Aberrant chromatin remodeling in gynecological cancer (Review)

  • Authors:
    • Ryuichiro Okawa
    • Kouji Banno
    • Miho Iida
    • Megumi Yanokura
    • Takashi Takeda
    • Moito Iijima
    • Haruko Kunitomi‑Irie
    • Kanako Nakamura
    • Masataka Adachi
    • Kiyoko Umene
    • Yuya Nogami
    • Kenta Masuda
    • Yusuke Kobayashi
    • Eiichiro Tominaga
    • Daisuke Aoki
  • View Affiliations

  • Published online on: September 6, 2017     https://doi.org/10.3892/ol.2017.6891
  • Pages: 5107-5113
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Epigenetic regulatory mechanisms are a current focus in studies investigating cancer. Chromatin remodeling alters chromatin structure and regulates gene expression, and aberrant chromatin remodeling is involved in carcinogenesis. AT‑rich interactive domain‑containing protein 1A (ARID1A) and SWItch/sucrose non‑fermentable‑related, matrix‑associated, actin‑dependent regulator of chromatin, subfamily a, member 4 are remodeling factors that are mutated in numerous types of cancer. In gynecological cancer, ARID1A mutations have been identified in 46‑57% of clear cell carcinoma and 30% of endometrioid carcinoma. Mutations of chromodomain helicase, DNA‑binding protein 4 have been detected in 17‑21% of endometrial serous cancer, and mutations of ARID1A and mixed‑lineage leukemia 3 occur in 36 and 27% of uterine carcinosarcoma, respectively. These data suggest that aberrant chromatin remodeling is a potential cause of cancer, and have led to the development of novel proteins targeting these processes. Additional accumulation of information on the mechanisms of chromatin remodeling and markers for these events may promote personalized anticancer therapies.

Introduction

Epigenetics involves the regulation of gene expression without a change in DNA sequence. Somatic cells retain and transfer epigenetic information based on DNA methylation, histone methylation, acetylation, ubiquitination, ADP ribosylation, histone modification, small RNAs unrelated to genetic codes and modification of chromatin structure through chromatin remodeling. The term ‘chromatin remodeling’ refers to the alteration of chromatin structure from a closed state to a loosened one, which is termed ‘euchromatin’ (1). There are a few types of chromatin remodeling complexes, including the SWItch/sucrose non-fermentable (SWI/SNF) complex, which has several subunits, including ARID1A and brahma homologue (BRM)-related gene 1 (BRG1; also referred to as SMARCA4) (2). Through the interaction between subunits, chromatin remodeling complexes change chromatin structure, and this determines gene expression levels via the regulation of the interaction between proteins with double-stranded DNA (3). This change in accessibility may be achieved by adenosine triphosphate (ATP)-dependent complexes modulating histone-DNA association and by covalent modification of core nucleosomal histones mediating the transcriptional activity (4). Epigenetics is also associated with intracellular communication (5). These are key events in cell growth, and thus epigenetic abnormalities may induce carcinogenesis, developmental defects and multifactorial disease. The association between aberrant chromatin remodeling with gynecological cancer is discussed in the present review.

Chromatin remodeling

In eukaryotes, almost all genomic DNA is packaged by core histones to form chromatin structures. These structures change in events such as transcription, replication, modification and recombination of genomic DNA (6). The requirement for different chromatin structures is fulfilled by chromatin remodeling, which is an important factor in the regulation of gene expression.

Chromatin remodeling is performed by two enzyme groups: Histone modifiers, which chemically alter histones; and ATP-dependent chromatin remodeling factors, which bind to nucleosome cores and surrounding DNA to change the chromatin structure. Using energy from ATP dephosphorylation, remodeling factors alter nucleosomal structure, transiently loosen binding with DNA, and coordinate with specific chaperones, exchanging specific or all nucleosome cores (4). The nucleosomal structure is dynamically changed by remodeling factors, resulting in prompt changes in the chromatin structure (6). Several types of ATP-dependent remodeling factors are known, including a number of high-molecular-weight protein complexes with >10 subunits (6). The activity of these complexes is regulated and they are transferred to specific DNA sites to regulate gene expression by changing the chromatin structure (6,7). ATP-dependent remodeling factors are classified into several families: SWI/SNF, imitation SWI (ISWI), INO80, SWR1, nucleosome remodeling deacetylase (NuRD)/Mi2/CHD and nucleosome remodeling factor (7).

Aberrant chromatin remodeling and cancer

Chromatin remodeling factors regulate epigenetic gene expression, and aberrations in this process may induce carcinogenesis. A large-scale study of genome sequences has identified mutations of genes encoding remodeling factors in a number of types of human cancer, including those for the SWI/SNF complex, which has led to the suggestion that SWI/SNF complexes are protective against cancer (7,8). Mutations in SWI/SNF-related, matrix-associated, actin-dependent regulator of chromatin, subfamily a, member 4 (SMRCA4/BRG1), which encodes the ATPase subunit of the SWI/SNF complex, has been detected in >30% of non-small cell lung carcinoma (NSCLC) (7). Similarly, mutations in the ARID1A, which encodes an additional subunit of the SWI/SNF complex, has been detected in 46–57% of clear cell carcinoma and 30% of endometrioid carcinoma in ovarian cancer (9). ARID1A mutations also occur in 13% of hepatocellular carcinoma (HCC), 9.6% of gastrointestinal adenocarcinoma and 2.5% of malignant melanoma (7). Chromodomain helicase, DNA-binding protein 4 (CHD4), which forms the nucleosome remodeling and deacetylase (NuRD) complex, is overexpressed or mutated in serous endometrial cancer, and metastasis-associated protein 1 overexpression has been detected in breast cancer (10).

