Characterization of a Germ-Line Deletion, Including the Entire INK4/ARF Locus, in a Melanoma-Neural System Tumor Family: Identification of ANRIL, an Antisense Noncoding RNA Whose Expression Coclusters with ARF (original) (raw)

Skip Nav Destination

Clinical Research| April 17 2007

Eric Pasmant;

1Laboratoire de Génétique Moléculaire-Institut National de la Sante et de la Recherche Medicale U745, Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris V;

Search for other works by this author on:

Ingrid Laurendeau;

1Laboratoire de Génétique Moléculaire-Institut National de la Sante et de la Recherche Medicale U745, Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris V;

Search for other works by this author on:

Delphine Héron;

2Département de Génétique, Hôpital de la Pitié-Salpêtrière, Paris, France; and

Search for other works by this author on:

Michel Vidaud;

1Laboratoire de Génétique Moléculaire-Institut National de la Sante et de la Recherche Medicale U745, Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris V;

Search for other works by this author on:

Dominique Vidaud;

1Laboratoire de Génétique Moléculaire-Institut National de la Sante et de la Recherche Medicale U745, Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris V;

Search for other works by this author on:

Ivan Bièche

1Laboratoire de Génétique Moléculaire-Institut National de la Sante et de la Recherche Medicale U745, Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris V;

3Laboratoire d'Oncogénétique-Institut National de la Sante et de la Recherche Medicale U735, Centre René Huguenin, St-Cloud, France

Search for other works by this author on:

Crossmark: Check for Updates

Requests for reprints: Ivan Bièche, Laboratoire de Génétique Moléculaire-Institut National de la Sante et de la Recherche Medicale U745, Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris 5, 4 Avenue de l'Observatoire, 75006 Paris, France. Phone: 33-1-53-73-97-25; Fax: 33-1-44-07-17-54; E-mail: ivan.bieche@univ-paris5.fr.

Received: June 01 2006

Revision Received: November 17 2006

Accepted: February 09 2007

Online ISSN: 1538-7445

Print ISSN: 0008-5472

©2007 American Association for Cancer Research.

2007

Cancer Res (2007) 67 (8): 3963–3969.

Article history

Revision Received:

November 17 2006

Accepted:

February 09 2007

Abstract

We have previously detected a large germ-line deletion, which included the entire p15/CDKN2B-p16/CDKN2A-p14/ARF gene cluster, in the largest melanoma-neural system tumor (NST) syndrome family known to date by means of heterozygosity mapping based on microsatellite markers. Here, we used gene dose mapping with sequence-tagged site real-time PCR to locate the deletion end points, which were then precisely characterized by means of long-range PCR and nucleotide sequencing. The deletion was exactly 403,231 bp long and included the entire p15/CDKN2B, p16/CDKN2A, and p14/ARF genes. We then developed a simple and rapid assay to detect the junction fragment and to serve as a direct predictive DNA test for this large French family. We identified a new large antisense noncoding RNA (named ANRIL) within the 403-kb germ-line deletion, with a first exon located in the promoter of the p14/ARF gene and overlapping the two exons of p15/CDKN2B. Expression of ANRIL mainly coclustered with p14/ARF both in physiologic (various normal human tissues) and in pathologic conditions (human breast tumors). This study points to the existence of a new gene within the p15/CDKN2B-p16/CDKN2A-p14/ARF locus putatively involved in melanoma-NST syndrome families and in melanoma-prone families with no identified p16/CDKN2A mutations as well as in somatic tumors. [Cancer Res 2007;67(8):3963–9]

Introduction

Hereditary cutaneous malignant melanoma (CMM) is a well-established syndrome (1). One of the main CMM loci has been mapped to chromosome arm 9p21. Three candidate tumor suppressor genes have been identified in this region: p15/CDKN2B encodes p15 protein, p16/CDKN2A encodes p16 protein, and p14/ARF encodes p14ARF protein (p19ARF in mice; ref. 2). p14ARF protein is encoded by an alternative exon 1 (1β) and by exons 2 and 3 of the p16/CDKN2A gene in a different reading frame. Consequently, p16 and p14ARF share no homology at the amino acid level and have significantly different functions. p15 and p16 are specific cyclin-dependent kinase (CDK) 4/CDK6 inhibitors in the retinoblastoma (Rb) pathway, and inactivation of p15 and/or p16 allows cells to escape cell cycle arrest in G1. p14ARF acts in both the p53 and Rb pathways by interacting with MDM2, a protein common to the two pathways. p14ARF binds MDM2 and promotes its degradation, resulting in p53 activation and G1 and G2 arrest. p16/CDKN2A is currently the most clinically relevant melanoma susceptibility gene in 9p21. By contrast, germ-line mutations in p14/ARF are rare (3, 4) and have not been described in p15/CDKN2B.

