ARTICLES Epigenetic Regulation Identifies RASEF as a Tumor-Suppressor in Uveal Melanoma

Willem Maat,1 Sigrid H. W. Beiboer,2 Martine J. Jager,1 Gre´ P. M. Luyten,1 Nelleke A. Gruis,3 and Pieter A. van der Velden1,3

5 PURPOSE. Recently, a segregation study in families with uveal Recently, Jo¨nsson et al. revealed a genetic component in three and cutaneous melanoma identified 9q21 as a potential locus such families, in which members are affected by either uveal or harboring a tumor-suppressor gene (TSG). One of the in cutaneous melanoma. Linkage analysis in these families identi- this area, RASEF, was then analyzed as a candidate TSG, but fied a potential uveal melanoma susceptibility locus on chro- lack of point mutations and copy number changes could not mosome 9, area q21. confirm this. In this study, the RASEF gene was investigated for This locus has a long history in melanoma that started with potential mutations and gene silencing by promoter methyl- detection of isochromosome 9q with cytogenetic analysis.6,7 ation in uveal melanoma. Loss of heterozygosity (LOH) of markers at 9q22 was subse- ETHODS quently frequently reported and was shown to be associated M . Eleven uveal melanoma cell lines and 35 primary 8,9 uveal melanoma samples were screened for mutations in the with proliferation and tumor progression. Recently, single RASEF gene by high-resolution melting-curve and digestion nucleotide polymorphism (SNP) analysis has confirmed the analysis. Expression of RASEF was determined by real-time LOH of this locus in melanoma, while genome-wide analysis in dizygotic twins for nevi numbers also showed linkage with this RT-PCR in all cell lines and 16 primary uveal melanoma sam- 10,11 ples, and the status of the promoter of the RASEF 9q region. In addition, a gene slightly distal to RASEF, gene was analyzed and confirmed by direct sequencing. RMI1, has recently been shown to be a risk factor for cutane- ous melanoma, whereas the locus for familial melanoma sus- RESULTS. Mutation screening revealed a known polymorphism 12,13 3 ceptibility is located on the short arm of 9. (R262C; C T) in exon 5 of the RASEF gene that displayed a Cutaneous melanomas are often characterized by loss of the normal frequency (54%). Of the primary uveal melanomas, 46% cell-cycle regulator p16 and/or activation of the RAS/RAF/ERK presented a heterozygous genotype, and 10 (91%) of 11 cell pathway.14,15 These hallmarks of melanoma are also recog- lines showed a homozygous genotype. Melting-curve analysis nized in uveal melanoma, although the underlying mechanisms indicated loss of heterozygosity in at least two primary tumors. differ.16,17 Whereas in cutaneous melanoma, p16 is commonly Low RASEF expression in the cell lines and primary tumors lost by chromosomal deletion of the CDKN2A gene, the pref- correlated with methylation of the RASEF promoter region. erential mechanism in uveal melanoma appears to be silencing Homozygosity and methylation of the RASEF gene in primary 18 ϭ of the p16-encoding CDKN2A promoter by methylation. Mu- tumors were associated with decreased survival (P 0.019). tations in BRAF, NRAS,orc-kit lead to constitutive ERK acti- CONCLUSIONS. Homozygosity, in combination with methylation, vation in most cutaneous melanomas.19,20 However, mutations appears to be the mechanism targeting RASEF in uveal mela- in BRAF have only rarely been reported in uveal melanoma, noma, and allelic imbalance at this locus supports a TSG role whereas activating NRAS and c-kit mutations have never been for RASEF.(Invest Ophthalmol Vis Sci. 2008;49:1291–1298) reported.21 Still, ERK activation is also present in uveal mela- DOI:10.1167/iovs.07-1135 noma, and this knowledge leads to the question of what causes ERK activation in the absence of activating mutations in BRAF, veal melanoma is the most common primary intraocular NRAS,orc-kit.16,17,21 Uneoplasm in adults, with an annual incidence of six to The RASEF (RAS and EF hand domain containing) gene is eight per million in Caucasian populations.1 In contrast to located on , area q21, and encodes a cutaneous melanoma, clustering of uveal melanoma in families with calcium-binding EF-hand and Ras GTPase (Rab family) is extremely rare.2–4 Occurrence of both uveal melanoma and motifs (http://www.genome.ucsc.edu/ provided in the public cutaneous melanoma in a single family has been observed.4 domain by the Genome Bioinformatics Group, University of Santa Cruz, CA); it is also known as RAB45 or FLJ31614.22 Based on the functional domains in RASEF, the gene product may be engaged in the RAS pathway and in combination with 1 3 From the Departments of Ophthalmology and Dermatology, evidence for linkage of the RASEF region with cutaneous and Leiden University Medical Center (LUMC), Leiden, The Netherlands; uveal melanoma, molecular analysis of this gene is warranted. and 2Hogeschool Leiden, Leiden, The Netherlands. In line with the analysis of cutaneous melanoma reported Supported by Dutch Cancer Society (KWF) Grant RUL 2001-2472. 5 Submitted for publication August 31, 2007; revised November 9, by Jo¨nsson et al., we therefore set out to analyze RASEF for 2007; accepted February 18, 2008. mutations and for expression of the gene in uveal melanoma. Disclosure: W. Maat, None; S.H.W. Beiboer, None; M.J. Jager, None; G.P.M. Luyten, None; N.A. Gruis, None; P.A. van der Velden, None MATERIALS AND METHODS The publication costs of this article were defrayed in part by page charge payment. This article must therefore be marked “advertise- Cell Lines and Primary Uveal ment” in accordance with 18 U.S.C. §1734 solely to indicate this fact. Melanoma Specimens Corresponding author: Pieter A. van der Velden, Skin Research Lab, Department of Dermatology, Leiden University Medical Center, In total, 11 cell lines derived from primary uveal melanomas (92.1; PO Box 9600, 2300 RC Leiden, The Netherlands; [email protected]. OCM-1, -3, and -8; and Mel-202, -270, -285, and -290) and uveal mela-

