Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

Proteomic and bioinformatic analysis of mammalian SWI/SNF complexes identifies extensive roles in human malignancy

Abstract

Subunits of mammalian SWI/SNF (mSWI/SNF or BAF) complexes have recently been implicated as tumor suppressors in human malignancies. To understand the full extent of their involvement, we conducted a proteomic analysis of endogenous mSWI/SNF complexes, which identified several new dedicated, stable subunits not found in yeast SWI/SNF complexes, including BCL7A, BCL7B and BCL7C, BCL11A and BCL11B, BRD9 and SS18. Incorporating these new members, we determined mSWI/SNF subunit mutation frequency in exome and whole-genome sequencing studies of primary human tumors. Notably, mSWI/SNF subunits are mutated in 19.6% of all human tumors reported in 44 studies. Our analysis suggests that specific subunits protect against cancer in specific tissues. In addition, mutations affecting more than one subunit, defined here as compound heterozygosity, are prevalent in certain cancers. Our studies demonstrate that mSWI/SNF is the most frequently mutated chromatin-regulatory complex (CRC) in human cancer, exhibiting a broad mutation pattern, similar to that of TP53. Thus, proper functioning of polymorphic BAF complexes may constitute a major mechanism of tumor suppression.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Identification of new, dedicated subunits of mSWI/SNF-like BAF complexes.
Figure 2: mSWI/SNF complex subunit genes are mutated in human cancers with high frequency.
Figure 3: mSWI/SNF complexes are more frequently mutated than other chromatin-modifying complexes in human cancer.
Figure 4: mSWI/SNF subunit genes are more frequently mutated than EP300, MLL and HDAC family genes.
Figure 5: Mutations in mSWI/SNF subunit genes occur in a broad spectrum of cancer types.
Figure 6: Occurrence of multiple mSWI/SNF mutations in human cancers.

References

  1. Lander, E.S. Initial impact of the sequencing of the human genome. Nature 470, 187–197 (2011).

    Article  CAS  PubMed  Google Scholar 

  2. Wang, W. et al. Purification and biochemical heterogeneity of the mammalian SWI-SNF complex. EMBO J. 15, 5370–5382 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Lessard, J. et al. An essential switch in subunit composition of a chromatin remodeling complex during neural development. Neuron 55, 201–215 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Ho, L. et al. An embryonic stem cell chromatin remodeling complex, esBAF, is essential for embryonic stem cell self-renewal and pluripotency. Proc. Natl. Acad. Sci. USA 106, 5181–5186 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Dunaief, J.L. et al. The retinoblastoma protein and BRG1 form a complex and cooperate to induce cell cycle arrest. Cell 79, 119–130 (1994).

    Article  CAS  PubMed  Google Scholar 

  6. Strobeck, M.W. et al. BRG-1 is required for RB-mediated cell cycle arrest. Proc. Natl. Acad. Sci. USA 97, 7748–7753 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Versteege, I. et al. Truncating mutations of hSNF5/INI1 in aggressive paediatric cancer. Nature 394, 203–206 (1998).

    Article  CAS  PubMed  Google Scholar 

  8. Sévenet, N. et al. Constitutional mutations of the hSNF5/INI1 gene predispose to a variety of cancers. Am. J. Hum. Genet. 65, 1342–1348 (1999).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Biegel, J.A. et al. Germ-line and acquired mutations of INI1 in atypical teratoid and rhabdoid tumors. Cancer Res. 59, 74–79 (1999).

    CAS  PubMed  Google Scholar 

  10. Taylor, M.D. et al. Familial posterior fossa brain tumors of infancy secondary to germline mutation of the hSNF5 gene. Am. J. Hum. Genet. 66, 1403–1406 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Klochendler-Yeivin, A. et al. The murine SNF5/INI1 chromatin remodeling factor is essential for embryonic development and tumor suppression. EMBO Rep. 1, 500–506 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Reisman, D.N. et al. Concomitant down-regulation of BRM and BRG1 in human tumor cell lines: differential effects on RB-mediated growth arrest vs CD44 expression. Oncogene 21, 1196–1207 (2002).

    Article  CAS  PubMed  Google Scholar 

  13. Wong, A.K. et al. BRG1, a component of the SWI-SNF complex, is mutated in multiple human tumor cell lines. Cancer Res. 60, 6171–6177 (2000).

    CAS  PubMed  Google Scholar 

  14. Rodriguez-Nieto, S. et al. Massive parallel DNA pyrosequencing analysis of the tumor suppressor BRG1/SMARCA4 in lung primary tumors. Hum. Mutat. 32, E1999–E2017 (2011).

