Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Jul 31.
Published in final edited form as: Nat Genet. 2011 Sep 28;43(10):925–926. doi: 10.1038/ng.956

Two new studies report germline mutations in BAP1

Alisa M Goldstein 1
PMCID: PMC4117248  NIHMSID: NIHMS343708  PMID: 21956388

Abstract

Two new studies describe germline mutations in BAP1 in putatively dissimilar cancer-related syndromes. Although the predominant neoplasms – melanocytic tumors with distinct clinical and histopathological features versus mesothelioma – differ, uveal melanoma occurs, albeit infrequently, in both disorders.


BAP1 is a tumor suppressor gene located on chromosome 3p21 in a region that shows loss or deletions in numerous cancers, including lung and breast cancer, as well as uveal melanoma and mesothelioma. Two recent studies reported high frequencies of somatic mutations in BAP1 in uveal melanoma1 and mesothelioma2. Harbour et al1 found inactivating somatic mutations of BAP1 in 47% (28/60) (Table 1) of uveal melanoma with a much higher frequency of somatic mutations (27/34, 79%) in metastasizing uveal melanoma. In addition, one tumor had a germline frameshift mutation, suggesting that this variant was a susceptibility allele. Bott et al2 identified somatic inactivating mutations in BAP1 in 23% (12/53) of a discovery set of malignant pleural mesotheliomas (MPM), half from patients who reported asbestos exposure, and in 18% (12/68) of an independent set of MPM [total: 20% (24/121)]. In this issue, Wiesner, Bastian, Speicher and colleagues3 and Testa, Carbone, and colleagues4 provide further links between BAP1, uveal melanoma and mesothelioma by reporting on germline mutations of BAP1 in two putatively distinct cancer-related syndromes characterized predominantly by melanocytic tumors or mesothelioma, respectively, plus uveal melanoma in both disorders.

Table 1.

Tumors with somatic mutations in BAP1

Tumor Type No. Tumors Somatic Mutations Reference
No. %
Melanocytic
 Common nevi 29 0 0.0 Wiesner et al3
 Spitz nevi 17 0 0.0 Wiesner et al3
 Atypical Spitz nevi 18 2* 11.1 Wiesner et al3
 Cutaneous melanoma 60 3 5.0 Wiesner et al3
Uveal melanoma, total 93 41 44.1 Wiesner et al3; Harbour et al1
 Wiesner et al3 study 33 13 39.4 Wiesner et al3
 Harbour et al1 study 60 28 46.7 Harbour et al1
  Low metastatic risk 26 1 3.8 Harbour et al1
  High metastatic risk 31 25 80.6 Harbour et al1
  Metastatic 3 2 66.7 Harbour et al1
Mesothelioma, total 139 28 20.1 Testa et al; Bott et al2
 Testa et al study4 18 4 22.2 Testa et al4
 Bott et al study2 121 24 19.8 Bott et al2
Breast 251 1 0.6 Forbes et al, 2008 & 20115,6
Lung 322 2 0.4 Forbes et al, 2008 & 20115,6
Ovary 59 2 3.4 Forbes et al, 2008 & 20115,6
Pancreas 30 0 0.0 Forbes et al, 2008 & 20115,6
*

These tumors had morphologic features similar to the melanocytic tumors observed in families 1 and 2 (Wiesner et al3)

Two new cancer-related syndromes?

Wiesner et al3 identified co-segregating germline mutations of BAP1 in two families (1 and 2) with multiple members with melanocytic tumors that ranged histopathologically from epithelioid nevi to atypical melanocytic proliferations that had features that overlapped with cutaneous melanoma. Both families had one member with uveal melanoma and family 2 had multiple members with cutaneous melanoma. Affected family members had many (5 to >50) of the clinically and histopathologically distinct melanocytic tumors, whereas few melanomas developed suggesting that the risk of malignant potential in individual tumors was low. Examination of a subset of the familial melanocytic tumors revealed that the majority of tumors showed biallelic inactivation of BAP1 by various somatic alterations.

