Skip to main content

Advertisement

Log in

Genetic alterations of PTEN in human melanoma

  • Review
  • Published:
Cellular and Molecular Life Sciences Aims and scope Submit manuscript

Abstract

The PTEN gene is one of the most frequently inactivated tumor suppressor genes in sporadic cancers. Inactivating mutations and deletions of the PTEN gene are found in many types of cancers, including melanoma. However, the exact frequency of PTEN alteration in melanoma is unknown. In this study, we comprehensively reviewed 16 studies on PTEN genetic changes in melanoma cell lines and tumor biopsies. To date, 76 PTEN alterations have been reported in melanoma cell lines and 38 PTEN alterations in melanoma biopsies. The rate of PTEN alterations in melanoma cell lines, primary melanoma, and metastatic melanoma is 27.6, 7.3, and 15.2%, respectively. Three mutations were found in both melanoma cell lines and biopsies. These mutations are scattered throughout the gene, with the exception of exon 9. A mutational hot spot is found in exon 5, which encodes the phosphatase activity domain. Evidence is also presented to suggest that numerous homozygous deletions and missense variants exist in the PTEN transcript. Studying PTEN functions and implications of its mutations and other genes could provide insights into the precise nature of PTEN function in melanoma and additional targets for new therapeutic approaches.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

References

  1. Li J, Yen C, Liaw D, Podsypanina K, Bose S, Wang SI, Puc J, Miliaresis C, Rodgers L, McCombie R, Bigner SH, Giovanella BC, Ittmann M, Tycko B, Hibshoosh H, Wigler MH, Parsons R (1997) PTEN, a putative protein tyrosine phosphatase gene mutated in human brain, breast, and prostate cancer. Science 275:1943–1947

    PubMed  CAS  Google Scholar 

  2. Steck PA, Pershouse MA, Jasser SA, Yung WK, Lin H, Ligon AH, Langford LA, Baumgard ML, Hattier T, Davis T, Frye C, Hu R, Swedlund B, Teng DH, Tavtigian SV (1997) Identification of a candidate tumour suppressor gene, MMAC1, at chromosome 10q23.3 that is mutated in multiple advanced cancers. Nat Genet 15:356–362

    PubMed  CAS  Google Scholar 

  3. Li DM, Sun H (1997) TEP1, encoded by a candidate tumor suppressor locus, is a novel protein tyrosine phosphatase regulated by transforming growth factor beta. Cancer Res 57:2124–2129

    PubMed  CAS  Google Scholar 

  4. Dahia PL, Marsh DJ, Zheng Z, Zedenius J, Komminoth P, Frisk T, Wallin G, Parsons R, Longy M, Larsson C, Eng C (1997) Somatic deletions and mutations in the Cowden disease gene, PTEN, in sporadic thyroid tumors. Cancer Res 57:4710–4713

    PubMed  CAS  Google Scholar 

  5. Guldberg P, thor Straten P, Birck A, Ahrenkiel V, Kirkin AF, Zeuthen J (1997) Disruption of the MMAC1/PTEN gene by deletion or mutation is a frequent event in malignant melanoma. Cancer Res 57:3660–3663

    PubMed  CAS  Google Scholar 

  6. Risinger JI, Hayes AK, Berchuck A, Barrett JC (1997) PTEN/MMAC1 mutations in endometrial cancers. Cancer Res 57:4736–4738

    PubMed  CAS  Google Scholar 

  7. Tashiro H, Blazes MS, Wu R, Cho KR, Bose S, Wang SI, Li J, Parsons R, Ellenson LH (1997) Mutations in PTEN are frequent in endometrial carcinoma but rare in other common gynecological malignancies. Cancer Res 57:3935–3940

    PubMed  CAS  Google Scholar 

  8. Halachmi N, Halachmi S, Evron E, Cairns P, Okami K, Saji M, Westra WH, Zeiger MA, Jen J, Sidransky D (1998) Somatic mutations of the PTEN tumor suppressor gene in sporadic follicular thyroid tumors. Genes Chromosomes Cancer 23:239–243

    PubMed  CAS  Google Scholar 

  9. Maxwell GL, Risinger JI, Gumbs C, Shaw H, Bentley RC, Barrett JC, Berchuck A, Futreal PA (1998) Mutation of the PTEN tumor suppressor gene in endometrial hyperplasias. Cancer Res 58:2500–2503

    PubMed  CAS  Google Scholar 

  10. Wang SI, Puc J, Li J, Bruce JN, Cairns P, Sidransky D, Parsons R (1997) Somatic mutations of PTEN in glioblastoma multiforme. Cancer Res 57:4183–4186

    PubMed  CAS  Google Scholar 

  11. Yang J, Ren Y, Wang L, Li B, Chen Y, Zhao W, Xu W, Li T, Dai F (2010) PTEN mutation spectrum in breast cancers and breast hyperplasia. J Cancer Res Clin Oncol 136:1303–1311

    PubMed  CAS  Google Scholar 

  12. Barbosa M, Henrique M, Pinto-Basto J, Claes K, Soares G (2011) Prostate cancer in Cowden syndrome: somatic loss and germline mutation of the PTEN gene. Cancer Genet 204:224–225

    PubMed  CAS  Google Scholar 

  13. Birck A, Ahrenkiel V, Zeuthen J, Hou-Jensen K, Guldberg P (2000) Mutation and allelic loss of the PTEN/MMAC1 gene in primary and metastatic melanoma biopsies. J Invest Dermatol 114:277–280

    PubMed  CAS  Google Scholar 

  14. Celebi JT, Shendrik I, Silvers DN, Peacocke M (2000) Identification of PTEN mutations in metastatic melanoma specimens. J Med Genet 37:653–657

    PubMed  CAS  Google Scholar 

  15. Liaw D, Marsh DJ, Li J, Dahia PL, Wang SI, Zheng Z, Bose S, Call KM, Tsou HC, Peacocke M, Eng C, Parsons R (1997) Germline mutations of the PTEN gene in Cowden disease, an inherited breast and thyroid cancer syndrome. Nat Genet 16:64–67