    Article  CAS  PubMed  Google Scholar 

  15. Weissman, B. & Knudsen, K.E. Hijacking the chromatin remodeling machinery: impact of SWI/SNF perturbations in cancer. Cancer Res. 69, 8223–8230 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Huang, J., Zhao, Y.L., Li, Y., Fletcher, J.A. & Xiao, S. Genomic and functional evidence for an ARID1A tumor suppressor role. Genes Chromosom. Cancer 46, 745–750 (2007).

    Article  CAS  PubMed  Google Scholar 

  17. Heebøll, S. et al. SMARCC1 expression is upregulated in prostate cancer and positively correlated with tumour recurrence and dedifferentiation. Histol. Histopathol. 23, 1069–1076 (2008).

    PubMed  Google Scholar 

  18. Takita, J. et al. Gene expression profiling and identification of novel prognostic marker genes in neuroblastoma. Genes Chromosom. Cancer 40, 120–132 (2004).

    Article  CAS  PubMed  Google Scholar 

  19. Hoyal, C.R. et al. Genetic polymorphisms in DPF3 associated with risk of breast cancer and lymph node metastases. J. Carcinog. 4, 13 (2005).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Khavari, P.A., Peterson, C.L., Tamkun, J.W., Mendel, D.B. & Crabtree, G.R. BRG1 contains a conserved domain of the SWI2/SNF2 family necessary for normal mitotic growth and transcription. Nature 366, 170–174 (1993).

    Article  CAS  PubMed  Google Scholar 

  21. Lemon, B., Inouye, C., King, D.S. & Tjian, R. Selectivity of chromatin-remodelling cofactors for ligand-activated transcription. Nature 414, 924–928 (2001).

    Article  CAS  PubMed  Google Scholar 

  22. Spitnik-Elson, P. & Atsmon, A. Detachment of ribosomal proteins by salt. I. Effect of conditions on the amount of protein detached. J. Mol. Biol. 45, 113–124 (1969).

    Article  CAS  PubMed  Google Scholar 

  23. Roberts, M.E. & Walker, I.O. Structural studies on Escherichia coli ribosomes. 3. Denaturation and sedimentation of ribosomal subunits unfolded in urea. Biochim. Biophys. Acta 199, 184–193 (1970).

    Article  CAS  PubMed  Google Scholar 

  24. Middeljans, E. et al. SS18 together with animal-specific factors defines human BAF-type SWI/SNF complexes. PLoS ONE 7, e33834 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Kaeser, M.D., Aslanian, A., Dong, M.Q., Yates, J.R. III & Emerson, B.M. BRD7, a novel PBAF-specific SWI/SNF subunit, is required for target gene activation and repression in embryonic stem cells. J. Biol. Chem. 283, 32254–32263 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Cancer Genome Atlas Network. Comprehensive molecular characterization of human colon and rectal cancer. Nature 487, 330–337 (2012).

  27. Seshagiri, S. et al. Recurrent R-spondin fusions in colon cancer. Nature 488, 660–664 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Cancer Genome Atlas Research Network. Integrated genomic analyses of ovarian carcinoma. Nature 474, 609–615 (2011).

  29. Jones, S. et al. Frequent mutations of chromatin remodeling gene ARID1A in ovarian clear cell carcinoma. Science 330, 228–231 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Jones, S. et al. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science 321, 1801–1806 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Biankin, A.V. et al. Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature 491, 399–405 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Dalgliesh, G.L. et al. Systematic sequencing of renal carcinoma reveals inactivation of histone modifying genes. Nature 463, 360–363 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Varela, I. et al. Exome sequencing identifies frequent mutation of the SWI/SNF complex gene PBRM1 in renal carcinoma. Nature 469, 539–542 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Li, M. et al. Inactivating mutations of the chromatin remodeling gene ARID2 in hepatocellular carcinoma. Nat. Genet. 43, 828–829 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Fujimoto, A. 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).

    Article  CAS  PubMed  Google Scholar 

  36. Guichard, C. et al. Integrated analysis of somatic mutations and focal copy-number changes identifies key genes and pathways in hepatocellular carcinoma. Nat. Genet. 44, 694–698 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Gui, Y. et al. Frequent mutations of chromatin remodeling genes in transitional cell carcinoma of the bladder. Nat. Genet. 43, 875–878 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Wang, K. et al. Exome sequencing identifies frequent mutation of ARID1A in molecular subtypes of gastric cancer. Nat. Genet. 43, 1219–1223 (2011).

