BRCA2

Source: Wikipedia, the free encyclopedia.
BRCA2
Gene ontology
Molecular function
Cellular component
Biological process
Sources:Amigo / QuickGO
Ensembl
UniProt
RefSeq (mRNA)

NM_000059

NM_001081001
NM_009765

RefSeq (protein)

NP_000050

NP_001074470
NP_033895

Location (UCSC)Chr 13: 32.32 – 32.4 MbChr 5: 150.45 – 150.49 Mb
PubMed search[3][4]
Wikidata
View/Edit HumanView/Edit Mouse

BRCA2 and BRCA2 (

Orthologs, styled Brca2 and Brca2, are common in other vertebrate species.[6][7] BRCA2 is a human tumor suppressor gene[8][9] (specifically, a caretaker gene), found in all humans; its protein, also called by the synonym breast cancer type 2 susceptibility protein, is responsible for repairing DNA.[10]

BRCA2 and BRCA1 are normally expressed in the cells of breast and other tissue, where they help repair damaged DNA or destroy cells if DNA cannot be repaired. They are involved in the repair of chromosomal damage with an important role in the error-free repair of DNA double strand breaks.[11][12] If BRCA1 or BRCA2 itself is damaged by a BRCA mutation, damaged DNA is not repaired properly, and this increases the risk for breast cancer.[13][14] BRCA1 and BRCA2 have been described as "breast cancer susceptibility genes" and "breast cancer susceptibility proteins". The predominant allele has a normal tumor suppressive function whereas high penetrance mutations in these genes cause a loss of tumor suppressive function, which correlates with an increased risk of breast cancer.[15]

The BRCA2 gene is located on the long (q) arm of

chromosome 13 at position 12.3 (13q12.3).[16] The human reference BRCA2 gene contains 27 exons, and the cDNA has 10,254 base pairs[17] coding for a protein of 3418 amino acids.[18][19]

Function

ATM (ATM) is a protein kinase that is recruited and activated by DNA double-strand breaks. DNA double-strand damages also activate the Fanconi anemia core complex (FANCA/B/C/E/F/G/L/M).[20] The FA core complex monoubiquitinates the downstream targets FANCD2 and FANCI.[21] ATM activates (phosphorylates) CHEK2 and FANCD2[22] CHEK2 phosphorylates BRCA1.[23] Ubiquinated FANCD2 complexes with BRCA1 and RAD51.[24] The PALB2 protein acts as a hub,[25] bringing together BRCA1, BRCA2 and RAD51 at the site of a DNA double-strand break, and also binds to RAD51C, a member of the RAD51 paralog complex RAD51B-RAD51C-RAD51D-XRCC2 (BCDX2). The BCDX2 complex is responsible for RAD51 recruitment or stabilization at damage sites.[26] RAD51 plays a major role in homologous recombinational
repair of DNA during double strand break repair. In this process, an ATP dependent DNA strand exchange takes place in which a single strand invades base-paired strands of homologous DNA molecules. RAD51 is involved in the search for homology and strand pairing stages of the process.

Although the structures of the BRCA1 and BRCA2 genes are very different, at least some functions are interrelated. The proteins made by both genes are essential for repairing damaged DNA (see Figure of recombinational repair steps). BRCA2 binds the single strand DNA and directly interacts with the recombinase RAD51 to stimulate[27] and maintain [28] strand invasion, a vital step of homologous recombination. The localization of RAD51 to the DNA double-strand break requires the formation of the BRCA1-PALB2-BRCA2 complex. PALB2 (Partner and localizer of BRCA2)[29] can function synergistically with a BRCA2 chimera (termed piccolo, or piBRCA2) to further promote strand invasion.[30] These breaks can be caused by natural and medical radiation or other environmental exposures, but also occur when chromosomes exchange genetic material during a special type of cell division that creates sperm and eggs (meiosis). Double strand breaks are also generated during repair of DNA cross links. By repairing DNA, these proteins play a role in maintaining the stability of the human genome and prevent dangerous gene rearrangements that can lead to hematologic and other cancers.