Investigative Ophthalmology & Visual Science, April 2008, Vol. 49, No. 4 Copyright © Association for Research in Vision and Ophthalmology 1291

Downloaded from iovs.arvojournals.org on 10/02/2021 1292 Maat et al. IOVS, April 2008, Vol. 49, No. 4

TABLE 1. Tumor Characteristics and Survival Data of 35 Uveal Melanoma Patients Sorted by Methylation Status and RASEF Genotype

Survival Hypermethylated Tumor ID Cell Type (mo) Present Status RASEF Genotype

UM1 Spindle 210 Alive Not present Hom C UM16 Epithelioid 115 Alive Not present Hom C UM29 Spindle 24 Died, due to metastases Not present Hom C UM3 Mixed 95 Died, due to metastases Not present Hom C UM33 Mixed 24 Died, due to metastases Not present Hom C UM12 Epithelioid 137 Died, due to metastases Not present Hom T UM19 Spindle 15 Died, due to metastases Not present Hom T UM26 Spindle 122 Alive Not present Hom T UM31 Mixed 34 Died, due to metastases Not present Hom T UM32 Epithelioid 63 Died, due to metastases Not present Hom T UM4 Spindle 31 Died, due to metastases Not present Hom T UM6 Epithelioid 57 Died, due to metastases Not present Hom T UM27 Mixed 23 Died, due to metastases Present Hom C UM28 Epithelioid 33 Died, due to metastases Present Hom C UM30 Spindle 113 Alive Present Hom C UM11 Mixed 13 Died, due to metastases Present Hom T UM18 Spindle 12 Died, due to metastases Present Hom T UM5 Spindle 50 Died, due to metastases Present Hom T UM35 Mixed 94 Alive Present Het/homT* UM21 Epithelioid 167 Died, due to metastases Present Het/homT* UM13 Epithelioid 30 Died, due to metastases Present Het/homT* UM15 Mixed 23 Died, due to metastases Present Het UM17 Epithelioid 33 Died, due to metastases Present Het UM2 Mixed 29 Died, due to metastases Not present Het UM14 Mixed 42 Died, due to metastases Not present Het UM25 Mixed 29 Died, other cause Not present Het UM7 Mixed 63 Died, other cause Not present Het UM23 Spindle 152 Died, unknown cause Not present Het UM22 Spindle 2 Lost to follow up Not present Het UM8 Spindle 191 Alive Not present Het UM9 Mixed 131 Alive Not present Het UM10 Mixed 136 Alive Not present Het UM20 Mixed 187 Alive Not present Het UM24 Mixed 143 Alive Not present Het UM34 Mixed 106 Alive Not present Het

* Loss of heterozygosity/allelic imbalance.

noma metastases (OMM-1, -2.3, and -2.5) were analyzed. All melanoma Zymo Research Corp., Orange, CA). Enzymatically methylated human cell lines were cultured in RPMI 1640 medium (Invitrogen-Gibco, DNA (Chemicon Europe Ltd., Hampshire, UK) was used as the positive Paisley, Scotland, UK) supplemented with 3 mM L-glutamine (Invitro- control in all experiments. DNA and RNA concentrations were deter- gen-Gibco), 2% penicillin-streptomycin, and 10% FBS (Hyclone, Logan, mined by spectrophotometer (model ND-1000; NanoDrop Technolo-