Deleted regions encoding mixed-lineage leukemia protein 3 (MLL3) produce chromosomal aberrations that are frequently associated with acute myeloid leukemia (AML) (11). Similar gene mutations are identified in medulloblastoma, HCC (12), bladder carcinoma (13), prostate cancer (14), colorectal cancer (15), gastric adenocarcinoma (16), NSCLC (17), breast cancer (18) and pancreatic cancer (19) and in AML (11). Je et al (20) revealed mutations causing a frameshift of MLL3 in 28.1% of cases of gastric cancer and 7.5% of cases of colon cancer.

Chromatin remodeling-associated gene mutations and carcinogenic mechanism

ARID1A is located at 1p35.3 and encodes an ~250-kD protein that is involved in interactions between numerous proteins, including the SWI/SNF complex. The SWI/SNF complex has multiple activities, including the following: The promotion of binding of transcription factors, coactivators and compressors; mobilization of histone-modifying enzymes; promotion of binding of nucleosomes with promoter and enhancer regions; and promotion of chromatin loop formation to induce interactions of enhancers and promoters (Fig. 1) (7,21). The SWI/SNF complex and ARID1A also regulate transcription to induce steroid hormones: It has been suggested that ARID1A may be involved in recruiting SWI/SNF to regulate genes through its ability to stimulate steroid hormone receptor-mediated transcriptional activation (22,23). Wu and Roberts (21) proposed three activities of ARID1A in the repression of tumors, namely, proliferation, differentiation and apoptosis. Gastrointestinal and breast cancer cells demonstrate a tendency to grow following ARID1A-knockdown, and growth rates decrease subsequent to re-expression of ARID1A. Ovarian epithelial cells and mouse preosteoblast cells indicated similar proliferation behaviors following ARID1A-knockdown. With regard to differentiation, ARID1A-knockdown eliminated self-renewal of ES cells and inhibited the differentiation of neurons and osteocytes in vitro. For apoptosis, the Fas apoptotic pathway in Jurkat cells was inhibited by knockdown of ARID1A. These results demonstrate that an ARID1A deficit has those three effects on tumor suppression (21). An ARID1A deficit has also been associated with the activation of the phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) signaling pathway, and with the amplification of zinc-finger protein 217 (ZNF217), which are involved in cancer development (24).

Dynamic regulation of chromatin structures to allow transcription factors to bind to DNA is necessary for gene transcription, duplication and repair. Two complexes, BRG1-associated factor (BAF) and polybromo-associated BAF (PBAF), in the SWI/SNF family, perform this role in eukaryotes (22). BRG1 and BRM are subunits containing ATPase domains that hydrolyze ATP to provide energy for translocation of nucleosomes and changes in chromatin structure (25). BRG1 binds to BRCA1 and regulates cluster of differentiation 44 expression as part of the epithelial-mesenchymal transition in cancer (25). BRG1 (also referred to as SMARCA4) is located at p13.2 on the short arm of chromosome 19 (19p13.2). BRG1 regulates DNA transcription and serves a role in tumor suppression due to remodeling of the chromatin structure. Therefore, mutations and deletions of this gene are identified in a number of cancer types, including ovarian small cell carcinoma, rhabdoid tumors (kidney and brain), medulloblastoma, lung adenocarcinoma, mantle cell lymphoma, Burkitt's lymphoma, HCC, esophageal adenocarcinoma, melanoma, non-melanoma skin cancer and intraductal papillary mucinous neoplasms of the pancreas (26).

Chromodomain helicase DNA-binding protein 4 (CHD4) is located on the short arm of chromosome 12 (12p13) and its transcription product is a molecule in the SNF2/RAD54 helicase family. CHD4 serves a key role in epigenetic transcription suppression, as it acts in nucleosome remodeling in an ATP-dependent manner, and is the major protein involved in the formation of a deacetylase complex. CHD4 exhibits tandem chromodomains in the N-terminal region and an ATPase-helicase domain in the central region. The chromodomains recognize and bind to nucleosomes and regulate interactions with chromatin, whereas the ATPase-helicase domain is involved in DNA transcription, duplication, recombination and repair (27). Mutations in these domains cause hyposegmentation in cells, indicating that the two domains are required for CHD4 function (27).

Mutations of CHD4 have been identified in several cancer types, and particularly in serous endometrial carcinoma: Zhao et al (28) revealed that 11/52 patients exhibited a heterozygous somatic CHD4 mutation. Le Gallo et al (29) also identified a somatic mutation in CHD4 in 17% of patients with serous endometrial cancer. CHD4 is characterized by ‘signature’ motifs that contain important amino acid residues required for ATP hydrolysis and helicase activity. The normal function of CHD4 is eliminated by R957Q, R1127G and R1162W mutations in these residues (30). In an immunohistological examination of lesion tissues in gastric cancer and colorectal cancer, Kim et al (30) identified no CHD4 expression in 56.4% of patients with gastric cancer and 55.7% with colorectal cancer. Insertion or deletion of 1 to 2 bases caused a somatic mutation in CHD4, with the resulting frameshift causing elimination of normal CHD4 expression (30).

