NIH Public Access Author Manuscript Crit Rev Eukaryot Gene Expr. Author manuscript; available in PMC 2014 July 21.

NIH-PA Author Manuscript

Published in final edited form as: Crit Rev Eukaryot Gene Expr. 2014 ; 24(2): 151–161.

SWI/SNF Chromatin Remodeling Enzymes in Melanocyte Differentiation and Melanoma A. Mehrotra, G. Mehta, S. Aras, A. Trivedi, and I.L. de la Serna* Department of Biochemistry and Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH

Abstract

NIH-PA Author Manuscript

Epidermal melanocytes are pigment-producing cells derived from the neural crest that protects skin from the damaging effects of solar radiation. Malignant melanoma, a highly aggressive cancer, arises from melanocytes. SWI/SNF enzymes are multiprotein complexes that remodel chromatin structure and have extensive roles in cellular differentiation. Components of the complex have been found to be mutated or lost in several human cancers. This review focuses on studies that implicate SWI/SNF enzymes in melanocyte differentiation and in melanoma.

Keywords melanoma; microphthalmia-associated transcription factor; SOX10; SWI/SNF chromatin remodeling enzymes; gene expression

I. INTRODUCTION

NIH-PA Author Manuscript

Melanocytes are pigment-producing cells found in the basal layer of epidermis, hair follicles, the inner ear, eye choroid, and heart.1 Making melanin to protect skin from the damaging effects of ultraviolet (UV) radiation from the sun is a key function of epidermal melanocytes. Melanocytes comprise approximately 2% of the cell population of the skin. They synthesize melanin in specialized organelles called melanosomes and transfer the melanosomes to keratinocytes, the predominant epidermal component. Exposure to UV radiation leads to DNA damage in melanocytes and surrounding cells of the skin and is the single most critical environmental agent that promotes the development of melanoma.2 During melanocyte differentiation, a number of key transcription factors contribute to the activation of melanocyte-specific genes, which have functions in melanin synthesis and melanosome biology, motility, and survival. Microphthalmia-associated transcription factor (MITF) is critically required for melanocyte specification, differentiation, and survival, as demonstrated by a loss of melanocytes in mice in which the expression of MITF is disrupted.3 Other transcriptional regulators including SOX10 also have crucial roles in

© 2014 Begell House, Inc. * Address all correspondence to: I.L. de la Serna, Department of Biochemistry and Cancer Biology, University of Toledo College of Medicine and Life Sciences, 3035 Arlington Ave, Toledo, OH 43614; Tel.: (419) 383-4111; Fax: (419) 383-6228; [email protected].

Mehrotra et al.

Page 2

NIH-PA Author Manuscript

melanocyte differentiation. 4 Interestingly, both SOX10 and MITF also have important roles in the oncogenic transformation of melanocytes to melanoma.5,6 How transcription factors that are intimately involved in differentiation, a process that often is associated with decreased proliferation, also contribute to oncogenic transformation is not well understood. An important consideration is the interaction of these transcription factors with chromatin remodeling enzymes to activate specific genes involved in melanocyte differentiation and melanoma. SWI/SNF chromatin remodeling enzymes have been shown to be important for the regulation of genes during cellular differentiation and in cancer.7,8

NIH-PA Author Manuscript

SWI/SNF enzymes are evolutionarily conserved, multisubunit complexes of 1–2 MDa that utilize the energy derived from adenosine triphosphate (ATP) hydrolysis to alter chromatin structure and regulate gene expression.7 Diverse SWI/SNF complexes contain a catalytic subunit—BRG1 or BRM—and 8–12 accessory proteins (BRG1/BRM-associated factors [BAFs]). Furthermore, complexes containing BRG1 can be further classified into polybromo-associated factor (PBAF) complexes, which uniquely contain BAF200 and BAF180,9 or BAF complexes, which uniquely contain BAF250A or BAF250B10 (Fig. 1). In vitro, chromatin remodeling is achieved by a core complex containing BRG1 or BRM, BAF47, BAF170, and BAF155.11 In vivo, additional BAFs are required for interactions with transcriptional activators and repressors.12,13 The BRG1 subunit and other SWI/SNF subunits, including BAF47, BAF60C, BAF155, BAF180, and BAF250A, have been shown to be essential for murine development. 14–20 Studies indicate that SWI/SNF enzymes are required for both embryonic stem cell pluripotency as well as cellular differentiation.21,22 SWI/SNF enzymes have been shown to interact with MITF and SOX10.23–25 In particular, the interaction between MITF and SWI/SNF enzymes has been demonstrated to be important for melanocyte differentiation and in melanoma.23,24 This review focuses on the studies implicating SWI/SNF enzymes in the transcriptional regulation of melanocyte differentiation and in melanoma and discusses evidence indicating that components of the SWI/SNF complex are disrupted in melanoma.

II. MELANOCYTE DIFFERENTIATION

NIH-PA Author Manuscript

Melanocytes are derived from multipotent neural crest cells that migrate from the embryonic neural tube in a dorsolateral path to populate the epidermis, hair follicles, and other regions.26 In contrast, neural crest cells that migrate ventrally give rise to neurons and Schwann cells. Some melanocytes also arise from Schwann cell precursors located in peripheral nerves.27 Melanoblasts, cells committed to the melanocyte lineage, are developmentally related to Schwann cells, each requiring the activity of SOX10.28 It is MITF, however, that commits precursors to the melanocyte lineage and is considered the master regulator of melanocyte differentiation. SOX10 is a Sry-related high-mobility group box transcriptional regulator that is required for neural crest pluripotency and survival as well as for the differentiation of several neural crest lineages, including Schwann cells and melanocytes.29 In melanocytes, SOX10 activates MITF expression and contributes to the expression of a subset of MITF target genes involved in melanin synthesis, such as tyrosinase, tyrosinase-related protein 1, and

Crit Rev Eukaryot Gene Expr. Author manuscript; available in PMC 2014 July 21.

Mehrotra et al.

Page 3

NIH-PA Author Manuscript

dopachrome tautomerase.30–33 SOX10 also is required for the differentiation of other neural crest lineages, including Schwann cells. SWI/SNF enzymes are required for SOX10 expression and for expression of SOX10 target genes during Schwann cell differentiation25,34,35 (Fig. 2). Interestingly, downregulation of the BRG1 subunit of SWI/SNF enzymes in melanoma cells compromises SOX10 expression.36 However, it is not known whether BRG1 or other components of the SWI/SNF complex interact with SOX10 during melanocyte differentiation or in melanoma.