Following the observation that patients with neural system tumors (NST) are at an increased risk of developing CMM, a large population-based survey showed an increased risk of NSTs in melanoma patients and also in their first- and second-degree relatives (5). This has been termed the melanoma-astrocytoma syndrome, owing to the presence of astrocytomas in the first family to be characterized (OMIM 155755; ref. 6). The authors suggested joint predisposition segregating as a Mendelian disorder. Three large deletions have now been reported in families with a hereditary predisposition to melanoma and NSTs: one involving p15/CDKN2B, p16/CDKN2A, and p14/ARF in a French family (7), one 21,826-pb deletion restricted to exon 1β of p14/ARF in a U.S. family (ref. 8, family B in ref. 9), and one probably involving only p14/ARF in a U.K. family (ref. 7, family D in ref. 9). Three additional large deletions have been reported in families with a hereditary predisposition to melanoma without NSTs: two involving only p14/ARF in a U.S. (10) and in a U.K. family (family A in ref. 9) and one restricted to p16/CDKN2A in a U.K. family (family C in ref. 9). It is noteworthy that Mistry et al. (9) suggested coancestry of families A (without NSTs) and B (with NSTs) and concluded that the susceptibility in these two families was primarily to melanoma. Finally, germ-line mutations in p16/CDKN2 have been reported in families with a hereditary predisposition to melanoma with NSTs (9).

Here, using sequence-tagged site (STS) real-time PCR-based gene dose mapping, we determined the precise size and end points of a large germ-line deletion removing the entire p15/CDKN2B-p16/CDKN2A-p14/ARF gene cluster initially detected in a large French melanoma-NST syndrome family (7).

We also identified, within the 403-kb germ-line deletion, a new large antisense noncoding RNA (named ANRIL) with a first exon located in the promoter of the p14/ARF gene and overlapping the two exons of p15/CDKN2B. Expression of this noncoding RNA coclustered mainly with p14/ARF in both physiologic and pathologic conditions.

Materials and Methods

Patients. The family with a cluster of CMM and NSTs (Fig. 1) has been investigated since 1977 at Hôpital Pitié-Salpêtrière in Paris, France. About 100 individuals have been surveyed. Pathologic materials initially investigated in 16 different laboratories were, when possible, reassessed by both a dermatopathologist and a neuropathologist. Extensive historical and clinical descriptions of this kindred are available elsewhere (11). Overall, eight members of this family have or had CMM and nine members have or had NSTs, ranging from astrocytoma of all grades to schwannoma, neurofibroma, and meningioma. Six relatives have both skin tumors and NSTs, resulting in statistical cosegregation of the skin tumor and NST traits. This family therefore illustrates a melanoma-NST syndrome probably inherited as a single autosomal dominant Mendelian trait. However, the possibility of other explanations cannot be ruled out for the constellation of tumors observed in this family, such as multiple tightly linked genes or a contiguous gene syndrome.

Figure 1.

Figure 1. Pedigree of the family with the melanoma-NST syndrome. Square and circle pedigree symbols, males and females, respectively. Diamonds, unaffected siblings of the two sexes. Number inside the symbol, multiple individuals. Asterisks, the three 403-kb deleted and affected patients tested. Patient III-16 had excision of what proved to be a neurofibroma of the tarsus. An intercostal neurofibroma was excised in patient III-23 at age 32 and a popliteal neurofibroma in her daughter, IV-20, at age 15. The six other NST-affected patients have or had astrocytoma, schwannoma, neurofibroma, or meningioma (11).

Pedigree of the family with the melanoma-NST syndrome. Square and circle pedigree symbols, males and females, respectively. Diamonds, unaffected siblings of the two sexes. Number inside the symbol, multiple individuals. Asterisks, the three 403-kb deleted and affected patients tested. Patient III-16 had excision of what proved to be a neurofibroma of the tarsus. An intercostal neurofibroma was excised in patient III-23 at age 32 and a popliteal neurofibroma in her daughter, IV-20, at age 15. The six other NST-affected patients have or had astrocytoma, schwannoma, neurofibroma, or meningioma (11).

Figure 1.

Figure 1. Pedigree of the family with the melanoma-NST syndrome. Square and circle pedigree symbols, males and females, respectively. Diamonds, unaffected siblings of the two sexes. Number inside the symbol, multiple individuals. Asterisks, the three 403-kb deleted and affected patients tested. Patient III-16 had excision of what proved to be a neurofibroma of the tarsus. An intercostal neurofibroma was excised in patient III-23 at age 32 and a popliteal neurofibroma in her daughter, IV-20, at age 15. The six other NST-affected patients have or had astrocytoma, schwannoma, neurofibroma, or meningioma (11).

Pedigree of the family with the melanoma-NST syndrome. Square and circle pedigree symbols, males and females, respectively. Diamonds, unaffected siblings of the two sexes. Number inside the symbol, multiple individuals. Asterisks, the three 403-kb deleted and affected patients tested. Patient III-16 had excision of what proved to be a neurofibroma of the tarsus. An intercostal neurofibroma was excised in patient III-23 at age 32 and a popliteal neurofibroma in her daughter, IV-20, at age 15. The six other NST-affected patients have or had astrocytoma, schwannoma, neurofibroma, or meningioma (11).

Close modal

DNA extraction. High-molecular-weight DNA was prepared by standard proteinase K digestion followed by phenol-chloroform extraction from whole blood leukocytes or lymphoblastoid cell lines from seven individuals, including three affected haplotype-positive relatives (III-16, III-23, and IV-20) and four healthy haplotype-negative relatives (II-9, III-10, III-14, and IV-9). The haplotypes have been defined elsewhere (11).