    Article  CAS  PubMed  Google Scholar 

  39. Zang, Z.J. et al. Exome sequencing of gastric adenocarcinoma identifies recurrent somatic mutations in cell adhesion and chromatin remodeling genes. Nat. Genet. 44, 570–574 (2012).

    Article  CAS  PubMed  Google Scholar 

  40. Shah, S.P. et al. The clonal and mutational evolution spectrum of primary triple-negative breast cancers. Nature 486, 395–399 (2012).

    Article  CAS  PubMed  Google Scholar 

  41. Banerji, S. et al. Sequence analysis of mutations and translocations across breast cancer subtypes. Nature 486, 405–409 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Ellis, M.J. et al. Whole-genome analysis informs breast cancer response to aromatase inhibition. Nature 486, 353–360 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Stephens, P.J. et al. The landscape of cancer genes and mutational processes in breast cancer. Nature 486, 400–404 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Cancer Genome Atlas Network. Comprehensive molecular portraits of human breast tumours. Nature 490, 61–70 (2012).

  45. Cancer Genome Atlas Research Network. Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature 455, 1061–1068 (2008).

  46. Parsons, D.W. et al. An integrated genomic analysis of human glioblastoma multiforme. Science 321, 1807–1812 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Parsons, D.W. et al. The genetic landscape of the childhood cancer medulloblastoma. Science 331, 435–439 (2011).

    Article  CAS  PubMed  Google Scholar 

  48. Pugh, T.J. et al. Medulloblastoma exome sequencing uncovers subtype-specific somatic mutations within a broad landscape of genetic heterogeneity. Nature 488, 106–110 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Robinson, G. et al. Novel mutations target distinct subgroups of medulloblastoma. Nature 488, 43–48 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Jones, D.T. et al. Dissecting the genomic complexity underlying medulloblastoma. Nature 488, 100–105 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Stransky, N. et al. The mutational landscape of head and neck squamous cell carcinoma. Science 333, 1157–1160 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Agrawal, N. et al. Exome sequencing of head and neck squamous cell carcinoma reveals inactivating mutations in NOTCH1. Science 333, 1154–1157 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Durinck, S. et al. Temporal dissection of tumorigenesis in primary cancers. Cancer Discov. 1, 137–143 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Wei, X. et al. Exome sequencing identifies GRIN2A as frequently mutated in melanoma. Nat. Genet. 43, 442–446 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Berger, M.F. et al. Melanoma genome sequencing reveals frequent PREX2 mutations. Nature 485, 502–506 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Hodis, E. et al. A landscape of driver mutations in melanoma. Cell 150, 251–263 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Cancer Genome Atlas Research Network. Comprehensive genomic characterization of squamous cell lung cancers. Nature 489, 519–525 (2012).

  58. Imielinski, M. et al. Mapping the hallmarks of lung adenocarcinoma with massively parallel sequencing. Cell 150, 1107–1120 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Govindan, R. et al. Genomic landscape of non–small cell lung cancer in smokers and never-smokers. Cell 150, 1121–1134 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Yan, X.J. et al. Exome sequencing identifies somatic mutations of DNA methyltransferase gene DNMT3A in acute monocytic leukemia. Nat. Genet. 43, 309–315 (2011).

    Article  CAS  PubMed  Google Scholar 

  61. Welch, J.S. et al. The origin and evolution of mutations in acute myeloid leukemia. Cell 150, 264–278 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Quesada, V. et al. Exome sequencing identifies recurrent mutations of the splicing factor SF3B1 gene in chronic lymphocytic leukemia. Nat. Genet. 44, 47–52 (2012).

    Article  CAS  Google Scholar 

  63. Pasqualucci, L. et al. Analysis of the coding genome of diffuse large B-cell lymphoma. Nat. Genet. 43, 830–837 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Lohr, J.G. et al. Discovery and prioritization of somatic mutations in diffuse large B-cell lymphoma (DLBCL) by whole-exome sequencing. Proc. Natl. Acad. Sci. USA 109, 3879–3884 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Morin, R.D. et al. Frequent mutation of histone-modifying genes in non-Hodgkin lymphoma. Nature 476, 298–303 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Love, C. et al. The genetic landscape of mutations in Burkitt lymphoma. Nat. Genet. 44, 1321–1325 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Richter, J. et al. Recurrent mutation of the ID3 gene in Burkitt lymphoma identified by integrated genome, exome and transcriptome sequencing. Nat. Genet. 44, 1316–1320 (2012).