NIH-PA Author Manuscript

MITF is a basic helix-loop-helix leucine zipper transcription factor that regulates gene expression by binding to canonical E box elements in the promoter region of its target genes either as homodimer or heterodimer with other related family members (TFE3, TFEB, and TFEC proteins).3,37 MITF plays a prominent role in the development of several different cell types, including melanocytes, osteoclasts, mast cells, and retinal pigment epithelial cells of the eye.38–40 In the melanocyte lineage, MITF promotes survival, proliferation, and differentiation of melanoblasts into melanocytes.3 It activates genes required for melanin synthesis and melanocyte function, including tyrosinase, TRP1, DCT,3 as well as genes required for survival and cell cycle regulation, including the genes encoding anti-apoptotic factors, BCL2 and melanoma inhibitor of apoptosis (ML-IAP), and cell cycle regulators (CDK2, p16INK4A, and p21WAF1/CIP1).41–45 SWI/SNF enzymes have been shown to be required for MITF-mediated activation of melanocyte specific genes as well as the expression of MITF and the activation of MITF target genes that regulate proliferation and survival23,24,46,47 (Fig. 2). Furthermore, SWI/SNF enzymes interact with MITF in osteoclasts to activate osteoclast-specific genes.48

III. MELANOMA

NIH-PA Author Manuscript

Melanoma is an extremely aggressive malignancy with a tendency for widespread metastasis. The aggressive nature of melanoma is thought to be attributed at least in part to the activity of lineage-specific transcription factors such as SOX10 and MITF. SOX10 has been demonstrated to promote the formation of giant congenital nevi that frequently progress to melanoma and to be required for melanoma proliferation and survival.6 MITF is considered a lineage addiction oncogene that promotes melanoma proliferation and survival.49 The MITF gene is amplified in approximately 10–20% of human melanomas and promotes oncogene-induced transformation of melanocytes and melanoma chemoresistance.50 There is evidence that SWI/SNF enzymes cooperate with MITF to promote melanoma tumorigenicity,24 as well as evidence that some SWI/SNF subunits are downregulated or mutated in melanoma.51–57 A. The SWI/SNF ATPases in Melanoma BRG1 has been shown to promote melanoma differentiation as well as to increase survival of melanoma cells that were exposed to a chemotherapeutic agent or to UV radiation.24,47 One mechanism by which BRG1 promotes survival from UV radiation is by activating the MITF target gene, ML-IAP (BIRC7, livin). ML-IAP is a potent inhibitor of apoptosis that is highly expressed in melanoma and promotes aggressive behavior and poor response to chemotherapeutic agents.58 BRG1 was found to remodel chromatin on the ML-IAP promoter and to facilitate MITF and coactivator binding. Thus, BRG1 may promote Crit Rev Eukaryot Gene Expr. Author manuscript; available in PMC 2014 July 21.

Mehrotra et al.

Page 4

NIH-PA Author Manuscript

melanoma survival after DNA damage, in part through interactions with MITF to activate the expression of this potent antiapoptotic gene. Interestingly, the requirement for BRG1 is not compensated by the alternative subunit BRM in several melanoma cell lines.47

NIH-PA Author Manuscript

There have been several reports of the status of BRG1 in melanoma. One study assayed BRG1 expression in 38 primary and metastatic melanoma samples by immunohistochemistry and found that BRG1 was lost in a significant number of these samples.51 In a larger study that compared 48 dysplastic nevi, 90 primary melanomas, and 47 metastatic melanomas, BRG1 expression was significantly higher in primary and metastatic melanoma compared to dysplastic nevi.59 A recent study reported heterogeneous staining of BRG1 in primary melanoma and occasional lack of detectable BRG1 expression in some tumors.36 Yet another study indicated that BRG1 messenger RNA levels were significantly higher in stage IV melanoma samples compared to normal and stage III melanoma samples.60 Interestingly, recent exome sequencing studies have detected BRG1 mutations at a low frequency in patient-derived melanoma samples53 (Table 1). These studies suggest that BRG1 function may be compromised in a small subset of melanomas. However, the concomitant loss of BRG1 and the alternative ATPase BRM has not been reported. The expression of either BRG1 or BRM is downregulated in a subset of melanoma cell lines, and knocking down both ATPases in melanoma decreases proliferation and survival, suggesting that either the BRG1 or BRM subunit is required for melanoma tumorigenticity.24,46 The requirement for SWI/SNF activity is associated with MITFdependent and -independent functions.24,36,46,47,60

NIH-PA Author Manuscript

SWI/SNF complexes containing BRM have chromatin remodeling activities similar to those of complexes containing BRG1 in vitro, but several studies suggest that they have distinct functional roles in vivo.61 In one study, BRG1 was found to promote osteoblast differentiation, whereas BRM was found to maintain the proliferating precursor state.62 Disruption of BRM in mice does not affect embryonic development, whereas disruption of BRG1 is embryonically lethal.14,63 Downregulation of BRM in BRG1-deficient melanoma cells significantly inhibits the expression of most MITF target genes, including the proproliferative and survival genes CDK2, TBX2, and BCL2, thus compromising tumorigenicity.24 However, BRM cannot compensate for BRG1 in the expression of all MITF target genes, indicating that BRM only partially compensates for BRG1 loss.47 Interestingly, disruption of BRM in mice does not result in an obvious pigmentary phenotype,63 but exacerbates skin photocarcinogenesis.64 A hotspot mutation in BRM has been reported in nonmelanoma skin cancers.65 In a survey of 7 melanoma cell lines, one cell line was determined to have a mutation in BRM54 (Table 1). However, BRM expression was found to be infrequently downregulated in patient-derived melanoma samples.36,51 B. BAFs in Melanoma BAF180 (polybromo or PBRM1) is an SWI/SNF subunit that is specific to the PBAF complex. It contains 6 bromodomains that interact with acetylated histones and potentially mediate other protein– protein interactions.66,67 BAF180 is thought to function as a targeting subunit for the PBAF complex to specific loci. Mice with BAF180 deletions have severe cardiac defects, with an impaired epithelial-to-mesenchymal transition, and die

Crit Rev Eukaryot Gene Expr. Author manuscript; available in PMC 2014 July 21.

Mehrotra et al.

Page 5

NIH-PA Author Manuscript

during embryogenesis.68,69 There is a requirement for BAF180 in gene activation by nuclear hormone receptors70 and for replication of damaged DNA.71 BAF180 is mutated in breast and renal cell carcinomas and is thought to function as a tumor suppressor by interacting with the p53 protein to promote cell cycle arrest and senescence.72–74 Interestingly, BAF180 expression was found to be low in a subset of melanoma cell lines and dispensable for MLIAP activation by BRG1.47 There have not yet been any reports of BAF180 mutations in melanoma. It is important to note that BAF180 stability is dependent on ARID2 (BAF200), another component of the PBAF complex.9 ARID2 (BAF200) is a subunit of the PBAF complex that contains a conserved N-terminal AT-rich DNA interaction domain and 2 conserved C-terminal acetylene zinc-finger motifs.75 Proteins containing ARID have been shown to be involved in a variety of domains biological processes including embryonic development.76 One report indicates that ARID2 is required for osteoblast differentiation.77 Interestingly, ARID2 can form a complex with other SWI/SNF subunits in the absence of BAF180 and is required for activation of a subset of interferon-α-inducible genes independent of BAF180. Thus, PBAF complexes containing or lacking BAF180 may regulate a distinct set of genes.

NIH-PA Author Manuscript

ARID2 mutations have been detected in hepatitis C virus–associated hepatocellular carcinomas,78 non-small-cell lung cancer,79 pancreatic cancer,80 and melanoma53,55–57 (Table 1). The studies of melanoma identified ARID2 as the SWI/SNF component that is most frequently mutated. In 2 of these studies, ARID2 mutations occurred at a significant frequency to be designated as driver mutations.53,57 There has not yet been any investigation into the biological function of ARID2 in melanocytes nor studies of how ARID2 might act as a tumor suppressor.

NIH-PA Author Manuscript

Two proteins containing an AT-rich DNA domain, ARID1A (BAF250A) and ARID1B (BAF250B), are highly homologous but mutually exclusive components of BAF complexes.10 Deletion of ARID1A in mice results in early embryonic lethality. ARID1A was found to be required for embryonic cell self-renewal as well as for differentiation into mesoderm-derived cardiomyocytes and adipocytes, but not for ectoderm-derived neurons. 20 Furthermore, deletion of the DNA-interacting domain of ARID1A in mice disrupts SWI/SNF binding to target sites and leads to embryonic lethality.81 Biallelic inactivation of ARID1B in embryonic stem cells results in reduced proliferation and self-renewal capacity.82 ARID1A and ARID1B have different expression patterns during embryonic development83 and opposing roles in cell cycle regulation during osteoblast differentiation. 84 Interestingly, ARID1A mutations have been reported in uterine and ovarian cancers,85,86 whereas ARID1B was found to be mutated at high frequency in breast cancer.87 Both ARID1A and ARID1B mutations have been reported in hepatocellular carcinoma88,89 and neuroblastoma90 as well as in melanoma, although at a low frequency53–56 (Table1). ARID1A and ARID1B were both highly expressed in 2 surveys of melanoma cell lines.24,46 There have not yet been any functional studies of ARID1A and ARID1B in melanocyte differentiation or in melanoma. BAF47 (SMARCB1) is a core component of SWI/SNF chromatin remodeling complexes that is required for optimum SWI/SNF chromatin remodeling activity in vitro.11 Deletion of the BAF47 gene results in early embryonic lethality.15–17 Interestingly, BAF47 heterozygous

Crit Rev Eukaryot Gene Expr. Author manuscript; available in PMC 2014 July 21.

Mehrotra et al.

Page 6

NIH-PA Author Manuscript

mice develop tumors with rhabdoid features that display loss of heterozygosity. Thus, BAF47 acts as a classic tumor suppressor. Human malignant rhabdoid tumors frequently have inactivating mutations in the BAF47 gene,91 and mutations in BAF47 have been detected in schwannomatosis. 92 In melanoma, one study indicates that downregulation of BAF47 expression occurs in a significant number of primary and metastatic melanomas and correlates with poor prognosis.52 Mutations of BAF47 also have been detected in melanoma cell lines (Table 1).55 Interestingly, BAF47 is required for oncogene-induced senescence of human melanocytes.93 BAF47 also has been shown to interact with MITF, suggesting that it may be required for melanocyte differentiation.24

IV. CONCLUSIONS AND FUTURE DIRECTIONS

NIH-PA Author Manuscript

Current studies suggest that SWI/SNF complexes have a key role in the regulation of gene expression during melanocyte differentiation and in melanoma. SWI/SNF enzymes have been shown to be required for the expression of 2 transcription factors, SOX10 and MITF, that promote melanoma tumorigenesis, as well as for activation of MITF target genes.24,36,46,47 Furthermore, SWI/SNF enzymes are required for melanoma proliferation and can contribute to chemoresistance.24,36,46,47 These observations conflict somewhat with evidence indicating loss of function mutations in a subset of SWI/SNF genes or loss of expression of SWI/SNF components.51–57 However, it seems that the ATPases BRG1 and BRM are rarely concomitantly disrupted, suggesting that SWI/SNF activity may be compromised but not completely ablated in melanoma. Rather than a decrease in SWI/SNF activity, the disruption of BAFs such as ARID2 would be predicted to alter SWI/SNF interactions with transcription factors and affect recruitment of the complex to specific locations. Elucidation of the role of SWI/SNF-associated subunits in the transcriptional regulation of proliferation and survival genes will increase our understanding of how disruption of SWI/SNF components alters gene expression in melanoma and provide insight into how some SWI/SNF components can act as tumor suppressors in this malignancy.

NIH-PA Author Manuscript

In addition to their function in transcriptional regulation, SWI/SNF chromatin remodeling enzymes have recently been implicated in DNA damage signaling and repair pathways. It was demonstrated that BRG1 is recruited to sites of UV-induced damage, promotes chromatin relaxation, and facilitates nucleotide excision repair.94,95 BAF47 was shown to promote recruitment of the nucleotide excision repair machinery and checkpoint factors to UV-induced lesions.96 Thus, the loss of some SWI/SNF components in UV-exposed melanocytes may drive melanomagenesis because of increased mutagenesis and genomic instability resulting from defective DNA repair. A better understanding of this aspect of SWI/SNF function in melanocytes may help illuminate the role that these enzymes have in the initiation and progression of melanoma.

Acknowledgments ILD was supported by grant no. R01(ARO59379) from the National Institute of Arthritis and Musculoskeletal and Skin Diseases.

Crit Rev Eukaryot Gene Expr. Author manuscript; available in PMC 2014 July 21.

Mehrotra et al.

