J Gynecol Oncol.  2013 Oct;24(4):376-381. 10.3802/jgo.2013.24.4.376.

The roles of ARID1A in gynecologic cancer

Affiliations
  • 1Department of Pathology, National Taiwan University College of Medicine, Taipei, Taiwan. tlmao@ntu.edu.tw
  • 2Departments of Gynecology and Obstetrics, Pathology and Oncology, Johns Hopkins Medical Institutions, Baltimore, MD, USA.

Abstract

One of the exciting findings in recent cancer genome studies is the discovery of somatic mutations in several chromatin remodeling genes. These studies not only illuminate the emerging roles of chromatin remodeling in the pathogenesis of human cancer but also provide molecular genetic basis of aberrant epigenomic regulation as one of the key mechanisms driving cancer development. This is because chromatin remodeling influences a variety of DNA activities such as replication, transcription, repair, methylation, and recombination. Among the mutated chromatin remodeling genes reported, ARID1A is frequently mutated in a variety of human cancers, especially in endometrium-related neoplasms including ovarian clear cell carcinoma, ovarian endometrioid carcinomas, and uterine endometrioid carcinomas, all of which arise from endometrial epithelium. This review will summarize the recent advances in studying the roles of ARID1A mutations in gynecologic cancers with special emphasis on how this new knowledge will further extend our understanding of the pathogenesis of endometrium-related carcinomas.

Keyword

ARID1A; BAF250a; Chromatin remodeling; Endometriosis; Ovarian cancer

MeSH Terms

Carcinoma, Endometrioid
Chromatin Assembly and Disassembly
DNA
Endometriosis
Epigenomics
Epithelium
Female
Genome
Humans
Methylation
Molecular Biology
Ovarian Neoplasms
Recombination, Genetic
DNA

Figure

  • Fig. 1 Immunohistochemical study of an endometrial endometrioid carcinoma with coexisting high-grade (A) and low-grade (B, C) components. The high-grade component is negative for ARID1A (D) while the low-grade component has areas with retained ARID1A staining (E) and areas with clonal loss of ARID1A (F) (A-F, ×200).


Cited by  1 articles

Loss of Tumor Suppressor ARID1A Protein Expression Correlates with Poor Prognosis in Patients with Primary Breast Cancer
Hyun Deuk Cho, Jong Eun Lee, Hae Yoen Jung, Mee-Hye Oh, Ji-Hye Lee, Si-Hyong Jang, Kyung-Ju Kim, Sun Wook Han, Sung Yong Kim, Han Jo Kim, Sang Byung Bae, Hyun Ju Lee
J Breast Cancer. 2015;18(4):339-346.    doi: 10.4048/jbc.2015.18.4.339.


