Cancer Letters

Cancer Letters

Volume 264, Issue 1, 8 June 2008, Pages 145-161
Cancer Letters

Genomic instability, mutations and expression analysis of the tumour suppressor genes p14ARF, p15INK4b, p16INK4a and p53 in actinic keratosis

https://doi.org/10.1016/j.canlet.2008.01.042Get rights and content

Abstract

Actinic keratosis (AK) is a well-established pre-cancerous skin lesion that has the potential to progress to squamous cell carcinoma (SCC). We investigated the involvement of the CDKN2A, CDKN2B and p53 genes in AK and in the progression of AK to SCC. Mutational analysis on exons 1a, 1b and 2 of the CDKN2A locus and exon 1 of the CDKN2B locus as well as allelic imbalance was performed in 26 AK specimens. Expression levels of the genes p14ARF, p15INK4b, p16INK4a and p53 were examined in 16 AKs and 12 SCCs by real-time RT-PCR. A previously described polymorphism of p16INK4a (Ala148Thr) was detected at an allelic frequency of 12%. Six samples carried novel mutations at codon 71 of the CDKN2A locus and one sample presented an additional mutation at codon 65. Two AK samples carried a not-previously described non-UV type missense mutation at codon 184 (Val184Glu) of exon 1b in the p14ARF gene. Regarding the CDKN2B locus a new mutation at codon 50 (Ala50Thr) and another at codon 24 (Arg24Arg), were detected. Microsatellite instability (MSI) was found in 15% of AKs in at least one marker, indicating that genetic instability has some implication in the development of AK. Down-regulation of p16INK4a and p53 mRNA levels was noted in SCC compared to AK. TSGs expression levels in sun-exposed morphologically normal-appearing skin, suggests that abnormal growth stimuli might exist in these tissues as well. Furthermore, we suggest a possible role of p15INK4b, independently from the intracellular pathway mediated by p16INK4a, and of p14ARF in AK development, as well as in the progression of AK to SCC. The deregulation of the expression profiles of the CDKN2A, CDKN2B and p53 genes may, independently of mutations and LOH at 9p21, play a significant role in AK and progression of AK to SCC.

Introduction

Actinic keratosis (AK) is a well-established pre-cancerous skin lesion that has the potential to progress to squamous cell carcinoma (SCC). SCCs of the skin are the most prevalent keratinocyte-derived tumours, second to basal cell carcinomas (BCCs) [1]. Clinically AKs appear primarily on chronically sun-exposed areas [2], and sites adjacent to AKs may contain significant histological alterations, suggesting extensive pre-neoplastic alterations in sun-damaged skin [3]. It has been found that 82.4% of SCCs arise either within (26.7%) or in close proximity to AKs (55.7%) [4], and the risk of AK progression to SCC has been reported as being between 0.025% and 16% [5]. This progression is thought to be due to chronic sun exposure, specifically ultraviolet B sunlight. Additionally, 72% of SCCs are associated with contiguous AKs [6].

UV radiation (UVR) is a prevailing factor implicated in the etiology of SCCs and AKs. Solar radiation (UVB and UVA) can mutagenise DNA, which often produces UV landmark mutations (two tandem CC:GG to TT:AA transitions and two C:G to T:A transitions at dipyrimidic sites). When these mutations affect the function of sufficient oncogenes, tumour suppressor genes (TSGs) and important housekeeping genes, they result in an uncontrolled cell cycle and the transformation of keratinocytes [7].

TSG inactivation is recognised as a pivotal event in the initiation and progression of neoplasia. The 9p21 chromosomal band, at which the INK4a/ARF (CDKN2A) and INK4b (CDKN2B) gene loci are located, is one of the most frequently altered genomic regions in human cancers [8], and the cyclin-dependent kinase inhibitor 2A (CDKN2A) locus is the second most commonly altered gene locus in human cancer after p53 [9]. The locus CDKN2A encodes two TSGs, p16INK4a and p14ARF, both involved in the negative control of cell proliferation. They share common exons 2 and 3, but have alternatively spliced first exons (exon 1a for p16INK4a and 1b for p14ARF). These first exons are under the control of distinct promoters and uniquely create two proteins that have no sequence homology at the amino acid level [10]. p16INK4a is a CDK4 inhibitor that specifically acts at the G1/S phase of the cell cycle by negatively controlling retinoblastoma (Rb) phosphorylation status [11], while p14ARF binds to MDM2 preventing both MDM2-mediated p53 degradation and MDM2-mediated Rb inactivation thus causing the arrest of the G1 and G2 phases of the cell [12]. Consequently, stabilised p53 can induce temporary and permanent growth arrest, DNA repair, terminal differentiation or apoptosis in response to oncogenic signals and DNA damage [13]. p15INK4b, another cyclin-dependent kinase inhibitor gene located in chromosome region 9p21.2, has a strong structural and functional homology to p16INK4a [10] and is transcriptionally activated by transforming growth factor-β (TGF-β) [14]. p15INK4b protein binds to the cdk4-Cyclin D complex, displacing p27 and freeing it to bind to and inhibit the cdk2-Cyclin E complex required for entry into the S phase of the cell cycle [15].