    Article  CAS  PubMed  Google Scholar 

  68. Chapman, M.A. et al. Initial genome sequencing and analysis of multiple myeloma. Nature 471, 467–472 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Zhang, J. et al. The genetic basis of early T-cell precursor acute lymphoblastic leukaemia. Nature 481, 157–163 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Gutierrez, A. et al. The BCL11B tumor suppressor is mutated across the major molecular subtypes of T-cell acute lymphoblastic leukemia. Blood 118, 4169–4173 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. De Keersmaecker, K. et al. The TLX1 oncogene drives aneuploidy in T cell transformation. Nat. Med. 16, 1321–1327 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Wiegand, K.C. et al. ARID1A mutations in endometriosis-associated ovarian carcinomas. N. Engl. J. Med. 363, 1532–1543 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Guan, B. et al. Mutation and loss of expression of ARID1A in uterine low-grade endometrioid carcinoma. Am. J. Surg. Pathol. 35, 625–632 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  74. Peña-Llopis, S. et al. BAP1 loss defines a new class of renal cell carcinoma. Nat. Genet. 44, 751–759 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  75. Puente, X.S. et al. Whole-genome sequencing identifies recurrent mutations in chronic lymphocytic leukaemia. Nature 475, 101–105 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. DelBove, J. et al. Identification of a core member of the SWI/SNF complex, BAF155/SMARCC1, as a human tumor suppressor gene. Epigenetics 6, 1444–1453 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Crew, A.J. et al. Fusion of SYT to two genes, SSX1 and SSX2, encoding proteins with homology to the Kruppel-associated box in human synovial sarcoma. EMBO J. 14, 2333–2340 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Clark, J. et al. Identification of novel genes, SYT and SSX, involved in the t(X;18)(p11.2;q11.2) translocation found in human synovial sarcoma. Nat. Genet. 7, 502–508 (1994).

    Article  CAS  PubMed  Google Scholar 

  79. de Leeuw, B., Balemans, M., Olde Weghuis, D. & Geurts van Kessel, A. Identification of two alternative fusion genes, SYT-SSX1 and SYT-SSX2, in t(X;18)(p11.2;q11.2)-positive synovial sarcomas. Hum. Mol. Genet. 4, 1097–1099 (1995).

    Article  CAS  PubMed  Google Scholar 

  80. Berger, R. et al. t(5;14)/HOX11L2-positive T-cell acute lymphoblastic leukemia. A collaborative study of the Groupe Francais de Cytogenetique Hematologique (GFCH). Leukemia 17, 1851–1857 (2003).

    Article  CAS  PubMed  Google Scholar 

  81. Satterwhite, E. et al. The BCL11 gene family: involvement of BCL11A in lymphoid malignancies. Blood 98, 3413–3420 (2001).

    Article  CAS  PubMed  Google Scholar 

  82. Kadoch, C. & Crabtree, G.R. Reversible disruption of mSWI/SNF (BAF) complexes by the SS18-SSX oncogenic fusion in synovial sarcoma. Cell 153, 71–85 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Morin, R.D. et al. Somatic mutations altering EZH2 (Tyr641) in follicular and diffuse large B-cell lymphomas of germinal-center origin. Nat. Genet. 42, 181–185 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Park, S.W., Chung, N.G., Eom, H.S., Yoo, N.J. & Lee, S.H. Mutational analysis of EZH2 codon 641 in non-Hodgkin lymphomas and leukemias. Leuk. Res. 35, e6–e7 (2011).

    Article  PubMed  Google Scholar 

  85. Bödör, C. et al. EZH2 Y641 mutations in follicular lymphoma. Leukemia 25, 726–729 (2011).

    Article  CAS  PubMed  Google Scholar 

  86. Ryan, R.J. et al. EZH2 codon 641 mutations are common in BCL2-rearranged germinal center B cell lymphomas. PLoS ONE 6, e28585 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Kannan, K. et al. Whole-exome sequencing identifies ATRX mutation as a key molecular determinant in lower-grade glioma. Oncotarget 3, 1194–1203 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  88. Schwartzentruber, J. et al. Driver mutations in histone H3.3 and chromatin remodelling genes in paediatric glioblastoma. Nature 482, 226–231 (2012).

    Article  CAS  PubMed  Google Scholar 

  89. Jiao, Y. et al. DAXX/ATRX, MEN1, and mTOR pathway genes are frequently altered in pancreatic neuroendocrine tumors. Science 331, 1199–1203 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Dawson, M.A. & Kouzarides, T. Cancer epigenetics: from mechanism to therapy. Cell 150, 12–27 (2012).

    Article  CAS  PubMed  Google Scholar 

  91. Atsmon, A., Spitnik-Elson, P. & Elson, D. Detachment of ribosomal proteins by salt. II. Some properties of protein-deficient particles formed by the detachment of ribosomal proteins. J. Mol. Biol. 45, 125–135 (1969).