BRCA2 has been shown to possess a crucial role in protection from the

MRE11-dependent nucleolytic degradation of the reversed forks that are forming during DNA replication fork stalling (caused by obstacles such as mutations, intercalating agents etc.).[31]

Like BRCA1, BRCA2 probably regulates the activity of other genes and plays a critical role in embryo development.

Clinical significance

Absolute risk of cancers in BRCA1 or BRCA2 mutation.[32]

Certain variations of the BRCA2 gene increase risks for breast cancer as part of a hereditary breast–ovarian cancer syndrome. Researchers have identified hundreds of mutations in the BRCA2 gene, many of which cause an increased risk of cancer. BRCA2 mutations are usually insertions or deletions of a small number of DNA base pairs in the gene. As a result of these mutations, the protein product of the BRCA2 gene is abnormal, and does not function properly. Researchers believe that the defective BRCA2 protein is unable to fix DNA damage that occurs throughout the genome. As a result, there is an increase in mutations due to error-prone translesion synthesis past un-repaired DNA damage, and some of these mutations can cause cells to divide in an uncontrolled way and form a tumor.

People who have two mutated copies of the BRCA2 gene have one type of Fanconi anemia. This condition is caused by extremely reduced levels of the BRCA2 protein in cells, which allows the accumulation of damaged DNA. Patients with Fanconi anemia are prone to several types of leukemia (a type of blood cell cancer); solid tumors, particularly of the head, neck, skin, and reproductive organs; and bone marrow suppression (reduced blood cell production that leads to anemia). Women having inherited a defective BRCA1 or BRCA2 gene have risks for breast and ovarian cancer that are so high and seem so selective that many mutation carriers choose to have prophylactic surgery. There has been much conjecture to explain such apparently striking tissue specificity. Major determinants of where BRCA1- and BRCA2-associated hereditary cancers occur are related to tissue specificity of the cancer pathogen, the agent that causes chronic inflammation, or the carcinogen. The target tissue may have receptors for the pathogen, become selectively exposed to carcinogens and an infectious process. An innate genomic deficit impairs normal responses and exacerbates the susceptibility to disease in organ targets. This theory also fits data for several tumor suppressors beyond BRCA1 or BRCA2. A major advantage of this model is that it suggests there are some options in addition to prophylactic surgery.[33]

In addition to breast cancer in men and women, mutations in BRCA2 also lead to an increased risk of ovarian, uterine tube, prostate and pancreatic cancer. In some studies, mutations in the central part of the gene have been associated with a higher risk of ovarian cancer and a lower risk of prostate cancer than mutations in other parts of the gene. Several other types of cancer have also been seen in certain families with BRCA2 mutations.

In general, strongly inherited gene mutations (including mutations in BRCA2) account for only 5-10% of breast cancer cases; the specific risk of getting breast or other cancer for anyone carrying a BRCA2 mutation depends on many factors.[34]

History

The BRCA2 gene was discovered in 1994.

Ashkenazi Jewish ancestry.[37][38][39][40]

The gene was first cloned by scientists at Myriad Genetics, Endo Recherche, Inc., HSC Research & Development Limited Partnership, and the University of Pennsylvania.[41]

Methods to diagnose the likelihood of a patient with mutations in

Association for Molecular Pathology v. Myriad Genetics lawsuit.[45]

Germline mutations and founder effect

All germline BRCA2 mutations identified to date have been inherited, suggesting the possibility of a large "founder" effect in which a certain mutation is common to a well-defined population group and can theoretically be traced back to a common ancestor. Given the complexity of mutation screening for BRCA2, these common mutations may simplify the methods required for mutation screening in certain populations. Analysis of mutations that occur with high frequency also permits the study of their clinical expression.[46] A striking example of a founder mutation is found in Iceland, where a single BRCA2 (999del5) mutation accounts for virtually all breast/ovarian cancer families.[47][48] This frame-shift mutation leads to a highly truncated protein product. In a large study examining hundreds of cancer and control individuals, this 999del5 mutation was found in 0.6% of the general population. Of note, while 72% of patients who were found to be carriers had a moderate or strong family history of breast cancer, 28% had little or no family history of the disease. This strongly suggests the presence of modifying genes that affect the phenotypic expression of this mutation, or possibly the interaction of the BRCA2 mutation with environmental factors. Additional examples of founder mutations in BRCA2 are given in the table below.