    PubMed  CAS  Google Scholar 

  16. Marsh DJ, Coulon V, Lunetta KL, Rocca-Serra P, Dahia PL, Zheng Z, Liaw D, Caron S, Duboue B, Lin AY, Richardson AL, Bonnetblanc JM, Bressieux JM, Cabarrot-Moreau A, Chompret A, Demange L, Eeles RA, Yahanda AM, Fearon ER, Fricker JP, Gorlin RJ, Hodgson SV, Huson S, Lacombe D, Eng C et al (1998) Mutation spectrum and genotype-phenotype analyses in Cowden disease and Bannayan-Zonana syndrome, two hamartoma syndromes with germline PTEN mutation. Hum Mol Genet 7:507–515

    PubMed  CAS  Google Scholar 

  17. Eng C, Hobert JA (2009) PTEN hamartoma tumor syndrome: an overview. Genet Med 11:687–694

    PubMed  Google Scholar 

  18. Eng C (2003) PTEN: one gene, many syndromes. Hum Mutat 22:183–198

    PubMed  CAS  Google Scholar 

  19. Waite KA, Eng C (2002) Protean PTEN: form and function. Am J Hum Genet 70:829–844

    PubMed  CAS  Google Scholar 

  20. Di Cristofano A, Pandolfi PP (2000) The multiple roles of PTEN in tumor suppression. Cell 100:387–390

    PubMed  Google Scholar 

  21. Mutter GL (2001) Pten, a protean tumor suppressor. Am J Pathol 158:1895–1898

    PubMed  CAS  Google Scholar 

  22. Furnari FB, Huang HJ, Cavenee WK (1998) The phosphoinositol phosphatase activity of PTEN mediates a serum-sensitive G1 growth arrest in glioma cells. Cancer Res 58:5002–5008

    PubMed  CAS  Google Scholar 

  23. Georgescu MM, Kirsch KH, Kaloudis P, Yang H, Pavletich NP, Hanafusa H (2000) Stabilization and productive positioning roles of the C2 domain of PTEN tumor suppressor. Cancer Res 60:7033–7038

    PubMed  CAS  Google Scholar 

  24. Hlobilkova A, Guldberg P, Thullberg M, Zeuthen J, Lukas J, Bartek J (2000) Cell cycle arrest by the PTEN tumor suppressor is target cell specific and may require protein phosphatase activity. Exp Cell Res 256:571–577

    PubMed  CAS  Google Scholar 

  25. Weng LP, Brown JL, Eng C (2001) PTEN coordinates G(1) arrest by down-regulating cyclin D1 via its protein phosphatase activity and up-regulating p27 via its lipid phosphatase activity in a breast cancer model. Hum Mol Genet 10:599–604

    PubMed  CAS  Google Scholar 

  26. Dey N, Crosswell HE, De P, Parsons R, Peng Q, Su JD, Durden DL (2008) The protein phosphatase activity of PTEN regulates SRC family kinases and controls glioma migration. Cancer Res 68:1862–1871

    PubMed  CAS  Google Scholar 

  27. Davidson L, Maccario H, Perera NM, Yang X, Spinelli L, Tibarewal P, Glancy B, Gray A, Weijer CJ, Downes CP, Leslie NR (2010) Suppression of cellular proliferation and invasion by the concerted lipid and protein phosphatase activities of PTEN. Oncogene 29:687–697

    PubMed  CAS  Google Scholar 

  28. Poon JS, Eves R, Mak AS (2010) Both lipid- and protein-phosphatase activities of PTEN contribute to the p53-PTEN anti-invasion pathway. Cell Cycle 9:4450–4454

    PubMed  CAS  Google Scholar 

  29. Fountain JW, Bale SJ, Housman DE, Dracopoli NC (1990) Genetics of melanoma. Cancer Surv 9:645–671

    PubMed  CAS  Google Scholar 

  30. Fults D, Pedone C (1993) Deletion mapping of the long arm of chromosome 10 in glioblastoma multiforme. Genes Chromosomes Cancer 7:173–177

    PubMed  CAS  Google Scholar 

  31. Isshiki K, Elder DE, Guerry D, Linnenbach AJ (1993) Chromosome 10 allelic loss in malignant melanoma. Genes Chromosomes Cancer 8:178–184

    PubMed  CAS  Google Scholar 

  32. Herbst RA, Weiss J, Ehnis A, Cavenee WK, Arden KC (1994) Loss of heterozygosity for 10q22–10qter in malignant melanoma progression. Cancer Res 54:3111–3114

    PubMed  CAS  Google Scholar 

  33. Healy E, Rehman I, Angus B, Rees JL (1995) Loss of heterozygosity in sporadic primary cutaneous melanoma. Genes Chromosomes Cancer 12:152–156

    PubMed  CAS  Google Scholar 

  34. Ittmann M (1996) Allelic loss on chromosome 10 in prostate adenocarcinoma. Cancer Res 56:2143–2147

    PubMed  CAS  Google Scholar 

  35. Parmiter AH, Balaban G, Clark WH Jr, Nowell PC (1988) Possible involvement of the chromosome region 10q24–q26 in early stages of melanocytic neoplasia. Cancer Genet Cytogenet 30:313–317

    PubMed  CAS  Google Scholar 

  36. Poetsch M, Lorenz G, Kleist B (2002) Detection of new PTEN/MMAC1 mutations in head and neck squamous cell carcinomas with loss of chromosome 10. Cancer Genet Cytogenet 132:20–24

    PubMed  CAS  Google Scholar 

  37. Tonks NK, Neel BG (1996) From form to function: signaling by protein tyrosine phosphatases. Cell 87:365–368

    PubMed  CAS  Google Scholar 

  38. Yuvaniyama J, Denu JM, Dixon JE, Saper MA (1996) Crystal structure of the dual specificity protein phosphatase VHR. Science 272:1328–1331

    PubMed  CAS  Google Scholar 

  39. Myers MP, Stolarov JP, Eng C, Li J, Wang SI, Wigler MH, Parsons R, Tonks NK (1997) P-TEN, the tumor suppressor from human chromosome 10q23, is a dual-specificity phosphatase. Proc Natl Acad Sci USA 94:9052–9057