UT). All cell cultures were incubated at 37°C in a humidified 5% CO2 gies Inc., Wilmington, DE). atmosphere. Archival frozen tumor specimens of primary uveal mela- noma came from 35 patients who attended the Leiden University Mutation Screening and Genotyping Medical Center between 1988 and 1996. All tumors were primary A 96-well light scanner (Idaho Technologies Inc., Salt Lake City, UT) for lesions with a tumor diameter greater than 12 mm, a prominence high-resolution melting-curve analysis was used to scan all amplicons greater than 6 mm, and no treatment before enucleation. The validity of the RASEF gene. The primers are shown in Table 2. DNA samples of the diagnosis of uveal melanoma was confirmed histologically in all were amplified with a double-stranded DNA-binding dye (LC Green cases, and clinical and survival data were listed for use in the study Plus; Idaho Technologies). Melting curves were analyzed in plots (Table 1). The research protocol followed the tenets of the current showing differences in fluorescence. The shift and curve shapes of version of the Declaration of Helsinki (World Medical Association melting profiles were used to distinguish between samples from con- Declaration of Helsinki 1964; Ethical Principles for Medical Research trol subjects and patients. PCR reaction with the green dye (LC Green; Involving Human Subjects). Idaho Technologies) contained PCR buffer (Invitrogen, Breda, The Netherlands), 1.5 mM MgCl ,40␮M dNTPs, 1:10 diluted green dye (LC DNA and RNA Extraction and 2 Green; Idaho Technologies), 0.4 ␮M of forward and reverse primers, Sodium-Bisulfite Modification and 1 unit Taq polymerase per 10-␮L reaction (Fast Start; Roche Using a column-based extraction kit (Genomic tip 100/G; Qiagen Diagnostics BV, Almere, The Netherlands). PCR consisted of an initial Benelux BV, Venlo, The Netherlands), DNA was extracted from the cell denaturation at 94°C for 6 minutes followed by 40 cycles consisting of lines and frozen tumor material, according to the manufacturer’s guide- 15 seconds at 96°C, 30 seconds at 58°C, and 60 seconds at 72°C, and lines. RNA was also extracted with a column-based extraction kit the PCR ended with a 1-minute denaturation at 94°C. After amplifica- (RNeasy mini kit; Qiagen Benelux) from tumors in which enough tion, the amplified fragments (exon 5) were digested using 4 units of frozen material was available (n ϭ16). RNA was converted to cDNA the restriction enzyme BstU1 (New England Biolabs, Beverly, MA) (iScript cDNA synthesis kit; Bio-Rad Laboratories BV, Veenendaal, the directly added to the PCR mixture. Analysis was performed by over- Netherlands), according to the manufacturer’s guidelines. Genomic night digestion of the amplified fragments at 60°C. The BstU1 enzyme DNA was modified with sodium bisulfite (EZ Methylation Gold kit; recognizes and cleaves the 5Ј-CG∧CG-3Ј sequence. PCR products were

Downloaded from iovs.arvojournals.org on 10/02/2021 IOVS, April 2008, Vol. 49, No. 4 RASEF as a Tumor-Suppressor Gene in Uveal Melanoma 1293

TABLE 2. RASEF Primers Used for Mutation Detection, Expression Analysis, and Methylation Analysis

Primer Sequence

Exon 1 Forward 3Ј-GGCAAGCAGCGGTGGACTC-5Ј Reverse 5Ј-GTAGGTGAAGGAAGACAAGCAACTC-3Ј Exon 2 Forward 3Ј-TCTTCCCTTCCTTCCGTTCATTCTG-5Ј Reverse 5Ј-GTCCACCTATATCATAGTGTGACAATGC-3Ј Exon 3 Forward 3Ј-TTCTCTTCATCTGTAATATATAGGGCTTAACG-5Ј Reverse 5Ј-CCCTCTCCGTAGAAACCACCTC-3Ј Exon 4 Forward 3Ј-TCACCTTCCCTGTGTAGGAGAAC-5Ј Reverse 5Ј-CTGAGATGCTGAGGCTGTTCC-3Ј Exon 5 Forward 3Ј-CAAAGCAATTCAAAGTGAGTTTGTAAGC-5Ј Reverse 5Ј-TGAGGATGTGGTCTAACAGGAAGTG-3Ј Exon 6 Forward 3Ј-GTGTGGGAGGGTGACAGGAC-5Ј Reverse 5Ј-AAATCATTAGAAAGTAAAGAAGATATTAGCAAAG-3Ј Exon 7 Forward 3Ј-AAAGGGTCTGGGAGGGTAGG-5Ј Reverse 5Ј-AAACAAGTGAAATGTAAATGTAATGAGC-3Ј Exon 8 Forward 3Ј-CCCAATGATACTTTCCTTGTCTCTCTTTC-5Ј Reverse 5Ј-ACTTACTTGAGGCTCTCCTTTAAGAAATTAC-3Ј Exon 9 Forward 3Ј-TAGTTACATTAGAAGTTTGAGTAGTGTGC-5Ј Reverse 5Ј-TTAACATACCTGTCATAGCCTAGAGG-3Ј Exon 10 Forward 3Ј-AGCCCTCAGGTAAATTGGTCTTCC-5Ј Reverse 5Ј-TGACAGATAGAAGGCAAATAAGGTGAC-3Ј Exon 11–12 Forward 3Ј-TGACATAAGGGATGAAGAGACATTTGG-5Ј Reverse 5Ј-TTATCAACCGAAATACGAGCCATACC-3Ј Exon 13 Forward 3Ј-CAATGGAATTATTTACATCGTGCTCTC-5Ј Reverse 5Ј-TTTGAGTATGAAGAACATCAAGTGG-3Ј Exon 14 Forward 3Ј-GGCAACACAAACTGACTGATGATG-5Ј Reverse 5Ј-TTTCTGTTTCTCCATTATGATTTCTTACCTC-3Ј Exon 15 Forward 3Ј-TGTTGCTGTTGTTCTGTGGTCATC-5Ј Reverse 5Ј-ACCGACTTCAAAGCCATTAAACCC-3Ј Exon 16 Forward 3Ј-AAGGGCTTCATTTAATTGTGTGTATTTC-5Ј Reverse 5Ј-CCACCATGACTGACAGATAAGAGAG-3Ј Exon 17 Forward 3Ј-TATGAAGATTAAGTCAAGACCTATAAAGC-5Ј Reverse 5Ј-GACTTTGTGGGTAACCTAATTCAGC-3Ј RASEF QPCR Forward 3Ј-ATCAGACTTCAAAGCACAGAAATGG-5Ј RASEF QPCR Reverse 5Ј-TTCCTCTTCCAACTCACTCAACTG-3Ј RASEF Bisulfite Forward 3Ј-GGGATGGAGGCGGATGGG-5Ј RASEF Bisulfite Reverse 5Ј-CCGCAACTCCGTACACAATACC-3Ј RASEF PAP Forward 5Ј-GGACGGAGAGGAGTTGGTTCGGTTG-ddC-3Ј RASEF PAP Reverse 5Ј-CCGCAACTCCGTACACAATACCCGAAA-ddC-3Ј

separated on a 2% agarose gel in 1ϫ TBE (0.09 M Tris-borate, 0.002 M Melting-Temperature Analysis EDTA; pH 8.2). A melting-temperature analysis was performed as described earlier.23

RASEF Expression Restriction Digestion Analysis and The expression of RASEF in 16 tumors for which RNA was available Sequence Analysis was analyzed with real-time RT-PCR and specific primers, which are After amplification with specific primers for bisulfite-converted DNA, 23 shown in Table 2. PCR was performed as described earlier. the PCR-amplified fragments were digested with 4 units of restriction enzyme HinfI (Fermentas GmbH, St. Leon Rot, Germany) directly Methylation Analysis added to the PCR mixture (under conditions specified by the manu- facturer). The HinfI enzyme recognizes and cleaves the 5Ј-G∧ANTC-3Ј We applied bisulfite modification of tumor DNA in combination with sequence. This sequence is not present in unmodified DNA and in PCR, as this introduces sequence differences between methylated and modified unmethylated DNA. The RASEF amplicon of methylated DNA unmethylated DNA that can be analyzed with several methods. The contains one HinfI recognition site and is dependent on both CT sequence differences were initially determined with melting-tempera- conversion and methylation of a CpG. The recognition site 5Ј- ture analysis, as this method provides both quantitative and qualitative G∧ANTC-3Ј appears only when the first C in the 5Ј-GANCC-3Ј se- measures of methylation (see Fig. 3). The methylation status of the quence is converted into thymine, whereas the second must be meth- RASEF promoter region was determined by polymerase chain reaction ylated and remains a cytosine. PCR products were separated on a 2% with specific primers and by melting-temperature analysis and was agarose gel in 1ϫ TBE (0.09 M Tris-borate, 0.002 M EDTA; pH 8.2). further validated with a restriction digestion analysis. DNA bands were excised from the gel, purified using a gel extraction Primers were designed on computer (Beacon Designer Software kit (Nucleospin Extract II; Macherey-Nagel, GmbH, Du¨ren, Germany) ver. 5.0; Premier Biosoft International, Palo Alto, CA) using bisulfite- and sequenced (Prism 3700 DNA sequencing system; Applied Biosys- converted DNA sequences and amplified a CpG island in the RASEF tems, Foster City, CA). gene promoter (Consensus CDS [Coding Sequence] accession number 6662.1; Gene ID 158158/ http://www.ncbi.nlm.nih.gov/ a genome database hosted by the National Center for Biotechnology Information, Pyrophosphorolysis-Activated Polymerization Bethesda, MD). The primers are shown in Table 2. PCR was performed In the pyrophosphorolysis-activated polymerization (PAP) reaction, exactly as described earlier.23 primers are used that contain a dideoxy-nucleotide (ddNTP) at their 3Ј

Downloaded from iovs.arvojournals.org on 10/02/2021 1294 Maat et al. IOVS, April 2008, Vol. 49, No. 4