Reference

1. Reisman D, Glaros S, Thompson EA. The SWI/SNF complex and cancer. Oncogene. 2009; 28:1653–1668.
2. Van Rechem C, Boulay G, Leprince D. HIC1 interacts with a specific subunit of SWI/SNF complexes, ARID1A/BAF250A. Biochem Biophys Res Commun. 2009; 385:586–590.
3. Gao X, Tate P, Hu P, Tjian R, Skarnes WC, Wang Z. ES cell pluripotency and germ-layer formation require the SWI/SNF chromatin remodeling component BAF250a. Proc Natl Acad Sci U S A. 2008; 105:6656–6661.
4. Ho L, Crabtree GR. Chromatin remodelling during development. Nature. 2010; 463:474–484.
5. Biegel JA, Kalpana G, Knudsen ES, Packer RJ, Roberts CW, Thiele CJ, et al. The role of INI1 and the SWI/SNF complex in the development of rhabdoid tumors: meeting summary from the workshop on childhood atypical teratoid/rhabdoid tumors. Cancer Res. 2002; 62:323–328.
6. Biegel JA, Pollack IF. Molecular analysis of pediatric brain tumors. Curr Oncol Rep. 2004; 6:445–452.
7. Bourdeaut F, Freneaux P, Thuille B, Lellouch-Tubiana A, Nicolas A, Couturier J, et al. hSNF5/INI1-deficient tumours and rhabdoid tumours are convergent but not fully overlapping entities. J Pathol. 2007; 211:323–330.
8. Reisman DN, Sciarrotta J, Wang W, Funkhouser WK, Weissman BE. Loss of BRG1/BRM in human lung cancer cell lines and primary lung cancers: correlation with poor prognosis. Cancer Res. 2003; 63:560–566.
9. Fukuoka J, Fujii T, Shih JH, Dracheva T, Meerzaman D, Player A, et al. Chromatin remodeling factors and BRM/BRG1 expression as prognostic indicators in non-small cell lung cancer. Clin Cancer Res. 2004; 10:4314–4324.
10. Chunder N, Mandal S, Basu D, Roy A, Roychoudhury S, Panda CK. Deletion mapping of chromosome 1 in early onset and late onset breast tumors: a comparative study in eastern India. Pathol Res Pract. 2003; 199:313–321.
11. Huang J, Zhao YL, Li Y, Fletcher JA, Xiao S. Genomic and functional evidence for an ARID1A tumor suppressor role. Genes Chromosomes Cancer. 2007; 46:745–750.
12. Wang X, Nagl NG Jr, Flowers S, Zweitzig D, Dallas PB, Moran E. Expression of p270 (ARID1A), a component of human SWI/SNF complexes, in human tumors. Int J Cancer. 2004; 112:636.
13. Jones S, Wang TL, Shih IeM, Mao TL, Nakayama K, Roden R, et al. Frequent mutations of chromatin remodeling gene ARID1A in ovarian clear cell carcinoma. Science. 2010; 330:228–231.
14. Wiegand KC, Shah SP, Al-Agha OM, Zhao Y, Tse K, Zeng T, et al. ARID1A mutations in endometriosis-associated ovarian carcinomas. N Engl J Med. 2010; 363:1532–1543.
15. Cornen S, Adelaide J, Bertucci F, Finetti P, Guille A, Birnbaum DJ, et al. Mutations and deletions of ARID1A in breast tumors. Oncogene. 2012; 31:4255–4256.
16. Jones S, Li M, Parsons DW, Zhang X, Wesseling J, Kristel P, et al. Somatic mutations in the chromatin remodeling gene ARID1A occur in several tumor types. Hum Mutat. 2012; 33:100–103.
17. Agrawal N, Jiao Y, Bettegowda C, Hutfless SM, Wang Y, David S, et al. Comparative genomic analysis of esophageal adenocarcinoma and squamous cell carcinoma. Cancer Discov. 2012; 2:899–905.
18. Streppel MM, Lata S, Delabastide M, Montgomery EA, Wang JS, Canto MI, et al. Next-generation sequencing of endoscopic biopsies identifies ARID1A as a tumor-suppressor gene in Barrett's esophagus. Oncogene. 2013; 01. 14. [Epub]. http://dx.doi.org/10.1038/onc.2012.586.
19. Zang ZJ, Cutcutache I, Poon SL, Zhang SL, McPherson JR, Tao J, et al. Exome sequencing of gastric adenocarcinoma identifies recurrent somatic mutations in cell adhesion and chromatin remodeling genes. Nat Genet. 2012; 44:570–574.
20. Gui Y, Guo G, Huang Y, Hu X, Tang A, Gao S, et al. Frequent mutations of chromatin remodeling genes in transitional cell carcinoma of the bladder. Nat Genet. 2011; 43:875–878.