Population or subgroup BRCA2 mutation(s)[46][49] Reference(s)
Ashkenazi Jewish 6174delT [50]
Dutch 5579insA [51]
Finns 8555T>G, 999del5, IVS23-2A>G [52][53]
French Canadians 8765delAG, 3398delAAAAG [54][55][56]
Hungarians 9326insA [57]
Icelanders 999del5 [47][48]
Italians 8765delAG [58]
Northern Irish 6503delTT [59]
Pakistanis 3337C>T [60]
Scottish 6503delTT [59]
Slovenians IVS16-2A>G [61]
Spanish 3034delAAAC(codon936), 9254del5 [62]
Swedish 4486delG [63]

Meiosis

In the plant Arabidopsis thaliana, loss of the BRCA2 homolog AtBRCA2 causes severe defects in both male meiosis and in the development of the female gametocyte.[64] AtBRCA2 protein is required for proper localization of the synaptonemal complex protein AtZYP1 and the recombinases AtRAD51 and AtDMC1. Furthermore, AtBRCA2 is required for proper meiotic synapsis. Thus AtBRCA2 is likely important for meiotic recombination. It appears that AtBRCA2 acts during meiosis to control the single-strand invasion steps mediated by AtRAD51 and AtDMC1 occurring during meiotic homologous recombinational repair of DNA damages.[64]

Homologs of BRCA2 are also essential for meiosis in the fungus

Ustilago maydis,[65] the worm Caenorhabditis elegans,[66][67] and the fruitfly Drosophila melanogaster.[68]

Mice that produce truncated versions of BRCA2 are viable but sterile.[69] BRCA2 mutant rats have a phenotype of growth inhibition and sterility in both sexes.[70] Aspermatogenesis in these mutant rats is due to a failure of homologous chromosome synapsis during meiosis.

BRC repeat sequences

DMC1 (DNA meiotic recombinase 1) is a meiosis specific homolog of RAD51 that mediates strand exchange during homologous recombinational repair. DMC1 promotes the formation of DNA strand invasion products (joint molecules) between homologous DNA molecules. Human DMC1 interacts directly with each of a series of repeat sequences in the BRCA2 protein (called BRC repeats) that stimulate joint molecule formation by DMC1.[71] BRC repeats conform to a motif consisting of a sequence of about 35 highly conserved amino acids that are present at least once in all BRCA2-like proteins. The BRCA2 BRC repeats stimulate joint molecule formation by promoting the interaction of single-stranded DNA (ssDNA) with DMC1.[71] The ssDNA complexed with DMC1 can pair with homologous ssDNA from another chromosome during the synopsis stage of meiosis to form a joint molecule, a central step in homologous recombination. Thus the BRC repeat sequences of BRCA2 appear to play a key role in recombinational repair of DNA damages during meiotic recombination.

Overall, it appears that homologous recombination during meiosis functions to repair DNA damages,[citation needed] and that BRCA2 plays a key role in performing this function.

Neurogenesis

BRCA2 is required in the mouse for neurogenesis and suppression of medulloblastoma.[72] ‘’BRCA2’’ loss profoundly affects neurogenesis, particularly during embryonic and postnatal neural development. These neurological defects arise from DNA damage.[72]

Epigenetic control

Epigenetic alterations in expression of BRCA2 (causing over-expression or under-expression) are very frequent in sporadic cancers (see Table below) while mutations in BRCA2 are rarely found.[73][74][75]

In non-small cell lung cancer, BRCA2 is epigenetically repressed by hypermethylation of the promoter.[76] In this case, promoter hypermethylation is significantly associated with low mRNA expression and low protein expression but not with loss of heterozygosity of the gene.