    PubMed  CAS  Google Scholar 

  40. Maehama T, Dixon JE (1998) The tumor suppressor, PTEN/MMAC1, dephosphorylates the lipid second messenger, phosphatidylinositol 3, 4, 5-trisphosphate. J Biol Chem 273:13375–13378

    PubMed  CAS  Google Scholar 

  41. Lee JO, Yang H, Georgescu MM, Di Cristofano A, Maehama T, Shi Y, Dixon JE, Pandolfi P, Pavletich NP (1999) Crystal structure of the PTEN tumor suppressor: implications for its phosphoinositide phosphatase activity and membrane association. Cell 99:323–334

    PubMed  CAS  Google Scholar 

  42. Manning BD, Cantley LC (2007) AKT/PKB signaling: navigating downstream. Cell 129:1261–1274

    PubMed  CAS  Google Scholar 

  43. Alvarez-Nunez F, Bussaglia E, Mauricio D, Ybarra J, Vilar M, Lerma E, de Leiva A, Matias-Guiu X (2006) PTEN promoter methylation in sporadic thyroid carcinomas. Thyroid 16:17–23

    PubMed  CAS  Google Scholar 

  44. Garcia JM, Silva J, Pena C, Garcia V, Rodriguez R, Cruz MA, Cantos B, Provencio M, Espana P, Bonilla F (2004) Promoter methylation of the PTEN gene is a common molecular change in breast cancer. Genes Chromosomes Cancer 41:117–124

    PubMed  CAS  Google Scholar 

  45. Ho CM, Lin MC, Huang SH, Huang CJ, Lai HC, Chien TY, Chang SF (2009) PTEN promoter methylation and LOH of 10q22–23 locus in PTEN expression of ovarian clear cell adenocarcinomas. Gynecol Oncol 112:307–313

    PubMed  CAS  Google Scholar 

  46. Kang YH, Lee HS, Kim WH (2002) Promoter methylation and silencing of PTEN in gastric carcinoma. Lab Invest 82:285–291

    PubMed  CAS  Google Scholar 

  47. Koul D (2008) PTEN signaling pathways in glioblastoma. Cancer Biol Ther 7:1321–1325

    PubMed  CAS  Google Scholar 

  48. Salvesen HB, MacDonald N, Ryan A, Jacobs IJ, Lynch ED, Akslen LA, Das S (2001) PTEN methylation is associated with advanced stage and microsatellite instability in endometrial carcinoma. Int J Cancer 91:22–26

    PubMed  CAS  Google Scholar 

  49. Soria JC, Lee HY, Lee JI, Wang L, Issa JP, Kemp BL, Liu DD, Kurie JM, Mao L, Khuri FR (2002) Lack of PTEN expression in non-small cell lung cancer could be related to promoter methylation. Clin Cancer Res 8:1178–1184

    PubMed  CAS  Google Scholar 

  50. Huse JT, Brennan C, Hambardzumyan D, Wee B, Pena J, Rouhanifard SH, Sohn-Lee C, le Sage C, Agami R, Tuschl T, Holland EC (2009) The PTEN-regulating microRNA miR-26a is amplified in high-grade glioma and facilitates gliomagenesis in vivo. Genes Dev 23:1327–1337

    PubMed  CAS  Google Scholar 

  51. Zhang JG, Wang JJ, Zhao F, Liu Q, Jiang K, Yang GH (2010) MicroRNA-21 (miR-21) represses tumor suppressor PTEN and promotes growth and invasion in non-small cell lung cancer (NSCLC). Clin Chim Acta 411:846–852

    PubMed  CAS  Google Scholar 

  52. Poliseno L, Salmena L, Riccardi L, Fornari A, Song MS, Hobbs RM, Sportoletti P, Varmeh S, Egia A, Fedele G, Rameh L, Loda M, Pandolfi PP (2010) Identification of the miR-106b ~ 25 microRNA cluster as a proto-oncogenic PTEN-targeting intron that cooperates with its host gene MCM7 in transformation. Sci Signal 3:ra29

    PubMed  Google Scholar 

  53. Tamguney T, Stokoe D (2007) New insights into PTEN. J Cell Sci 120:4071–4079

    PubMed  CAS  Google Scholar 

  54. Wang X, Jiang X (2008) Post-translational regulation of PTEN. Oncogene 27:5454–5463

    PubMed  CAS  Google Scholar 

  55. Trotman LC, Wang X, Alimonti A, Chen Z, Teruya-Feldstein J, Yang H, Pavletich NP, Carver BS, Cordon-Cardo C, Erdjument-Bromage H, Tempst P, Chi SG, Kim HJ, Misteli T, Jiang X, Pandolfi PP (2007) Ubiquitination regulates PTEN nuclear import and tumor suppression. Cell 128:141–156

    PubMed  CAS  Google Scholar 

  56. Poliseno L, Salmena L, Zhang J, Carver B, Haveman WJ, Pandolfi PP (2010) A coding-independent function of gene and pseudogene mRNAs regulates tumour biology. Nature 465:1033–1038

    PubMed  CAS  Google Scholar 

  57. Nardella C, Carracedo A, Salmena L, Pandolfi PP (2010) Faithfull modeling of PTEN loss driven diseases in the mouse. Curr Top Microbiol Immunol 347:135–168

    PubMed  CAS  Google Scholar 

  58. Suzuki H, Freije D, Nusskern DR, Okami K, Cairns P, Sidransky D, Isaacs WB, Bova GS (1998) Interfocal heterogeneity of PTEN/MMAC1 gene alterations in multiple metastatic prostate cancer tissues. Cancer Res 58:204–209

    PubMed  CAS  Google Scholar 

  59. Podsypanina K, Ellenson LH, Nemes A, Gu J, Tamura M, Yamada KM, Cordon-Cardo C, Catoretti G, Fisher PE, Parsons R (1999) Mutation of Pten/Mmac1 in mice causes neoplasia in multiple organ systems. Proc Natl Acad Sci USA 96:1563–1568

    PubMed  CAS  Google Scholar 

  60. Di Cristofano A, Pesce B, Cordon-Cardo C, Pandolfi PP (1998) Pten is essential for embryonic development and tumour suppression. Nat Genet 19:348–355

