Stanford Home
Ovarian Kaleidoscope Database (OKdb)

Home

History

Transgenic Mouse Models

INFORGRAPHICS

Search
Submit
Update
Chroms
Browse
Admin

Hsueh lab

HPMR

Visits
since 01/2001:
176557

BRCA2 DNA repair associated OKDB#: 19
 Symbols: BRCA2 Species: human
 Synonyms: FAD, FACD, FAD1, GLM3, BRCC2, FANCD, PNCA2, FANCD1, XRCC11, BROVCA2  Locus: 13q13.1 in Homo sapiens


For retrieval of Nucleotide and Amino Acid sequences please go to: OMIM Entrez Gene
Mammalian Reproductive Genetics   Endometrium Database Resource   Orthologous Genes   UCSC Genome Browser   GEO Profiles new!   Amazonia (transcriptome data) new!

R-L INTERACTIONS   MGI

DNA Microarrays
SHOW DATA ...
link to BioGPS
General Comment The breast tumor suppressor gene BRCA2 was identified by positional cloning methods. Mutations of this gene also leads to ovarian cancer in some families (Tavtigian et al., 1996; Sharan et al., 1997). In the ovary, Brca1 and Brca2 exhibited a comparable hormone-independent pattern of expression in oocytes, granulosa cells and thecal cells of developing follicles (Blackshear et al., 1998).

NCBI Summary: Inherited mutations in BRCA1 and this gene, BRCA2, confer increased lifetime risk of developing breast or ovarian cancer. Both BRCA1 and BRCA2 are involved in maintenance of genome stability, specifically the homologous recombination pathway for double-strand DNA repair. The largest exon in both genes is exon 11, which harbors the most important and frequent mutations in breast cancer patients. The BRCA2 gene was found on chromosome 13q12.3 in human. The BRCA2 protein contains several copies of a 70 aa motif called the BRC motif, and these motifs mediate binding to the RAD51 recombinase which functions in DNA repair. BRCA2 is considered a tumor suppressor gene, as tumors with BRCA2 mutations generally exhibit loss of heterozygosity (LOH) of the wild-type allele. [provided by RefSeq, May 2020]
General function Chromosome organization, Cell death/survival, Cell cycle regulation, Tumor suppressor
Comment
Cellular localization Nuclear
Comment
Ovarian function Follicle development, Oogenesis, Oocyte maturation
Comment Ovarian Aging in Women with BRCA Germline Mutations. Lin W et al. (2017) Recent clinical and laboratory studies suggested that women with BRCA mutations have lower ovarian reserve and their primordial follicle oocytes may be more prone to DNA damage but the direct proof is lacking. To determine whether women with germline BRCA mutations have reduced primordial follicle reserve and increased oocyte DNA damage. A laboratory comparative study of ovarian tissue obtained from the risk-reducing salpingo- oophorectomy (RRSO) samples of unaffected BRCA mutation carrier (BMC) women vs. age-matched organ donor cadavers. Two academic centers. Of the 230 ovarian specimens from BMC, eighteen met the study inclusion criteria. Healthy ovaries from twelve organ donor cadavers served as controls. Histology and immunohistochemical analysis on paraffin-embedded ovarian sections. Primordial follicle density and the percentage of DNA double-strand break (DSB) positive primordial follicle oocytes. Ovaries from BMC had significantly lower primordial follicle densities compared to the controls (11.2±2.0 vs. 44.2±6.2 follicles/mm3, p=0.0002). Moreover, based on γH2AX expression, BRCA mutations were associated with increased DNA DSBs in primordial follicle oocytes (62±5.2% vs. 36±3.4%, p=0.0005). In subgroup analysis, both BRCA1 and BRCA2 mutations were associated with lower primordial follicle density (p=0.0001 and 0.0030, respectively) and BRCA1 mutations were associated with higher DNA DSBs (p=0.0003) compared to controls. On comparison of linear regression curves by multivariate analysis, the rates of follicle decline (R2=0.74, p=0.0001) and DNA DSB accumulation (R2=0.70, p=0.0001) appeared to be accelerated particularly in primordial follicle oocytes of BMC over age 30. We provided the first direct evidence of diminished ovarian reserve as well as accelerated primordial follicle loss and oocyte DNA damage in women with BRCA mutations. In addition to furthering our understanding of ovarian aging, our findings may be useful when counseling BMC women.////////////////// Brca2/Pds5 complexes mobilize persistent meiotic recombination sites to the nuclear envelope. Kusch T et al. (2015) Homologous recombination is required for reciprocal exchange between homologous chromosome arms during meiosis. Only select meiotic recombination events become chromosomal crossovers; the majority of recombination outcomes are noncrossovers. Growing evidence suggests that crossovers are repaired after noncrossovers. Here, I report that persisting recombination sites are mobilized to the nuclear envelope of Drosophila pro-oocytes during mid-pachytene. Their number correlates with the average crossover rate per meiosis. Proteomic and interaction studies reveal that the recombination mediator, Brca2, associates with lamin and the cohesion factor, Pds5, to secure persistent recombination sites at the nuclear envelope. In Rad51 females, all persistent DNA breaks are directed to the nuclear envelope. By contrast, a reduction of Pds5 or Brca2 levels abolishes the movement and causes a reduction of crossovers rates. The data suggest that persistent meiotic DNA double-strand breaks might correspond to crossovers, which are mobilized to the nuclear envelope for their repair. The identification of Brca2/Pds5 complexes as key mediators of this process provides a first mechanistic explanation for the contribution of lamins and cohesins to meiotic recombination.//////////////////
Expression regulated by
Comment
Ovarian localization Oocyte, Granulosa, Theca
Comment
Follicle stages Antral
Comment
Phenotypes POF (premature ovarian failure)
Mutations 5 mutations