Page 7

ABBREVIATIONS NIH-PA Author Manuscript

ATP

adenosine triphosphate

BAF

BRG1/BRM-associated factor

MITF

microphthalmia-associated transcription factor

BAF

ML-IAP, melanoma inhibitor of apoptosis

PBAF

polybromo-associated factor

UV

ultraviolet

References

NIH-PA Author Manuscript NIH-PA Author Manuscript

1. Goding CR. Mitf from neural crest to melanoma: signal transduction and transcription in the melanocyte lineage. Genes Dev. 2000; 14(14):1712–28. [PubMed: 10898786] 2. Abdel-Malek ZA, Kadekaro AL, Swope VB. Stepping up melanocytes to the challenge of UV exposure. Pigment Cell Melanoma Res. 2010; 23(2):171–86. [PubMed: 20128873] 3. Steingrimsson E, Copeland NG, Jenkins NA. Melanocytes and the microphthalmia transcription factor network. Annu Rev Genet. 2004; 38:365–411. [PubMed: 15568981] 4. Mollaaghababa R, Pavan WJ. The importance of having your SOX on: role of SOX10 in the development of neural crest-derived melanocytes and glia. Oncogene. 2003; 22(20):3024–34. [PubMed: 12789277] 5. Tsao H, Chin L, Garraway LA, Fisher DE. Melanoma: from mutations to medicine. Genes Dev. 2012; 26(11):1131–55. [PubMed: 22661227] 6. Shakhova O, Zingg D, Schaefer SM, Hari L, Civenni G, Blunschi J, Claudinot S, Okoniewski M, Beermann F, Mihic-Probst D, Moch H, Wegner M, Dummer R, Barrandon Y, Cinelli P, Sommer L. Sox10 promotes the formation and maintenance of giant congenital naevi and melanoma. Nat Cell Biol. 2012; 14(8):882–90. [PubMed: 22772081] 7. de la Serna IL, Ohkawa Y, Imbalzano AN. Chromatin remodelling in mammalian differentiation: lessons from ATP-dependent remodellers. Nat Rev Genet. 2006; 7(6):461–73. [PubMed: 16708073] 8. Shain AH, Pollack JR. The spectrum of SWI/SNF mutations, ubiquitous in human cancers. PLoS One. 2013; 8(1):e55119. [PubMed: 23355908] 9. Yan Z, Cui K, Murray DM, Ling C, Xue Y, Gerstein A, Parsons R, Zhao K, Wang W. PBAF chromatin-remodeling complex requires a novel specificity subunit, BAF200, to regulate expression of selective interferonresponsive genes. Genes Dev. 2005; 19(14):1662–7. [PubMed: 15985610] 10. Wang X, Nagl NG, Wilsker D, Van Scoy M, Pacchione S, Yaciuk P, Dallas PB, Moran E. Two related ARID family proteins are alternative subunits of human SWI/SNF complexes. Biochem J. 2004; 383(Pt 2):319–25. [PubMed: 15170388] 11. Phelan ML, Sif S, Narlikar GJ, Kingston RE. Reconstitution of a core chromatin remodeling complex from SWI/SNF subunits. Mol Cell. 1999; 3(2):247–53. [PubMed: 10078207] 12. Belandia B, Orford RL, Hurst HC, Parker MG. Targeting of SWI/SNF chromatin remodelling complexes to estrogen- responsive genes. EMBO J. 2002; 21(15):4094–103. [PubMed: 12145209] 13. Hsiao PW, Fryer CJ, Trotter KW, Wang W, Archer TK. BAF60a mediates critical interactions between nuclear receptors and the BRG1 chromatin-remodeling complex for transactivation. Mol Cell Biol. 2003; 23(17):6210–20. [PubMed: 12917342] 14. Bultman S, Gebuhr T, Yee D, La Mantia C, Nicholson J, Gilliam A, Randazzo F, Metzger D, Chambon P, Crabtree G, Magnuson T. A Brg1 null mutation in the mouse reveals functional differences among mammalian SWI/SNF complexes. Mol Cell. 2000; 6(6):1287–95. [PubMed: 11163203] 15. Klochendler-Yeivin A, Fiette L, Barra J, Muchardt C, Babinet C, Yaniv M. The murine SNF5/INI1 chromatin remodeling factor is essential for embryonic development and tumor suppression. EMBO Rep. 2000; 1(6):500–6. [PubMed: 11263494]

Crit Rev Eukaryot Gene Expr. Author manuscript; available in PMC 2014 July 21.

Mehrotra et al.

Page 8

NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript

16. Guidi CJ, Sands AT, Zambrowicz BP, Turner TK, Demers DA, Webster W, Smith TW, Imbalzano AN, Jones SN. Disruption of Ini1 leads to peri-implantation lethality and tumorigenesis in mice. Mol Cell Biol. 2001; 21(10):3598–603. [PubMed: 11313485] 17. Roberts CW, Galusha SA, McMenamin ME, Fletcher CD, Orkin SH. Haploinsufficiency of Snf5 (integrase interactor 1) predisposes to malignant rhabdoid tumors in mice. Proc Natl Acad Sci U S A. 2000; 97(25):13796– 800. [PubMed: 11095756] 18. Lickert H, Takeuchi JK, Von Both I, Walls JR, McAuliffe F, Adamson SL, Henkelman RM, Wrana JL, Rossant J, Bruneau BG. Baf60c is essential for function of BAF chromatin remodelling complexes in heart development. Nature. 2004; 432(7013):107–12. [PubMed: 15525990] 19. Han D, Jeon S, Sohn DH, Lee C, Ahn S, Kim WK, Chung H, Seong RH. SRG3, a core component of mouse SWI/SNF complex, is essential for extra-embryonic vascular development. Dev Biol. 2008; 315(1):136–46. [PubMed: 18206867] 20. 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(18):6656–61. [PubMed: 18448678] 21. Keenen B, de la Serna IL. Chromatin remodeling in embryonic stem cells: regulating the balance between pluripotency and differentiation. J Cell Physiol. 2009; 219(1):1–7. [PubMed: 19097034] 22. Saladi SV, de la Serna IL. ATP dependent chromatin remodeling enzymes in embryonic stem cells. Stem Cell Rev. 2010; 6(1):62–73. [PubMed: 20148317] 23. de la Serna IL, Ohkawa Y, Higashi C, Dutta C, Osias J, Kommajosyula N, Tachibana T, Imbalzano AN. The microphthalmia-associated transcription factor requires SWI/SNF enzymes to activate melanocyte-specific genes. J Biol Chem. 2006; 281(29):20233–41. [PubMed: 16648630] 24. Keenen B, Qi H, Saladi SV, Yeung M, de la Serna IL. Heterogeneous SWI/SNF chromatin remodeling complexes promote expression of microphthalmia-associated transcription factor target genes in melanoma. Oncogene. 2010; 29(1):81–92. [PubMed: 19784067] 25. Weider M, Kuspert M, Bischof M, Vogl MR, Hornig J, Loy K, Kosian T, Müller J, Hillgärtner S, Tamm ER, Metzger D, Wegner M. Chromatin-remodeling factor Brg1 is required for Schwann cell differentiation and myelination. Dev Cell. 2012; 23(1):193–201. [PubMed: 22814607] 26. Erickson CA. From the crest to the periphery: control of pigment cell migration and lineage segregation. Pigment Cell Res. 1993; 6(5):336–47. [PubMed: 8302772] 27. Adameyko I, Lallemend F, Aquino JB, Pereira JA, Topilko P, Muller T, Fritz N, Beljajeva A, Mochii M, Liste I, Usoskin D, Suter U, Birchmeier C, Ernfors P. Schwann cell precursors from nerve innervation are a cellular origin of melanocytes in skin. Cell. 2009; 139(2):366–79. [PubMed: 19837037] 28. Graham A. Melanocyte production: dark side of the Schwann cell. Curr Biol. 2009; 19(24):R1116– 7. [PubMed: 20064420] 29. Britsch S, Goerich DE, Riethmacher D, Peirano RI, Rossner M, Nave KA, Birchmeier C, Wegner M. The transcription factor Sox10 is a key regulator of peripheral glial development. Genes Dev. 2001; 15(1):66–78. [PubMed: 11156606] 30. Huber WE, Price ER, Widlund HR, Du J, Davis IJ, Wegner M, Fisher DE. A tissue-restricted cAMP transcriptional response: SOX10 modulates alpha-melanocyte- stimulating hormonetriggered expression of microphthalmia-associated transcription factor in melanocytes. J Biol Chem. 2003; 278(46):45224–30. [PubMed: 12944398] 31. Murisier F, Guichard S, Beermann F. A conserved transcriptional enhancer that specifies Tyrp1 expression to melanocytes. Dev Biol. 2006; 298(2):644–55. [PubMed: 16934245] 32. Murisier F, Guichard S, Beermann F. The tyrosinase enhancer is activated by Sox10 and Mitf in mouse melanocytes. Pigment Cell Res. 2007; 20(3):173–84. [PubMed: 17516925] 33. Jiao Z, Mollaaghababa R, Pavan WJ, Antonellis A, Green ED, Hornyak TJ. Direct interaction of Sox10 with the promoter of murine dopachrome tautomerase (Dct) and synergistic activation of Dct expression with Mitf. Pigment Cell Res. 2004; 17(4):352–62. [PubMed: 15250937] 34. Limpert AS, Bai S, Narayan M, Wu J, Yoon SO, Carter BD, Lu QR. NF-kappaB forms a complex with the chromatin remodeler BRG1 to regulate schwann cell differentiation. J Neurosci. 2013; 33(6):2388–97. [PubMed: 23392668]

Crit Rev Eukaryot Gene Expr. Author manuscript; available in PMC 2014 July 21.

