Hostname: page-component-848d4c4894-pjpqr Total loading time: 0 Render date: 2024-06-24T18:38:26.244Z Has data issue: false hasContentIssue false

Review of: The inhibitor of cyclin-dependent kinase 4a/alternative reading frame (INK4a/ARF) locus encoded proteins p16INK4a and p19ARF repress cyclin D1 transcription through distinct cis elements

Published online by Cambridge University Press:  05 April 2005

C. Scafoglio
Affiliation:
Dipartimento di Patologia Generale, Seconda Universita' degli Studi di Napoli, Napoli, Italy.
A. Weisz
Affiliation:
Dipartimento di Patologia Generale, Seconda Universita' degli Studi di Napoli, Napoli, Italy.

Abstract

Type
Journals Club
Copyright
2005 Cambridge University Press

Citation of original article:

D'Amico M., Wu K., Fu M., Rao M., Albanese C., Russell R. G., Lian H., Bregman D., White M. A., Pestell R. G. Cancer Research 2004 Jun 15; 64(12): 4122–4130.

Abstract of the original article

The Ink4a/Arf locus encodes two structurally unrelated tumour suppressor proteins, p16(INK4a) and p14(ARF) (murine p19(ARF)). Invariant inactivation of either the p16(INK4a)–cyclin D/CDK–pRb pathway and/or p53–p14(ARF) pathway occurs in most human tumours. Cyclin D1 is frequently over-expressed in breast cancer cells contributing an alternate mechanism inactivating the p16(INK4a)/pRb pathway. Targeted over-expression of cyclin D1 to the mammary gland is sufficient for tumorigenesis, and cyclin D1 −/− mice are resistant to Ras-induced mammary tumours. Recent studies suggest cyclin D1 and p16(INK4a) expression are reciprocal in human breast cancers. Herein, reciprocal regulation of cyclin D1 and p16(INK4a) was observed in tissues of mice mutant for the Ink4a/Arf locus. p16(INK4a) and p19(ARF) inhibited DNA synthesis in MCF-7 cells. p16(INK4a) repressed cyclin D1 expression and transcription. Repression of cyclin D1 by p16(INK4a) occurred independently of the p16(INK4a)–CDK4-binding function and required a cAMP-response element/activating transcription factor-2-binding site. p19(ARF) repressed cyclin D1 through a novel distal cis element at −1137, which bound p53 in chromatin-immunoprecipitation assays. Transcriptional repression of the cyclin D1 gene through distinct DNA sequences may contribute to the tumour suppressor function of the Ink4a/Arf locus.

Review

Cyclin D1 is the regulatory subunit of the cyclin-dependent kinase (CDK) 4/6 holoenzymes, protein kinases whose activity is strictly controlled during the cell cycle and represents a limiting step for G1–S transition, mainly acting upon the retinoblastoma tumour-suppressor gene product pRb [1]. Activation of CCND1, the gene encoding cyclin D1, is a crucial step for G1 phase progression and completion. All the mitogenic pathways activated by both extra-cellular and intra-cellular signals, including female sex steroids, increase cyclin D1 levels, a required step for cell transition through the G1–S restriction point [24]. The cyclin D1 gene is over-expressed in 50% of breast cancers and its activity is required for breast carcinogenesis [56] and responsiveness of breast epithelia to ovarian steroids [7]. Nonetheless, CCND1 amplification occurs only in 13–15% of breast cancer specimens [8], indicating that over-expression of this gene in breast cancer is mainly due to dysfunctions of its transcriptional and post-transcriptional controls.

In their recent report, D'Amico et al. [9] highlight a novel mechanism for CCND1 gene regulation by the two proteins encoded by the CDKN2A (cyclin-dependent kinase inhibitor 2A) tumour suppressor locus: p16INK4a and p17ARF. These two cell cycle inhibitors are translated from the same mRNA but from different reading frames, depending upon alternative splicing of exon 1, and share the sequence encoded in exons 2 and 3 [10]. Starting from the observation that in human breast cancers cyclin D1 expression is reciprocal to that of p16INK4a, the authors went on to observe an increased proliferation rate of embryonic fibroblasts (MEFs) from INK4a/ARF−/− mutant mice, with respect to their normal counterparts, concomitant to cyclin D1 over-expression. On the other hand, they found that high levels of either p16INK4a or p19ARF hamper G1–S transition in human MCF-7 breast cancer cells and, most notably, reduce cyclin D1 expression. Considering the possibility that the CCND1 gene itself might be targeted by the two tumour suppressor proteins, resulting in transcriptional repression, the authors demonstrated by transient transfection assays in MCF-7 cells that over-expression of either one of the two proteins indeed induces inhibition of CCND1 promoter activity. Molecular and genetic analysis of the mechanisms underlying this effect of the inhibitors revealed that p16INK4a-dependent gene suppression requires a functional CRE (cAMP Response Element) in the CCND1 promoter, and is probably mediated by interference with DNA-bound CREB–ATF-2 complex interactions with components of the basal transcription machinery, while promoter silencing by p19ARF might depend on an atypical p53 response element, thereafter named ARE (ARF Response Element).

Cyclin-dependent kinase inhibitors play a fundamental role in regulating cellular proliferation and often show tumour suppressor actions. The two proteins encoded by the CDKN2A locus are able to block cellular proliferation in two different ways and in response to different stimuli. p16INK4a inhibits the kinase activity of the cyclin D1-CDK4/6 complexes, blocking E2F1 release from pRb suppression and thereby acting as a modulator of the G1–S transition; the observation that it also prevents CCND1 transcription unveils a novel mechanism for interfering with the cyclin/retinoblastoma protein pathway, that is, prevention of cyclin D1 synthesis. On the other hand, p19ARF is activated by cellular stresses and enhances p53 stability by inhibiting MDM2-dependent degradation. As a consequence, the two products of the CDKN2A locus influence the activity of two main cell cycle regulators: pRb and p53. Furthermore, the observation that both proteins inhibit CCND1 transcription through distinct cis-acting genetic elements highlights an additional point of intersection between the INK4a–cyclin D/CDKs–pRb and the ARF–MDM2–p53 pathways, and underlines the importance of CCND1 regulation for cellular proliferation control. p16INK4a and p19ARF, in fact, appear to be effectors of growth inhibitory circuits also independently from their ability to inhibit CDKs, exerting each in its own way a direct control on CCND1 promoter activity.

The CCND1 gene, thus, emerges once again as a true cellular sensor, capable of integrating the complexity of extra-cellular (growth factors and growth inhibitors, hormones, inter-cellular signals, etc.) and intra-cellular (cell stress, in its many variants, etc.) signals onto functional changes of the cell cycle machinery. Interestingly, also cyclin D1 can control transcription of its own gene, suggesting a way for signal amplification. In human breast cancer cells, CCND1 activation by female sex steroids occurs stepwise. During early G1, gene activation in response to these hormones occurs via direct interaction (tethering) of the oestrogen and progesterone receptors with the AP-1 complex bound to the distal CCND1 regulatory region, resulting in recruitment of the basal transcriptional machinery to the promoter; later in the cell cycle sustained gene transcription rate is supported, instead, by recruitment of cyclin D1 to a separate regulatory site of the promoter [11].

These observations, indicating that three main regulators of the ‘cell cycle clock’, namely INK4a, ARF and cyclin D1, converge on CCND1 confirms the importance of transcriptional regulation of this gene in breast cancer cells, both for signal integration and maintenance of specific cellular phenotypes, including aberrant cell proliferation and survival.

References

Kato J, Matsushime H, Hiebert SW, et al. Direct binding of cyclin D to the retinoblastoma gene product (pRb) and pRb phosphorylation by the cyclin D-dependent kinase CDK4. Genes Dev 1993; 7: 331342.Google Scholar
Wang C, Li Z, Fu M, Bouras T, Pestell RG. Signal transduction mediated by cyclin D1: from mitogens to cell proliferation: a molecular target with therapeutic potential. Cancer Treat Res 2004; 119: 217237.Google Scholar
Altucci L, Addeo R, Cicatiello L, et al. 17beta-Estradiol induces cyclin D1 gene transcription, p36D1–p34cdk4 complex activation and p105Rb phosphorylation during mitogenic stimulation of G(1)-arrested human breast cancer cells. Oncogene 1996; 12: 23152324.Google Scholar
Musgrove EA, Hamilton JA, Lee CS, et al. Growth factor, steroid, and steroid antagonist regulation of cyclin gene expression associated with changes in T-47D human breast cancer cell cycle progression. Mol Cell Biol 1993; 13: 35773587.Google Scholar
Wang TC, Cardiff RD, Zukerberg L, et al. Mammary hyperplasia and carcinoma in MMTV-cyclin D1 transgenic mice. Nature 1994; 369: 669671.Google Scholar
Yu Q, Geng Y, Sicinski P. Specific protection against breast cancers by cyclin D1 ablation. Nature 2001; 411: 10171021.Google Scholar
Sicinski P, Donaher JL, Parker SB, et al. Cyclin D1 provides a link between development and oncogenesis in the retina and breast. Cell 1995; 82: 621630.Google Scholar
Fantl V, Richards MA, Smith R, et al. Gene amplification on chromosome band 11q13 and oestrogen receptor status in breast cancer. Eur J Cancer 1990; 26: 423429.Google Scholar
D'Amico M, Wu K, Fu M, et al. The inhibitor of cyclin-dependent kinase 4a/alternative reading frame (INK4a/ARF) locus encoded proteins p16INK4a and p19ARF repress cyclin D1 transcription through distinct cis elements. Cancer Res 2004; 64: 41224130.Google Scholar
Sherr CJ. The INK4a/ARF network in tumour suppression. Nat Rev Mol Cell Biol 2001; 2: 731737.Google Scholar
Cicatiello L, Addeo R, Sasso A, et al. Estrogens and progesterone promote persistent CCND1 gene activation during G1 by inducing transcriptional derepression via c-Jun/c-Fos/estrogen receptor (progesterone receptor) complex assembly to a distal regulatory element and recruitment of cyclin D1 to its own gene promoter. Mol Cell Biol 2004; 24: 72607274.Google Scholar