Species: None
Mutation name: None
type: null mutation
fertility: infertile - ovarian defect
Comment: brca2 in zebrafish ovarian development, spermatogenesis, and tumorigenesis. Shive HR et al. Humans with inherited mutations in BRCA2 are at increased risk for developing breast and ovarian cancer; however, the relationship between BRCA2 mutation and these cancers is not understood. Studies of Brca2 mutation by gene targeting in mice are limited, given that homozygous Brca2 mutation typically leads to early embryonic lethality. We established a zebrafish line with a nonsense mutation in brca2 exon 11 (brca2(Q658X)), a mutation similar in location and type to BRCA2 mutations found in humans with hereditary breast and ovarian cancer. brca2(Q658X) homozygous zebrafish are viable and survive to adulthood; however, juvenile homozygotes fail to develop ovaries during sexual differentiation. Instead, brca2(Q658X) homozygotes develop as infertile males with meiotic arrest in spermatocytes. Germ cell migration to the embryonic gonadal ridge is unimpaired in brca2(Q658X) homozygotes; thus, failure of ovarian development is not due to defects in early establishment of the embryonic gonad. Homozygous tp53 mutation rescues ovarian development in brca2(Q658X) homozygous zebrafish, reflecting the importance of germ cell apoptosis in gonad morphogenesis. Adult brca2(Q658X) homozygous zebrafish are predisposed to testicular neoplasias. In addition, tumorigenesis in multiple tissues is significantly accelerated in combination with homozygous tp53 mutation in both brca2(Q658X) homozygous and brca2(Q658X) heterozygous zebrafish. These studies reveal critical roles for brca2 in ovarian development and tumorigenesis in reproductive tissues.

Species: human
Mutation name:
type: naturally occurring
fertility: subfertile
Comment: BRCA2 in Ovarian Development and Function. Reply. Zangen D et al. (2019)we have recently identified additional compound heterozygous BRCA2 mutations in Chinese patients with premature ovarian insufficiency: c.68-1G→C plus c.4440T→G(p.Y1480X) in two affected sisters and c.8168A→T(p.D2723V) plus c.9697_9700del(p.C3233Wfs*15) in a woman whose immediate family members were unaffected. //////////////////