Knockout mice experiments suggest a possible role for p14ARF in the initiation of cutaneous SCC development [8], [11] and for p16INK4a in the determination of the differentiation status of skin squamous tumours [16]. In primary cutaneous SCC, loss of heterozygosity (LOH) at the 9p21 locus and CDKN2A mutations have been reported, but with disparate result [17], [18], [19], [20]. Although AK shares many of the same loci as SCC, LOH in the CDKN2A locus appears to be less common in AK lesions than it is in SCC [21].

Inactivation of the Rb and p53 tumour suppressor pathways is observed in most human cancers [8]. By virtue of its close involvement in both pathways, the p14ARF–p15INK4b–p16INK4a gene cluster at chromosome 9p21 may be a nexus in the cellular-growth-control network, the inactivation of which results in the collapse of the tumour suppression system [28]. The loss of the CDKN2A, CDKN2B and p53 genes at the gene level (point mutation, deletion or promoter hypermethylation) or at the mRNA expression level in pre-tumour and tumour cells is, perhaps, the key event that provides considerable growth stimuli leading to uncontrolled proliferation and destabilisation.

In this study, we investigated the integrity of the CDKN2A and CDKN2B loci in 26 AK samples, by analysing the presence of allelic imbalance/loss of heterozygosity (AI/LOH) in microsatellite markers and observed a low percentage of LOH (4%) and a significant incidence of MSI (15%) in at least one marker of the 9p21 locus. Furthermore, exons 1a, 1b and 2 of the CDKN2A and exon 1 of the CDKN2B loci were screened for mutations in 26 AK samples, and novel mutations apart from previously reported nucleotide changes were detected. Mutation analysis and genomic instability in SCC has been extensively studied in the past. Since many of our patients with AK presented contiguous SCC, we considered it interesting to study the expression levels of the TSGs p14ARF, p15INK4b, p16INK4a and p53 in AK and SCC, in comparison with normal adjacent tissue. Our results suggest that p16INK4a and p53 mRNA down-regulation in SCC compared to AK, might lead in collapse of the tumour suppressor system. Furthermore, we suggest a possible role of p15INK4b, independently from the intracellular pathway mediated by p16INK4a, and of p14ARF in AK development, as well as in the progression of AK to SCC. Apart from mutations and LOH/MSI at 9p21, the deregulation of the expression profile of the TSGs may play a significant role in AK appearance and progression of AK to SCC.

Section snippets

Tumour specimens and DNA extraction

Twenty-six AK with 22 adjacent normal tissues and 12 SCC samples with six adjacent normal tissues, were obtained from patients treated at ‘A. Sygros’ Hospital (Athens, Greece) with the approval of the donors and the institute’s ethical committee. Patients with AK lesions were between the ages of 63 and 92 years (average 76; SD ± 7.2; 16 males and 10 females). Patients with SCC lesions were between the ages of 63 and 88 years (average 75; SD ± 7.5; 7 males and 5 females). A total of eight AK patients

Results

Mutation analysis and genomic instability for the genes p14ARF, p15INK4B, p16INK4a and p53 in SCC, has been extensively studied in the past [19], [20], [21], [25], [26], [27], [28], [29]. Considering the fact that many of our AK patients had contiguous SCCs, and that so far few reports have focused on the expression of these TSGs in SCC, it was prompting also to evaluate their mRNA levels.

Discussion

Inactivation of the Rb and p53 tumour suppressor pathways is observed in most human cancers [8]. Due to its close participation in both pathways, the p14ARF–p15INK4b–p16INK4a gene cluster at chromosome 9p21 may be a nexus of the cellular-growth-control network, the inactivation of which results in the collapse of the tumour suppression system [28]. To date, a vast amount of data has demonstrated multiple types of genetic alterations in the 9p21 region, the prevalence of which varies with the

References (70)

  • R.M. MacKie et al.

    CDKN2A germline mutations in U.K. patients with familial melanoma and multiple primary melanomas

    J. Invest. Dermatol.

    (1998)
  • A. Heidenreich et al.

    Molecular analysis of P16(Ink4)/CDKN2 and P15(INK4B)/MTS2 genes in primary human testicular germ cell tumors

    J. Urol.

    (1998)
  • Y. Zhang et al.

    Mutations in human ARF exon 2 disrupt its nucleolar localization and impair its ability to block nuclear export of MDM2 and p53

    Mol. Cell

    (1999)
  • H. Rizos et al.

    Mutations in the INK4a/ARF melanoma susceptibility locus functionally impair p14ARF

    J. Biol. Chem.

    (2001)
  • J.J. Scarisbrick et al.

    Frequent abnormalities of the p15 and p16 genes in mycosis fungoides and sezary syndrome

    J. Invest. Dermatol.

    (2002)
  • F.J. Child et al.

    Inactivation of tumor suppressor genes p15(INK4b) and p16(INK4a) in primary cutaneous B cell lymphoma

    J. Invest. Dermatol.

    (2002)
  • A. Hodges et al.

    Immunohistochemical comparison of p16 expression in actinic keratoses and squamous cell carcinomas of the skin

    Mod. Pathol.