21. Shain AH, Giacomini CP, Matsukuma K, Karikari CA, Bashyam MD, Hidalgo M, et al. Convergent structural alterations define SWItch/Sucrose NonFermentable (SWI/SNF) chromatin remodeler as a central tumor suppressive complex in pancreatic cancer. Proc Natl Acad Sci U S A. 2012; 109:E252–E259.
22. Huang J, Deng Q, Wang Q, Li KY, Dai JH, Li N, et al. Exome sequencing of hepatitis B virus-associated hepatocellular carcinoma. Nat Genet. 2012; 44:1117–1121.
23. Fujimoto A, Totoki Y, Abe T, Boroevich KA, Hosoda F, Nguyen HH, et al. Whole-genome sequencing of liver cancers identifies etiological influences on mutation patterns and recurrent mutations in chromatin regulators. Nat Genet. 2012; 44:760–764.
24. Sausen M, Leary RJ, Jones S, Wu J, Reynolds CP, Liu X, et al. Integrated genomic analyses identify ARID1A and ARID1B alterations in the childhood cancer neuroblastoma. Nat Genet. 2013; 45:12–17.
25. Giulino-Roth L, Wang K, MacDonald TY, Mathew S, Tam Y, Cronin MT, et al. Targeted genomic sequencing of pediatric Burkitt lymphoma identifies recurrent alterations in antiapoptotic and chromatin-remodeling genes. Blood. 2012; 120:5181–5184.
26. Guan B, Wang TL, Shih IeM. ARID1A, a factor that promotes formation of SWI/SNF-mediated chromatin remodeling, is a tumor suppressor in gynecologic cancers. Cancer Res. 2011; 71:6718–6727.
27. Guan B, Gao M, Wu CH, Wang TL, Shih IeM. Functional analysis of in-frame indel ARID1A mutations reveals new regulatory mechanisms of its tumor suppressor functions. Neoplasia. 2012; 14:986–993.
28. Maeda D, Mao TL, Fukayama M, Nakagawa S, Yano T, Taketani Y, et al. Clinicopathological significance of loss of ARID1A immunoreactivity in ovarian clear cell carcinoma. Int J Mol Sci. 2010; 11:5120–5128.
29. Guan B, Mao TL, Panuganti PK, Kuhn E, Kurman RJ, Maeda D, et al. Mutation and loss of expression of ARID1A in uterine low-grade endometrioid carcinoma. Am J Surg Pathol. 2011; 35:625–632.
30. Veras E, Mao TL, Ayhan A, Ueda S, Lai H, Hayran M, et al. Cystic and adenofibromatous clear cell carcinomas of the ovary: distinctive tumors that differ in their pathogenesis and behavior: a clinicopathologic analysis of 122 cases. Am J Surg Pathol. 2009; 33:844–853.
31. Yamamoto S, Tsuda H, Takano M, Tamai S, Matsubara O. Loss of ARID1A protein expression occurs as an early event in ovarian clear-cell carcinoma development and frequently coexists with PIK3CA mutations. Mod Pathol. 2012; 25:615–624.
32. Ayhan A, Mao TL, Seckin T, Wu CH, Guan B, Ogawa H, et al. Loss of ARID1A expression is an early molecular event in tumor progression from ovarian endometriotic cyst to clear cell and endometrioid carcinoma. Int J Gynecol Cancer. 2012; 22:1310–1315.
33. Kuo KT, Mao TL, Jones S, Veras E, Ayhan A, Wang TL, et al. Frequent activating mutations of PIK3CA in ovarian clear cell carcinoma. Am J Pathol. 2009; 174:1597–1601.
34. Yamamoto S, Tsuda H, Takano M, Iwaya K, Tamai S, Matsubara O. PIK3CA mutation is an early event in the development of endometriosis-associated ovarian clear cell adenocarcinoma. J Pathol. 2011; 225:189–194.
35. Yamamoto S, Tsuda H, Takano M, Tamai S, Matsubara O. PIK3CA mutations and loss of ARID1A protein expression are early events in the development of cystic ovarian clear cell adenocarcinoma. Virchows Arch. 2012; 460:77–87.
36. Wu CH, Mao TL, Vang R, Ayhan A, Wang TL, Kurman RJ, et al. Endocervical-type mucinous borderline tumors are related to endometrioid tumors based on mutation and loss of expression of ARID1A. Int J Gynecol Pathol. 2012; 31:297–303.
37. Maeda D, Shih IeM. Pathogenesis and the role of ARID1A mutation in endometriosis-related ovarian neoplasms. Adv Anat Pathol. 2013; 20:45–52.