FIGURE 1. (A) Melting-curve analy- sis of the RASEF R262C polymor- phism in control and primary uveal melanoma and cell lines. Red: het- erozygous control and tumor sam- ples; blue: heterozygous primary uveal melanoma samples with a low- ered difference plot that is not seen in control samples. (B) A calibration curve was created with dilutions of the T allele in a constant background of the C allele. Based on this curve it was estimated that the relative abun- dance of the alleles in the primary uveal melanoma samples 13 and 21 was decreased at least 10-fold. (C–E) RASEF exon 5 sequence analysis of primary uveal melanomas shows the R262C polymorphism at position 37. The cytosine from the consensus se- quence is substituted for a thymine.

terminus and hence will not be extended. A polymerase with pyro- quently in the population, with reported frequencies between phosphorolysis activity can remove the dideoxy-cytosine and thereby 50% and 58% of the Caucasian population.22 In 10 of the 11 cell activate polymerization. Since this pyrophosphorolysis activity is de- lines, a homozygous genotype of the T allele was observed. pendent on double-stranded DNA, only primers that perfectly match The primary uveal melanomas (n ϭ 35) displayed a normal the template will be activated. The specificity of pyrophosphorolysis frequency of the SNP (54%), with 16 uveal melanomas present- allows us to amplify specifically the minute amounts of methylated ing a heterozygous genotype (Table 1). However, both melting- RASEF DNA in the background of unmethylated DNA. The PAP prod- curve analysis and restriction enzyme analysis revealed imbal- ucts can be further validated with sequence analysis for internal CpGs. ance of the alleles in two samples. Whereas in gel analysis of The primers are shown in Table 2. The amplification was performed in BstUI digested RASEF, exon 5 PCR indicated the presence of a final volume of 25 ␮L containing 5 ␮L5ϫ PAP buffer (prepared as the C-allele, melting-curve analysis indicated that the relative described by Liu and Sommer.24), 0.3 ␮L (10 picomoles/␮L) of each concentration of the C-allele was at least 10-fold lower than the primer, 0.5 ␮L Taq polymerase (KLENtaq; DNA Polymerase Technol- T-allele in UM13 and -21 (Fig. 1). ␮ ␮ 25 ogy, Inc., St. Louis, MO), 17.9 LH2O, and 1 L DNA sample. Amplification was initiated by hot start, followed by 40 cycles at 94°C Expression analysis for 15 seconds, 60°C for 40 seconds, 64°C for 40 seconds (T-anneal); 68°C for 40 seconds (pyrophosphorolysis activity); and 72°C for 40 The allelic imbalance observed in the primary uveal mela- seconds (elongation), and using a final melting curve from 70°C to noma was followed up by RASEF RT-PCR expression analy- 97°C with an increase in temperature of 0.2°C every 10 seconds. PCR sis. In the cell lines, two groups were distinguishable, based products were separated on a 2% agarose gel in 1ϫ TBE (0.09 M on expression levels. In 6 of 11 uveal melanoma cell lines Tris-borate, 0.002 M EDTA; pH 8.2). (92.1; Mel 202 and -270; and OMM-1, -2.3, and -2.5) an approximately 30- to 100-fold reduced expression of RASEF Statistical Analysis was displayed compared with the other cell lines (OCM1, -3, Survival analysis for RASEF promoter methylation was performed using and -8; Mel285 and -290; Fig. 2). Cell lines Mel270 and a Kaplan Meier analysis and log rank test (SPSS ver. 14.0 for Windows; OMM-2.3 and -2.5 are derived from the same patient and fell SPSS Inc., Chicago, IL). For a comparison between the presence or into the same group. Among the uveal melanomas cell lines absence of RASEF methylation and metastatic disease and tumor char- with low RASEF expression, a homozygous (TT) genotype acteristics, the ␹2 test and analysis of variance were performed. prevailed. When primary tumors were analyzed, variable levels of RASEF expression were also observed, but cluster- ing into two groups was not as marked, and the absolute RESULTS expression levels differed even more. One sample failed in the expression analysis (UM15). Correlating the expression Mutation Screening in Uveal Melanoma levels with the genotypes of the tumors revealed that the To analyze RASEF as a tumor-suppressor gene (TSG) candidate, homozygous tumors tended to present a lower RASEF ex- we first investigated the gene for mutations. Mutation screen- pression (P ϭ 0.015), as was the case in one tested uveal ing was performed in two steps: first, the 17 exons were melanoma presenting allelic imbalance (UM13). prescreened by high-resolution melting-curve analysis. Though melting-curve analysis showed few variations, we nevertheless Methylation Analysis generated sequences for 2 tumor samples of each exon both sequenced with the forward and the backward PCR primer. Because we did not detect mutations that could explain the We detected a sequence variation, which was a known poly- low RASEF expression in the primary uveal melanomas and the morphism in exon 5 of RASEF encoding a R262C (C3T; cell lines, we considered epigenetic regulation as the possible Arginine3Cysteine) substitution (Fig. 1). This SNP occurs fre- mechanism of downregulation. All five RASEF-expressing cell

Downloaded from iovs.arvojournals.org on 10/02/2021 IOVS, April 2008, Vol. 49, No. 4 RASEF as a Tumor-Suppressor Gene in Uveal Melanoma 1295