Species: human
Mutation name:
type: naturally occurring
fertility: subfertile
Comment: Essential Role of BRCA2 in Ovarian Development and Function. Weinberg-Shukron A et al. (2018) The causes of ovarian dysgenesis remain incompletely understood. Two sisters with XX ovarian dysgenesis carried compound heterozygous truncating mutations in the BRCA2 gene that led to reduced BRCA2 protein levels and an impaired response to DNA damage, which resulted in chromosomal breakage and the failure of RAD51 to be recruited to double-stranded DNA breaks. The sisters also had microcephaly, and one sister was in long-term remission from leukemia, which had been diagnosed when she was 5 years old. Drosophila mutants that were null for an orthologue of BRCA2 were sterile, and gonadal dysgenesis was present in both sexes. These results revealed a new role for BRCA2 and highlight the importance to ovarian development of genes that are critical for recombination during meiosis. (Funded by the Israel Science Foundation and others.).//////////////////

Species: mouse
Mutation name:
type: null mutation
fertility: subfertile
Comment: BRCA2 deficiency is a potential driver for human primary ovarian insufficiency. Miao Y et al. (2019) Reproductive problem has been one of the top issues for women health worldwide in recent decades. As a typical female disease, primary ovarian insufficiency (POI) results in a loss of ovarian follicles and oocytes that thus destroys women fertility. However, due to the complex of POI etiology and rare resource of human POI oocytes, few biomarkers have been identified in clinics and no effective strategy could be applied to treat POI patients. In the search of possible association between DNA damage and POI by Smart-Seq2 and RT2 profiler PCR array, we find that BRCA2, a core DNA repair gene for homologous recombination shows significantly lower expression in two POI patient oocytes. In line with this, we generated oocyte-specific knockout mouse model driven by Gdf9-Cre. The Brca2-deficient mice are infertile because of the arrested follicle development and defective oocyte quality caused by the accumulation of DNA damage. Notably, ectopic expression of Brca2 in Brca2-deficient oocytes could partially restore the oocyte maturation and chromosome stability. Collectively, our data assign a definite deficiency to BRCA2 as a POI driver during follicle development and oocyte maturation, and provide a potential fertility treatment strategy for POI patients induced by BRCA2 deficiency.//////////////////

Species: human
Mutation name:
type: naturally occurring
fertility: subfertile
Comment: Homozygous hypomorphic BRCA2 variant in primary ovarian insufficiency without cancer or Fanconi anaemia trait. Caburet S et al. (2020) Primary ovarian insufficiency (POI) affects 1% of women under 40 years and is a public health problem. The genetic causes of POI are highly heterogeneous with isolated or syndromic forms. Recently, variants in genes involved in DNA repair have been shown to cause POI. Notably, syndromic POI with Fanconi anaemia (FA) traits related to biallelic BRCA2 truncated variants has been reported. Here, we report a novel phenotype of isolated POI with a BRCA2 variant in a consanguineous Turkish family. Exome sequencing (ES) was performed in the patient. We also performed functional studies, including a homologous recombination (HR) test, cell proliferation, radiation-induced RAD51 foci formation assays and chromosome breakage studies in primary and lymphoblastoid immortalised cells. The expression of BRCA2 in human foetal ovaries was studied. ES identified a homozygous missense c.8524C>T/p.R2842C-BRCA2 variant. BRCA2 defects induce cancer predisposition and FA. Remarkably, neither the patient nor her family exhibited somatic pathologies. The patient's cells showed intermediate levels of chromosomal breaks, cell proliferation and radiation-induced RAD51 foci formation compared with controls and FA cells. R2842C-BRCA2 only partially complemented HR efficiency compared with wild type-BRCA2. BRCA2 is expressed in human foetal ovaries in pachytene stage oocytes, when meiotic HR occurs. We describe the functional assessment of a homozygous hypomorphic BRCA2 variant in a patient with POI without cancer or FA trait. Our findings extend the phenotype of BRCA2 biallelic alterations to fully isolated POI. This study has a major impact on the management and genetic counselling of patients with POI.//////////////////

Genomic Region show genomic region
Phenotypes and GWAS show phenotypes and GWAS
Links
OMIM (Online Mendelian Inheritance in Man: an excellent source of general gene description and genetic information.)
OMIM \ Animal Model
KEGG Pathways
Recent Publications
None
Search for Antibody


created: July 22, 1999, midnight by: Hsueh   email:
home page:
last update: June 4, 2020, 10:45 a.m. by: hsueh    email:



Use the back button of your browser to return to the Gene List.

Click here to return to gene search form