Testa et al4 discovered co-segregating germline mutations in BAP1 in two families (L and W) with 5 or more members with mesothelioma. The families had modest levels of asbestos exposure from having lived in asbestos-containing houses but did not have occupational asbestos exposure. The families also had multiple members with various malignancies including two members with uveal melanoma in family L. Somatic alterations of the familial mesothelioma tumors indicated biallelic inactivation of BAP1. Testa et al4 also sequenced BAP1 in germline DNAs from 26 sporadic mesothelioma cases and uncovered two inactivating frameshift mutations in patients subsequently found to have had uveal melanoma. Given the rarity of both uveal melanoma and mesothelioma in the United States, the authors concluded that it was extremely unlikely for these two malignancies to have occurred in the same individuals by chance.

Cancers and BAP1 mutations

Besides uveal melanoma, mesothelioma, and the distinct melanocytic tumors, numerous additional tumors were observed in the families3,4. Several cancers including cutaneous melanoma (n=3), non-melanoma skin (n=2), ovarian (n=1), breast (n=1), renal (n=1), and pancreatic (n=1) cancer occurred in family members who inherited their family’s inactivating BAP1 mutation. Whether these cancers are part of the spectrum of tumors related to germline BAP1 mutations requires additional study.

Data from COSMIC (Catalogue Of Somatic Mutations In Cancer)5,6 showed low frequencies of somatic BAP1 mutations in several of the observed tumors including breast (0.4%), ovarian (3.4%), and pancreatic (0.0%) cancer (table 1). In addition, Wiesner et al3 showed that five percent (3/60) of primary melanomas originating from acral skin (n=15), mucosa (n=15), or skin with (n=15) or without chronic sun damage had somatic BAP1 mutations. Finally, lung cancer, which was one of the first cancers originally reported to have BAP1 somatic mutations7, also had very few somatic mutations (0.6%)5,6 suggesting that another gene in the 3p21 region may be implicated in some of the cancers that show loss/deletion in this region810. Alternatively, a different mechanism of inactivation of BAP1 may be involved. In contrast to the low frequency of somatic mutations for the tumor types described above, 44% (41/93) of uveal melanoma and 20% (28/139) of sporadic mesotheliomas had somatic mutations of BAP114 (table 1). For most of the tumors evaluated, BAP1 showed biallelic loss. Harbour et al1 hypothesized that biallelic loss of BAP1 was a key event in uveal melanoma metastasis. The current studies suggest a potentially more complex scenario that may depend on various combinations of the tissue of origin, environmental exposures, functional consequences of the BAP1 mutations, and mechanisms of inactivation of the second BAP1 allele.

BAP1 (ubiquitin carboxy-terminal hydrolase/BRCA1-associated protein 1), a 729 amino acid nuclear-localized deubiquitinating enzyme, was originally identified as an ubiquitin hydrolase that binds to the RING finger domain of BRCA17,11. BAP1 contains numerous functional domains including the ubiquitin carboxy-terminal hydrolase (UCH) domain, a host cell factor-1 (HCF-1) binding domain, and binding domains for BRCA1 and BARD1. BAP1 has been functionally implicated in numerous biologic processes including chromatin, DNA damage response, and regulation of the cell cycle and cell growth1,2,7,1115. Given its complexity, different germline mutations in BAP1 may predispose to divergent tumor types.

Future Directions

These studies raise several provocative questions. Are these disorders two distinct syndromes or a single syndrome with a wide phenotypic range? For what tumor types do germline mutations of BAP1 increase susceptibility? What role do exposures (e.g. asbestos, ultraviolet radiation) have on the tumor types that develop? Are there genotype-phenotype correlations between specific germline (or somatic) mutations and the resultant tumors? Answers to these questions will require extensive additional clinical, molecular genetic, genetic epidemiologic and functional studies. Since its original discovery in 19987, BAP1 has captivated and beguiled scientists. The studies by Wiesner, Bastian, Speicher and colleagues3 and Testa, Carbone, and colleagues4 continue this trend.