In sporadic ovarian cancer, an opposite effect is found. BRCA2 promoter and 5'-UTR regions have relatively few or no methylated CpG dinucleotides in the tumor DNA compared with that of non-tumor DNA, and a significant correlation is found between hypomethylation and a >3-fold over-expression of BRCA2.[77] This indicates that hypomethylation of the BRCA2 promoter and 5'-UTR regions leads to over-expression of BRCA2 mRNA.

One report indicated some epigenetic control of BRCA2 expression by the microRNAs miR-146a and miR-148a.[78]

BRCA2 expression in cancer

In eukaryotes, BRCA2 protein has an important role in homologous recombinational repair. In mice and humans, BRCA2 primarily mediates orderly assembly of RAD51 on single-stranded (ss) DNA, the form that is active for homologous pairing and strand invasion.[79] BRCA2 also redirects RAD51 from double-stranded DNA and prevents dissociation from ssDNA.[79] In addition, the four paralogs of RAD51, consisting of RAD51B (RAD51L1), RAD51C (RAD51L2), RAD51D (RAD51L3), XRCC2 form a complex called the BCDX2 complex (see Figure: Recombinational repair of DNA). This complex participates in RAD51 recruitment or stabilization at damage sites.[26] The BCDX2 complex appears to act by facilitating the assembly or stability of the RAD51 nucleoprotein filament. RAD51 catalyses strand transfer between a broken sequence and its undamaged homologue to allow re-synthesis of the damaged region (see homologous recombination models).

Some studies of cancers report over-expressed BRCA2 whereas other studies report under-expression of BRCA2. At least two reports found over-expression in some sporadic breast tumors and under-expression in other sporadic breast tumors.[80][81] (see Table).

Many cancers have epigenetic deficiencies in various DNA repair genes (see Frequencies of epimutations in DNA repair genes in cancers). These repair deficiencies likely cause increased unrepaired DNA damages. The over-expression of BRCA2 seen in many cancers may reflect compensatory BRCA2 over-expression and increased homologous recombinational repair to at least partially deal with such excess DNA damages. Egawa et al.[82] suggest that increased expression of BRCA2 can be explained by the genomic instability frequently seen in cancers, which induces BRCA2 mRNA expression due to an increased need for BRCA2 for DNA repair.

Under-expression of BRCA2 would itself lead to increased unrepaired DNA damages. Replication errors past these damages (see translesion synthesis) would lead to increased mutations and cancer.

BRCA2 expression in sporadic cancers
Cancer Over or Under expression Frequency of altered expression Evaluation method Ref.
Sporadic ovarian cancer Over-expression 80% messenger RNA [77]
Sporadic ovarian cancer Under-expression 42% immunohistochemistry [83]
(recurrent cancer in study above) Increased-expression 71% immunohistochemistry [83]
Non-small cell lung cancer Under-expression 34% immunohistochemistry [76]
Breast cancer Over-expression 66% messenger RNA [82]
Breast cancer Over-expression 20% messenger RNA [80]
(same study as above) Under-expression 11% messenger RNA [80]
Breast cancer Over-expression 30% immunohistochemistry [81]
(same study as above) Under-expression 30% immunohistochemistry [81]
Triple negative breast cancer Under-expression 90% immunohistochemistry [84]

Interactions

BRCA2 has been shown to interact with

Domain architecture

BRCA2 repeat
SCOP2
1n0w / SCOPe / SUPFAM
Available protein structures:
Pfam  structures / ECOD  
PDBRCSB PDB; PDBe; PDBj
PDBsumstructure summary

BRCA2 contains a number of 39 amino acid repeats that are critical for binding to RAD51 (a key protein in DNA recombinational repair) and resistance to methyl methanesulphonate treatment.[101][108][109][117]

The BRCA2 helical domain adopts a

genes that map to a 1.5-Mb locus deleted in an inherited developmental malformation syndrome.[115]

The BRCA OB1 domain assumes an OB fold, which consists of a highly curved five-stranded

DSS1 (deleted in split-hand/split foot syndrome) protein.[115]

The BRCA OB3 domain assumes an OB fold, which consists of a highly curved five-stranded

The Tower domain adopts a

homeodomain transcription factors. The Tower domain has an important role in the tumour suppressor function of BRCA2, and is essential for appropriate binding of BRCA2 to DNA.[115] Studies shown that conformation of this tower domain is allosterically controlled by a small protein "DSS1", which interacts with helical, OB1 and OB2 domains of BRCA2.[118]

Patents, enforcement, litigation, and controversy

A patent application for the isolated

The patents begin to expire in 2014.

Peter Meldrum, CEO of Myriad Genetics, has acknowledged that Myriad has "other competitive advantages that may make such [patent] enforcement unnecessary" in Europe.[120]

Legal decisions surrounding the BRCA1 and BRCA2 patents will affect the field of genetic testing in general.[121] In June 2013, in Association for Molecular Pathology v. Myriad Genetics (No. 12-398), the US Supreme Court unanimously ruled that, "A naturally occurring DNA segment is a product of nature and not patent eligible merely because it has been isolated," invalidating Myriad's patents on the BRCA1 and BRCA2 genes. However, the Court also held that manipulation of a gene to create something not found in nature could still be eligible for patent protection.[122] The Federal Court of Australia came to the opposite conclusion, upholding the validity of an Australian Myriad Genetics patent over the BRCA1 gene in February 2013,[123] but this decision is being appealed and the appeal will include consideration of the US Supreme Court ruling.[124]

References

  1. ^ a b c GRCh38: Ensembl release 89: ENSG00000139618Ensembl, May 2017
  2. ^ a b c GRCm38: Ensembl release 89: ENSMUSG00000041147Ensembl, May 2017
  3. ^ "Human PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  4. ^ "Mouse PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  5. ^ Hamel PJ (2007-05-29). "BRCA1 and BRCA2: No Longer the Only Troublesome Genes Out There". HealthCentral. Retrieved 2010-07-02.
  6. ^ "OrthoMaM phylogenetic marker: BRCA2 coding sequence". Archived from the original on 2016-03-03. Retrieved 2010-02-19.
  7. ^ "BRCA2 gene tree". Ensembl. May 2021
  8. PMID 10193517
    .
  9. .
  10. ^ Check W (2006-09-01). "BRCA: What we know now". College of American Pathologists. Retrieved 2010-08-23.
  11. PMID 17683622
    .
  12. ^ Friedenson B (2008-06-08). "Breast cancer genes protect against some leukemias and lymphomas" (video). SciVee.
  13. ^ "Breast and Ovarian Cancer Genetic Screening". Palo Alto Medical Foundation. Archived from the original on 4 October 2008. Retrieved 2008-10-11.
  14. PMID 17683622
    .
  15. .
  16. ^ .
  17. ^ "BRCA2 breast cancer 2, early onset [Homo sapiens]". EntrezGene. National Center for Biotechnology Information, U.S. National Library of Medicine.
  18. ^ "Breast cancer type 2 susceptibility protein - Homo sapiens (Human)". P51587. UniProt.
  19. ^ Williams-Jones B (2002). Genetic testing for sale: Implications of commercial brca testing in Canada (Ph.D.). The University of British Columbia.
  20. PMID 20484397
    .
  21. .
  22. .
  23. .
  24. .
  25. .
  26. ^ .
  27. .
  28. .
  29. ^ .
  30. .
  31. .
  32. .
  33. .
  34. ^ "High-Penetrance Breast and/or Ovarian Cancer Susceptibility Genes". National Cancer Institute. Retrieved 7 December 2012.
  35. S2CID 4346791
    .
  36. ^ High-Impact Science: Tracking down the BRCA genes (Part 2) Archived 2012-03-03 at the Wayback Machine - Cancer Research UK science blog, 2012
  37. ^ "Kenneth Offit | Breast Cancer Research Foundation | BCRF". Bcrfcure.org. 23 June 2014. Retrieved 2015-07-16.
  38. ISSN 0362-4331
    . Retrieved 2015-07-02.
  39. ^ "20 Years of Progress in Understanding Breast Cancer" (JPG). Mskcc.org. Retrieved 2015-07-17.