Mehrotra et al.

Page 9

NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript

35. Marathe HG, Mehta G, Zhang X, Datar I, Mehrotra A, Yeung KC, de la Sema IL. SWI/SNF enzymes promote SOX10-mediated activation of myelin gene expression. PLoS One. 2013; 8(7):e69037. [PubMed: 23874858] 36. Ondrusova L, Vachtenheim J, Reda J, Zakova P, Benkova K. MITF-independent pro-survival role of BRG1-containing SWI/SNF complex in melanoma cells. PLoS One. 2013; 8(1):e54110. [PubMed: 23349796] 37. Levy C, Khaled M, Fisher DE. MITF: master regulator of melanocyte development and melanoma oncogene. Trends Mol Med. 2006; 12(9):406–14. [PubMed: 16899407] 38. Hodgkinson CA, Moore KJ, Nakayama A, Steingrimsson E, Copeland NG, Jenkins NA, Arnheiter H. Mutations at the mouse microphthalmia locus are associated with defects in a gene encoding a novel basic-helix-loop-helix- zipper protein. Cell. 1993; 74(2):395–404. [PubMed: 8343963] 39. Steingrimsson E, Tessarollo L, Pathak B, Hou L, Arnheiter H, Copeland NG, Jenkins NA. Mitf and Tfe3, two members of the Mitf-Tfe family of bHLH-Zip transcription factors, have important but functionally redundant roles in osteoclast development. Proc Natl Acad Sci U S A. 2002; 99(7): 4477–82. [PubMed: 11930005] 40. Ito A, Morii E, Maeyama K, Jippo T, Kim DK, Lee YM, Ogihara H, Hashimoto K, Kitamura Y, Nojima H. Systematic method to obtain novel genes that are regulated by mi transcription factor: impaired expression of granzyme B and tryptophan hydroxylase in mi/mi cultured mast cells. Blood. 1998; 91(9):3210–21. [PubMed: 9558376] 41. McGill GG, Horstmann M, Widlund HR, Du J, Motyckova G, Nishimura EK, Lin YL, Ramaswamy S, Avery W, Ding HF, Jordan SA, Jackson IJ, Korsmeyer SJ, Golub TR, Fisher DE. Bcl2 regulation by the melanocyte master regulator Mitf modulates lineage survival and melanoma cell viability. Cell. 2002; 109(6):707–18. [PubMed: 12086670] 42. Dynek JN, Chan SM, Liu J, Zha J, Fairbrother WJ, Vucic D. Microphthalmia-associated transcription factor is a critical transcriptional regulator of melanoma inhibitor of apoptosis in melanomas. Cancer Res. 2008; 68(9):3124–32. [PubMed: 18451137] 43. Du J, Widlund HR, Horstmann MA, Ramaswamy S, Ross K, Huber WE, Nishimura EK, Golub TR, Fisher DE. Critical role of CDK2 for melanoma growth linked to its melanocyte-specific transcriptional regulation by MITF. Cancer Cell. 2004; 6(6):565–76. [PubMed: 15607961] 44. Loercher AE, Tank EM, Delston RB, Harbour JW. MITF links differentiation with cell cycle arrest in melanocytes by transcriptional activation of INK4A. J Cell Biol. 2005; 168(1):35–40. [PubMed: 15623583] 45. Carreira S, Goodall J, Aksan I, La Rocca SA, Galibert MD, Denat L, Larue L, Goding CR. Mitf cooperates with Rb1 and activates p21Cip1 expression to regulate cell cycle progression. Nature. 2005; 433(7027):764–9. [PubMed: 15716956] 46. Vachtenheim J, Ondrusova L, Borovansky J. SWI/SNF chromatin remodeling complex is critical for the expression of microphthalmia-associated transcription factor in melanoma cells. Biochem Biophys Res Commun. 2010; 392(3):454–9. [PubMed: 20083088] 47. Saladi SV, Wong PG, Trivedi AR, Marathe HG, Keenen B, Aras S, Liew ZQ, Setaluri V, de la Serna IL. BRG1 promotes survival of UV-irradiated melanoma cells by cooperating with MITF to activate the melanoma inhibitor of apoptosis gene. Pigment Cell Melanoma Res. 2013; 26(3):377– 91. [PubMed: 23480510] 48. Sharma SM, Bronisz A, Hu R, Patel K, Mansky KC, Sif S, Ostrowski MC. MITF and PU.1 recruit p38 MAPK and NFATc1 to target genes during osteoclast differentiation. J Biol Chem. 2007; 282(21):15921–9. [PubMed: 17403683] 49. Garraway LA, Sellers WR. Lineage dependency and lineage-survival oncogenes in human cancer. Nat Rev Cancer. 2006; 6(8):593–602. [PubMed: 16862190] 50. Garraway LA, Widlund HR, Rubin MA, Getz G, Berger AJ, Ramaswamy S, Beroukhim R, Milner DA, Granter SR, Du J, Lee C, Wagner SN, Li C, Golub TR, Rimm DL, Meyerson ML, Fisher DE, Sellers WR. Integrative genomic analyses identify MITF as a lineage survival oncogene amplified in malignant melanoma. Nature. 2005; 436(7047):117–22. [PubMed: 16001072] 51. Becker TM, Haferkamp S, Dijkstra MK, Scurr LL, Frausto M, Diefenbach E, Scolyer RA, Reisman DN, Mann GJ, Kefford RF, Rizos H. The chromatin remodelling factor BRG1 is a novel binding partner of the tumor suppressor p16INK4a. Mol Cancer. 2009; 8:4. [PubMed: 19149898]

Crit Rev Eukaryot Gene Expr. Author manuscript; available in PMC 2014 July 21.

Mehrotra et al.