MLL3 belongs to a gene cluster of the MLL family and is also called lysine N-methyltransferase 2C (KMT2C). MLL3 exhibits a histone methyltransferase SET domain, a HMG-binding domain, a nuclear receptor binding domain and 5 zinc fingers, and acts as a nuclear receptor coactivator in mammals (11). The MLL family transfers 1, 2 or 3 methyl groups to lysine K4 of methyl histone H3, and MLL3 particularly methylates H3K4 in enhancer regions (11). In a study of familial nasopharyngeal carcinoma, Sasaki et al (31) proposed that the mechanism of carcinogenesis involves the action of acquired factors such as somatic mutation and Epstein-Barr virus infection in regions containing germline mutations that frequently cause a stop codon in MLL3. In an analysis of gene mutations in patients with Lynch syndrome, Villacis et al (32) also suggested that a MLL3 mutation increases the risk of colorectal cancer.

Enhancer of zeste 2 polycomb repressive complex 2 subunit (EZH2) encodes proteins in the polycomb group (PcG) family and is located on chromosome 7 (7q35-q36). PcG proteins contribute to the epigenetic regulation of gene expression, for example: EZH2 methylates histone H3 core protein lysine 27 and inhibits gene transcription (33). EZH2 demonstrates high expression in numerous types of cancer, including breast cancer, melanoma and lung cancer (33). In gynecological cancer, high EZH2 expression occurs in uterine fibroids and cervical lesions. Yang et al (34) proposed a mechanism in which EZH2 inhibits the expression of the DNA mismatch repair gene Mutator S protein homolog 2 (MSH2) and develops uterine fibroids. Cai et al (35) revealed that EZH2 was expressed more frequently in cervical cancer tissues compared with normal tissues, and that cisplatin resistance in cervical cancer was increased by the inhibition of endogenous EZH2 expression with short hairpin RNA. Furthermore, an overexpression of EZH2 has been identified in 66% of tumors and 67% of endothelial cells of tumor vessels in patients with ovarian cancer (36). Patients with high expression of EZH2 in tumors exhibited a significantly poorer prognosis compared with those without high expression. The inactivation of EZH2 expression increases apoptosis of cancer cells, decreases the number of vessels in tumor tissues and reduces the growth of ovarian cancer cells (36).

Aberrant chromatin remodeling and ovarian cancer

Ovarian clear cell carcinoma (OCCC) is a chemoresistant cancer due to delayed cell division (37). OCCC exhibits two carcinogenic pathways, which are referred to as the adenofibroma-carcinoma and endometriosis-carcinoma sequences (38,39). The differences in the genetic backgrounds of these two pathways are unclear, but the ARID1A mutation has been suggested to be involved in the onset of OCCC via the endometriosis-carcinoma sequence, rather than via the adenofibroma-carcinoma sequence (38,39). Jones et al (9) detected ARID1A mutations in 24 (57%) of 42 patients with OCCC, and concluded that ARID1A is a tumor suppressor gene and that ARID1A mutation inactivates gene products through the aberrant chromatin remodeling associated with OCCC pathogenesis. ARID1A encodes a component of the SWI/SNF complex, which regulates cell growth, controls cell cycle regulation and cell division and repairs DNA (40,41).

Wiegand et al (42) detected an ARID1A mutation in 55 (46%) of 119 patients with OCCC and identified a deficit in BAF250a, a protein encoded by ARID1A, in 36% of these patients (Table I). BAF250a gives specificity to the SWI/SNF complex and enables regulation of gene expression (22). Furthermore, ARID1A mutations and BAF250a deficits were identified in OCCC and adjacent endometriotic lesions, but not in distant lesions, which suggests that this mutation and resultant BAF250a deficit are events in the early stage of neoplastic transformation of endometriosis (2,42). A previous study confirmed that an ARID1A deficit was also an early phenomenon in endometriosis-associated ovarian cancer (EAOC) and endometriotic ovarian cysts, together with AKT protein activation and a histone H2A variant (γH2AX) (43). An ARID1A deficit has also been identified as a poor prognostic factor in patients with stage I/II OCCC, and may be a useful biological marker for the prediction of prognosis (42).

Table I.

Aberrant chromatin remodeling-associated genes in cancer.

Table I.

Aberrant chromatin remodeling-associated genes in cancer.

Gene nameMutation ratio, %Gene abnormalityType of cancer(Refs.)
ARID1A46–57MS, NS, FSOvarian clear cell carcinoma(9)
30MS, NS, FSOvarian endometrioid carcinoma(2)
13MS, NS, FSHCC(7)
9.6MS, NS, FSGastrointestinal adenocarcinoma(7)
2.5MS, NS, FSMalignant melanoma(7)
36MutationEndometrial serous carcinoma(29)
36MutationUterine CS(57)
CHD456.4FSGastric cancer(30)
55.7FSColorectal cancer(30)
21MutationEndometrial serious carcinoma(28)
7OEEndometrial carcinoma(29)
4OEEndometrial clear cell carcinoma(29)
EZH266OEOvarian cancer(36)
UnknownOEMelanoma, BC, lung cancer, cervical cancer(33,34)
MLL3<5MS, NS, FSBladder carcinoma(13)
8MS, NS, FSProstate cancer(14)
13MS, NS, FSGastric adenocarcinoma(16)
27MutationUterine CS(57)
14FSColorectal cancer(15)
28.1FSGastric cancer(20)
7.5FSColon cancer(20)
UnknownDeletionAML(21)
UnknownMS, NS, FSBC, medulloblastoma, pancreatic cancer, HCC, NSCLC(12,1719)
SMARCA4/10GM, NS, FSLung adenocarcinoma(49)
BRG131.3GM, NS, FSLung large cell carcinoma(49)
36.4GM, NS, FSLung pleomorphic carcinoma(49)
94GM, NS, FSSCCOHT(26,4547)
<30MutationNSCLC(7)
10–20MutationMelanoma, esophageal adenocarcinoma, intraductal papillary mucinous neoplasms of the pancreas(26)
UnknownMutationOSCC, rhabdoid tumor, mantle cell lymphoma, Burkitt lymphoma, non-melanoma skin cancer(26)

[i] MS, missense mutation; NS, nonsense mutation; FS, frameshift mutation; OE, overexpression; GM, germline mutation; HCC, hepatocellular carcinoma; BC, breast cancer; NSCLC, non-small cell lung carcinoma; AML, acute myeloid leukemia; OSCC, ovarian small cell carcinoma; CS, carcinosarcoma; SCCOHT, small cell carcinoma of the ovary hypercalcemic type; SMARCA4/BRG1, SWI/SNF-related, matrix-associated, actin-dependent regulator of chromatin, subfamily a, member 4; MLL3, mixed-lineage leukemia 3; EZH2, enhancer of zeste 2 polycomb repressive complex 2 subunit; CHD4, chromodomain helicase, DNA-binding protein 4; ARID1A, AT-rich interaction domain 1A.

Small cell carcinoma of the ovary, hypercalcemic type (SCCOHT) associated with hypercalcemia is a rare disease and is considered to be a rhabdoid tumor (26). SCCOHT is a poorly differentiated tumor associated with a poor prognosis that develops in young females (44). In an immunohistological study, Conlon et al (44) measured loss of SMARCA4 expression in 94% of patients with SCCOHT, whereas loss of SMARCA4 expression is usually identified in <5% of patients with ovarian cancer (44). Therefore, these data are considered to be specific to SCCOHT (44). In SCCOHT, germline mutations have been revealed in one allele of SMARCA4, and expression is deleted due to an inactivating germline mutation and frameshift and nonsense mutations in the other allele (26,4547). Rhabdoid tumors that develop in organs other than the ovary, including the kidney and brain, have germline and somatic expression of SMARCA4 (48). Immunostaining for the expression of SMARCA4 in tumor tissues of patients with lung cancer revealed downregulation of SMARCA4 in no patients with squamous cell carcinoma, in 10% with adenocarcinoma, in 31.3% with large cell carcinoma and in 36.4% with pleomorphic carcinoma (49), and somatic mutation and deletion of SMARCA4 are present in these types of cancer (26). SMARCA4 is a subunit of the BAF and PBAF complexes, and mutation and deletion produces incomplete complexes and abnormal subunits that may cause dysregulation of genes and induce disease (50).

Aberrant chromatin remodeling and endometrial cancer

Endometrial cancer includes endometrioid carcinoma and serous carcinoma, which is less common compared with endometrioid carcinoma and has a relatively poor prognosis (51). Almost all serous carcinomas are poorly differentiated type 2 endometrial cancer with myometrial, vascular and extrauterine invasion (51). In exome sequencing of endometrial serous carcinomas in 53 patients, Le Gallo et al (29) detected CHD4 mutations in 9 (17%) cases, and identified mutation of chromatin remodeling genes, including ARID1A, in 19 (36%) (29) (Table I). Similarly, Zhao et al (28) identified CHD4 mutations in 11 (21%) of 52 patients with endometrial serous carcinomas. CHD4 is a catalytic subunit of the NuRD complex that inhibits transcription and repairs DNA damage (52). CHD4 overexpression has also been revealed in 7% of endometrioid carcinomas and 4% of endometrial clear cell carcinomas, with half of CHD4 mutations affecting the ATPase/helicase domain or helicase domain, which is suspected to be the cause of endometrial cancer (29).

Carcinosarcoma (CS) is an extremely rare gynecological disease with a poor prognosis (53). Histological results of CS demonstrate mixed epithelial carcinoma and non-epithelial sarcoma (53). CS occurs commonly in the uterine body, but has also been identified in the ovary, uterine cervix and vagina (5456). The incidence in the United States is 2 per 100,000, and the 5-year survival rates are 35–65% in the early stage and ~10% in stage IV (53). In 22 patients with uterine CS, Jones et al (57) revealed ARID1A mutations in 8 (36%) cases, mutations of histone methyltransferase MLL3 in 6 (27%) cases, mutations of speckle-type POZ protein (SPOP), which is involved in chromatin remodeling, in 3 (14%) cases, and mutations of chromatin remodeling-associated genes in 14 (64%) cases (57). ARID1A serves an important role in the regulation of cell growth, and MLL3 is a coactivator of tumor protein p53 (TP53), a tumor suppressor p53 gene (2,58). SPOP is a transcriptional repressor of p53 via the bric-a-brac/tramtrack/broad complex protein (59). Jones et al (57) suggested that a specific tissue-type of uterine CS depends on aberrant chromatin remodeling. Therefore, a complete understanding of genetic mutations in this cancer will be useful for diagnosis, early detection and treatment.

Therapy targeting aberrant chromatin remodeling

Cancer cells with an ARID1A deficit are highly sensitive to small molecule inhibitors in the PI3K/AKT signal transduction pathway. Therefore, drugs that inhibit this pathway are effective in patients with cancer with an ARID1A deficit (60). Therapy targeting epigenetic regulatory mechanisms in cancer cells is also under development. Bitler et al (61) focused on the activity of EZH2, a methylation factor in cancer with ARID1A mutation, and identified that proliferation of cells with an ARID1A mutation was selectively inhibited by the administration of an EZH2 inhibitor. This suggests that EZH2 inactivation is a potential therapy for cancer with ARID1A mutation, and EZH2 inhibition has been demonstrated to reduce the number of ovarian tumors with ARID1A mutations in vivo. Therefore, pharmacological inhibition of EZH2 expression may be a therapeutic strategy for cancer with an ARID1A mutation (61).