FIGURE 2. Expression analysis for RASEF in cell lines and primary uveal melanoma (UM), measured with real- time RT-PCR. Expression was nor- malized with the control gene RPS11. The change (x-fold) of ex- pression is calculated compared with the median expression level. The RASEF genotypes of the samples are indicated in the graph. UM13 dis- plays loss of the C allele indicated by a lowercase c. UM15 failed in the expression analysis.

lines contained an unmethylated promoter while hypermeth- ples 15 (failed in expression analysis) and 21 displayed the ylation of all CpGs within the amplicon was present in all six highest methylation but also contained an equimolar level of cell lines that lacked RASEF expression. The analysis of meth- unmethylated RASEF. In the other uveal melanoma samples ylation with melting temperature was confirmed by sequence with methylated RASEF, a minor fraction of the CpGs was analysis (Fig. 3). In primary uveal melanomas, methylation was methylated. Still, there was a correlation between methylation much more heterogeneous and never reached the level of and expression of RASEF in the primary tumor samples, al- methylation observed in the cell lines. Uveal melanoma sam- though not as obvious as in the cell lines.

FIGURE 3. (A) Melting-temperature analysis of amplified RASEF product reveals the methylation of primary uveal melanoma samples and cell lines. Blue: methylated samples; green and purple: samples with a mixed methylation pattern; red: un- methylated samples. (B) Methylation in the promotor region of the RASEF gene in primary uveal melanoma sample 25, as shown by sequence analysis after PAP. After bisulfite treatment and PCR, unmethylated cy- tosines converted into thymidine. Methylated cytosines remained unchanged.

Downloaded from iovs.arvojournals.org on 10/02/2021 1296 Maat et al. IOVS, April 2008, Vol. 49, No. 4

FIGURE 4. Kaplan-Meier analysis and log rank test showed the difference in survival according to genotype and presence or absence of methylation of the promotor region of the RASEF gene (P ϭ 0.019).

To validate methylation in primary tumors, we used restric- In line with the findings of Jo¨nsson et al.,5 we did not detect tion-enzyme analysis. By HinfI digestion, we were able to any mutations in the RASEF gene other than a known SNP.5,22 confirm RASEF methylation in primary uveal melanoma (data Using this SNP, we detected allelic imbalance in some of the not shown). Next, we set out to isolate the methylated fraction tumors that were heterozygous for this marker (UM13 and -21). and applied PAP. By applying PAP, we were able to show Because the imbalances were not complete, we suspect tumor completely methylated alleles and thereby validate melting- heterogeneity in the primary tumors in contrast to the cell temperature analysis in five tumors that had shown a methyl- lines, all of which, with one exception, displayed a homozy- ated fraction in the background of unmethylated DNA. In one gous genotype. Gene expression analysis revealed that 5 of 11 sample, a methylated allele was detected in a tumor that had uveal melanoma cell lines had high RASEF expression, whereas shown a normal curve with melting-temperature analysis, sug- the others hardly showed expression. As almost all the low- gesting a very low level of the methylated allele (Fig. 3). expression cell lines displayed the homozygous T-allele, there appears to be an association between expression and geno- Survival type. This apparent association, however, could also be based on the small number of cell lines that were tested and the fact The mean follow-up of the 35 patients was 78 months (2–210 that three cell lines were derived from the same patient (Mel months), and 20 patients had died of tumor-related metastasis 270, OMM 2.3, and OMM 2.5). In the primary tumors, expres- at the time of analysis. Two patients had died of a metastasis sion varied widely and often exceeded the expression seen in from another primary tumor (UM7 and UM25), one patient was the cell lines. Among the uveal melanomas with low RASEF lost to follow-up (UM22) after 2 months, and two patients had expression a homozygous genotype prevailed, but this fact died of unknown causes. The presence of methylation within does not favor a specific allele. This finding may indicate that the RASEF promoter region correlated with death due to met- there is no risk factor linked to either allele and that the low astatic disease (P ϭ 0.024; log rank test). The genotype of the expression is more likely due to a somatic alteration. As we had 35 tumors did not correlate to cell type, methylation status, or not observed any mutations in the cell lines, we subsequently the development of metastatic disease (P ϭ 0.441; Pearson ␹2). RASEF Although the genotype itself was not associated with met- considered epigenetic modifications as the cause of low astatic death, patients with a homozygous genotype and meth- expression. Indeed, all cell lines that did not express RASEF ylation of the RASEF gene (n ϭ 7) had a significantly higher contained a methylated promoter, whereas all cell lines with risk of development of metastasis than did patients with a expression lacked this methylation, confirming our hypothesis. heterozygous genotype and no methylation (survival 51 Ϯ 15.5 Hereafter, we performed demethylation experiments with vs. 161 Ϯ 19.0 months; P ϭ 0.019). 5-azacytidine, which revealed a highly induced expression in a cell line with methylated RASEF. Demethylation of an unmeth- ylated cell line resulted in the opposite effect. The demethyl- DISCUSSION ating agent is highly toxic and may explain downregulation of RASEF expression in the unmethylated cell line. Toxicity of Linkage analysis in uveal and cutaneous melanoma families 5-azacytidine and demethylation of all the other genes during identified the 9q21 region as a locus for a potential TSG treatment are the reasons that we reserve functional analysis involved in the development of melanoma. In addition, LOH using genetically modified cell lines for follow-up research. analysis in two uveal melanomas from members of the families The primary uveal melanomas displayed heterogeneity for in which linkage was identified indicated 9q21 to be the RASEF methylation but never reached levels above ϳ50% possible region for a TSG.5 The 9q21 region harbors the RASEF methylation, and most commonly only a part of the CpGs gene, which is potentially involved in the RAS pathway prom- present in the promotor region was methylated. Furthermore, inent in the development of melanoma.26,27 As patients with methylation not only coincided with low expression but also melanoma from the family just mentioned had been analyzed with a homozygous genotype, which suggests a combination for RASEF mutations, we set out to analyze sporadic uveal of methylation and LOH being the mechanism of loss of ex- melanoma and uveal melanoma cell lines. pression. The additional effect of LOH seems to be associated