Footnotes

Competing Financial Interests

The author declares no competing financial interest

References

  • 1.Harbour JW, et al. Frequent mutation of BAP1 in metastasizing uveal melanomas. Science. 2010;330:1410–3. doi: 10.1126/science.1194472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Bott M, et al. The nuclear deubiquitinase BAP1 is commonly inactivated by somatic mutations and 3p21.1 losses in malignant pleural mesothelioma. Nat Genet. 2011;43:668–72. doi: 10.1038/ng.855. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Wiesner T, Obenauf AC, Murali R. Germline mutations in BAP1 predispose to melanocytic tumors. Nat Genet. 2011 doi: 10.1038/ng.910. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Testa JR, Cheung M, Pei J. Germline BAP1 mutations predispose to malignant mesothelioma. Nat Genet. 2011 doi: 10.1038/ng.912. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Forbes SA, et al. The Catalogue of Somatic Mutations in Cancer (COSMIC) Curr Protoc Hum Genet. 2008;Chapter 10(Unit 10):11. doi: 10.1002/0471142905.hg1011s57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Forbes SA, et al. COSMIC: mining complete cancer genomes in the Catalogue of Somatic Mutations in Cancer. Nucleic Acids Res. 2011;39:D945–50. doi: 10.1093/nar/gkq929. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Jensen DE, et al. BAP1: a novel ubiquitin hydrolase which binds to the BRCA1 RING finger and enhances BRCA1-mediated cell growth suppression. Oncogene. 1998;16:1097–112. doi: 10.1038/sj.onc.1201861. [DOI] [PubMed] [Google Scholar]
  • 8.Buchhagen DL, Qiu L, Etkind P. Homozygous deletion, rearrangement and hypermethylation implicate chromosome region 3p14.3–3p21.3 in sporadic breast-cancer development. Int J Cancer. 1994;57:473–9. doi: 10.1002/ijc.2910570406. [DOI] [PubMed] [Google Scholar]
  • 9.Thiberville L, et al. Frequency and prognostic evaluation of 3p21–22 allelic losses in non-small-cell lung cancer. Int J Cancer. 1995;64:371–7. doi: 10.1002/ijc.2910640604. [DOI] [PubMed] [Google Scholar]
  • 10.Wood LD, et al. The genomic landscapes of human breast and colorectal cancers. Science. 2007;318:1108–13. doi: 10.1126/science.1145720. [DOI] [PubMed] [Google Scholar]
  • 11.Ventii KH, et al. BRCA1-associated protein-1 is a tumor suppressor that requires deubiquitinating activity and nuclear localization. Cancer Res. 2008;68:6953–62. doi: 10.1158/0008-5472.CAN-08-0365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Machida YJ, Machida Y, Vashisht AA, Wohlschlegel JA, Dutta A. The deubiquitinating enzyme BAP1 regulates cell growth via interaction with HCF-1. J Biol Chem. 2009;284:34179–88. doi: 10.1074/jbc.M109.046755. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Matsuoka S, et al. ATM and ATR substrate analysis reveals extensive protein networks responsive to DNA damage. Science. 2007;316:1160–6. doi: 10.1126/science.1140321. [DOI] [PubMed] [Google Scholar]
  • 14.Scheuermann JC, et al. Histone H2A deubiquitinase activity of the Polycomb repressive complex PR-DUB. Nature. 2010;465:243–7. doi: 10.1038/nature08966. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Stokes MP, et al. Profiling of UV-induced ATM/ATR signaling pathways. Proc Natl Acad Sci U S A. 2007;104:19855–60. doi: 10.1073/pnas.0707579104. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES