with the wild-type p21 promoter-reporter deletion construct p21P 93-S or with mutant p21P 93-S constructs and lovastatin-induced luciferase activity was measured. RLU, relative luciferase units.

    DISCUSSION
Top
Abstract
Introduction
Materials & Methods
Results
Discussion
References

The hormone refractory prostate cancer cell line studied here represents the most advanced and least treatable form of adult carcinoma (29, 30). Progressive malignant tranformation in epithelial tumors has been associated with a variety of genetic changes (31). Recent data suggest that mutations in cell cycle genes are the most common genetic change in tumor cells (32). Although inactivation or deletion of the RB gene itself is limited to a subset of human tumors (33), it has been suggested that continued molecular characterization may reveal that virtually all tumors harbor a mutation in one of the genes responsible for regulating G1 progression (32) and that the final common target of the genetic changes in G1 regulatory proteins is the functional inactivation of pRB (13).

Passage from G1 to S phase is regulated by D-type cyclins and their cognate kinases Cdk4 and Cdk6, which act by phosphorylating and inactivating pRB prior to the restriction point at which cells commit to DNA synthesis (34), and by cyclin E-Cdk2, which acts later in G1 (35). Genetic changes associated with G1 cyclins and their Cdks in human cancers include overexpression of cyclin D1, overexpression or mutation of Cdk4, loss of expression of the cyclin D-directed Cdk inhibitor p16, mutation or deletion of p53 and RB, and overexpression of cyclin E (36-39). The data presented here demonstrate that in the human androgen-independent prostate carcinoma cell line PC-3-M, the mevalonate pathway inhibitor lovastatin induces G1 arrest, blocks pRB phosphorylation, and increases in vivo binding of pRB to E2F-1, consistent with restoration of the growth-regulatory activity of pRB. Furthermore, lovastatin blocks cyclin E-Cdk2 kinase activity, in association with enhanced expression of the Cdk inhibitor p21.

Lovastatin has been shown previously to increase the protein level of the Cdk inhibitor p27KIP1 in HeLa cells, HL60 cells, and normal fibroblasts (40, 41). There was no effect on p27KIP1 in the PC-3-M cells (data not shown), and thus G1 arrest appears to be independent of p27KIP1 in the prostate cancer cells studied.

Inhibition of the mevalonate pathway has many ramifications including loss of the protein isoprenylation modifications farnesylation and geranylgeranylation, loss of sterol synthesis, and potential perturbation of N-linked glycosylation through inhibition of dolichol synthesis (42). To begin to map the target of lovastatin action within the mevalonate pathway, we incubated PC-3-M cells with the squalene synthase inhibitor zaragozic acid (43). We found that zaragozic acid treatment had no effect on p21 levels or cell cycle distribution (data not shown). Because lovastatin effects on the cell cycle were completely reversible by mevalonate and not mimicked by zaragozic acid, the critical point in the mevalonate pathway appears to be between HMG-CoA reductase and squalene synthase. One possible mechanism of lovastatin action is through Ras inactivation due to inhibition of Ras isoprenylation. Evidence suggesting that Ras may not be the mediator of the lovastatin response was presented by De Clue et al. (44) who showed that lovastatin arrests the growth of Ras-transformed cells even when the transforming Ras protein utilizes a myristate-dependent rather than farnesyl-dependent membrane-targeting moiety, and thus is not dependent on the mevalonate pathway. These data do not, however, eliminate Ras inactivation as the mechanism of lovastatin-induced G1 arrest, and further experimental approaches, including studies with specific farnesyltransferase inhibitors, will be required.

Although lovastatin itself may not be the optimal HMG-CoA reductase inhibitor for cancer treatment, it has been shown to induce growth arrest and cell death in tumor cells in vitro and in athymic mice in vivo (44-47). A phase I study of lovastatin was completed recently at the National Cancer Institute, in which it was demonstrated that doses as high as 25 times the standard anti-hypercholesterolemic dose were well tolerated (48). More precise identification of the target of lovastatin action would aid in developing new anticancer drugs that act through perturbation of the HMG-CoA reductase pathway.

In addition to p53, recent studies have demonstrated activation of the p21 promoter by TGF-beta (49, 50), vitamin D3 (51), phorbol ester, okadaic acid (52-54), STAT1 (55), and butyrate (28). Activation by vitamin D3 and STAT1 occurs more than 500 base pairs upstream of the lovastatin-responsive region (51, 55). In the U937 myeloid leukemia cell line, it has been demonstrated that activation of p21 transcription by phorbol esters and okadaic acid is mediated by the promoter region between -154 and +16 and that Sp1 plays a critical role in this response. In K562, SW480, and HepG2 cells the activation of p21 transcription by phorbol ester was mapped to the region between -95 and -121, and AP2 was shown to act as a positive regulator of the phorbol ester response. The Tbeta RE is located between -71 and -86 relative to the p21 transcription start site (49, 50). We found that the lovastatin response region maps between -64 and -93. Furthermore, mutation of the Tbeta RE abrogated lovastatin-induced p21 promoter activation. Thus the lovastatin response site corresponds with the Tbeta RE. One possible mechanism for the induction of p21 by lovastatin in PC-3-M cells would be induction of TGF-beta by lovastatin and activation of p21 transcription by TGF-beta . It is likely, however, that lovastatin effects on p21 are unrelated to TGF-beta because PC-3-M cells have an attenuated response to TGF-beta , and direct addition of TGF-beta 1 to PC-3-M cells did not activate the p21 promoter reporter construct (data not shown). Recently it was reported that butyrate activates the p21 promoter and that this response maps to the Tbeta RE (28). Sp1 is required for p21 promoter activation in response to both TGF-beta and butyrate (28, 50). We have shown that mutation of an Sp1 binding site also blocks lovastatin activation of the p21 promoter. Several studies have shown that Sp1 is a critical factor in regulating transcriptional responses to pRB. Sp1 binds to the pRB-responsive element, and Sp1 binding is required and sufficient for promoter activation in response to pRB (56-58). Although the overall level of pRB protein decreases in PC-3-M cells in response to lovastatin, that decrease is predominantly or entirely in the hyperphosphorylated form (Fig. 1A and data not shown), and there is a substantial increase in active pRB as assessed by the markedly enhanced E2F-pRB binding in the in vivo co-immunoprecipitation experiment (Fig. 1B). These data suggest that both pRB and Sp1 may play a role in lovastatin-induced activation of the p21 promoter.

The data presented here demonstrate that lovastatin induces p21 transcription in the absence of wild-type p53. The most frequently mutated tumor suppressor in human cancer is p53. Therefore, p53-independent induction of p21 presents an attractive target for anticancer drug development (59). The data presented here suggest that perturbation of the mevalonate pathway may offer a new approach to transcriptional control of this critical negative growth-regulatory gene.

    ACKNOWLEDGEMENTS

We thank Drs. Bert Vogelstein and Todd Waldman, The Johns Hopkins Oncology Center, for kindly providing the p21 promoter constructs; Dr. David Givol for kindly providing the human p21WAF1/CIP1 DNA probe; and Dr. Frederic Kaye, NCI, National Institutes of Health, for kindly providing the cDNA encoding the GST-RB fusion protein containing amino acids 379-928 of human pRB. We thank Alfred W. Alberts, Merck, for generously providing lovastatin. We are grateful to Drs. Konrad Huppi and Frederic Kaye for critical review of the manuscript.

    FOOTNOTES

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

To whom correspondence should be addressed: Medicine Branch, NCI, Bldg. 10, Rm. 12N226, National Institutes of Health, Bethesda, MD 20892. Tel.: 301-496-1547; Fax: 301-402-0172; E-mail: trepel{at}helix.nih.gov.

1 The abbreviations used are: Cdk, cyclin-dependent kinase; pRB, retinoblastoma protein; GST, glutathione S-transferase; TGF-beta , transforming growth factor-beta ; Tbeta RE, transforming growth factor-beta -responsive element; kb, kilobase pair; HMG-CoA, 3-hydroxy-3-methylglutaryl-coenzyme A.

2 W.-K. Kang, M. J. Ha, S. J. Lee, P. Nguyen, F. Pirnia, M. M. Borner, E. Solary, Y. Pommier, C. E. Myers, and J. B. Trepel, manuscript in preparation.