38. Vercellini P, Crosignani P, Somigliana E, Vigano P, Buggio L, Bolis G, et al. The 'incessant menstruation' hypothesis: a mechanistic ovarian cancer model with implications for prevention. Hum Reprod. 2011; 26:2262–2273.
39. Yamaguchi K, Mandai M, Toyokuni S, Hamanishi J, Higuchi T, Takakura K, et al. Contents of endometriotic cysts, especially the high concentration of free iron, are a possible cause of carcinogenesis in the cysts through the iron-induced persistent oxidative stress. Clin Cancer Res. 2008; 14:32–40.
40. Wiegand KC, Lee AF, Al-Agha OM, Chow C, Kalloger SE, Scott DW, et al. Loss of BAF250a (ARID1A) is frequent in high-grade endometrial carcinomas. J Pathol. 2011; 224:328–333.
41. Liang H, Cheung LW, Li J, Ju Z, Yu S, Stemke-Hale K, et al. Whole-exome sequencing combined with functional genomics reveals novel candidate driver cancer genes in endometrial cancer. Genome Res. 2012; 22:2120–2129.
42. Llobet D, Pallares J, Yeramian A, Santacana M, Eritja N, Velasco A, et al. Molecular pathology of endometrial carcinoma: practical aspects from the diagnostic and therapeutic viewpoints. J Clin Pathol. 2009; 62:777–785.
43. Cheung LW, Hennessy BT, Li J, Yu S, Myers AP, Djordjevic B, et al. High frequency of PIK3R1 and PIK3R2 mutations in endometrial cancer elucidates a novel mechanism for regulation of PTEN protein stability. Cancer Discov. 2011; 1:170–185.
44. McConechy MK, Ding J, Cheang MC, Wiegand KC, Senz J, Tone AA, et al. Use of mutation profiles to refine the classification of endometrial carcinomas. J Pathol. 2012; 228:20–30.
45. Bosse T, Ter Haar NT, Seeber LM, Diest PJ, Hes FJ, Vasen HF, et al. Loss of ARID1A expression and its relationship with PI3K-Akt pathway alterations, TP53 and microsatellite instability in endometrial cancer. Mod Pathol. 2013; 05. 24. [Epub]. http://dx.doi.org/10.1038/modpathol.2013.96.
46. Allo G, Bernardini MQ, Wu RC, Shih IM, Kalloger S, Pollett A, et al. ARID1A loss correlates with mismatch repair deficiency and intact p53 expression in high-grade endometrial carcinomas. Mod Pathol. 2013; 07. 26. [Epub]. http://dx.doi.org/10.1038/modpathol.2013.144.
47. Mao TL, Ardighieri L, Ayhan A, Kuo KT, Wu CH, Wang TL, et al. Loss of ARID1A expression correlates with stages of tumor progression in uterine endometrioid carcinoma. Am J Surg Pathol. 2013; 37:1342–1348.
48. Lowery WJ, Schildkraut JM, Akushevich L, Bentley R, Marks JR, Huntsman D, et al. Loss of ARID1A-associated protein expression is a frequent event in clear cell and endometrioid ovarian cancers. Int J Gynecol Cancer. 2012; 22:9–14.
49. Fadare O, Renshaw IL, Liang SX. Does the loss of ARID1A (BAF-250a) expression in endometrial clear cell carcinomas have any clinicopathologic significance? A pilot assessment. J Cancer. 2012; 3:129–136.
50. Fadare O, Gwin K, Desouki MM, Crispens MA, Jones HW 3rd, Khabele D, et al. The clinicopathologic significance of p53 and BAF-250a (ARID1A) expression in clear cell carcinoma of the endometrium. Mod Pathol. 2013; 26:1101–1110.
51. Katagiri A, Nakayama K, Rahman MT, Rahman M, Katagiri H, Nakayama N, et al. Loss of ARID1A expression is related to shorter progression-free survival and chemoresistance in ovarian clear cell carcinoma. Mod Pathol. 2012; 25:282–288.
52. Werner HM, Berg A, Wik E, Birkeland E, Krakstad C, Kusonmano K, et al. ARID1A loss is prevalent in endometrial hyperplasia with atypia and low-grade endometrioid carcinomas. Mod Pathol. 2013; 26:428–434.
53. Wu JN, Roberts CW. ARID1A mutations in cancer: another epigenetic tumor suppressor? Cancer Discov. 2013; 3:35–43.
54. Papamichos-Chronakis M, Peterson CL. Chromatin and the genome integrity network. Nat Rev Genet. 2013; 14:62–75.
Full Text Links
  • JGO
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr