Rare missense variants in POT1 predispose to familial cutaneous malignant melanoma

Jianxin Shi, Xiaohong R Yang, Bari Ballew, Melissa Rotunno, Donato Calista, Maria Concetta Fargnoli, Paola Ghiorzo, Brigitte Bressac-de Paillerets, Eduardo Nagore, Marie Francoise Avril, Neil E Caporaso, Mary L McMaster, Michael Cullen, Zhaoming Wang, Xijun Zhang, NCI DCEG Cancer Sequencing Working Group, NCI DCEG Cancer Genomics Research Laboratory, French Familial Melanoma Study Group, William Bruno, Lorenza Pastorino, Paola Queirolo, Jose Banuls-Roca, Zaida Garcia-Casado, Amaury Vaysse, Hamida Mohamdi, Yasser Riazalhosseini, Mario Foglio, Fanélie Jouenne, Xing Hua, Paula L Hyland, Jinhu Yin, Haritha Vallabhaneni, Weihang Chai, Paola Minghetti, Cristina Pellegrini, Sarangan Ravichandran, Alexander Eggermont, Mark Lathrop, Ketty Peris, Giovanna Bianchi Scarra, Giorgio Landi, Sharon A Savage, Joshua N Sampson, Ji He, Meredith Yeager, Lynn R Goldin, Florence Demenais, Stephen J Chanock, Margaret A Tucker, Alisa M Goldstein, Yie Liu, Maria Teresa Landi, Jianxin Shi, Xiaohong R Yang, Bari Ballew, Melissa Rotunno, Donato Calista, Maria Concetta Fargnoli, Paola Ghiorzo, Brigitte Bressac-de Paillerets, Eduardo Nagore, Marie Francoise Avril, Neil E Caporaso, Mary L McMaster, Michael Cullen, Zhaoming Wang, Xijun Zhang, NCI DCEG Cancer Sequencing Working Group, NCI DCEG Cancer Genomics Research Laboratory, French Familial Melanoma Study Group, William Bruno, Lorenza Pastorino, Paola Queirolo, Jose Banuls-Roca, Zaida Garcia-Casado, Amaury Vaysse, Hamida Mohamdi, Yasser Riazalhosseini, Mario Foglio, Fanélie Jouenne, Xing Hua, Paula L Hyland, Jinhu Yin, Haritha Vallabhaneni, Weihang Chai, Paola Minghetti, Cristina Pellegrini, Sarangan Ravichandran, Alexander Eggermont, Mark Lathrop, Ketty Peris, Giovanna Bianchi Scarra, Giorgio Landi, Sharon A Savage, Joshua N Sampson, Ji He, Meredith Yeager, Lynn R Goldin, Florence Demenais, Stephen J Chanock, Margaret A Tucker, Alisa M Goldstein, Yie Liu, Maria Teresa Landi

Abstract

Although CDKN2A is the most frequent high-risk melanoma susceptibility gene, the underlying genetic factors for most melanoma-prone families remain unknown. Using whole-exome sequencing, we identified a rare variant that arose as a founder mutation in the telomere shelterin gene POT1 (chromosome 7, g.124493086C>T; p.Ser270Asn) in five unrelated melanoma-prone families from Romagna, Italy. Carriers of this variant had increased telomere lengths and numbers of fragile telomeres, suggesting that this variant perturbs telomere maintenance. Two additional rare POT1 variants were identified in all cases sequenced in two separate Italian families, one variant per family, yielding a frequency for POT1 variants comparable to that for CDKN2A mutations in this population. These variants were not found in public databases or in 2,038 genotyped Italian controls. We also identified two rare recurrent POT1 variants in US and French familial melanoma cases. Our findings suggest that POT1 is a major susceptibility gene for familial melanoma in several populations.