Page 10

NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript

52. Lin H, Wong RP, Martinka M, Li G. Loss of SNF5 expression correlates with poor patient survival in melanoma. Clin Cancer Res. 2009; 15(20):6404–11. [PubMed: 19808872] 53. Hodis E, Watson IR, Kryukov GV, Arold ST, Imielinski M, Theurillat JP, ickerson E, Auclair D, Li L, Place C, Dicara D, Ramos AH, Lawrence MS, Cibulskis K, Sivachenko A, Voet D, Saksena G, Stransky N, Onofrio RC, Winckler W, Ardlie K, Wagle N, Wargo J, Chong K, Morton DL, Stemke-Hale K, Chen G, Noble M, Meyerson M, Ladbury JE, Davies MA, Gershenwald JE, Wagner SN, Hoon DS, Schadendorf D, Lander ES, Gabriel SB, Getz G, Garraway LA, Chin L. A landscape of driver mutations in melanoma. Cell. 2012; 150(2):251–63. [PubMed: 22817889] 54. Nikolaev SI, Sotiriou SK, Pateras IS, Santoni F, Sougioultzis S, Edgren H, Almusa H, Robyr D, Guipponi M, Saarela J, Gorgoulis VG, Antonarakis SE, Halazonetis TD. A single-nucleotide substitution mutator phenotype revealed by exome sequencing of human colon adenomas. Cancer Res. 2012; 72(23):6279–89. [PubMed: 23204322] 55. Wei X, Walia V, Lin JC, Teer JK, Prickett TD, Gartner J, Davis S, Stemke-Hale K, Davies MA, Gershenwald JE, Robinson W, Robinson S, Rosenberg SA, Samuels Y. NISC Comparative Sequencing Program. Exome sequencing identifies GRIN2A as frequently mutated in melanoma. Nat Genet. 2011; 43(5):442–6. [PubMed: 21499247] 56. Stark MS, Woods SL, Gartside MG, Bonazzi VF, Dutton-Regester K, Aoude LG, Chow D, Sereduk C, Niemi NM, Tang N, Ellis JJ, Reid J, Zismann V, Tyagi S, Muzny D, Newsham I, Wu Y, Palmer JM, Pollak T, Youngkin D, Brooks BR, Lanagan C, Schmidt CW, Kobe B, MacKeigan JP, Yin H, Brown KM, Gibbs R, Trent J, Hayward NK. Frequent somatic mutations in MAP3K5 and MAP3K9 in metastatic melanoma identified by exome sequencing. Nat Genet. 2012; 44(2): 165–9. [PubMed: 22197930] 57. Krauthammer M, Kong Y, Ha BH, Evans P, Bacchiocchi A, McCusker JP, Cheng E, Davis MJ, Goh G, Choi M, Ariyan S, Narayan D, Dutton-Regester K, Capatana A, Holman EC, Bosenberg M, Sznol M, Kluger HM, Brash DE, Stern DF, Materin MA, Lo RS, Mane S, Ma S, Kidd KK, Hayward NK, Lifton RP, Schlessinger J, Boggon TJ, Halaban R. Exome sequencing identifies recurrent somatic RAC1 mutations in melanoma. Nat Genet. 2012; 44(9):1006–14. [PubMed: 22842228] 58. Lazar I, Perlman R, Lotem M, Peretz T, Ben-Yehuda D, Kadouri L. The clinical effect of the inhibitor of apopotosis protein livin in melanoma. Oncology. 2012; 82(4):197–204. [PubMed: 22441029] 59. Lin H, Wong RP, Martinka M, Li G. BRG1 expression is increased in human cutaneous melanoma. Br J Dermatol. 2010; 163(3):502–10. [PubMed: 20491765] 60. Saladi SV, Keenen B, Marathe HG, Qi H, Chin KV, de la Serna IL. Modulation of extracellular matrix/adhesion molecule expression by BRG1 is associated with increased melanoma invasiveness. Mol Cancer. 2010; 9:280–97. [PubMed: 20969766] 61. Sif S, Saurin AJ, Imbalzano AN, Kingston RE. Purification and characterization of mSin3Acontaining Brg1 and hBrm chromatin remodeling complexes. Genes Dev. 2001; 15(5):603–18. [PubMed: 11238380] 62. Flowers S, Nagl NG Jr, Beck GR Jr, Moran E. Antagonistic roles for BRM and BRG1 SWI/SNF complexes in differentiation. J Biol Chem. 2009; 284(15):10067–75. [PubMed: 19144648] 63. Reyes JC, Barra J, Muchardt C, Camus A, Babinet C, Yaniv M. Altered control of cellular proliferation in the absence of mammalian brahma (SNF2alpha). EMBO J. 1998; 17(23):6979–91. [PubMed: 9843504] 64. Halliday GM, Zhou Y, Sou PW, Huang XX, Rana S, Bugeja MJ, Painter N, Scolyer RA, Muchardt C, Di Girolamo N, Lyons JG. The absence of Brm exacerbates photocarcinogenesis. Exp Dermatol. 2012; 21(8):599–604. [PubMed: 22775994] 65. Moloney FJ, Lyons JG, Bock VL, Huang XX, Bugeja MJ, Halliday GM. Hotspot mutation of Brahma in non-melanoma skin cancer. J Invest Dermatol. 2009; 129(4):1012–5. [PubMed: 18923443] 66. Charlop-Powers Z, Zeng L, Zhang Q, Zhou MM. Structural insights into selective histone H3 recognition by the human Polybromo bromodomain 2. Cell Res. 2010; 20(5):529–38. [PubMed: 20368734] 67. Filippakopoulos P, Picaud S, Mangos M, Keates T, Lambert JP, Barsyte-Lovejoy D, Felletar I, Volkmer R, Müller S, Pawson T, Gingras AC, Arrowsmith CH, Knapp S. Histone recognition and Crit Rev Eukaryot Gene Expr. Author manuscript; available in PMC 2014 July 21.

Mehrotra et al.