    REFERENCES
Top
Abstract
Introduction
Materials & Methods
Results
Discussion
References

  1. Sherr, C. J. (1994) Cell 79, 551-555[Medline] [Order article via Infotrieve]
  2. Draetta, G. F. (1994) Curr. Opin. Cell Biol. 6, 842-846[Medline] [Order article via Infotrieve]
  3. Hunter, T., and Pines, J. (1994) Cell 79, 573-582[Medline] [Order article via Infotrieve]
  4. King, R. W., Jackson, P. K., and Kirschner, M. W. (1994) Cell 79, 563-571[Medline] [Order article via Infotrieve]
  5. Scherr, C. J., and Roberts, J. M. (1995) Genes Dev. 9, 1149-1163[CrossRef][Medline] [Order article via Infotrieve]
  6. Noda, A., Ning, Y., Venable, S. F., Pereira-Smith, O. M., and Smith, J. R. (1994) Exp. Cell Res. 211, 90-98[CrossRef][Medline] [Order article via Infotrieve]
  7. Polyak, K., Kato, J.-Y., Solomon, J. M., Sherr, C. J., Massague, J., and Roberts, J. M. (1994) Genes Dev. 8, 9-22[Abstract]
  8. Halevy, O., Novitch, B. G., Spicer, D. B., Skapek, S. X., Rhee, J., Hannon, G. J., Beach, D., and Lassar, A. B. (1995) Science 267, 1018-1021[Medline] [Order article via Infotrieve]
  9. Waldman, T., Kinzler, K. W., and Vogelstein, B. (1995) Cancer Res. 55, 5187-5190[Abstract]
  10. Waga, S., Hannon, G. J., Beach, D., and Stillman, B. (1994) Nature 369, 574-578[CrossRef][Medline] [Order article via Infotrieve]
  11. Chellappan, S. P., Hiebert, S., Mudryj, M., Horowitz, J. M., and Nevins, J. R. (1991) Cell 65, 1053-1061[Medline] [Order article via Infotrieve]
  12. LaThangue, N. B. (1994) Trends Biochem. Sci. 19, 108-114[CrossRef][Medline] [Order article via Infotrieve]
  13. Weinberg, R. A. (1995) Cell 81, 323-330[Medline] [Order article via Infotrieve]
  14. Borner, M. M., Myers, C. E., Sartor, O., Sei, Y., Toko, T., Trepel, J. B., and Schneider, E. (1995) Cancer Res. 55, 2122-2128[Abstract]
  15. Goldstein, J. L., Helgeson, J. A. S., and Brown, M. S. (1979) J. Biol. Chem. 254, 5403-5409[Medline] [Order article via Infotrieve]
  16. Alberts, A. W., Chen, J., Kuron, G., Hunt, V., Huff, J., Hoffman, C., Rothrock, J., Lopez, M., Joshua, H., Harris, E., Patchett, A., Monaghaan, R., Currie, S., Stapley, E., Albers-Schonberg, G., Hensens, O., Hirshfield, J., Hoogsteen, K., Liesch, J., and Springer, J. (1980) Proc. Natl. Acad. Sci. U. S. A. 77, 3957-3961[Abstract]
  17. Keyomarsi, K., Sandoval, L., Band, V., and Pardee, A. B. (1991) Cancer Res. 51, 3602-3609[Abstract]
  18. Issacs, W. B., Carter, B. S., and Ewing, C. M. (1991) Cancer Res. 51, 4716-4720[Abstract]
  19. Gray-Bablin, J., Rao, S., and Keyomarsi, K. (1997) Cancer Res. 57, 604-609[Abstract]
  20. Kozlowski, J. M., Fidler, I. J., Campbell, D., Xu, Z.-L., Kaighn, M. E., and Hart, I. R. (1984) Cancer Res. 44, 3522-3529[Abstract]
  21. Fang, W.-G., Pirnia, F., Bang, Y.-J., Myers, C. E., and Trepel, J. B. (1992) J. Clin. Invest. 89, 191-196[Medline] [Order article via Infotrieve]