    PubMed  Google Scholar 

  61. Yilmaz OH, Valdez R, Theisen BK, Guo W, Ferguson DO, Wu H, Morrison SJ (2006) Pten dependence distinguishes haematopoietic stem cells from leukaemia-initiating cells. Nature 441:475–482

    PubMed  CAS  Google Scholar 

  62. Zhang J, Grindley JC, Yin T, Jayasinghe S, He XC, Ross JT, Haug JS, Rupp D, Porter-Westpfahl KS, Wiedemann LM, Wu H, Li L (2006) PTEN maintains haematopoietic stem cells and acts in lineage choice and leukaemia prevention. Nature 441:518–522

    PubMed  CAS  Google Scholar 

  63. Chen Z, Trotman LC, Shaffer D, Lin HK, Dotan ZA, Niki M, Koutcher JA, Scher HI, Ludwig T, Gerald W, Cordon-Cardo C, Pandolfi PP (2005) Crucial role of p53-dependent cellular senescence in suppression of Pten-deficient tumorigenesis. Nature 436:725–730

    PubMed  CAS  Google Scholar 

  64. Chalhoub N, Baker SJ (2009) PTEN and the PI3-kinase pathway in cancer. Annu Rev Pathol 4:127–150

    PubMed  CAS  Google Scholar 

  65. Orloff MS, Eng C (2008) Genetic and phenotypic heterogeneity in the PTEN hamartoma tumour syndrome. Oncogene 27:5387–5397

    PubMed  CAS  Google Scholar 

  66. Tsao H, Zhang X, Benoit E, Haluska FG (1998) Identification of PTEN/MMAC1 alterations in uncultured melanomas and melanoma cell lines. Oncogene 16:3397–3402

    PubMed  CAS  Google Scholar 

  67. Tsao H, Zhang X, Fowlkes K, Haluska FG (2000) Relative reciprocity of NRAS and PTEN/MMAC1 alterations in cutaneous melanoma cell lines. Cancer Res 60:1800–1804

    PubMed  CAS  Google Scholar 

  68. Tsao H, Goel V, Wu H, Yang G, Haluska FG (2004) Genetic interaction between NRAS and BRAF mutations and PTEN/MMAC1 inactivation in melanoma. J Invest Dermatol 122:337–341

    PubMed  CAS  Google Scholar 

  69. Jonsson G, Dahl C, Staaf J, Sandberg T, Bendahl PO, Ringner M, Guldberg P, Borg A (2007) Genomic profiling of malignant melanoma using tiling-resolution arrayCGH. Oncogene 26:4738–4748

    PubMed  CAS  Google Scholar 

  70. Alimonti A, Carracedo A, Clohessy JG, Trotman LC, Nardella C, Egia A, Salmena L, Sampieri K, Haveman WJ, Brogi E, Richardson AL, Zhang J, Pandolfi PP (2010) Subtle variations in Pten dose determine cancer susceptibility. Nat Genet 42:454–458

    PubMed  CAS  Google Scholar 

  71. Shen-Li H, Koujak S, Szablocs M, Parsons R (2010) Reduction of Pten dose leads to neoplastic development in multiple organs of Pten (shRNA) mice. Cancer Biol Ther 10:1194–1200

    PubMed  Google Scholar 

  72. Kim DK, Myung SJ, Yang SK, Hong SS, Kim KJ, Byeon JS, Lee GH, Kim JH, Min YI, Lee SM, Jeong JY, Song K, Jung SA (2005) Analysis of PTEN gene mutations in Korean patients with Cowden syndrome and polyposis syndrome. Dis Colon Rectum 48:1714–1722

    PubMed  Google Scholar 

  73. Sato K, Tamura G, Tsuchiya T, Endoh Y, Sakata K, Motoyama T, Usuba O, Kimura W, Terashima M, Nishizuka S, Zou T, Meltzer SJ (2002) Analysis of genetic and epigenetic alterations of the PTEN gene in gastric cancer. Virchows Arch 440:160–165

    PubMed  CAS  Google Scholar 

  74. Nelen MR, van Staveren WC, Peeters EA, Hassel MB, Gorlin RJ, Hamm H, Lindboe CF, Fryns JP, Sijmons RH, Woods DG, Mariman EC, Padberg GW, Kremer H (1997) Germline mutations in the PTEN/MMAC1 gene in patients with Cowden disease. Hum Mol Genet 6:1383–1387

    PubMed  CAS  Google Scholar 

  75. Keniry M, Parsons R (2008) The role of PTEN signaling perturbations in cancer and in targeted therapy. Oncogene 27:5477–5485

    PubMed  CAS  Google Scholar 

  76. Daikoku T, Hirota Y, Tranguch S, Joshi AR, DeMayo FJ, Lydon JP, Ellenson LH, Dey SK (2008) Conditional loss of uterine Pten unfailingly and rapidly induces endometrial cancer in mice. Cancer Res 68:5619–5627

    PubMed  CAS  Google Scholar 

  77. Li G, Robinson GW, Lesche R, Martinez-Diaz H, Jiang Z, Rozengurt N, Wagner KU, Wu DC, Lane TF, Liu X, Hennighausen L, Wu H (2002) Conditional loss of PTEN leads to precocious development and neoplasia in the mammary gland. Development 129:4159–4170

    PubMed  CAS  Google Scholar 

  78. Wang S, Gao J, Lei Q, Rozengurt N, Pritchard C, Jiao J, Thomas GV, Li G, Roy-Burman P, Nelson PS, Liu X, Wu H (2003) Prostate-specific deletion of the murine Pten tumor suppressor gene leads to metastatic prostate cancer. Cancer Cell 4:209–221

    PubMed  CAS  Google Scholar 

  79. Suzuki A, Yamaguchi MT, Ohteki T, Sasaki T, Kaisho T, Kimura Y, Yoshida R, Wakeham A, Higuchi T, Fukumoto M, Tsubata T, Ohashi PS, Koyasu S, Penninger JM, Nakano T, Mak TW (2001) T cell-specific loss of Pten leads to defects in central and peripheral tolerance. Immunity 14:523–534