Downloaded from iovs.arvojournals.org on 10/02/2021 IOVS, April 2008, Vol. 49, No. 4 RASEF as a Tumor-Suppressor Gene in Uveal Melanoma 1297

References 1. Egan KM, Seddon JM, Glynn RJ, Gragoudas ES, Albert DM. Epide- miologic aspects of uveal melanoma. Surv Ophthalmol. 1988;32: 239–251. 2. Kodjikian L, Nguyen K, Lumbroso L, et al. Familial uveal melanoma: a report on two families and a review of the literature. Acta Ophthalmol Scand. 2003;81:389–395. 3. Smith JH, Padnick-Silver L, Newlin A, Rhodes K, Rubinstein WS. Genetic study of familial uveal melanoma: association of uveal and cutaneous melanoma with cutaneous and ocular nevi. Ophthal- mology. 2007;114:774–779. 4. van Hees CL, Jager MJ, Bleeker JC, Kemme H, Bergman W. Occur- rence of cutaneous and uveal melanoma in patients with uveal melanoma and their first degree relatives. Melanoma Res. 1998;8: 175–180. 5. Jo¨nsson G, Bendahl PO, Sandberg T, et al. Mapping of a novel ocular and cutaneous malignant melanoma susceptibility locus to chromosome 9q21.32. J Natl Cancer Inst. 2005;97:1377–1382. 6. Kopf I, Stierner U, Islam Q, Delle U, Kindblom LG, Martinsson T. Characterization of four melanoma cell lines with electron micros- copy, immunocytochemistry, cytogenetics, flow cytometry, and southern analysis. Cancer Genet Cytogenet. 1992;62:111–123. 7. Albino AP, Sozzi G, Nanus DM, Jhanwar SC, Houghton AN. Malig- nant transformation of human melanocytes: induction of a com- plete melanoma phenotype and genotype. . 1992;7: 2315–2321. 8. Kumar R, Smeds J, Lundh RB, Hemminki K. Loss of heterozygosity at chromosome 9p21 (INK4–p14ARF locus): homozygous dele- tions and mutations in the p16 and p14ARF genes in sporadic primary melanomas. Melanoma Res. 1999;9:138–147. 9. Boni R, Vortmeyer AO, Burg G, Hofbauer G, Zhuang Z. The PTEN tumour suppressor gene and malignant melanoma. Melanoma Res. 1998;8:300–302. 10. Zhu G, Montgomery GW, James MR, et al. A genome-wide scan for FIGURE 5. Digestion analysis of DNA from a patient in whom the naevus count: linkage to CDKN2A and to other chromosome tumor (UM35, right) showed a nearly complete homozygous T-allele regions. Eur J Hum Genet. 2007;15:94–102. genotype revealed a heterozygous genotype in leukocyte DNA (left), 11. Stark M, Hayward N. Genome-wide loss of heterozygosity and confirming the mechanism of LOH in tumor tissue. copy number analysis in melanoma using high-density single-nu- cleotide polymorphism arrays. Cancer Res. 2007;67:2632–2642. 12. Broberg K, Hoglund M, Gustafsson C, et al. Genetic variant of the with the aggressiveness of the tumor, because homozygous human homologous recombination-associated gene RMI1 (S455N) tumors with a methylated RASEF promoter region tended to have impacts the risk of AML/MDS and malignant melanoma. Cancer a decreased survival compared with heterozygous tumors with- Lett. 2007;258:38–44. out methylation (P ϭ 0.019; Fig. 4). To confirm the suggested 13. Cannon-Albright LA, Goldgar DE, Meyer LJ, et al. Assignment of a mechanism, we compared the RASEF homozygous genotype of locus for familial melanoma, MLM, to chromosome 9p13–p22. four tumors with the genotype of their peripheral blood leu- Science. 1992;258:1148–1152. kocytes, also obtained at time of enucleation. One person, in 14. Cohen C, Zavala-Pompa A, Sequeira JH, et al. Mitogen-activated whom the tumor showed a nearly complete homozygous T- protein kinase activation is an early event in melanoma progres- allele genotype, revealed a heterozygous genotype in leukocyte sion. Clin Cancer Res. 2002;8:3728–3733. DNA, confirming the mechanism of LOH in tumor tissue (Fig. 15. Satyamoorthy K, Li G, Gerrero MR, et al. Constitutive mitogen- 5). Of interest, in the tumor tissue of this patient, the RASEF activated protein kinase activation in melanoma is mediated by gene was methylated, furthermore confirming our conclusion. both BRAF mutations and autocrine growth factor stimulation. We conclude that homozygosity in combination with meth- Cancer Res. 2003;63:756–759. ylation is the mechanism that targets RASEF in uveal mela- 16. Calipel A, Mouriaux F, Glotin AL, Malecaze F, Faussat AM, Mas- noma, appointing RASEF as a bona fide tumor suppressor that carelli F. Extracellular signal-regulated kinase-dependent prolifera- is epigenetically silenced in uveal melanoma. Allelic imbalance tion is mediated through the protein kinase A/B-Raf pathway in at this locus supports a tumor-suppressor role for RASEF; human uveal melanoma cells. J Biol Chem. 2006;281:9238–9250. however, analysis of RASEF in proliferation, survival, and mi- 17. Weber A, Hengge UR, Urbanik D, et al. Absence of mutations of gration of uveal melanoma is needed to confirm this. the BRAF gene and constitutive activation of extracellular-regu- lated kinase in malignant melanomas of the uvea. Lab Invest. 2003;83:1771–1776. Acknowledgments 18. van der Velden PA, Metzelaar-Blok JA, Bergman W, et al. Promoter hypermethylation: a common cause of reduced p16(INK4a) ex- The authors thank Bruce R. Ksander (Schepens Eye Institute, Harvard pression in uveal melanoma. Cancer Res. 2001;61:5303–5306. Medical School, Boston, MA) for cell lines Mel-202, -270, -285, and -290, 19. Davies H, Bignell GR, Cox C, et al. Mutations of the BRAF gene in and OMM-2.3 and -2.5, June Kan-Mitchell (Karmanos Cancer Institute, human cancer. Nature. 2002;417:949–954. Wayne State University School of Medicine, Detroit, MI) for cell lines 20. van Elsas A, Zerp SF, van der Flier S, et al. Relevance of ultraviolet- OCM-1, -3, and -8, and Rolf H. A. M. Vossen (Department of Human and induced N-ras oncogene point mutations in development of pri- Clinical Genetics, LUMC, Leiden, The Netherlands) for technical assis- mary human cutaneous melanoma. Am J Pathol. 1996;149:883– tance. 893.

Downloaded from iovs.arvojournals.org on 10/02/2021 1298 Maat et al. IOVS, April 2008, Vol. 49, No. 4

21. Zuidervaart W, van Nieuwpoort F, Stark M, et al. Activation of the 24. Liu Q, Sommer SS. Pyrophosphorolysis by Type II DNA MAPK pathway is a common event in uveal melanomas although it polymerases: implications for pyrophosphorolysis-activated poly- rarely occurs through mutation of BRAF or RAS. Br J Cancer. merization. Anal Biochem. 2004;324:22–28. 2005;92:2032–2038. 25. Liu Q, Sommer SS. PAP: detection of ultra rare mutations depends 22. Sweetser DA, Peniket AJ, Haaland C, et al. Delineation of the on P* oligonucleotides: “sleeping beauties” awakened by the kiss minimal commonly deleted segment and identification of candi- of pyrophosphorolysis. Hum Mutat. 2004;23:426–436. date tumor-suppressor genes in del(9q) acute myeloid leukemia. 26. Padua RA, Barrass N, Currie GA. A novel transforming gene in a Genes Cancer. 2005;44:279–291. human malignant melanoma cell line. Nature. 1984;311:671– 23. Maat W, van der Velden PA, Out-Luiting C, et al. Epigenetic inac- 673. tivation of RASSF1a in uveal melanoma. Invest Ophthalmol Vis Sci. 27. Rimoldi D, Salvi S, Lienard D, et al. Lack of BRAF mutations in uveal 2007;48:486–490. melanoma. Cancer Res. 2003;63:5712–5715.

ERRATUM

Erratum in: “Visual Impairment, Causes of Vision Loss, and Falls: The Singapore Malay Eye Study” by Lamoureux et al. (Invest Ophthalmol Vis Sci. 2008;49:528–533.) The correct spelling of the first author’s name is Ecosse L. Lamoureux.

Downloaded from iovs.arvojournals.org on 10/02/2021