Figures

Figure 1. Pedigrees of cutaneous malignant melanoma…
Figure 1. Pedigrees of cutaneous malignant melanoma (CMM)-prone families with the Ser270Asn variant (g.7:124493086 C>T) in POT1
Solid squares and circles: CMM cases; Circle with a dot in the center: obligate gene carrier; Circles: females; Squares: males. “y” indicates a variant carrier and “n” indicates a non-carrier. “age” is the age at diagnosis for CMM cases and age at exam for unaffected family members.
Figure 2. Structural illustrations of rare variants…
Figure 2. Structural illustrations of rare variants in POT1
(a) Homology-based 3D model of hPOT1 (UniProt; Q9NUX5, POTE1_HUMAN) constructed using Phyre2,. Secondary structural domains are schematically shown as cylinders (alpha helix), arrows (beta-sheet) and tubes (loop regions). The top templates used for modeling are 1XJV_A, 1ph4_A, 1jb7_A, and 1k8g_C,-. The N-terminal sequence end (blue) is the same as the experimentally derived structure (1XJV_A) and the C-terminal end regions (red) are highly similar to the telomere binding proteins 1ph4 and 1jb7. (b) Schematic representation of the N-terminal region (OB1 and OB2 domains) of hPOT1 protein (Research Collaboratory for Structural Bioinformatics (RCSB) Protein Data Bank (PDB) ID, 1XJV). Helical motifs are shown as cylinders (red) and beta-sheets as arrows pointing in the direction of C-terminus. The coil and the turn region segments are displayed as tubes. The three residues in the OB1/OB2 domains containing missense variants are shown in Corey-Pauling-Koltun (CPK) mode. The two affected amino acids (Ser270Asn and Asp224Asn) are in close proximity to the bound telomeric single-stranded DNA (ssDNA) decamer. Discovery Studio (v. 3.5, Accelrys Inc.) was used for visualization and display. (c) Schematic of POT1 genomic structure and conserved OB domains. POT1 is composed of 19 exons spanning approximately 108,000 bases of genomic sequence on chromosome 7q31.33. (d) Amino acid conservation among POT1 homologs. Higher percent identity at a given amino acid position is indicated by a deeper purple color. The positions of the variants identified in this study are indicated relative to NP_056265.
Figure 3. Telomere length in PBMC from…
Figure 3. Telomere length in PBMC from individuals with POT1-Ser270Asn variant
(a) telomere restriction fragment (TRF) analysis of PBMC. A representative blot shows telomere restriction fragments in each POT1 variant carrier (n=4) and the age-matched control (C, melanoma cases without POT1 variants, n=3). A labeled molecular weight marker (shown in kilobases) is used to determine telomere length. DNA was separated in a CHEF DR-II pulsed-field apparatus at 5 V/cm at an angle of 120° with switching times ramped from 1 to 15 s for 15 hours. Telomere length and heterogeneity are demonstrated by telomere signal intensities at higher and lower molecular weight. (b) Q-FISH analysis of ex vivo stimulated PBMC. Representative metaphase spreads of age-matched control and POT1 variant carrier showing DAPI staining (blue) and telomere fluorescence signals (red). Arrows: fragile telomeres (enlarged view demonstrated in the box). Left panel: quantitative measurement of telomere signal intensity in a jitter plot displaying complete distribution of telomeres with diverse signal intensity. Purple bars denote mean telomere signal intensity in a single control (C2) and a single Ser270Asn carrier (Ser270Asn-3). Additional variant carriers are described in Supplementary Fig. 6. Telomere signal intensity is depicted in arbitrary units (A.U). Right panel: Percentage of fragile telomeres in C2 and Ser270Asn-3. At least 30 metaphases / sample were counted. Error bars indicate standard deviation. P values were obtained from the Wilcoxon rank-sum test. The experiments were repeated three times on each sample.

References

    1. Goldstein AM, Tucker MA. Genetic epidemiology of cutaneous melanoma: a global perspective. Arch Dermatol. 2001;137:1493–6.
    1. Cannon-Albright LA, et al. Assignment of a locus for familial melanoma, MLM, to chromosome 9p13-p22. Science. 1992;258:1148–52.
    1. Hussussian CJ, et al. Germline p16 mutations in familial melanoma. Nat Genet. 1994;8:15–21.
    1. Gruis NA, et al. Homozygotes for CDKN2 (p16) germline mutation in Dutch familial melanoma kindreds. Nat Genet. 1995;10:351–3.
    1. Goldstein AM. Familial melanoma, pancreatic cancer and germline CDKN2A mutations. Hum Mutat. 2004;23:630.
    1. Eliason MJ, et al. Population-based prevalence of CDKN2A mutations in Utah melanoma families. J Invest Dermatol. 2006;126:660–6.
    1. Zuo L, et al. Germline mutations in the p16INK4a binding domain of CDK4 in familial melanoma. Nat Genet. 1996;12:97–9.
    1. Puntervoll HE, et al. Melanoma prone families with CDK4 germline mutation: phenotypic profile and associations with MC1R variants. J Med Genet. 2013;50:264–70.
    1. Wiesner T, et al. Germline mutations in BAP1 predispose to melanocytic tumors. Nat Genet. 2011;43:1018–21.
    1. Horn S, et al. TERT promoter mutations in familial and sporadic melanoma. Science. 2013;339:959–61.
    1. Loayza D, De Lange T. POT1 as a terminal transducer of TRF1 telomere length control. Nature. 2003;423:1013–8.
    1. Palm W, de Lange T. How shelterin protects mammalian telomeres. Annu Rev Genet. 2008;42:301–34.
    1. Lei M, Podell ER, Baumann P, Cech TR. DNA self-recognition in the structure of Pot1 bound to telomeric single-stranded DNA. Nature. 2003;426:198–203.
    1. Lei M, Podell ER, Cech TR. Structure of human POT1 bound to telomeric single-stranded DNA provides a model for chromosome end-protection. Nat Struct Mol Biol. 2004;11:1223–9.
    1. Forbes SA, et al. COSMIC: mining complete cancer genomes in the Catalogue of Somatic Mutations in Cancer. Nucleic Acids Res. 2011;39:D945–50.
    1. Ramsay AJ, et al. POT1 mutations cause telomere dysfunction in chronic lymphocytic leukemia. Nat Genet. 2013;45:526–30.
    1. Taboski MA, et al. Long telomeres bypass the requirement for telomere maintenance in human tumorigenesis. Cell Rep. 2012;1:91–8.
    1. Kendellen MF, Barrientos KS, Counter CM. POT1 association with TRF2 regulates telomere length. Mol Cell Biol. 2009;29:5611–9.
    1. Martinez P, et al. Increased telomere fragility and fusions resulting from TRF1 deficiency lead to degenerative pathologies and increased cancer in mice. Genes Dev. 2009;23:2060–75.
    1. Sfeir A, et al. Mammalian telomeres resemble fragile sites and require TRF1 for efficient replication. Cell. 2009;138:90–103.
    1. Badie S, et al. BRCA2 acts as a RAD51 loader to facilitate telomere replication and capping. Nat Struct Mol Biol. 2010;17:1461–9.
    1. Gu P, et al. CTC1 deletion results in defective telomere replication, leading to catastrophic telomere loss and stem cell exhaustion. EMBO J. 2012;31:2309–21.
    1. Takai H, Smogorzewska A, de Lange T. DNA damage foci at dysfunctional telomeres. Curr Biol. 2003;13:1549–56.
    1. Wang F, et al. The POT1-TPP1 telomere complex is a telomerase processivity factor. Nature. 2007;445:506–10.
    1. Xin H, et al. TPP1 is a homologue of ciliate TEBP-beta and interacts with POT1 to recruit telomerase. Nature. 2007;445:559–62.
    1. Robles-Espinoza CD, et al. POT1 loss-of-function variants predispose to familial melanoma. Nat Genet. 2014
    1. Landi MT, et al. Genetic susceptibility in familial melanoma from northeastern Italy. J Med Genet. 2004;41:557–66.
    1. Horvath MP, Schultz SC. DNA G-quartets in a 1.86 A resolution structure of an Oxytricha nova telomeric protein-DNA complex. J Mol Biol. 2001;310:367–77.
    1. Theobald DL, Schultz SC. Nucleotide shuffling and ssDNA recognition in Oxytricha nova telomere end-binding protein complexes. EMBO J. 2003;22:4314–24.
    1. Classen S, Ruggles JA, Schultz SC. Crystal structure of the N-terminal domain of Oxytricha nova telomere end-binding protein alpha subunit both uncomplexed and complexed with telomeric ssDNA. J Mol Biol. 2001;314:1113–25.
    1. Tucker MA, et al. A natural history of melanomas and dysplastic nevi: an atlas of lesions in melanoma-prone families. Cancer. 2002;94:3192–209.
    1. Liang XS, et al. Common genetic variants in candidate genes and risk of familial lymphoid malignancies. Br J Haematol. 2009;146:418–23.
    1. Chaudru V, et al. Influence of genes, nevi, and sun sensitivity on melanoma risk in a family sample unselected by family history and in melanoma-prone families. J Natl Cancer Inst. 2004;96:785–95.
    1. Bodelon C, et al. On the interplay of telomeres, nevi and the risk of melanoma. PLoS One. 2012;7:e52466.
    1. Maccioni L, et al. Variants at chromosome 20 (ASIP locus) and melanoma risk. Int J Cancer. 2013;132:42–54.
    1. Landi MT, et al. A genome-wide association study of lung cancer identifies a region of chromosome 5p15 associated with risk for adenocarcinoma. Am J Hum Genet. 2009;85:679–91.
    1. Landi MT, et al. Environment And Genetics in Lung cancer Etiology (EAGLE) study: an integrative population-based case-control study of lung cancer. BMC Public Health. 2008;8:203.
    1. Ballew BJ, et al. Germline mutations of regulator of telomere elongation helicase 1, RTEL1, in Dyskeratosis congenita. Hum Genet. 2013;132:473–80.
    1. DePristo MA, et al. A framework for variation discovery and genotyping using next-generation DNA sequencing data. Nat Genet. 2011;43:491–8.
    1. Purcell S, et al. PLINK: a tool set for whole-genome association and population-based linkage analyses. Am J Hum Genet. 2007;81:559–75.
    1. Browning BL, Browning SR. A unified approach to genotype imputation and haplotype-phase inference for large data sets of trios and unrelated individuals. Am J Hum Genet. 2009;84:210–23.
    1. Kong A, et al. Detection of sharing by descent, long-range phasing and haplotype imputation. Nat Genet. 2008;40:1068–75.
    1. Genin E, Tullio-Pelet A, Begeot F, Lyonnet S, Abel L. Estimating the age of rare disease mutations: the example of Triple-A syndrome. J Med Genet. 2004;41:445–9.
    1. Krumm N, et al. Copy number variation detection and genotyping from exome sequence data. Genome Res. 2012;22:1525–32.
    1. Notredame C, Higgins DG, Heringa J. T-Coffee: A novel method for fast and accurate multiple sequence alignment. J Mol Biol. 2000;302:205–17.
    1. Waterhouse AM, Procter JB, Martin DM, Clamp M, Barton GJ. Jalview Version 2--a multiple sequence alignment editor and analysis workbench. Bioinformatics. 2009;25:1189–91.
    1. Hathcock KS, Hodes RJ, Weng NP. Analysis of telomere length and telomerase activity. Curr Protoc Immunol. 2004;Chapter 10:30. Unit 10.
    1. Zijlmans JM, et al. Telomeres in the mouse have large inter-chromosomal variations in the number of T2AG3 repeats. Proc Natl Acad Sci U S A. 1997;94:7423–8.
    1. Wang Y, et al. An increase in telomere sister chromatid exchange in murine embryonic stem cells possessing critically shortened telomeres. Proc Natl Acad Sci U S A. 2005;102:10256–60.
    1. Bailey SM, Goodwin EH, Cornforth MN. Strand-specific fluorescence in situ hybridization: the CO-FISH family. Cytogenet Genome Res. 2004;107:14–7.
    1. Vallabhaneni H, O'Callaghan N, Sidorova J, Liu Y. Defective repair of oxidative base lesions by the DNA glycosylase Nth1 associates with multiple telomere defects. PLoS Genet. 2013;9:e1003639.
    1. Chai W, Shay JW, Wright WE. Human telomeres maintain their overhang length at senescence. Mol Cell Biol. 2005;25:2158–68.
    1. Liu Y, et al. The telomerase reverse transcriptase is limiting and necessary for telomerase function in vivo. Curr Biol. 2000;10:1459–62.

Source: PubMed

Подписаться