    PubMed  CAS  Google Scholar 

  80. Horie Y, Suzuki A, Kataoka E, Sasaki T, Hamada K, Sasaki J, Mizuno K, Hasegawa G, Kishimoto H, Iizuka M, Naito M, Enomoto K, Watanabe S, Mak TW, Nakano T (2004) Hepatocyte-specific Pten deficiency results in steatohepatitis and hepatocellular carcinomas. J Clin Invest 113:1774–1783

    PubMed  CAS  Google Scholar 

  81. Tsuruta H, Kishimoto H, Sasaki T, Horie Y, Natsui M, Shibata Y, Hamada K, Yajima N, Kawahara K, Sasaki M, Tsuchiya N, Enomoto K, Mak TW, Nakano T, Habuchi T, Suzuki A (2006) Hyperplasia and carcinomas in Pten-deficient mice and reduced PTEN protein in human bladder cancer patients. Cancer Res 66:8389–8396

    PubMed  CAS  Google Scholar 

  82. Yanagi S, Kishimoto H, Kawahara K, Sasaki T, Sasaki M, Nishio M, Yajima N, Hamada K, Horie Y, Kubo H, Whitsett JA, Mak TW, Nakano T, Nakazato M, Suzuki A (2007) Pten controls lung morphogenesis, bronchioalveolar stem cells, and onset of lung adenocarcinomas in mice. J Clin Invest 117:2929–2940

    PubMed  CAS  Google Scholar 

  83. Marsh V, Winton DJ, Williams GT, Dubois N, Trumpp A, Sansom OJ, Clarke AR (2008) Epithelial Pten is dispensable for intestinal homeostasis but suppresses adenoma development and progression after Apc mutation. Nat Genet 40:1436–1444

    PubMed  CAS  Google Scholar 

  84. Hwang PH, Yi HK, Kim DS, Nam SY, Kim JS, Lee DY (2001) Suppression of tumorigenicity and metastasis in B16F10 cells by PTEN/MMAC1/TEP1 gene. Cancer Lett 172:83–91

    PubMed  CAS  Google Scholar 

  85. Bonneau D, Longy M (2000) Mutations of the human PTEN gene. Hum Mutat 16:109–122

    PubMed  CAS  Google Scholar 

  86. Teng DH, Hu R, Lin H, Davis T, Iliev D, Frye C, Swedlund B, Hansen KL, Vinson VL, Gumpper KL, Ellis L, El-Naggar A, Frazier M, Jasser S, Langford LA, Lee J, Mills GB, Pershouse MA, Pollack RE, Tornos C, Troncoso P, Yung WK, Fujii G, Berson A, Steck PA et al (1997) MMAC1/PTEN mutations in primary tumor specimens and tumor cell lines. Cancer Res 57:5221–5225

    PubMed  CAS  Google Scholar 

  87. Pollock PM, Walker GJ, Glendening JM, Que Noy T, Bloch NC, Fountain JW, Hayward NK (2002) PTEN inactivation is rare in melanoma tumours but occurs frequently in melanoma cell lines. Melanoma Res 12:565–575

    PubMed  CAS  Google Scholar 

  88. Boni R, Vortmeyer AO, Burg G, Hofbauer G, Zhuang Z (1998) The PTEN tumour suppressor gene and malignant melanoma. Melanoma Res 8:300–302

    PubMed  CAS  Google Scholar 

  89. Reifenberger J, Wolter M, Bostrom J, Buschges R, Schulte KW, Megahed M, Ruzicka T, Reifenberger G (2000) Allelic losses on chromosome arm 10q and mutation of the PTEN (MMAC1) tumour suppressor gene in primary and metastatic malignant melanomas. Virchows Arch 436:487–493

    PubMed  CAS  Google Scholar 

  90. Poetsch M, Dittberner T, Woenckhaus C (2001) PTEN/MMAC1 in malignant melanoma and its importance for tumor progression. Cancer Genet Cytogenet 125:21–26

    PubMed  CAS  Google Scholar 

  91. Abdel-Rahman MH, Yang Y, Zhou XP, Craig EL, Davidorf FH, Eng C (2006) High frequency of submicroscopic hemizygous deletion is a major mechanism of loss of expression of PTEN in uveal melanoma. J Clin Oncol 24:288–295

    PubMed  CAS  Google Scholar 

  92. Robertson GP, Furnari FB, Miele ME, Glendening MJ, Welch DR, Fountain JW, Lugo TG, Huang HJ, Cavenee WK (1998) In vitro loss of heterozygosity targets the PTEN/MMAC1 gene in melanoma. Proc Natl Acad Sci USA 95:9418–9423

    PubMed  CAS  Google Scholar 

  93. Herbst RA, Podewski EK, Mommert S, Kapp A, Weiss J (1999) PTEN and MXI1 allelic loss on chromosome 10q is rare in melanoma in vivo. Arch Dermatol Res 291:567–569

    PubMed  CAS  Google Scholar 

  94. Cairns P, Polascik TJ, Eby Y, Tokino K, Califano J, Merlo A, Mao L, Herath J, Jenkins R, Westra W et al (1995) Frequency of homozygous deletion at p16/CDKN2 in primary human tumours. Nat Genet 11:210–212

    PubMed  CAS  Google Scholar 

  95. Zhou XP, Gimm O, Hampel H, Niemann T, Walker MJ, Eng C (2000) Epigenetic PTEN silencing in malignant melanomas without PTEN mutation. Am J Pathol 157:1123–1128

    PubMed  CAS  Google Scholar 

  96. Mirmohammadsadegh A, Marini A, Nambiar S, Hassan M, Tannapfel A, Ruzicka T, Hengge UR (2006) Epigenetic silencing of the PTEN gene in melanoma. Cancer Res 66:6546–6552

    PubMed  CAS  Google Scholar 

  97. Lahtz C, Stranzenbach R, Fiedler E, Helmbold P, Dammann RH (2010) Methylation of PTEN as a prognostic factor in malignant melanoma of the skin. J Invest Dermatol 130:620–622

    PubMed  CAS  Google Scholar 

  98. Mikhail M, Velazquez E, Shapiro R, Berman R, Pavlick A, Sorhaindo L, Spira J, Mir C, Panageas KS, Polsky D, Osman I (2005) PTEN expression in melanoma: relationship with patient survival, Bcl-2 expression, and proliferation. Clin Cancer Res 11:5153–5157

    PubMed  CAS  Google Scholar 

  99. Wang Y, Digiovanna JJ, Stern JB, Hornyak TJ, Raffeld M, Khan SG, Oh KS, Hollander MC, Dennis PA, Kraemer KH (2009) Evidence of ultraviolet type mutations in xeroderma pigmentosum melanomas. Proc Natl Acad Sci USA 106:6279–6284

    PubMed  CAS  Google Scholar 

  100. Inoue-Narita T, Hamada K, Sasaki T, Hatakeyama S, Fujita S, Kawahara K, Sasaki M, Kishimoto H, Eguchi S, Kojima I, Beermann F, Kimura T, Osawa M, Itami S, Mak TW, Nakano T, Manabe M, Suzuki A (2008) Pten deficiency in melanocytes results in resistance to hair graying and susceptibility to carcinogen-induced melanomagenesis. Cancer Res 68:5760–5768

    PubMed  CAS  Google Scholar 

  101. You MJ, Castrillon DH, Bastian BC, O’Hagan RC, Bosenberg MW, Parsons R, Chin L, DePinho RA (2002) Genetic analysis of Pten and Ink4a/Arf interactions in the suppression of tumorigenesis in mice. Proc Natl Acad Sci USA 99:1455–1460

    PubMed  CAS  Google Scholar 

  102. Nikolaou V, Kang X, Stratigos A, Gogas H, Latorre MC, Gabree M, Plaka M, Njauw C, Kypreou K, Mirmigi I, Stefanaki I, Tsao H (2011) Comprehensive mutational analysis of CDKN2A and CDK4 in Greek patients with cutaneous melanoma. Br J Dermatol. doi:10.1111/j.1365-2133.2011.10551.x

  103. Si L, Kong Y, Xu X, Flaherty KT, Sheng X, Cui C, Chi Z, Li S, Mao L, Guo J (2011) Prevalence of BRAF V600E mutation in Chinese melanoma patients: Large scale analysis of BRAF and NRAS mutations in a 432-case cohort. Eur J Cancer. doi:10.1016/j.ejca.2011.06.056

  104. Liu L, Lassam NJ, Slingerland JM, Bailey D, Cole D, Jenkins R, Hogg D (1995) Germline p16INK4A mutation and protein dysfunction in a family with inherited melanoma. Oncogene 11:405–412

    PubMed  CAS  Google Scholar 

  105. Flores JF, Walker GJ, Glendening JM, Haluska FG, Castresana JS, Rubio MP, Pastorfide GC, Boyer LA, Kao WH, Bulyk ML, Barnhill RL, Hayward NK, Housman DE, Fountain JW (1996) Loss of the p16INK4a and p15INK4b genes, as well as neighboring 9p21 markers, in sporadic melanoma. Cancer Res 56:5023–5032

    PubMed  CAS  Google Scholar 

  106. Haluska FG, Hodi FS (1998) Molecular genetics of familial cutaneous melanoma. J Clin Oncol 16:670–682

    PubMed  CAS  Google Scholar 

  107. Serrano M, Lin AW, McCurrach ME, Beach D, Lowe SW (1997) Oncogenic ras provokes premature cell senescence associated with accumulation of p53 and p16INK4a. Cell 88:593–602

    PubMed  CAS  Google Scholar 

  108. Ikeda T, Yoshinaga K, Suzuki A, Sakurada A, Ohmori H, Horii A (2000) Anticorresponding mutations of the KRAS and PTEN genes in human endometrial cancer. Oncol Rep 7:567–570

    PubMed  CAS  Google Scholar 

  109. Davies MA, Stemke-Hale K, Lin E, Tellez C, Deng W, Gopal YN, Woodman SE, Calderone TC, Ju Z, Lazar AJ, Prieto VG, Aldape K, Mills GB, Gershenwald JE (2009) Integrated molecular and clinical analysis of AKT activation in metastatic melanoma. Clin Cancer Res 15:7538–7546

    PubMed  CAS  Google Scholar 

  110. Nogueira C, Kim KH, Sung H, Paraiso KH, Dannenberg JH, Bosenberg M, Chin L, Kim M (2010) Cooperative interactions of PTEN deficiency and RAS activation in melanoma metastasis. Oncogene 29:6222–6232

    PubMed  CAS  Google Scholar 

  111. Chin L, Pomerantz J, Polsky D, Jacobson M, Cohen C, Cordon-Cardo C, Horner JW 2nd, DePinho RA (1997) Cooperative effects of INK4a and ras in melanoma susceptibility in vivo. Genes Dev 11:2822–2834

    PubMed  CAS  Google Scholar 

  112. Dankort D, Curley DP, Cartlidge RA, Nelson B, Karnezis AN, Damsky WE Jr, You MJ, DePinho RA, McMahon M, Bosenberg M (2009) Braf(V600E) cooperates with Pten loss to induce metastatic melanoma. Nat Genet 41:544–552

    PubMed  CAS  Google Scholar 

  113. Hocker T, Tsao H (2007) Ultraviolet radiation and melanoma: a systematic review and analysis of reported sequence variants. Hum Mutat 28:578–588

    PubMed  CAS  Google Scholar 

  114. Mograbi B, Bocciardi R, Bourget I, Busca R, Rochet N, Farahi-Far D, Juhel T, Rossi B (2001) Glial cell line-derived neurotrophic factor-stimulated phosphatidylinositol 3-kinase and Akt activities exert opposing effects on the ERK pathway: importance for the rescue of neuroectodermic cells. J Biol Chem 276:45307–45319

    PubMed  CAS  Google Scholar 

  115. Moelling K, Schad K, Bosse M, Zimmermann S, Schweneker M (2002) Regulation of Raf-Akt Cross-talk. J Biol Chem 277:31099–31106