    Article  CAS  PubMed  Google Scholar 

  92. Robinson, A. & Sykes, J. A study of the atypical ribosomal RNA components of Rhodopseudomonas spheroides. Biochim. Biophys. Acta 238, 99–115 (1971).

    Article  CAS  PubMed  Google Scholar 

  93. Cairns, B.R. et al. RSC, an essential, abundant chromatin-remodeling complex. Cell 87, 1249–1260 (1996).

    Article  CAS  PubMed  Google Scholar 

  94. Mizuguchi, G. et al. ATP-driven exchange of histone H2AZ variant catalyzed by SWR1 chromatin remodeling complex. Science 303, 343–348 (2004).

    Article  CAS  PubMed  Google Scholar 

  95. Wang, W. et al. Diversity and specialization of mammalian SWI/SNF complexes. Genes Dev. 10, 2117–2130 (1996).

    Article  CAS  PubMed  Google Scholar 

  96. Zhao, K. et al. Rapid and phosphoinositol-dependent binding of the SWI/SNF-like BAF complex to chromatin after T lymphocyte receptor signaling. Cell 95, 625–636 (1998).

    Article  CAS  PubMed  Google Scholar 

  97. Olave, I., Wang, W., Xue, Y., Kuo, A. & Crabtree, G.R. Identification of a polymorphic, neuron-specific chromatin remodeling complex. Genes Dev. 16, 2509–2517 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Yoo, A.S., Staahl, B.T., Chen, L. & Crabtree, G.R. MicroRNA-mediated switching of chromatin-remodelling complexes in neural development. Nature 460, 642–646 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Yoo, A.S. et al. MicroRNA-mediated conversion of human fibroblasts to neurons. Nature 476, 228–231 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Uno, K. et al. Aberrations of the hSNF5/INI1 gene are restricted to malignant rhabdoid tumors or atypical teratoid/rhabdoid tumors in pediatric solid tumors. Genes Chromosom. Cancer 34, 33–41 (2002).

    Article  CAS  PubMed  Google Scholar 

  101. Roberts, C.W., Galusha, S.A., McMenamin, M.E., Fletcher, C.D. & Orkin, S.H. Haploinsufficiency of Snf5 (integrase interactor 1) predisposes to malignant rhabdoid tumors in mice. Proc. Natl. Acad. Sci. USA 97, 13796–13800 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Durum, S.K. Bcl11: sibling rivalry in lymphoid development. Nat. Immunol. 4, 512–514 (2003).

    Article  CAS  PubMed  Google Scholar 

  103. Sankaran, V.G. et al. Human fetal hemoglobin expression is regulated by the developmental stage-specific repressor BCL11A. Science 322, 1839–1842 (2008).

    Article  CAS  PubMed  Google Scholar 

  104. Ho, L. et al. esBAF facilitates pluripotency by conditioning the genome for LIF/STAT3 signalling and by regulating polycomb function. Nat. Cell Biol. 13, 903–913 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Kia, S.K., Gorski, M.M., Giannakopoulos, S. & Verrijzer, C.P. SWI/SNF mediates polycomb eviction and epigenetic reprogramming of the INK4b-ARF-INK4a locus. Mol. Cell Biol. 28, 3457–3464 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported in part by US National Institutes of Health (NIH) grants NS046789 and CA163915 (both to G.R.C.). G.R.C. is an Investigator of the Howard Hughes Medical Institute. C.K. is supported by the National Science Foundation (Graduate Research Fellowship Program). D.C.H. is supported by a Helen Hay Whitney Foundation Fellowship. C.H. is supported by Eunice Kennedy Shriver National Institute of Child Health and Human Development Fellowship F32HD072627.

Author information

Authors and Affiliations

Authors

Contributions

C.K. performed and interpreted experiments. D.C.H. and C.H. performed genomic data analysis and interpretation. L.E., L.H. and J.R. performed proteomic mass spectrometry data collection and analysis. C.K., D.C.H. and G.R.C. conceived of and wrote the manuscript.

Corresponding author

Correspondence to Gerald R Crabtree.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Tables 1–4, 6 and 7, Supplementary Figures 1 and 2, Supplementary Note (PDF 2950 kb)

Supplementary Table 5

Mutation Frequencies (separate Excel Workbook) (XLSX 65 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Kadoch, C., Hargreaves, D., Hodges, C. et al. Proteomic and bioinformatic analysis of mammalian SWI/SNF complexes identifies extensive roles in human malignancy. Nat Genet 45, 592–601 (2013). https://doi.org/10.1038/ng.2628

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.2628

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research