STS real-time PCR-based gene dose mapping. In this method, quantitative values are obtained from the threshold cycle number at which the increase in the signal associated with exponential growth of PCR products begins to be detected by PE Biosystems analysis software (Perkin-Elmer Applied Biosystems, Foster City, CA), used as recommended in the manufacturer's manuals. The precise amount of genomic DNA added to each reaction mix (based on absorbance) and its quality (i.e., lack of extensive degradation) are both difficult to assess. We therefore also quantified the ALB gene (encoding albumin and mapping to chromosome region 4q11-q13) as an endogenous DNA control, and each sample was normalized based on its ALB content. The relative STS marker copy number was also normalized to a calibrator, or 1× sample, consisting of genomic DNA from a normal subject.

Final results, expressed as N-fold differences in the target STS marker copy number relative to the ALB gene and the calibrator, and termed “N_STS_,” were determined as follows: N_STS_ = 2(Δ_C_tsample − Δ_C_tcalibrator), where Δ_C_t values of the sample and calibrator are determined by subtracting the average _C_t value of the target STS marker from the average C_t value of the ALB gene. Given the target STS marker, samples with N_STS values of 0.5 and 1.0 were considered deleted and normal, respectively. Primers for ALB and the 26 9p21-linked STS markers were chosen with the assistance of the computer program Oligo 4.0 (National Biosciences, Plymouth, MN). We conducted BLASTN searches against nr (the nonredundant set of the Genbank, European Molecular Biology Laboratory, and DNA Data Bank of Japan database sequences) to confirm the total STS specificity of the nucleotide sequences chosen for the primers. The primer pairs for each STS were selected to be unique in the human genome; in particular, they were located outside repetitive DNA sequences (Alus, LINEs, etc.). All PCRs were done with an ABI Prism 7700 Sequence Detection System (Perkin-Elmer Applied Biosystems) and the SYBR Green PCR Core Reagents kit (Perkin-Elmer Applied Biosystems). The thermal cycling conditions comprised an initial denaturation step at 95°C for 10 min and 50 cycles at 95°C for 15 s and 65°C for 1 min. Experiments were done with triplicates for each data point.

Fine characterization of the 403-kb germ-line deletion. Long-range PCR was done with the Expand Long Template PCR System (Boehringer Mannheim, Mannheim, Germany) as recommended by the manufacturer. Forward primer STS20 and reverse primer STS17 amplified an ∼1.5-kb mutant genomic fragment, the normal fragment being too large (405 kb) to be amplified. PCR products were sequenced with the Applied Biosystems Prism Dye Terminator Cycle Sequencing Ready Reaction kit (Perkin-Elmer Applied Biosystems) and an Applied Biosystems DNA Sequencer Model 310 (Perkin-Elmer Applied Biosystems).

Real-time reverse transcription-PCR. The theoretical and practical aspects of real-time quantitative reverse transcription-PCR (RT-PCR) using the ABI Prism 7700 Sequence Detection System have been described in detail elsewhere (12). Briefly, results, expressed as N-fold differences in target gene (p15/CDKN2B, p16/CDKN2A, p14/ARF, or ANRIL gene) expression relative to an endogenous RNA control (PPIA gene), termed N_target_, are determined by the following formula: N_target_ = 2Δ_C_tsample, where the Δ_C_t value of the sample is determined by subtracting the C_t value of the target gene from the C_t value of the PPIA gene. The N_target values of the samples are subsequently normalized such that the N_target value of the normal human tissue (or of the breast tumor), which contains the smallest amount of target gene mRNA, is 1. The nucleotide sequences of the primers for PPIA and for the four target genes are shown in Table 1. To avoid amplification of contaminating genomic DNA, one of the two primers was placed at the junction between two exons. The thermal cycling conditions comprised an initial denaturation step at 95°C for 10 min and 50 cycles at 95°C for 15 s and 65°C for 1 min.

Table 1.

Oligonucleotide primer sequences used

Genes Oligonucleotide Sequence
ANRIL Upper primer 5′-CAACATCCACCACTGGATCTTAACA-3′
Lower primer 5′-AGCTTCGTATCCCCAATGAGATACA-3′
p14/ARF Upper primer 5′-GGTTTTCGTGGTTCACATCCC-3′
Lower primer 5′-CCCATCATCATGACCTGGTCTT-3′
p16/CDKN2A Upper primer 5′-CATGGAGCCTTCGGCTGACT-3′
Lower primer 5′-CCATCATCATGACCTGGATCG-3′
p15/CDKN2B Upper primer 5′-GAATGCGCGAGGAGAACAAG-3′
Lower primer 5′-CCATCATCATGACCTGGATCG-3′
PPIA Upper primer 5′-GTCAACCCCACCGTGTTCTT-3′
Lower primer 5′-CTGCTGTCTTTGGGACCTTGT-3′
Genes Oligonucleotide Sequence
ANRIL Upper primer 5′-CAACATCCACCACTGGATCTTAACA-3′
Lower primer 5′-AGCTTCGTATCCCCAATGAGATACA-3′
p14/ARF Upper primer 5′-GGTTTTCGTGGTTCACATCCC-3′
Lower primer 5′-CCCATCATCATGACCTGGTCTT-3′
p16/CDKN2A Upper primer 5′-CATGGAGCCTTCGGCTGACT-3′
Lower primer 5′-CCATCATCATGACCTGGATCG-3′
p15/CDKN2B Upper primer 5′-GAATGCGCGAGGAGAACAAG-3′
Lower primer 5′-CCATCATCATGACCTGGATCG-3′
PPIA Upper primer 5′-GTCAACCCCACCGTGTTCTT-3′
Lower primer 5′-CTGCTGTCTTTGGGACCTTGT-3′