    PubMed  CAS  Google Scholar 

  116. Tsao H, Mihm MC Jr, Sheehan C (2003) PTEN expression in normal skin, acquired melanocytic nevi, and cutaneous melanoma. J Am Acad Dermatol 49:865–872

    PubMed  Google Scholar 

  117. Singh RS, Diwan AH, Zhang PS, Prieto VG (2007) Phosphoinositide 3-kinase is not overexpressed in melanocytic lesions. J Cutan Pathol 34:220–225

    PubMed  Google Scholar 

  118. Curtin JA, Fridlyand J, Kageshita T, Patel HN, Busam KJ, Kutzner H, Cho KH, Aiba S, Brocker EB, LeBoit PE, Pinkel D, Bastian BC (2005) Distinct sets of genetic alterations in melanoma. N Engl J Med 353:2135–2147

    PubMed  CAS  Google Scholar 

  119. Fecher LA, Cummings SD, Keefe MJ, Alani RM (2007) Toward a molecular classification of melanoma. J Clin Oncol 25:1606–1620

    PubMed  CAS  Google Scholar 

  120. Wan PT, Garnett MJ, Roe SM, Lee S, Niculescu-Duvaz D, Good VM, Jones CM, Marshall CJ, Springer CJ, Barford D, Marais R (2004) Mechanism of activation of the RAF-ERK signaling pathway by oncogenic mutations of B-RAF. Cell 116:855–867

    PubMed  CAS  Google Scholar 

  121. Rubinstein JC, Sznol M, Pavlick AC, Ariyan S, Cheng E, Bacchiocchi A, Kluger HM, Narayan D, Halaban R (2010) Incidence of the V600K mutation among melanoma patients with BRAF mutations, and potential therapeutic response to the specific BRAF inhibitor PLX4032. J Transl Med 8:67

    PubMed  Google Scholar 

  122. Sun W, Schuchter LM (2001) Metastatic melanoma. Curr Treat Options Oncol 2:193–202

    PubMed  CAS  Google Scholar 

  123. Flaherty KT, Puzanov I, Kim KB, Ribas A, McArthur GA, Sosman JA, O’Dwyer PJ, Lee RJ, Grippo JF, Nolop K, Chapman PB (2010) Inhibition of mutated, activated BRAF in metastatic melanoma. N Engl J Med 363:809–819

    PubMed  CAS  Google Scholar 

  124. Chapman PB, Hauschild A, Robert C, Haanen JB, Ascierto P, Larkin J, Dummer R, Garbe C, Testori A, Maio M, Hogg D, Lorigan P, Lebbe C, Jouary T, Schadendorf D, Ribas A, O’Day SJ, Sosman JA, Kirkwood JM, Eggermont AM, Dreno B, Nolop K, Li J, Nelson B, Hou J, Lee RJ, Flaherty KT, McArthur GA (2011) Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med 364:2507–2516

    PubMed  CAS  Google Scholar 

  125. Solit DB, Garraway LA, Pratilas CA, Sawai A, Getz G, Basso A, Ye Q, Lobo JM, She Y, Osman I, Golub TR, Sebolt-Leopold J, Sellers WR, Rosen N (2006) BRAF mutation predicts sensitivity to MEK inhibition. Nature 439:358–362

    PubMed  CAS  Google Scholar 

  126. Smalley KS, Contractor R, Haass NK, Lee JT, Nathanson KL, Medina CA, Flaherty KT, Herlyn M (2007) Ki67 expression levels are a better marker of reduced melanoma growth following MEK inhibitor treatment than phospho-ERK levels. Br J Cancer 96:445–449

    PubMed  CAS  Google Scholar 

  127. Adjei AA, Cohen RB, Franklin W, Morris C, Wilson D, Molina JR, Hanson LJ, Gore L, Chow L, Leong S, Maloney L, Gordon G, Simmons H, Marlow A, Litwiler K, Brown S, Poch G, Kane K, Haney J, Eckhardt SG (2008) Phase I pharmacokinetic and pharmacodynamic study of the oral, small-molecule mitogen-activated protein kinase kinase 1/2 inhibitor AZD6244 (ARRY-142886) in patients with advanced cancers. J Clin Oncol 26:2139–2146

    PubMed  CAS  Google Scholar 

  128. Packer LM, East P, Reis-Filho JS, Marais R (2009) Identification of direct transcriptional targets of (V600E)BRAF/MEK signalling in melanoma. Pigment Cell Melanoma Res 22:785–798

    PubMed  CAS  Google Scholar 

  129. Pollock PM, Harper UL, Hansen KS, Yudt LM, Stark M, Robbins CM, Moses TY, Hostetter G, Wagner U, Kakareka J, Salem G, Pohida T, Heenan P, Duray P, Kallioniemi O, Hayward NK, Trent JM, Meltzer PS (2003) High frequency of BRAF mutations in nevi. Nat Genet 33:19–20

    PubMed  CAS  Google Scholar 

  130. Wu H, Goel V, Haluska FG (2003) PTEN signaling pathways in melanoma. Oncogene 22:3113–3122

    PubMed  CAS  Google Scholar 

  131. Smalley KS, Flaherty KT (2009) Integrating BRAF/MEK inhibitors into combination therapy for melanoma. Br J Cancer 100:431–435

    PubMed  CAS  Google Scholar 

  132. Villanueva J, Vultur A, Lee JT, Somasundaram R, Fukunaga-Kalabis M, Cipolla AK, Wubbenhorst B, Xu X, Gimotty PA, Kee D, Santiago-Walker AE, Letrero R, D’Andrea K, Pushparajan A, Hayden JE, Brown KD, Laquerre S, McArthur GA, Sosman JA, Nathanson KL, Herlyn M (2010) Acquired resistance to BRAF inhibitors mediated by a RAF kinase switch in melanoma can be overcome by cotargeting MEK and IGF-1R/PI3K. Cancer Cell 18:683–695

    PubMed  CAS  Google Scholar 

  133. Paraiso KH, Xiang Y, Rebecca VW, Abel EV, Chen YA, Munko AC, Wood E, Fedorenko IV, Sondak VK, Anderson AR, Ribas A, Palma MD, Nathanson KL, Koomen JM, Messina JL, Smalley KS (2011) PTEN loss confers BRAF inhibitor resistance to melanoma cells through the suppression of BIM expression. Cancer Res 71:2750–2760