  22. Schnier, J. B., Nishi, K., Goodrich, D. W., and Bradbury, E. M. (1996) Proc. Natl. Acad. Sci. U. S. A. 93, 5941-5946[Abstract/Free Full Text]
  23. Bang, Y.-J., Pirnia, F., Fang, W.-G., Kang, W. K., Sartor, O., Whitesell, L., Ha, M. J., Tsokos, M., Sheahan, M. D., Nguyen, P., Niklinski, W. T., Myers, C. E., and Trepel, J. B. (1994) Proc. Natl. Acad. Sci. U. S. A. 91, 5330-5334[Abstract]
  24. Bang, Y.-J., Kim, S.-J., Danielpour, D., O'Reilly, M. A., Kim, K. Y., Myers, C. E., and Trepel, J. B. (1992) Proc. Natl. Acad. Sci. U. S. A. 89, 3556-3560[Abstract]
  25. Kaelin, W. G., Jr., Pallas, D. C., Decaprio, J. A., Kaye, F. J., and Livingston, D. M. (1991) Cell 64, 521-532[Medline] [Order article via Infotrieve]
  26. Datto, M. B., Yu, Y., and Wang, X.-F. (1995) J. Biol. Chem. 270, 28623-28628[Abstract/Free Full Text]
  27. DeCaprio, J. A., Ludlow, J. W., Lynch, D., Furukawa, Y., Griffin, J., Piwnica-Worms, H., Huang, C.-M., and Livingston, D. M. (1989) Cell 58, 1085-1095[Medline] [Order article via Infotrieve]
  28. Nakano, K., Mizuno, T., Sowa, Y., Orita, T., Yoshino, T., Okuyama, Y., Fujita, T., Ohtani-Fujita, N., Matsukawa, Y., Tokino, T., Yamagishi, H., Oka, T., Nomura, H., and Sakai, T. (1997) J. Biol. Chem. 272, 22199-22206[Abstract/Free Full Text]
  29. Coffey, D. S. (1993) Cancer 71, Suppl. 3, 880-886
  30. Zhang, R. D., Fidler, I. J., and Price, J. E. (1991) Invasion Metastasis 11, 204-215[Medline] [Order article via Infotrieve]
  31. Fearon, E. R., and Vogelstein, B. (1990) Cell 61, 759-767[Medline] [Order article via Infotrieve]
  32. Clurman, B. E., and Roberts, J. M. (1995) J. Natl. Cancer Inst. 87, 1499-1501[Medline] [Order article via Infotrieve]
  33. Horowitz, J. M., Park, S. H., Bogenmann, E., Cheng, J. C., Yandell, D. W., Kaye, F. J., Minna, J. D., Dryja, T. P., and Weinberg, R. A. (1990) Proc. Natl. Acad. Sci. U. S. A. 87, 2775-2779[Abstract]
  34. Pardee, A. B. (1989) Science 246, 603-608[Medline] [Order article via Infotrieve]
  35. Resnitzky, D., and Reed, S. I. (1995) Mol. Cell. Biol. 15, 3463-3469[Abstract]
  36. He, J., Allen, J. R., Collins, V. P., Allalunis-Turner, M. J., Godbout, R., Day, R. S., III, and James, C. D. (1994) Cancer Res. 54, 5804-5807[Abstract]
  37. Keyomarsi, K., and Pardee, A. B. (1993) Proc. Natl. Acad. Sci. U. S. A. 90, 1112-1116[Abstract]
  38. Nobori, T., Miura, K., Wu, D. J., Lois, A., Takbayashi, K., and Carson, D. A. (1994) Nature 368, 753-756[CrossRef][Medline] [Order article via Infotrieve]
  39. Wolfel, T., Hauer, M., Schneider, J., Serrano, M., Wolfel, C., Klehmann-Hieb, E., De Plaen, E., Hankeln, T., Meyer zum Buschenfelde, K. H., and Beach, D. (1995) Science 269, 1281-1284[Medline] [Order article via Infotrieve]
  40. Hengst, L., Dulic, V., Slingerland, J. M., Lees, M., and Reed, S. I. (1994) Proc. Natl. Acad. Sci. U. S. A. 91, 5291-5295[Abstract]