Results and Discussion

In a French family with a history of melanoma and NSTs, we have previously identified a large germ-line deletion, including the entire p15/CDKN2B-p16/CDKN2A-p14/ARF gene cluster (7). This family is the largest known melanoma-NST syndrome family, eight members having CMM and nine members having NSTs (six members have both skin tumors and NSTs), ranging from astrocytoma of all grades to schwannoma, neurofibroma, and meningioma (Fig. 1). Microsatellite polymorphism analysis showed heterozygosity at both the centromeric marker D9S976 and the telomeric marker D9S736, suggesting that the deleted region was smaller than 700 kb (Fig. 2). It is noteworthy that a fourth gene, MTAP, which encodes methylthioadenosine phosphorylase, is located in this 700-kb region. Behrmann et al. (13) has suggested that MTAP inactivation could be an important step in the development of melanomas.

Figure 2.

Figure 2. STS real-time PCR-based gene dose mapping assay. Multiple primers were designed to be regularly spaced every ∼70 kb (step 1), ∼5 kb (step 2), and ∼1 kb (step 1) within breakpoint regions. Long-range PCR with the forward primer of the STS20 marker and the reverse primer of the STS17 marker generates a PCR product of ∼1.5 kb.

STS real-time PCR-based gene dose mapping assay. Multiple primers were designed to be regularly spaced every ∼70 kb (step 1), ∼5 kb (step 2), and ∼1 kb (step 1) within breakpoint regions. Long-range PCR with the forward primer of the STS20 marker and the reverse primer of the STS17 marker generates a PCR product of ∼1.5 kb.

Figure 2.

Figure 2. STS real-time PCR-based gene dose mapping assay. Multiple primers were designed to be regularly spaced every ∼70 kb (step 1), ∼5 kb (step 2), and ∼1 kb (step 1) within breakpoint regions. Long-range PCR with the forward primer of the STS20 marker and the reverse primer of the STS17 marker generates a PCR product of ∼1.5 kb.

STS real-time PCR-based gene dose mapping assay. Multiple primers were designed to be regularly spaced every ∼70 kb (step 1), ∼5 kb (step 2), and ∼1 kb (step 1) within breakpoint regions. Long-range PCR with the forward primer of the STS20 marker and the reverse primer of the STS17 marker generates a PCR product of ∼1.5 kb.

Close modal

To determine the precise size and end points of this germ-line deletion, we estimated the number of copies (one or two) by real-time PCR-based gene dosage assay for various STS located between the markers D9S976 and D9S736.

We first analyzed 11 STS markers regularly spaced (every ∼70 kb) within the deleted region. Two additional steps, using carefully chosen additional STS markers (eight and seven markers, respectively), allowed us to refine the centromeric and telomeric breakpoint regions (∼1 kb) sufficiently for long-range PCR. The strategy and results are summarized in Fig. 2. Long-range PCR was done on genomic DNA from affected patient by using the forward primer of the retained STS20 marker and the reverse primer of the retained STS17 marker, generating a PCR product of ∼1.5 kb. The normal fragment was too large (∼405 kb) to be amplified. Sequencing of this PCR product revealed that a sequence between nucleotides 22,329,545 and 21,926,315 [numbered in the Build 34 National Center for Biotechnology Information (NCBI) Map Viewer]4

was deleted in the PCR product (Fig. 3). Thus, the deletion was exactly 403,231 bp long (∼403 kb). There were no highly homologous sequences between the two deletion boundaries, and only a 1-bp (adenine) identical sequence at the deletion end points, making it unlikely that the deletion resulted from an unequal homologous recombination event. It is noteworthy, however, that the centromeric breakpoint is located within a truncated LINE element.

Figure 3.

Figure 3. Schematic presentation of the large deletion that includes the entire p15/CDKN2B-p16/CDKN2A-p14/ARF gene cluster. Solid and broken horizontal lines, retained and deleted regions, respectively. Arrows, genes. Nucleotide sequences at the deletion end points. Lowercase letters, deleted sequences. Underlined, a 1-bp repeat sequence (adenine) at the deletion end. Numbers, nucleotide positions in the Build 34 NCBI Map Viewer.

Schematic presentation of the large deletion that includes the entire p15/CDKN2B-p16/CDKN2A-p14/ARF gene cluster. Solid and broken horizontal lines, retained and deleted regions, respectively. Arrows, genes. Nucleotide sequences at the deletion end points. Lowercase letters, deleted sequences. Underlined, a 1-bp repeat sequence (adenine) at the deletion end. Numbers, nucleotide positions in the Build 34 NCBI Map Viewer.

Figure 3.

Figure 3. Schematic presentation of the large deletion that includes the entire p15/CDKN2B-p16/CDKN2A-p14/ARF gene cluster. Solid and broken horizontal lines, retained and deleted regions, respectively. Arrows, genes. Nucleotide sequences at the deletion end points. Lowercase letters, deleted sequences. Underlined, a 1-bp repeat sequence (adenine) at the deletion end. Numbers, nucleotide positions in the Build 34 NCBI Map Viewer.

Schematic presentation of the large deletion that includes the entire p15/CDKN2B-p16/CDKN2A-p14/ARF gene cluster. Solid and broken horizontal lines, retained and deleted regions, respectively. Arrows, genes. Nucleotide sequences at the deletion end points. Lowercase letters, deleted sequences. Underlined, a 1-bp repeat sequence (adenine) at the deletion end. Numbers, nucleotide positions in the Build 34 NCBI Map Viewer.