Guan et al (62) demonstrated that an ARID1A in-frame mutation prevented ARID1A transport from the nucleus to the cytoplasm (62). The ARID1A protein was then degraded by the ubiquitin-proteasome system and was not available downstream, resulting in the onset of cancer. Thus, ARID1A degradation may be inhibited by targeting the ubiquitin-proteasome system in cells with an ARID1A mutation, with potential recovery of the original cancer inhibitory effect (62).

ARID1B has recently been identified as an ARID1A homolog (63). In cells with an ARID1A deficit, ARID1B is independently expressed and its proliferation is enhanced, which suggests that ARID1A and ARID1B may interact in promoting carcinogenesis. However, blocking the mechanism of ARID1B in cells with an ARID1A deficit destabilizes the SWI/SNF complex and inhibits cell proliferation. Therefore, ARID1B is also a therapeutic target in cancer with ARID1A mutation (Fig. 2) (63). Immunohistochemical detection of ARID1A expression may be a useful marker for the evaluation of malignancy, prognosis and treatment effect (64).

Conclusion

ARID1A mutation is involved in gynecological cancer types such as OCCC and uterine cancer through the induction of aberrant chromatin remodeling and promotion of tumorigenesis. Germline mutations and epigenetic regulatory mechanisms, including chromatin remodeling, are involved in carcinogenesis. Therefore, there is a requirement for methods for identifying chromatin remodeling-associated gene mutations, including ARID1A and BRG1, and for therapy targeting the carcinogenic mechanisms of aberrant chromatin remodeling.

Acknowledgements

The authors would like to thank Dr S. Fujiwara and Dr K. Hoshi (Keio University School of Medicine, Tokyo, Japan) for their assistance, and are grateful for support from the Keio Gijuku Academic Development Fund.

References

1 

Weaver IC, Korgan AC, Lee K, Wheeler RV, Hundert AS and Goguen D: Stress and the emerging roles of chromatin remodeling in signal integration and stable transmission of reversible phenotypes. Front Behav Neurosci. 11:412017. View Article : Google Scholar : PubMed/NCBI

2 

Takeda T, Banno K, Okawa R, Yanokura M, Iijima M, Irie-Kunitomi H, Nakamura K, Iida M, Adachi M, Umene K, et al: ARID1A gene mutation in ovarian and endometrial cancers (Review). Oncol Rep. 35:607–613. 2016. View Article : Google Scholar : PubMed/NCBI

3 

Clapier CR and Cairns BR: The biology of chromatin remodeling complexes. Annu Rev Biochem. 78:273–304. 2009. View Article : Google Scholar : PubMed/NCBI

4 

Ronan JL, Wu W and Crabtree GR: From neural development to cognition: Unexpected roles for chromatin. Nat Rev Genet. 14:347–359. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Huang B, Jiang C and Zhang R: Epigenetics: The language of the cell? Epigenomics. 6:73–88. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Alberts B, Johnson A, Lewis J, Raff M, Roberts K and Walter P: Molecular biology of the cell, 5th edition. Science. 215–216. 2008.PubMed/NCBI

7 

Wilson BG and Roberts CW: SWI/SNF nucleosome remodellers and cancer. Nat Rev Cancer. 11:481–492. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Oike T, Ogiwara H, Nakano T, Yokota J and Kohno T: Inactivating mutations in SWI/SNF chromatin remodeling genes in human cancer. Jpn J Clin Oncol. 43:849–855. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Jones S, Wang TL, Shih IeM, Mao TL, Nakayama K, Roden R, Glas R, Slamon D, Diaz LA Jr, Vogelstein B, et al: Frequent mutations of chromatin remodeling gene ARID1A in ovarian clear cell carcinoma. Science. 330:228–231. 2010. View Article : Google Scholar : PubMed/NCBI

10 

Mayes K, Qiu Z, Alhazmi A and Landry JW: ATP-dependent chromatin remodeling complexes as novel targets for cancer therapy. Adv Cancer Res. 121:183–233. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Li WD, Li QR, Xu SN, Wei FJ, Ye ZJ, Cheng JK and Chen JP: Exome sequencing identifies an MLL3 gene germ line mutation in a pedigree of colorectal cancer and acute myeloid leukemia. Blood. 121:1478–1479. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Fujimoto A, Totoki Y, Abe T, Boroevich KA, Hosoda F, Nguyen HH, Aoki M, Hosono N, Kubo M, Miya F, et al: Whole-genome sequencing of liver cancers identifies etiological influences on mutation patterns and recurrent mutations in chromatin regulators. Nat Genet. 44:760–764. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Gui Y, Guo G, Huang Y, Hu X, Tang A, Gao S, Wu R, Chen C, Li X, Zhou L, et al: Frequent mutations of chromatin remodeling genes in transitional cell carcinoma of the bladder. Nat Genet. 43:875–878. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Lindberg J, Mills IG, Klevebring D, Liu W, Neiman M, Xu J, Wikström P, Wiklund P, Wiklund F, Egevad L and Grönberg H: The mitochondrial and autosomal mutation landscapes of prostate cancer. Eur Urol. 63:702–708. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Watanabe Y, Castoro RJ, Kim HS, North B, Oikawa R, Hiraishi T, Ahmed SS, Chung W, Cho MY, Toyota M, et al: Frequent alteration of MLL3 frameshift mutations in microsatellite deficient colorectal cancer. PLoS One. 6:e233202011. View Article : Google Scholar : PubMed/NCBI

16 

Zang ZJ, Cutcutache I, Poon SL, Zhang SL, McPherson JR, Tao J, Rajasegaran V, Heng HL, Deng N, Gan A, et al: Exome sequencing of gastric adenocarcinoma identifies recurrent somatic mutations in cell adhesion and chromatin remodeling genes. Nat Genet. 44:570–574. 2012. View Article : Google Scholar : PubMed/NCBI

17 

Liu P, Morrison C, Wang L, Xiong D, Vedell P, Cui P, Hua X, Ding F, Lu Y, James M, et al: Identification of somatic mutations in non-small cell lung carcinomas using whole-exome sequencing. Carcinogenesis. 33:1270–1276. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Ellis MJ, Ding L, Shen D, Luo J, Suman VJ, Wallis JW, Van Tine BA, Hoog J, Goiffon RJ, Goldstein TC, et al: Whole-genome analysis informs breast cancer response to aromatase inhibition. Nature. 486:353–360. 2012.PubMed/NCBI

19 

Biankin AV, Waddell N, Kassahn KS, Gingras MC, Muthuswamy LB, Johns AL, Miller DK, Wilson PJ, Patch AM, Wu J, et al: Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature. 491:399–405. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Je EM and Lee SH, Yoo NJ and Lee SH: Mutational and expressional analysis of MLL genes in gastric and colorectal cancers with microsatellite instability. Neoplasma. 60:188–195. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Wu JN and Roberts CW: ARID1A mutations in cancer: Another epigenetic tumor suppressor? Cancer Discov. 3:35–43. 2013. View Article : Google Scholar : PubMed/NCBI

22 

Trotter KW, Fan HY, Ivey ML, Kingston RE and Archer TK: The HSA domain of BRG1 mediates critical interactions required for glucocorticoid receptor-dependent transcriptional activation in vivo. Mol Cell Biol. 28:1413–1426. 2008. View Article : Google Scholar : PubMed/NCBI

23 

Inoue H, Furukawa T, Giannakopoulos S, Zhou S, King DS and Tanese N: Largest subunits of the human SWI/SNF chromatin-remodeling complex promote transcriptional activation by steroid hormone receptors. J Biol Chem. 277:41674–41685. 2002. View Article : Google Scholar : PubMed/NCBI

24 

Huang HN, Lin MC, Huang WC, Chiang YC and Kuo KT: Loss of ARID1A expression and its relationship with PI3K-Akt pathway alterations and ZNF217 amplification in ovarian clear cell carcinoma. Mod Pathol. 27:983–990. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Biegel JA, Busse TM and Weissman BE: SWI/SNF chromatin remodeling complexes and cancer. Am J Med Genet C Semin Med Genet. 166C:350–366. 2014. View Article : Google Scholar : PubMed/NCBI

26 

Reisman D, Glaros S and Thompson EA: The SWI/SNF complex and cancer. Oncogene. 28:1653–1668. 2009. View Article : Google Scholar : PubMed/NCBI

27 

Yamada M, Sato N, Ikeda S, Arai T, Sawabe M, Mori S, Yamada Y, Muramatsu M and Tanaka M: Association of the chromodomain helicase DNA-binding protein 4 (CHD4) missense variation p.D140E with cancer: Potential interaction with smoking. Genes Chromosomes Cancer. 54:122–128. 2015. View Article : Google Scholar : PubMed/NCBI

28 

Zhao S, Choi M, Overton JD, Bellone S, Roque DM, Cocco E, Guzzo F, English DP, Varughese J, Gasparrini S, et al: Landscape of somatic single-nucleotide and copy-number mutations in uterine serous carcinoma. Proc Natl Acad Sci USA. 110:2916–2921. 2013. View Article : Google Scholar : PubMed/NCBI

29 

Le Gallo M, O'Hara AJ, Rudd ML, Urick ME, Hansen NF, O'Neil NJ, Price JC, Zhang S, England BM, Godwin AK, et al: Exome sequencing of serous endometrial tumors identifies recurrent somatic mutations in chromatin-remodeling and ubiquitin ligase complex genes. Nat Genet. 44:1310–1315. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Kim MS, Chung NG, Kang MR, Yoo NJ and Lee SH: Genetic and expressional alterations of CHD genes in gastric and colorectal cancers. Histopathology. 58:660–668. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Sasaki MM, Skol AD, Bao R, Rhodes LV, Chambers R, Vokes EE, Cohen EE and Onel K: Integrated genomic analysis suggests MLL3 is a novel candidate susceptibility gene for familial nasopharyngeal carcinoma. Cancer Epidemiol Biomarkers Prev. 24:1222–1228. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Villacis RA, Miranda PM, Gomy I, Santos EM, Carraro DM, Achatz MI, Rossi BM and Rogatto SR: Contribution of rare germline copy number variations and common susceptibility loci in Lynch syndrome patients negative for mutations in the mismatch repair genes. Int J Cancer. 138:1928–1935. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Kim KH and Roberts CW: Targeting EZH2 in cancer. Nat Med. 22:128–134. 2016. View Article : Google Scholar : PubMed/NCBI

34 

Yang Q, Laknaur A, Elam L, Ismail N, Gavrilova-Jordan L, Lue J, Diamond MP and Al-Hendy A: Identification of polycomb group protein EZH2-mediated DNA mismatch repair gene MSH2 in human uterine fibroids. Reprod Sci. 23:1314–1325. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Cai L, Wang Z and Liu D: Interference with endogenous EZH2 reverses the chemotherapy drug resistance in cervical cancer cells partly by up-regulating Dicer expression. Tumour Biol. 37:6359–6369. 2016. View Article : Google Scholar : PubMed/NCBI

36 

Lu C, Han HD, Mangala LS, Ali-Fehmi R, Newton CS, Ozbun L, Armaiz-Pena GN, Hu W, Stone RL, Munkarah A, et al: Regulation of tumor angiogenesis by EZH2. Cancer Cell. 18:185–197. 2010. View Article : Google Scholar : PubMed/NCBI

37 

Crotzer DR, Sun CC, Coleman RL, Wolf JK, Levenback CF and Gershenson DM: Lack of effective systemic therapy for recurrent clear cell carcinoma of the ovary. Gynecol Oncol. 105:404–408. 2007. View Article : Google Scholar : PubMed/NCBI

38 

Viganó P, Somigliana E, Chiodo I, Abbiati A and Vercellini P: Molecular mechanisms and biological plausibility underlying the malignant transformation of endometriosis: A critical analysis. Hum Reprod Update. 12:77–89. 2006. View Article : Google Scholar : PubMed/NCBI

39 

Nishikimi K, Kiyokawa T, Tate S, Iwamoto M and Shozu M: ARID1A expression in ovarian clear cell carcinoma with an adenofibromatous component. Histopathology. 67:866–871. 2015. View Article : Google Scholar : PubMed/NCBI

40 

Nagl NG Jr, Wang X, Patsialou A, Van Scoy M and Moran E: Distinct mammalian SWI/SNF chromatin remodeling complexes with opposing roles in cell-cycle control. EMBO J. 26:752–763. 2007. View Article : Google Scholar : PubMed/NCBI

41 

Weissman B and Knudsen KE: Hijacking the chromatin remodeling machinery: Impact of SWI/SNF perturbations in cancer. Cancer Res. 69:8223–8230. 2009. View Article : Google Scholar : PubMed/NCBI

42 

Wiegand KC, Shah SP, Al-Agha OM, Zhao Y, Tse K, Zeng T, Senz J, McConechy MK, Anglesio MS, Kalloger SE, et al: ARID1A mutations in endometriosis-associated ovarian carcinomas. N Engl J Med. 363:1532–1543. 2010. View Article : Google Scholar : PubMed/NCBI

43 

Itamochi H, Oumi N, Oishi T, Shoji T, Fujiwara H, Sugiyama T, Suzuki M, Kigawa J and Harada T: Loss of ARID1A expression is associated with poor prognosis in patients with stage I/II clear cell carcinoma of the ovary. Int J Clin Oncol. 20:967–973. 2015. View Article : Google Scholar : PubMed/NCBI

44 

Conlon N, Silva A, Guerra E, Jelinic P, Schlappe BA, Olvera N, Mueller JJ, Tornos C, Jungbluth AA, Young RH, et al: Loss of SMARCA4 expression is both sensitive and specific for the diagnosis of small cell carcinoma of ovary, hypercalcemic type. Am J Surg Pathol. 40:395–403. 2016. View Article : Google Scholar : PubMed/NCBI

45 

Jelinic P, Mueller JJ, Olvera N, Dao F, Scott SN, Shah R, Gao J, Schultz N, Gonen M, Soslow RA, et al: Recurrent SMARCA4 mutations in small cell carcinoma of the ovary. Nat Genet. 46:424–426. 2014. View Article : Google Scholar : PubMed/NCBI

46 

Ramos P, Karnezis AN, Craig DW, Sekulic A, Russell ML, Hendricks WP, Corneveaux JJ, Barrett MT, Shumansky K, Yang Y, et al: Small cell carcinoma of the ovary, hypercalcemic type, displays frequent inactivating germline and somatic mutations in SMARCA4. Nat Genet. 46:427–429. 2014. View Article : Google Scholar : PubMed/NCBI

47 

Witkowski L, Carrot-Zhang J, Albrecht S, Fahiminiya S, Hamel N, Tomiak E, Grynspan D, Saloustros E, Nadaf J, Rivera B, et al: Germline and somatic SMARCA4 mutations characterize small cell carcinoma of the ovary, hypercalcemic type. Nat Genet. 46:438–443. 2014. View Article : Google Scholar : PubMed/NCBI

48 

Schneppenheim R, Frühwald MC, Gesk S, Hasselblatt M, Jeibmann A, Kordes U, Kreuz M, Leuschner I, Subero Martin JI, Obser T, et al: Germline nonsense mutation and somatic inactivation of SMARCA4/BRG1 in a family with rhabdoid tumor predisposition syndrome. Am J Hum Genet. 86:279–284. 2010. View Article : Google Scholar : PubMed/NCBI

49 

Yoshimoto T, Matsubara D, Nakano T, Tamura T, Endo S, Sugiyama Y and Niki T: Frequent loss of the expression of multiple subunits of the SWI/SNF complex in large cell carcinoma and pleomorphic carcinoma of the lung. Pathol Int. 65:595–602. 2015. View Article : Google Scholar : PubMed/NCBI

50 

Helming KC, Wang X and Roberts CW: Vulnerabilities of mutant SWI/SNF complexes in cancer. Cancer Cell. 26:309–317. 2014. View Article : Google Scholar : PubMed/NCBI

51 

Sherman ME: Theories of endometrial carcinogenesis: A multidisciplinary approach. Mod Pathol. 13:295–308. 2000. View Article : Google Scholar : PubMed/NCBI