    (2002)
  • J. Pomerantz et al.

    The Ink4a tumor suppressor gene product, p19Arf, interacts with MDM2 and neutralizes MDM2’s inhibition of p53

    Cell

    (1998)
  • T. Fujiwara et al.

    Accumulated clonal genetic alterations in familial and sporadic colorectal carcinomas with widespread instability in microsatellite sequences

    Am. J. Pathol.

    (1998)
  • Y. Kushida et al.

    Loss of heterozygosity in actinic keratosis, squamous cell carcinoma and sun-exposed normal-appearing skin in Japanese: difference between Japanese and Caucasians

    Cancer Lett.

    (1999)
  • A.G. Quinn et al.

    Microsatellite instability in human non-melanoma and melanoma skin cancer

    J. Invest. Dermatol.

    (1995)
  • A. Rubben et al.

    Analysis of tumor cell evolution in a melanoma: evidence of mutational and selective pressure for loss of p16ink4 and for microsatellite instability

    J. Invest. Dermatol.

    (2000)
  • E. Alvino et al.

    High-frequency microsatellite instability is associated with defective DNA mismatch repair in human melanoma

    J. Invest. Dermatol.

    (2002)
  • A.J. Sober et al.

    Precursors to skin cancer

    Cancer

    (1995)
  • R. Marks

    Solar keratoses

    Br. J. Dermatol.

    (1990)
  • M.A. Mittelbronn et al.

    Frequency of pre-existing actinic keratosis in cutaneous squamous cell carcinoma

    Int. J. Dermatol.

    (1998)
  • R.G. Glogau

    The risk of progression to invasive disease

    J. Am. Acad. Dermatol.

    (2000)
  • D. Czarnecki et al.

    The majority of cutaneous squamous cell carcinomas arise in actinic keratoses

    J. Cutan. Med. Surg.

    (2002)
  • M. Kramer et al.

    Radiation-induced activation of transcription factors in mammalian cells

    Radiat. Environ. Biophys.

    (1990)
  • W.H. Liggett et al.

    Role of the p16 tumor suppressor gene in cancer

    J. Clin. Oncol.

    (1998)
  • P. Jiang et al.

    Comparative analysis of Homo sapiens and Mus musculus cyclin-dependent kinase (CDK) inhibitor genes p16 (MTS1) and p15 (MTS2)

    J. Mol. Evol.

    (1995)
  • K. Ranade et al.

    Mutations associated with familial melanoma impair p16INK4 function

    Nat. Genet.

    (1995)
  • F.J. Stott et al.

    The alternative product from the human CDKN2A locus, p14(ARF), participates in a regulatory feedback loop with p53 and MDM2

    EMBO J.

    (1998)
  • G.J. Hannon et al.

    p15INK4B is a potential effector of TGF-beta-induced cell cycle arrest

    Nature

    (1994)
  • C.J. Sherr

    The INK4a/ARF network in tumour suppression

    Nat. Rev. Mol. Cell Biol.

    (2001)
  • Cited by (69)

    • Guidelines of care for the management of actinic keratosis

      2021, Journal of the American Academy of Dermatology
      Citation Excerpt :

      The specific DNA mutations that have been characterized in AKs include those seen in keratinocyte carcinoma, including mutations in TP53, the gene encoding the p53 protein, known to have an important role in tumor suppression for a variety of malignancies, including SCC.36,37 Additional mutations that are thought to be pathogenic in the development of AK include mutations in p16, ras family members, NFKB, CDKN2A, telomerase, and TNF alpha.38-43 Estimates of the risk of progression of AK to SCC vary from less than 0.1% to 20%.27,30,44,45

    • The Genomic Landscape of Actinic Keratosis

      2021, Journal of Investigative Dermatology
      Citation Excerpt :

      Apart from an early study that suggested that AKs have more LOH than cSCC (Rehman et al., 1996), our findings are contrary to those of more recent studies that found that AKs have relatively few CNAs (García-Díez et al., 2019; Hameetman et al., 2013). However, most studies agree on the most common sites of chromosomal instability, with 9p loss—the region where CDKN2A is located—ubiquitous among AK studies (Ashton et al., 2003; García-Díez et al., 2019; Kanellou et al., 2008; Rehman et al., 1996). Interrogating published expression datasets against our SMG list found KIF24, KCNK5, EPB41L2, and ABI3BP downregulated in AK compared with those in the normal skin, suggesting tumor suppressor roles.

    • DNA Methylation in Epidermal Differentiation, Aging, and Cancer

      2020, Journal of Investigative Dermatology
    • DNA damage and telomere length shortening in the peripheral blood leukocytes of 20 years SM-exposed veterans

      2018, International Immunopharmacology
      Citation Excerpt :

      OGG1 primers were designed by primer 3 software. The p16INK4a primers were as described by Kanellou et al. [23], and Phosphoglycerate kinase (PGK) gene expression was also selected as normalizer as described by Eghtedardoost et al. [24], Table 1. Samples were run in duplicates, and the results were calculated by the ∆∆Ct method, and presented as fold changes in expression compared to the SM-unexposed healthy group.

    View all citing articles on Scopus
    View full text