Close modal

The 403-kb germ-line deletion included the entire p15/CDKN2B-p16/CDKN2A-p14/ARF gene cluster but unambiguously excluded additional well-characterized genes, namely MTAP (NM_002451) located in the telomeric region and DMRTA1 (NM_022160) in the centromeric region (Fig. 3).

To develop a direct predictive test for this large French family with the melanoma-NST syndrome associated with a 403-kb germ-line deletion, additional primers (Δ403-U: 5′-CCAGTCTATCGTTGTTGGACAT-3′ and Δ403-L: 5′-TGTATTTGATGGCATTCCACA-3′) were designed to obtain a short PCR product (210 bp) in a simple and rapid assay designed to detect the junction fragment. Our results indicate that PCR-based detection of the junction fragment is an effective and direct method for the identification of subjects with an individual risk in this family (Fig. 4). Indeed, the three affected individuals tested yielded the 210-bp fragment, whereas the four healthy relatives tested did not.

Figure 4.

Figure 4. A, PCR detection of the 403-kb germ-line deletion. Only the affected relatives (III-16, III-23, and IV-20) yielded a 210-bp fragment (arrow) with the primers Δ403-U (5′-CCAGTCTATCGTTGTTGGACAT-3′) and Δ403-L (5′-TGTATTTGATGGCATTCCACA-3′), whereas the healthy relatives (II-9, III-10, III-14, and IV-9) yielded no PCR product, the wild-type allele being too large (403,441 bp) to be amplified. N, high-quality normal human genomic DNA. MW, molecular weight marker. B, PCR control of the amount of genomic DNA added to each reaction. Genomic DNAs were amplified with a primer pair located in exon 4 of the HFE gene (NM_000410), generating a 151-bp fragment (arrow) in all patients tested.

A, PCR detection of the 403-kb germ-line deletion. Only the affected relatives (III-16, III-23, and IV-20) yielded a 210-bp fragment (arrow) with the primers Δ403-U (5′-CCAGTCTATCGTTGTTGGACAT-3′) and Δ403-L (5′-TGTATTTGATGGCATTCCACA-3′), whereas the healthy relatives (II-9, III-10, III-14, and IV-9) yielded no PCR product, the wild-type allele being too large (403,441 bp) to be amplified. N, high-quality normal human genomic DNA. MW, molecular weight marker. B, PCR control of the amount of genomic DNA added to each reaction. Genomic DNAs were amplified with a primer pair located in exon 4 of the HFE gene (NM_000410), generating a 151-bp fragment (arrow) in all patients tested.

Figure 4.

Figure 4. A, PCR detection of the 403-kb germ-line deletion. Only the affected relatives (III-16, III-23, and IV-20) yielded a 210-bp fragment (arrow) with the primers Δ403-U (5′-CCAGTCTATCGTTGTTGGACAT-3′) and Δ403-L (5′-TGTATTTGATGGCATTCCACA-3′), whereas the healthy relatives (II-9, III-10, III-14, and IV-9) yielded no PCR product, the wild-type allele being too large (403,441 bp) to be amplified. N, high-quality normal human genomic DNA. MW, molecular weight marker. B, PCR control of the amount of genomic DNA added to each reaction. Genomic DNAs were amplified with a primer pair located in exon 4 of the HFE gene (NM_000410), generating a 151-bp fragment (arrow) in all patients tested.

A, PCR detection of the 403-kb germ-line deletion. Only the affected relatives (III-16, III-23, and IV-20) yielded a 210-bp fragment (arrow) with the primers Δ403-U (5′-CCAGTCTATCGTTGTTGGACAT-3′) and Δ403-L (5′-TGTATTTGATGGCATTCCACA-3′), whereas the healthy relatives (II-9, III-10, III-14, and IV-9) yielded no PCR product, the wild-type allele being too large (403,441 bp) to be amplified. N, high-quality normal human genomic DNA. MW, molecular weight marker. B, PCR control of the amount of genomic DNA added to each reaction. Genomic DNAs were amplified with a primer pair located in exon 4 of the HFE gene (NM_000410), generating a 151-bp fragment (arrow) in all patients tested.

Close modal

A search of the dbEST database, using the Gapped BLAST program (14), allowed us to identify several human expressed sequence tags (EST) of an unidentified transcript (Genbank accession nos. CN277071, AA807394, BF002332, BI765545, BX100299, AI825844, AW169296, and BC038540). Some of these ESTs contained a polyadenylation site, suggesting that this gene is transcribed in the direction opposite from the p15/CDKN2B-p16/CDKN2A-p14/ARF gene cluster. RT-PCR amplification of a cDNA obtained from normal human testis mRNA, using various primers placed in the identified ESTs, and sequence analysis of the PCR products, identified a large cDNA 3,834 bp long. This cDNA seems to be untranslated, as it does not possess a clear open reading frame. We chose to name this novel gene ANRIL, for “antisense noncoding RNA in the INK4 locus” (Genbank accession no. DQ485453). Alignment of the ANRIL 3,834-bp cDNA with genomic sequence led to the conclusion that the ANRIL gene contains 19 exons, with exon/intron junctions corresponding to the consensus sequences of donor/acceptor splice sites and a polyadenylation site in the last exon. The ANRIL genomic sequence spans a region of 126.3 kb. We also characterized a short alternatively spliced ANRIL mRNA species 2,659 bp long, composed of the first 12 ANRIL exons and an additional 3′ exon (located in intron 12), including a second polyadenylation site (Genbank accession no. DQ485454).

The ANRIL gene overlaps the two exons of p15/CDKN2B but does not share any nucleotide sequences (Fig. 5A). Moreover, the 5′ end of the first exon of the ANRIL gene is located about −300 bp upstream of the transcription start site of the p14/ARF gene, suggesting that these two genes may share a bidirectional promoter (Fig. 5A). Global transcriptome analysis recently provided evidence that the bulk of the mammalian genome can produce transcripts from both stands, with a large number of novel noncoding RNAs (15). The latter authors also showed the existence of a large number of transcription units (paired sense/antisense transcripts) in the mammalian genome and suggested that antisense transcription contributes to controlling transcriptional outputs (16). For example, these authors identified 1,092 coding-noncoding pairs overlapping head to head on the promoter in the mouse genome.

Figure 5.

Figure 5. A, genomic organization of the p15/CDKN2B-p16/CDKN2A-p14/ARF gene cluster. Boxes, location of exons (approximately to scale). Exons 1α, 2, and 3 of p16/CDKN2A encode p16 protein, whereas exon 1β, spliced to exons 2 and 3 of p16/CDKN2A in a different reading frame and transcribed using a different promoter, encodes p14ARF protein. The ANRIL gene overlaps the two exons of p15/CDKN2B and is transcribed in the orientation opposite to the p15/CDKN2B-p16/CDKN2A-p14/ARF gene cluster. Exon 1 of ANRIL is present about −300 bp upstream of the transcription start site of p14/ARF (exon 1β). B, compiled literature on physical and family deletion maps of the p15/CDKN2B-p16/CDKN2A-p14/ARF gene cluster region. Black rectangles, deleted regions in families A, B, C, D, E, and F. Gray parts of the rectangles, in families C and D, the limits of the candidate regions of the breakpoints.

A, genomic organization of the p15/CDKN2B-p16/CDKN2A-p14/ARF gene cluster. Boxes, location of exons (approximately to scale). Exons 1α, 2, and 3 of p16/CDKN2A encode p16 protein, whereas exon 1β, spliced to exons 2 and 3 of p16/CDKN2A in a different reading frame and transcribed using a different promoter, encodes p14ARF protein. The ANRIL gene overlaps the two exons of p15/CDKN2B and is transcribed in the orientation opposite to the p15/CDKN2B-p16/CDKN2A-p14/ARF gene cluster. Exon 1 of ANRIL is present about −300 bp upstream of the transcription start site of p14/ARF (exon 1β). B, compiled literature on physical and family deletion maps of the p15/CDKN2B-p16/CDKN2A-p14/ARF gene cluster region. Black rectangles, deleted regions in families A, B, C, D, E, and F. Gray parts of the rectangles, in families C and D, the limits of the candidate regions of the breakpoints.

Figure 5.

Figure 5. A, genomic organization of the p15/CDKN2B-p16/CDKN2A-p14/ARF gene cluster. Boxes, location of exons (approximately to scale). Exons 1α, 2, and 3 of p16/CDKN2A encode p16 protein, whereas exon 1β, spliced to exons 2 and 3 of p16/CDKN2A in a different reading frame and transcribed using a different promoter, encodes p14ARF protein. The ANRIL gene overlaps the two exons of p15/CDKN2B and is transcribed in the orientation opposite to the p15/CDKN2B-p16/CDKN2A-p14/ARF gene cluster. Exon 1 of ANRIL is present about −300 bp upstream of the transcription start site of p14/ARF (exon 1β). B, compiled literature on physical and family deletion maps of the p15/CDKN2B-p16/CDKN2A-p14/ARF gene cluster region. Black rectangles, deleted regions in families A, B, C, D, E, and F. Gray parts of the rectangles, in families C and D, the limits of the candidate regions of the breakpoints.

A, genomic organization of the p15/CDKN2B-p16/CDKN2A-p14/ARF gene cluster. Boxes, location of exons (approximately to scale). Exons 1α, 2, and 3 of p16/CDKN2A encode p16 protein, whereas exon 1β, spliced to exons 2 and 3 of p16/CDKN2A in a different reading frame and transcribed using a different promoter, encodes p14ARF protein. The ANRIL gene overlaps the two exons of p15/CDKN2B and is transcribed in the orientation opposite to the p15/CDKN2B-p16/CDKN2A-p14/ARF gene cluster. Exon 1 of ANRIL is present about −300 bp upstream of the transcription start site of p14/ARF (exon 1β). B, compiled literature on physical and family deletion maps of the p15/CDKN2B-p16/CDKN2A-p14/ARF gene cluster region. Black rectangles, deleted regions in families A, B, C, D, E, and F. Gray parts of the rectangles, in families C and D, the limits of the candidate regions of the breakpoints.