  41. Hengst, L., and Reed, S. I. (1996) Science 271, 1861-1864[Abstract]
  42. Goldstein, J. L., and Brown, M. S. (1990) Nature 343, 425-430[CrossRef][Medline] [Order article via Infotrieve]
  43. Bergstrom, J. D., Kurtz, M. M., Rew, D. J., Amend, A. M., Karkas, J. D., Bostedor, R. G., Bansal, V. S., Dufresne, C., Van Middlesworth, F. L., Hensens, O. D., Liesch, J. M., Zink, D. L., Wilson, K. E., Onishi, J., Milligan, J. A., Bills, G., Kaplan, L., Nallin Omstead, M., Jenkins, R. G., Huang, L., Meinz, M. S., Quinn, L., Burg, R. W., Kong, Y. L., Mochales, S., Mojena, M., Martin, I., Pelaez, F., Diez, M. T., and Alberts, A. W. (1993) Proc. Natl. Acad. Sci. U. S. A. 90, 80-84[Abstract]
  44. DeClue, J. E., Vass, W. C., Papageorge, A. G., Lowy, D. R., and Willumsen, B. M. (1991) Cancer Res. 51, 712-717[Abstract]
  45. Maltese, W. A., Defendini, R., Green, R. A., Sheridan, K. M., and Donley, D. K. (1985) J. Clin. Invest. 76, 1748-1754[Medline] [Order article via Infotrieve]
  46. Bansal, N., Houle, A., and Melnykovych, G. (1991) FASEB J. 5, 211-216[Abstract/Free Full Text]
  47. Perez-Sala, D., Collado-Escobar, D., and Mollinedo, F. (1995) J. Biol. Chem. 270, 6235-6242[Abstract/Free Full Text]
  48. Thibault, A., Samid, D., Tompkins, A. C., Cooper, M. R., Figg, W. D., Hohl, R. J., Trepel, J. B., Liang, B., Patronas, N., Venzon, D. J., Reed, E., and Myers, C. E. (1996) Clin. Cancer Res. 2, 483-491[Abstract]
  49. Datto, M. B., Li, Y., Panus, J. F., Howe, D. J., Xiong, Y., and Wang, X.-F. (1995) Proc. Natl. Acad. Sci. U. S. A. 92, 5545-5549[Abstract]
  50. Datto, M. B., Yu, Y., and Wang, X.-F. (1995) J. Biol. Chem. 270, 28623-28628[Abstract/Free Full Text]
  51. Liu, M., Lee, M. H., Cohen, M., Bommakanti, M., and Freedman, L. P. (1996) Genes Dev. 10, 142-153[Abstract]
  52. Biggs, J. R., Kudlow, J. E., and Kraft, A. S. (1996) J. Biol. Chem. 271, 901-906[Abstract/Free Full Text]
  53. Zeng, Y.-X., and El-Deiry, S. W. (1996) Oncogene 12, 1557-1564[Medline] [Order article via Infotrieve]
  54. Zeng, Y.-X., Somasundaram, K., and El-Deiry, W. S. (1997) Nat. Genet. 15, 78-82[Medline] [Order article via Infotrieve]
  55. Chin, Y. E., Kitagawa, M., Su, W.-C. S., You, Z. H., Iwamoto, Y., and Fu, X.-Y. (1996) Science 272, 719-722[Abstract]
  56. Udvadia, A. J., Rogers, K. T., Higgins, P. D. R., Murata, Y., Martin, K. H., Humphrey, P. A., and Horowitz, J. M. (1993) Proc. Natl. Acad. Sci. U. S. A. 90, 3265-3269[Abstract]
  57. Kim, S.-J., Lee, H.-D., Robbins, P. D., Busam, K., Sporn, M. B., and Roberts, A. B. (1991) Proc. Natl. Acad. Sci. U. S. A. 88, 3052-3056[Abstract]
  58. Kim, S.-J., Onwuta, U. S., Lee, Y. I., Li, R., Botchan, M. R., and Robbins, P. D. (1992) Mol. Cell. Biol. 12, 2455-2463[Abstract]
  59. Kinzler, K. W., and Vogelstein, B. (1994) N. Engl. J. Med. 331, 49-50[Free Full Text]


Copyright © 1998 by The American Society for Biochemistry and Molecular Biology, Inc.