  40. ISSN 0362-4331
    . Retrieved 2015-07-07.
  41. ^ a b US patent 5837492, Tavtigian SV, Kamb A, Simard J, Couch F, Rommens JM, Weber BL, "Chromosome 13-linked breast cancer susceptibility gene", issued 1998-11-17, assigned to Myriad Genetics, Inc., Endo Recherche, Inc., HSC Research & Development Limited Partnership, Trustees of the University of Pennsylvania 
  42. ^ a b US patent 5747282, Skolnick HS, Goldgar DE, Miki Y, Swenson J, Kamb A, Harshman KD, Shattuck-Eidens DM, Tavtigian SV, Wiseman RW, Futreal PA, "7Q-linked breast and ovarian cancer susceptibility gene", issued 1998-05-05, assigned to Myriad Genetics, Inc., The United States of America as represented by the Secretary of Health and Human Services, and University of Utah Research Foundation 
  43. ^ US patent 5837492, Tavtigian SV, Kamb A, Simard J, Couch F, Rommens JM, Weber BL, "Chromosome 13-linked breast cancer susceptibility gene", issued 1998-11-17, assigned to Myriad Genetics, Inc., Endo Recherche, Inc., HSC Research & Development Limited Partnership, Trustees of the University of Pennsylvania 
  44. ^ a b Myriad Investor Page—see "Myriad at a glance" Archived 2012-10-18 at the Wayback Machine accessed October 2012
  45. ^ a b c Schwartz J (2009-05-12). "Cancer Patients Challenge the Patenting of a Gene". Health. New York Times.
  46. ^
    S2CID 23327569
    .
  47. ^ .
  48. ^ .
  49. .
  50. .
  51. .
  52. .
  53. .
  54. .
  55. .
  56. .
  57. .
  58. .
  59. ^
    PMID 12698193.{{cite journal}}: CS1 maint: numeric names: authors list (link
    )
  60. .
  61. .
  62. .
  63. .
  64. ^ .
  65. .
  66. .
  67. .
  68. .
  69. .
  70. .
  71. ^ .
  72. ^ .
  73. .
  74. .
  75. .
  76. ^ .
  77. ^ .
  78. .
  79. ^ .
  80. ^ .
  81. ^ .
  82. ^ .
  83. ^ .
  84. .
  85. ^ .
  86. .
  87. ^ .
  88. ^ .
  89. ^ .
  90. .
  91. .
  92. .
  93. .
  94. .
  95. .
  96. .
  97. .
  98. .
  99. .
  100. .
  101. ^ .
  102. ^ "Entrez Gene: PALB2 partner and localizer of BRCA2".
  103. ^
    PMID 12815053
    .
  104. .
  105. .
  106. .
  107. .
  108. ^ .
  109. ^ .
  110. .
  111. .
  112. .
  113. .
  114. .
  115. ^ .
  116. .
  117. .
  118. .
  119. ^ Conley J, Vorhous D, Cook-Deegan J (2011-03-01). "How Will Myriad Respond to the Next Generation of BRCA Testing?". Robinson, Bradshaw, and Hinson. Retrieved 2012-12-09.
  120. ^ "Genetics and Patenting". Human Genome Project Information. U.S. Department of Energy Genome Programs. 2010-07-07.
  121. ^ Liptak A (13 June 2013). "Supreme Court Rules Human Genes May Not Be Patented". New York Times. Retrieved 13 June 2013.
  122. Sydney Morning Herald
    . Retrieved June 14, 2013.
  123. Sydney Morning Herald
    . Retrieved June 14, 2013.

Further reading

External links

This article incorporates text from the public domain Pfam and InterPro: IPR002093
This article incorporates text from the public domain Pfam and InterPro: IPR015252
This article incorporates text from the public domain Pfam and InterPro: IPR015187
This article incorporates text from the public domain Pfam and InterPro: IPR015205

|}

This page is based on the copyrighted Wikipedia article: BRCA2. Articles is available under the CC BY-SA 3.0 license; additional terms may apply.Privacy Policy