    PubMed  CAS  Google Scholar 

  134. Gopal YN, Deng W, Woodman SE, Komurov K, Ram P, Smith PD, Davies MA (2010) Basal and treatment-induced activation of AKT mediates resistance to cell death by AZD6244 (ARRY-142886) in Braf-mutant human cutaneous melanoma cells. Cancer Res 70:8736–8747

    PubMed  CAS  Google Scholar 

  135. Xing F, Persaud Y, Pratilas CA, Taylor BS, Janakiraman M, She QB, Gallardo H, Liu C, Merghoub T, Hefter B, Dolgalev I, Viale A, Heguy A, De Stanchina E, Cobrinik D, Bollag G, Wolchok J, Houghton A, Solit DB (2011) Concurrent loss of the PTEN and RB1 tumor suppressors attenuates RAF dependence in melanomas harboring (V600E)BRAF. Oncogene. doi:10.1038/onc.2011.250

  136. Raftopoulou M, Etienne-Manneville S, Self A, Nicholls S, Hall A (2004) Regulation of cell migration by the C2 domain of the tumor suppressor PTEN. Science 303:1179–1181

    PubMed  CAS  Google Scholar 

  137. Torres J, Pulido R (2001) The tumor suppressor PTEN is phosphorylated by the protein kinase CK2 at its C terminus. Implications for PTEN stability to proteasome-mediated degradation. J Biol Chem 276:993–998

    PubMed  CAS  Google Scholar 

  138. Torres J, Rodriguez J, Myers MP, Valiente M, Graves JD, Tonks NK, Pulido R (2003) Phosphorylation-regulated cleavage of the tumor suppressor PTEN by caspase-3: implications for the control of protein stability and PTEN-protein interactions. J Biol Chem 278:30652–30660

    PubMed  CAS  Google Scholar 

  139. Myers MP, Tonks NK (1997) PTEN: sometimes taking it off can be better than putting it on. Am J Hum Genet 61:1234–1238

    PubMed  CAS  Google Scholar 

  140. Rodrigues GA, Falasca M, Zhang Z, Ong SH, Schlessinger J (2000) A novel positive feedback loop mediated by the docking protein Gab1 and phosphatidylinositol 3-kinase in epidermal growth factor receptor signaling. Mol Cell Biol 20:1448–1459

    PubMed  CAS  Google Scholar 

  141. Mahimainathan L, Choudhury GG (2004) Inactivation of platelet-derived growth factor receptor by the tumor suppressor PTEN provides a novel mechanism of action of the phosphatase. J Biol Chem 279:15258–15268

    PubMed  CAS  Google Scholar 

  142. Myers MP, Pass I, Batty IH, Van der Kaay J, Stolarov JP, Hemmings BA, Wigler MH, Downes CP, Tonks NK (1998) The lipid phosphatase activity of PTEN is critical for its tumor suppressor function. Proc Natl Acad Sci USA 95:13513–13518

    PubMed  CAS  Google Scholar 

  143. Steelman LS, Pohnert SC, Shelton JG, Franklin RA, Bertrand FE, McCubrey JA (2004) JAK/STAT, Raf/MEK/ERK, PI3K/Akt and BCR-ABL in cell cycle progression and leukemogenesis. Leukemia 18:189–218

    PubMed  CAS  Google Scholar 

  144. Shen WH, Balajee AS, Wang J, Wu H, Eng C, Pandolfi PP, Yin Y (2007) Essential role for nuclear PTEN in maintaining chromosomal integrity. Cell 128:157–170

    PubMed  CAS  Google Scholar 

  145. Song MS, Carracedo A, Salmena L, Song SJ, Egia A, Malumbres M, Pandolfi PP (2011) Nuclear PTEN regulates the APC-CDH1 tumor-suppressive complex in a phosphatase-independent manner. Cell 144:187–199

    PubMed  CAS  Google Scholar 

  146. Qiao X, Zhang L, Gamper AM, Fujita T, Wan Y (2010) APC/C-Cdh1: from cell cycle to cellular differentiation and genomic integrity. Cell Cycle 9:3904–3912

    PubMed  CAS  Google Scholar 

  147. Gast A, Scherer D, Chen B, Bloethner S, Melchert S, Sucker A, Hemminki K, Schadendorf D, Kumar R (2010) Somatic alterations in the melanoma genome: a high-resolution array-based comparative genomic hybridization study. Genes Chromosomes Cancer 49:733–745

    PubMed  CAS  Google Scholar 

  148. Daniotti M, Oggionni M, Ranzani T, Vallacchi V, Campi V, Di Stasi D, Torre GD, Perrone F, Luoni C, Suardi S, Frattini M, Pilotti S, Anichini A, Tragni G, Parmiani G, Pierotti MA, Rodolfo M (2004) BRAF alterations are associated with complex mutational profiles in malignant melanoma. Oncogene 23:5968–5977

    PubMed  CAS  Google Scholar 

  149. Packer L, Pavey S, Parker A, Stark M, Johansson P, Clarke B, Pollock P, Ringner M, Hayward N (2006) Osteopontin is a downstream effector of the PI3-kinase pathway in melanomas that is inversely correlated with functional PTEN. Carcinogenesis 27:1778–1786

    PubMed  CAS  Google Scholar 

Download references

Acknowledgments

This work is funded by research Grants from the Canadian Institutes of Health Research (MOP-84559, MOP-93810 and MOP-110974), Canadian Cancer Society Research Institute (2011-700714) and Canadian Dermatology Foundation to G.L. A.A.T. receives postdoctoral fellowship from Canadian Institutes of Health Research Skin Research Training Centre.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Gang Li.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Aguissa-Touré, AH., Li, G. Genetic alterations of PTEN in human melanoma. Cell. Mol. Life Sci. 69, 1475–1491 (2012). https://doi.org/10.1007/s00018-011-0878-0

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00018-011-0878-0

Keywords

Navigation