Close modal

To test the possible coordinated transcriptional regulation of ANRIL and p14/ARF, we quantified ANRIL, p14/ARF, p16/CDKN2A, and p15/CDKN2B mRNA in 22 normal human tissues (Supplementary Fig. S1_A_). The highest expression of ANRIL was observed in normal ovary, and the lowest expression was observed in skeletal muscle (100-fold lower than in ovary). ANRIL coclustered with p14/ARF, p16/CDKN2A, and p15/CDKN2B in these normal human tissues, with a stronger positive correlation between ANRIL and p14/ARF (Spearman rank correlation test: r = +0.949; P < 10−7) than between ANRIL and p16/CDKN2A (r = +0.573; P = 0.004) or between ANRIL and p15/CDKN2B (r = +0.561; P = 0.005). In a series of 12 breast tumor tissues showing a wide range of p14/ARF, p16/CDKN2A, and p15/CDKN2B mRNA levels, we also observed a stronger positive correlation of ANRIL with p14/ARF (Spearman rank correlation test: r = +0.965; P = 0.000001) than with p16/CDKN2A (r = +0.902; P = 0.00008) or p15/CDKN2B (r = +0.769; P = 0.004; Supplementary Fig. S1_B_). Finally, in an additional series of NF1-associated tumors in tissues derived from the embryonic neural crest (the Schwann cells are considered to be the progenitors of these tumors), we confirmed the positive link between the expression of ANRIL with p14/ARF as well as with p16/CDKN2A and p15/CDKN2B (data not shown) in a second tumor type.

These data suggest coordinated transcriptional regulation of ANRIL and p14/ARF, and possibly also p16/CDKN2A and p15/CDKN2B, in both physiologic and pathologic conditions.

At the moment, it is difficult to propose a mechanism of action for noncoding RNA. Indeed, the puzzle of noncoding RNA in eukaryotes is just starting to be assembled, and certainly, further unexpected pieces will be identified in the near future. To date, it likely seems that noncoding RNAs constitute a critical hidden layer of gene regulation in complex organisms, and their understanding requires new approaches in functional assays (17). We can hypothesize that ANRIL (antisense gene) could regulate p15/CDKN2B-p16/CDKN2A-p14/ARF locus (sense genes) by various novel mechanisms, such as RNA interference, gene cosuppression, gene silencing, chromatin structure maintenance, imprinting, and DNA demethylation (18). Moreover, we cannot rule out the possibility that ANRIL gene could be able to produce mature miRNAs (19) or encode a small peptide (20). Additional studies are thus necessary to elucidate the putative role via a genetic (or epigenetic) mechanism of ANRIL gene on the regulation (deregulation in tumorigenesis) of the p15/CDKN2B-p16/CDKN2A-p14/ARF locus.

Further studies of ANRIL and/or p14/ARF and/or p15/CDKN2B will be necessary to explain the additional tumor spectrum (i.e., NSTs) in this French family. Indeed, p15/CDKN2B and p14/ARF, like p16/CDKN2A, are altered during NST progression (2123), but no germ-line mutation of p15/CDKN2B or p14/ARF has been clearly implicated in inherited predisposition to NSTs (24). With regard to the ANRIL gene, we reanalyzed (Fig. 5B) the large 9p21 germ-line deletions previously reported in families with a hereditary predisposition to melanoma with or without NSTs (710). ANRIL was entirely deleted in our French family with NSTs (family F in Fig. 5B). ANRIL exon 1 was deleted in two U.K. families, one with and one without NSTs (families B and A, respectively), whereas its promoter was unambiguously deleted in one U.S. family without NSTs (families E) and probably deleted in another U.S. family with NSTs (families D). However, it could be hypothesized that these two U.S. families D and E, like the two U.K. families A and B (see ref. 9), could have an identical deletion of coancestral origin. Finally, the ANRIL gene was not deleted in a second U.K. family without NSTs (family C). Location of deletion end points of each large deletion reported in these different families needs to be fully characterized to further incriminate ANRIL as a putative candidate gene in melanoma-NST syndrome susceptibility.

The identification of this large antisense noncoding RNA could be important in cancer pathology as in hereditary predisposition to melanoma or in somatic alteration of the p15/CDKN2B-p16/CDKN2A-p14/ARF cluster observed in a large proportion of cancers, but also important in physiology area, the p15/CDKN2B-p16/CDKN2A-p14/ARF locus having a key role in the replicative senescence. Recent studies suggest that alteration of an antisense RNA can alter the expression of the corresponding sense mRNAs (25). Alteration of this noncoding RNA could be obtained by large deletion (including the entire ANRIL gene as in our family), deletion of one or several exons, deletion of a part of the promoter, splice sites mutations, aberrant DNA methylation (only in the somatic tumor), etc.

In conclusion, by applying STS real-time PCR-based gene dose mapping to a French melanoma-NST syndrome family, we identified a germ-line deletion encompassing a new putative transcriptional regulator gene (i.e., ANRIL) and the entire p15/CDKN2B-p16/CDKN2A-p14/ARF gene cluster. Our data suggest coordinated transcriptional regulation of ANRIL and p14/ARF (and, to a lesser extent, p16/CDKN2A and p15/CDKN2B) in both physiologic and pathologic conditions. Further studies are necessary to elucidate the role of the ANRIL gene in melanoma-NST syndrome families, in melanoma-prone families with no identified p16/CDKN2A mutations, and in somatic tumors.

Our new simple and rapid gene dose mapping assay should prove useful for characterizing other large germ-line deletions in research settings.

Acknowledgments

Grant support: Association pour la Recherche sur le Cancer and Ministère de l'Enseignement Supérieur et de la Recherche.