Page 11

NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript

large-scale structural analysis of the human bromodomain family. Cell. 2012; 149(1):214–31. [PubMed: 22464331] 68. Wang Z, Zhai W, Richardson JA, Olson EN, Meneses JJ, Firpo MT, Kang C, Skarnes WC, Tjian R. Polybromo protein BAF180 functions in mammalian cardiac chamber maturation. Genes Dev. 2004; 18(24):3106–16. [PubMed: 15601824] 69. Huang X, Gao X, Diaz-Trelles R, Ruiz-Lozano P, Wang Z. Coronary development is regulated by ATP-dependent SWI/SNF chromatin remodeling component BAF180. Dev Biol. 2008; 319(2): 258–66. [PubMed: 18508041] 70. Lemon B, Inouye C, King DS, Tjian R. Selectivity of chromatin-remodelling cofactors for ligandactivated transcription. Nature. 2001; 414(6866):924–8. [PubMed: 11780067] 71. Niimi A, Chambers AL, Downs JA, Lehmann AR. A role for chromatin remodellers in replication of damaged DNA. Nucleic Acids Res. 2012; 40(15):7393–403. [PubMed: 22638582] 72. Xia W, Nagase S, Montia AG, Kalachikov SM, Keniry M, Su T, Memeo L, Hibshoosh H, Parsons R. BAF180 is a critical regulator of p21 induction and a tumor suppressor mutated in breast cancer. Cancer Res. 2008; 68(6):1667–74. [PubMed: 18339845] 73. Varela I, Tarpey P, Raine K, Huang D, Ong CK, Stephens P, Davies H, Jones D, Lin ML, Teague J, Bignell G, Butler A, Cho J, Dalgliesh GL, Galappaththige D, Greenman C, Hardy C, Jia M, Latimer C, Lau KW, Marshall J, McLaren S, Menzies A, Mudie L, Stebbings L, Largaespada DA, Wessels LF, Richard S, Kahnoski RJ, Anema J, Tuveson DA, Perez-Mancera PA, Mustonen V, Fischer A, Adams DJ, Rust A, Chan-on W, Subimerb C, Dykema K, Furge K, Campbell PJ, Teh BT, Stratton MR, Futreal PA. Exome sequencing identifies frequent mutation of the SWI/SNF complex gene PBRM1 in renal carcinoma. Nature. 2011; 469(7331):539–42. [PubMed: 21248752] 74. Burrows AE, Smogorzewska A, Elledge SJ. Polybromo-associated BRG1-associated factor components BRD7 and BAF180 are critical regulators of p53 required for induction of replicative senescence. Proc Natl Acad Sci U S A. 2010; 107(32):14280–5. [PubMed: 20660729] 75. Zhang X, Azhar G, Zhong Y, Wei JY. Zipzap/p200 is a novel zinc finger protein contributing to cardiac gene regulation. Biochem Biophys Res Commun. 2006; 346(3):794–801. [PubMed: 16782067] 76. Wilsker D, Patsialou A, Dallas PB, Moran E. ARID proteins: a diverse family of DNA binding proteins implicated in the control of cell growth, differentiation, and development. Cell Growth Differ. 2002; 13(3):95–106. [PubMed: 11959810] 77. Xu F, Flowers S, Moran E. Essential role of ARID2 protein-containing SWI/SNF complex in tissue-specific gene expression. J Biol Chem. 2012; 287(7):5033–41. [PubMed: 22184115] 78. Li M, Zhao H, Zhang X, Wood LD, Anders RA, Choti MA, Pawlik TM, Daniel HD, Kannangai R, Offerhaus GJ, Velculescu VE, Wang L, Zhou S, Vogelstein B, Hruban RH, Papadopoulos N, Cai J, Torbenson MS, Kinzler KW. Inactivating mutations of the chromatin remodeling gene ARID2 in hepatocellular carcinoma. Nat Genet. 2011; 43(9):828–9. [PubMed: 21822264] 79. Manceau G, Letouze E, Guichard C, Didelot A, Cazes A, Corte H, Fabre E, Pallier K, Imbeaud S, Le Pimpec-Barthes F, Zucman-Rossi J, Laurent-Puig P, Blons H. Recurrent inactivating mutations of ARID2 in non-small cell lung carcinoma. Int J Cancer. 2013; 132(9):2217–21. [PubMed: 23047306] 80. Biankin AV, Waddell N, Kassahn KS, Gingras MC, Muthuswamy LB, Johns AL, Miller DK, Wilson PJ, Patch AM, Wu J, Chang DK, Cowley MJ, Gardiner BB, Song S, Harliwong I, Idrisoglu S, Nourse C, Nourbakhsh E, Manning S, Wani S, Gongora M, Pajic M, Scarlett CJ, Gill AJ, Pinho AV, Rooman I, Anderson M, Holmes O, Leonard C, Taylor D, Wood S, Xu Q, Nones K, Fink JL, Christ A, Bruxner T, Cloonan N, Kolle G, Newell F, Pinese M, Mead RS, Humphris JL, Kaplan W, Jones MD, Colvin EK, Nagrial AM, Humphrey ES, Chou A, Chin VT, Chantrill LA, Mawson A, Samra JS, Kench JG, Lovell JA, Daly RJ, Merrett ND, Toon C, Epari K, Nguyen NQ, Barbour A, Zeps N, Kakkar N, Zhao F, Wu YQ, Wang M, Muzny DM, Fisher WE, Brunicardi FC, Hodges SE, Reid JG, Drummond J, Chang K, Han Y, Lewis LR, Dinh H, Buhay CJ, Beck T, Timms L, Sam M, Begley K, Brown A, Pai D, Panchal A, Buchner N, De Borja R, Denroche RE, Yung CK, Serra S, Onetto N, Mukhopadhyay D, Tsao MS, Shaw PA, Petersen GM, Gallinger S, Hruban RH, Maitra A, Iacobuzio-Donahue CA, Schulick RD, Wolfgang CL, Morgan RA, Lawlor RT, Capelli P, Corbo V, Scardoni M, Tortora G, Tempero MA, Mann KM, Jenkins NA, Perez-Mancera PA, Adams DJ, Largaespada DA, Wessels LF, Rust AG, Stein LD, Tuveson DA, Copeland NG,

Crit Rev Eukaryot Gene Expr. Author manuscript; available in PMC 2014 July 21.

Mehrotra et al.

Page 12

NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript

Musgrove EA, Scarpa A, Eshleman JR, Hudson TJ, Sutherland RL, Wheeler DA, Pearson JV, McPherson JD, Gibbs RA, Grimmond SM. Australian Pancreatic Cancer Genome Initiative. Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature. 2012; 491(7424):399–405. [PubMed: 23103869] 81. Chandler RL, Brennan J, Schisler JC, Serber D, Patterson C, Magnuson T. ARID1a-DNA interactions are required for promoter occupancy by SWI/SNF. Mol Cell Biol. 2013; 33(2):265– 80. [PubMed: 23129809] 82. Yan Z, Wang Z, Sharova L, Sharov AA, Ling C, Piao Y, Aiba K, Matoba R, Wang W, Ko MS. BAF250B-associated SWI/SNF chromatin-remodeling complex is required to maintain undifferentiated mouse embryonic stem cells. Stem Cells. 2008; 26(5):1155–65. [PubMed: 18323406] 83. Flores-Alcantar A, Gonzalez-Sandoval A, Escalante-Alcalde D, Lomeli H. Dynamics of expression of ARID1A and ARID1B subunits in mouse embryos and in cells during the cell cycle. Cell Tissue Res. 2011; 345(1):137–48. [PubMed: 21647563] 84. Nagl NG Jr, Wang X, Patsialou A, Van Scoy M, Moran E. Distinct mammalian SWI/SNF chromatin remodeling complexes with opposing roles in cell-cycle control. EMBO J. 2007; 26(3): 752–63. [PubMed: 17255939] 85. Kandoth C, Schultz N, Cherniack AD, Akbani R, Liu Y, Shen H, Robertson AG, Pashtan I, Shen R, Benz CC, Yau C, Laird PW, Ding L, Zhang W, Mills GB, Kucherlapati R, Mardis ER, Levine DA. Integrated genomic characterization of endometrial carcinoma. Nature. 2013; 497(7447):67– 73. [PubMed: 23636398] 86. Wiegand KC, Shah SP, Al-Agha OM, Zhao Y, Tse K, Zeng T, Senz J, McConechy MK, Anglesio MS, Kalloger SE, Yang W, Heravi-Moussavi A, Giuliany R, Chow C, Fee J, Zayed A, Prentice L, Melnyk N, Turashvili G, Delaney AD, Madore J, Yip S, McPherson AW, Ha G, Bell L, Fereday S, Tam A, Galletta L, Tonin PN, Provencher D, Miller D, Jones SJ, Moore RA, Morin GB, Oloumi A, Boyd N, Aparicio SA, Shih IeM, Mes-Masson AM, Bowtell DD, Hirst M, Gilks B, Marra MA, Huntsman DG. ARID1A mutations in endometriosis-associated ovarian carcinomas. N Engl J Med. 2010; 363(16):1532–43. [PubMed: 20942669] 87. Stephens PJ, Tarpey PS, Davies H, Van Loo P, Greenman C, Wedge DC, Nik-Zainal S, Martin S, Varela I, Bignell GR, Yates LR, Papaemmanuil E, Beare D, Butler A, Cheverton A, Gamble J, Hinton J, Jia M, Jayakumar A, Jones D, Latimer C, Lau KW, McLaren S, McBride DJ, Menzies A, Mudie L, Raine K, Rad R, Chapman MS, Teague J, Easton D, Langerød A, Lee MT, Shen CY, Tee BT, Huimin BW, Broeks A, Vargas AC, Turashvili G, Martens J, Fatima A, Miron P, Chin SF, Thomas G, Boyault S, Mariani O, Lakhani SR, van de Vijver M, van ‘t Veer L, Foekens J, Desmedt C, Sotiriou C, Tutt A, Caldas C, Reis-Filho JS, Aparicio SA, Salomon AV, BørresenDale AL, Richardson AL, Campbell PJ, Futreal PA, Stratton MR. Oslo Breast Cancer Consortium (OSBREAC). The landscape of cancer genes and mutational processes in breast cancer. Nature. 2012; 486(7403):400–4. [PubMed: 22722201] 88. Fujimoto A, Totoki Y, Abe T, Boroevich KA, Hosoda F, Nguyen HH, Aoki M, Hosono N, Kubo M, Miya F, Arai Y, Takahashi H, Shirakihara T, Nagasaki M, Shibuya T, Nakano K, WatanabeMakino K, Tanaka H, Nakamura H, Kusuda J, Ojima H, Shimada K, Okusaka T, Ueno M, Shigekawa Y, Kawakami Y, Arihiro K, Ohdan H, Gotoh K, Ishikawa O, Ariizumi S, Yamamoto M, Yamada T, Chayama K, Kosuge T, Yamaue H, Kamatani N, Miyano S, Nakagama H, Nakamura Y, Tsunoda T, Shibata T, Nakagawa H. Whole-genome sequencing of liver cancers identifies etiological influences on mutation patterns and recurrent mutations in chromatin regulators. Nat Genet. 2012; 44(7):760–4. [PubMed: 22634756] 89. Guichard C, Amaddeo G, Imbeaud S, Ladeiro Y, Pelletier L, Maad IB, Calderaro J, Bioulac-Sage P, Letexier M, Degos F, Clément B, Balabaud C, Chevet E, Laurent A, Couchy G, Letouzé E, Calvo F, Zucman-Rossi J. Integrated analysis of somatic mutations and focal copy-number changes identifies key genes and pathways in hepatocellular carcinoma. Nat Genet. 2012; 44(6): 694–8. [PubMed: 22561517] 90. Sausen M, Leary RJ, Jones S, Wu J, Reynolds CP, Liu X, Blackford A, Parmigiani G, Diaz LA Jr, Papadopoulos N, Vogelstein B, Kinzler KW, Velculescu VE, Hogarty MD. Integrated genomic analyses identify ARID1A and ARID1B alterations in the childhood cancer neuroblastoma. Nat Genet. 2013; 45(1):12–7. [PubMed: 23202128]