52 

Polo SE, Kaidi A, Baskcomb L, Galanty Y and Jackson SP: Regulation of DNA-damage responses and cell-cycle progression by the chromatin remodelling factor CHD4. EMBO J. 29:3130–3139. 2010. View Article : Google Scholar : PubMed/NCBI

53 

Duman BB, Kara IO, Günaldi M and Ercolak V: Malignant mixed Mullerian tumor of the ovary with two cases and review of the literature. Arch Gynecol Obstet. 283:1363–1368. 2011. View Article : Google Scholar : PubMed/NCBI

54 

Sharma NK, Sorosky JI, Bender D, Fletcher MS and Sood AK: Malignant mixed mullerian tumor (MMMT) of the cervix. Gynecol Oncol. 97:442–445. 2005. View Article : Google Scholar : PubMed/NCBI

55 

Ahuja A, Safaya R, Prakash G, Kumar L and Shukla NK: Primary mixed mullerian tumor of the vagina - a case report with review of the literature. Pathol Res Pract. 207:253–255. 2011. View Article : Google Scholar : PubMed/NCBI

56 

George EM, Herzog TJ, Neugut AI, Lu YS, Burke WM, Lewin SN, Hershman DL and Wright JD: Carcinosarcoma of the ovary: Natural history, patterns of treatment, and outcome. Gynecol Oncol. 131:42–45. 2013. View Article : Google Scholar : PubMed/NCBI

57 

Jones S, Stransky N, McCord CL, Cerami E, Lagowski J, Kelly D, Angiuoli SV, Sausen M, Kann L, Shukla M, et al: Genomic analyses of gynaecologic carcinosarcomas reveal frequent mutations in chromatin remodelling genes. Nat Commun. 5:50062014. View Article : Google Scholar : PubMed/NCBI

58 

Lee J, Kim DH, Lee S, Yang QH, Lee DK, Lee SK, Roeder RG and Lee JW: A tumor suppressive coactivator complex of p53 containing ASC-2 and histone H3-lysine-4 methyltransferase MLL3 or its paralogue MLL4. Proc Natl Acad Sci USA. 106:8513–8518. 2009. View Article : Google Scholar : PubMed/NCBI

59 

Kwon JE, La M, Oh KH, Oh YM, Kim GR, Seol JH, Baek SH, Chiba T, Tanaka K, Bang OS, et al: BTB domain-containing speckle-type POZ protein (SPOP) serves as an adaptor of Daxx for ubiquitination by Cul3-based ubiquitin ligase. J Biol Chem. 281:12664–12672. 2006. View Article : Google Scholar : PubMed/NCBI

60 

Samartzis EP, Gutsche K, Dedes KJ, Fink D, Stucki M and Imesch P: Loss of ARID1A expression sensitizes cancer cells to PI3K- and AKT-inhibition. Oncotarget. 5:5295–5303. 2014. View Article : Google Scholar : PubMed/NCBI

61 

Bitler BG, Aird KM, Garipov A, Li H, Amatangelo M, Kossenkov AV, Schultz DC, Liu Q, Shih IeM, Conejo-Garcia JR, et al: Synthetic lethality by targeting EZH2 methyltransferase activity in ARID1A-mutated cancers. Nat Med. 21:231–238. 2015.PubMed/NCBI

62 

Guan B, Gao M, Wu CH, Wang TL and Shih IeM: Functional analysis of in-frame indel ARID1A mutations reveals new regulatory mechanisms of its tumor suppressor functions. Neoplasia. 14:986–993. 2012. View Article : Google Scholar : PubMed/NCBI

63 

Helming KC, Wang X, Wilson BG, Vazquez F, Haswell JR, Manchester HE, Kim Y, Kryukov GV, Ghandi M, Aguirre AJ, et al: ARID1B is a specific vulnerability in ARID1A-mutant cancers. Nat Med. 20:251–254. 2014. View Article : Google Scholar : PubMed/NCBI

64 

Nagymanyoki Z, Mutter GL, Hornick JL and Cibas ES: ARID1A is a useful marker of malignancy in peritoneal washings for endometrial carcinoma. Cancer Cytopathol. 123:253–257. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2017
Volume 14 Issue 5

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Okawa R, Banno K, Iida M, Yanokura M, Takeda T, Iijima M, Kunitomi‑Irie H, Nakamura K, Adachi M, Umene K, Umene K, et al: Aberrant chromatin remodeling in gynecological cancer (Review). Oncol Lett 14: 5107-5113, 2017
APA
Okawa, R., Banno, K., Iida, M., Yanokura, M., Takeda, T., Iijima, M. ... Aoki, D. (2017). Aberrant chromatin remodeling in gynecological cancer (Review). Oncology Letters, 14, 5107-5113. https://doi.org/10.3892/ol.2017.6891
MLA
Okawa, R., Banno, K., Iida, M., Yanokura, M., Takeda, T., Iijima, M., Kunitomi‑Irie, H., Nakamura, K., Adachi, M., Umene, K., Nogami, Y., Masuda, K., Kobayashi, Y., Tominaga, E., Aoki, D."Aberrant chromatin remodeling in gynecological cancer (Review)". Oncology Letters 14.5 (2017): 5107-5113.
Chicago
Okawa, R., Banno, K., Iida, M., Yanokura, M., Takeda, T., Iijima, M., Kunitomi‑Irie, H., Nakamura, K., Adachi, M., Umene, K., Nogami, Y., Masuda, K., Kobayashi, Y., Tominaga, E., Aoki, D."Aberrant chromatin remodeling in gynecological cancer (Review)". Oncology Letters 14, no. 5 (2017): 5107-5113. https://doi.org/10.3892/ol.2017.6891