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

References

1

Hayward NK. Genetics of melanoma predisposition.

Oncogene

2003

;

22

:

3053

–62.

2

Lowe SW, Sherr CJ. Tumor suppression by Ink4a-Arf: progress and puzzles.

Curr Opin Genet Dev

2003

;

13

:

77

–83.

3

Rizos H, Darmanian AP, Holland EA, Mann GJ, Kefford RF. Mutations in the INK4a/ARF melanoma susceptibility locus functionally impair p14ARF.

J Biol Chem

2001

;

276

:

41424

–34.

4

Hewitt C, Lee Wu C, Evans G, et al. Germline mutation of ARF in a melanoma kindred.

Hum Mol Genet

2002

;

11

:

1273

–9.

5

Azizi E, Friedman J, Pavlotsky F, et al. Familial cutaneous malignant melanoma and tumors of the nervous system. A hereditary cancer syndrome.

Cancer

1995

;

76

:

1571

–8.

6

Kaufman DK, Kimmel DW, Parisi JE, Michels VV. A familial syndrome with cutaneous malignant melanoma and cerebral astrocytoma.

Neurology

1993

;

43

:

1728

–31.

7

Bahuau M, Vidaud D, Jenkins RB, et al. Germ-line deletion involving the INK4 locus in familial proneness to melanoma and nervous system tumors.

Cancer Res

1998

;

58

:

2298

–303.

8

Randerson-Moor JA, Harland M, Williams S, et al. A germline deletion of p14(ARF) but not CDKN2A in a melanoma-neural system tumour syndrome family.

Hum Mol Genet

2001

;

10

:

55

–62.

9

Mistry SH, Taylor C, Randerson-Moor JA, et al. Prevalence of 9p21 deletions in UK melanoma families.

Genes Chromosomes Cancer

2005

;

44

:

292

–300.

10

Laud K, Marian C, Avril MF, et al. Comprehensive analysis of CDKN2A (p16INK4A/p14ARF) and CDKN2B genes in 53 melanoma index cases considered to be at heightened risk of melanoma.

J Med Genet

2006

;

43

:

39

–47.

11

Bahuau M, Vidaud D, Kujas M, et al. Familial aggregation of malignant melanoma/dysplastic naevi and tumours of the nervous system: an original syndrome of tumour proneness. Familial aggregation of malignant melanoma/dysplastic naevi and tumours of the nervous system: an original syndrome of tumour proneness.

Ann Genet

1997

;

40

:

78

–91.

12

Bieche I, Parfait B, Le Doussal V, et al. Identification of CGA as a novel estrogen receptor-responsive gene in breast cancer: an outstanding candidate marker to predict the response to endocrine therapy.

Cancer Res

2001

;

61

:

1652

–8.

13

Behrmann I, Wallner S, Komyod W, et al. Characterization of methylthioadenosin phosphorylase (MTAP) expression in malignant melanoma.

Am J Pathol

2003

;

163

:

683

–90.

14

Altschul SF, Madden TL, Schäffer AA, et al. Gapped BLAST and PSI-BLAST: a new generation of protein database search programs.

Nucleic Acids Res

1997

;

25

:

3389

–402.

15

Carninci P, Kasukawa T, Katayama S, et al. The transcriptional landscape of the mammalian genome.

Science

2005

;

309

:

1559

–63.

16

Katayama S, Tomaru Y, Kasukawa T, et al. Antisense transcription in the mammalian transcriptome.

Science

2005

;

309

:

1564

–6.

17

Mattick JS. The functional genomics of noncoding RNA.

Science

2005

;

239

:

1527

–8.

18

Costa F. Non-coding RNAs: new players in eukaryotic biology.

Gene

2005

;

357

:

83

–94.

19

Calin GA, Croce CM. MicroRNA-cancer connection: the beginning of a new tale.

Cancer Res

2006

;

64

:

7390

–4.

20

Chooniedass-Kothari S, Emberley E, Hamedani MK, et al. The steroid receptor RNA activator is the first functional RNA encoding a protein.

FEBS Lett

2004

;

566

:

43

–7.

21

Jen J, Harper JW, Bigner SH, et al. Deletion of p16 and p15 genes in brain tumors.

Cancer Res

1994

;

54

:

6353

–8.

22

Izumoto S, Arita N, Ohnishi T, Hiraga S, Taki T, Hayakawa T. Homozygous deletions of p16INK4A/MTS1 and p15INK4B/MTS2 genes in glioma cells and primary glioma tissues.

Cancer Lett

1995

;

97

:

241

–7.

23

Bostrom J, Meyer-Puttlitz B, Wolter M, et al. Alterations of the tumor suppressor genes CDKN2A (p16(INK4a)), p14(ARF), CDKN2B (p15(INK4b)), and CDKN2C (p18(INK4c)) in atypical and anaplastic meningiomas.

Am J Pathol

2001

;

159

:

661

–9.

24

Gao L, Liu L, van Meyel D, et al. Lack of germ-line mutations of CDK4, p16(INK4A), and p15(INK4B) in families with glioma.

Clin Cancer Res

1997

;

3

:

977

–81.

25

Wahlestedt C. Natural antisense and noncoding RNA transcripts as potential drug targets.

Drug Discov Today

2006

;

11

:

503

–8.

©2007 American Association for Cancer Research.

2007

Supplementary data

1,345 Views

518 Web of Science

Citing articles via

Email alerts