Crit Rev Eukaryot Gene Expr. Author manuscript; available in PMC 2014 July 21.

Mehrotra et al.

Page 13

NIH-PA Author Manuscript

91. Versteege I, Sevenet N, Lange J, Rousseau-Merck MF, Ambros P, Handgretinger R, Aurias A, Delattre O. Truncating mutations of hSNF5/INI1 in aggressive paediatric cancer. Nature. 1998; 394(6689):203–6. [PubMed: 9671307] 92. Hulsebos TJ, Plomp AS, Wolterman RA, Robanus-Maandag EC, Baas F, Wesseling P. Germline mutation of INI1/SMARCB1 in familial schwannomatosis. Am J Hum Genet. 2007; 80(4):805–10. [PubMed: 17357086] 93. Wajapeyee N, Serra RW, Zhu X, Mahalingam M, Green MR. Oncogenic BRAF induces senescence and apoptosis through pathways mediated by the secreted protein IGFBP7. Cell. 2008; 132(3):363–74. [PubMed: 18267069] 94. Zhao Q, Wang QE, Ray A, Wani G, Han C, Milum K, Wani AA. Modulation of nucleotide excision repair by mammalian SWI/SNF chromatin-remodeling complex. J Biol Chem. 2009; 284(44):30424–32. [PubMed: 19740755] 95. Zhang L, Zhang Q, Jones K, Patel M, Gong F. The chromatin remodeling factor BRG1 stimulates nucleotide excision repair by facilitating recruitment of XPC to sites of DNA damage. Cell Cycle. 2009; 8(23):3953–9. [PubMed: 19901545] 96. Ray A, Mir SN, Wani G, Zhao Q, Battu A, Zhu Q, Wang QE, Wani AA. Human SNF5/INI1, a component of the human SWI/SNF chromatin remodeling complex, promotes nucleotide excision repair by influencing ATM recruitment and downstream H2AX phosphorylation. Mol Cell Biol. 2009; 29(23):6206–19. [PubMed: 19805520]

NIH-PA Author Manuscript NIH-PA Author Manuscript Crit Rev Eukaryot Gene Expr. Author manuscript; available in PMC 2014 July 21.

Mehrotra et al.

Page 14

NIH-PA Author Manuscript NIH-PA Author Manuscript

FIG. 1.

Heterogeneous SWI/SNF complexes containing either BRG1 or BRM and associated factors exist in mammalian cells: PBAF (a), BAF(BRG1) (b), and BAF(BRM) (c).

NIH-PA Author Manuscript Crit Rev Eukaryot Gene Expr. Author manuscript; available in PMC 2014 July 21.

Mehrotra et al.

Page 15

NIH-PA Author Manuscript

FIG. 2.

SWI/SNF interacts with SOX10 and MITF to activate melanocyte- and Schwann cell– specific genes.

NIH-PA Author Manuscript NIH-PA Author Manuscript Crit Rev Eukaryot Gene Expr. Author manuscript; available in PMC 2014 July 21.

Mehrotra et al.

Page 16

TABLE 1

NIH-PA Author Manuscript

List of Mutations in SWI/SNF Components That Have Been Reported to Occur in Melanoma Gene

Aliases

Chromosome

Mutation

Reference

1/121 Nonsense

53

SMARCA4

BRG1

19

3/7 Nonsynonymous

54

1/14 Nonsynonymous

55

SMARCA2

BRM

9

1/7 Nonsynonymous

54

ARID1A

ARID1B

NIH-PA Author Manuscript

ARID2

SMARCB1

BAF250A, SMARCF1

BAF250B

BAF200

BAF47, INI1, SNF5

3/121 Nonsense

53

2/7 Nonsense

54

1/7 Nonsynonymous

54

1/8 Nonsynonymous

56

1/14 Nonsynonymous

55

3/121 Nonsense

53

1

6

12

22

3/121 Nonsense

53

6/121 Nonsense

53

2/121 Splice site

53

1/121 Frameshift

53

6/97 Nonsense

57

5/97 Frameshift

57

1/8 Nonsense

56

2/14 Nonsense

55

2/14 Nonsynonymous

55

NIH-PA Author Manuscript Crit Rev Eukaryot Gene Expr. Author manuscript; available in PMC 2014 July 21.

SNF chromatin remodeling enzymes in melanocyte differentiation and melanoma.

Epidermal melanocytes are pigment-producing cells derived from the neural crest that protects skin from the damaging effects of solar radiation. Malig...
230KB Sizes 0 Downloads 4 Views