BRCA1 and BRCA2: Different Roles in A Common Pathway of Genome Protection

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

PERSPECTIVES

ask from what type of DNA damage are


G E N O M I C I N S TA B I L I T Y I N C A N C E R — O P I N I O N
these proteins protecting the cell? How do
the two proteins communicate in a com-
BRCA1 and BRCA2: different roles mon pathway of genome protection? And
why are breast and ovarian epithelial cells
in a common pathway of genome preferentially susceptible to tumorigenesis?
We also discuss the association of breast

protection cancer subtypes with BRCA1 and BRCA2


deficiency.

Rohini Roy, Jarin Chun and Simon N. Powell Maintaining genome integrity
One aspect of maintaining genomic integ-
Abstract | The proteins encoded by the two major breast cancer susceptibility rity is mediated by a cellular network of
genes, BRCA1 and BRCA2, work in a common pathway of genome protection. signalling events (the DDR) that is trig-
However, the two proteins work at different stages in the DNA damage response gered in response to genotoxic stress. The
(DDR) and in DNA repair. BRCA1 is a pleiotropic DDR protein that functions in both DDR to DSBs involves sensors that can
detect broken ends, effectors that execute
checkpoint activation and DNA repair, whereas BRCA2 is a mediator of the core repair and mediators that facilitate inter-
mechanism of homologous recombination. The links between the two proteins are actions between sensors and effectors
not well understood, but they must exist to explain the marked similarity of human (FIG. 1). The DDR also includes the acti-
cancer susceptibility that arises with germline mutations in these genes. As vation of checkpoints that delay the cell
discussed here, the proteins work in concert to protect the genome from cycle before or during replication (G1/S
or intra‑S‑phase checkpoints) or before
double-strand DNA damage during DNA replication.
cell division (G2/M checkpoint) to ensure
that genetic errors are not transmitted to
The greatest risk factor for breast and ovar- the major mechanism for protecting the subsequent generations by allowing time
ian cancer is inheritance of a mutation integrity of the genome in proliferating for DNA repair. DSBs are considered to be
in one of the breast cancer susceptibility cells, because other DSB repair pathways the most threatening form of DNA damage,
genes, BRCA1 or BRCA2. BRCA1 and are error-prone and generate chromo- as the integrity of both strands of the DNA
BRCA2 are tumour suppressor genes, the some deletions and translocations2. A duplex is compromised simultaneously.
coding regions of which show no homology curious feature of HBOC syndrome is that DSBs can occur as by-products of DNA
to previously described proteins or to each BRCA1‑associated breast cancer is more replication or during exposure to ionizing
other. If one copy of either gene is mutated often oestrogen-receptor (ER) negative, radiation and other genotoxic compounds.
in the germ line, the result is hereditary whereas BRCA2‑associated breast cancers In mammalian cells, DSBs are repaired by
breast and ovarian cancer (HBOC) syn- have the same distribution of cancer sub- HR (which is mostly error-free), or by non-
drome, which is inherited in an autosomal- types as found sporadically. In hereditary homologous end-joining (NHEJ; which is
dominant manner. This syndrome is breast cancer for which there is no evidence error-prone). The genome is particularly
associated with not only early-onset breast of a BRCA1 or BRCA2 mutation, muta- susceptible to DNA damage during repli-
cancer but also an increased risk of ovarian, tions in the DNA damage response (DDR) cation because damage on a single strand
pancreatic, stomach, laryngeal, fallopian kinases CHK2 or ataxia-telangiectasia can be converted to double-strand damage
tube and prostate cancer. HBOC syndrome mutated (ATM) might account for the and lead to replication fork collapse. In
accounts for 5–7% of all cases of breast breast cancer predisposition3,4. Additional the absence of an intact HR pathway, these
cancer, and individuals with HBOC syn- inherited mutations may also occur in replication-associated DSBs can result in
drome have a lifetime risk of developing other members of the BRCA1–BRCA2–HR chromosome rearrangements and hence
breast cancer of 50–80%, and of 30–50% pathway, such as partner and localizer of genomic instability.
for ovarian cancer (TABLE 1). The estimated BRCA2 (PALB2) and BRCA1‑interacting HR repairs DSBs during the S and G2
frequency of developing other common protein C‑terminal helicase 1 (BRIP1; also phases of the cell cycle, when an intact sister
malignancies associated with a mutation in known as BACH1 or FANCJ), but currently chromatid can serve as a template for repair;
BRCA2 is only 0.1% for prostate cancer and the frequency of mutations in these genes it is also pivotal in maintaining replication
0.5% for pancreatic cancer, although the in hereditary breast cancer is low. In the fidelity. The protection of the genome by
relative risk is significantly increased (up to absence of known germline predisposi- HR involves damage recognition by the
20‑fold for prostate cancer and 10‑fold for tion for breast cancer, mutations in BRCA1 kinases ATM and ataxia telangiectasia and
pancreatic cancer)1. and BRCA2 are uncommon in sporadic Rad3-related (ATR), signal mediation by
In addition to having similar disease breast cancer. CHK2 and BRCA1, and initiation of repair
phenotypes, both proteins are known to Linking the biochemistry of BRCA1 and by the effectors BRCA2 and RAD51. There
function in homologous recombination BRCA2 function to a common pathway are also several facilitators of the HR path-
(HR), a vital DNA repair process that uses of genome protection often creates more way, such as PALB2 and BRIP1, and each
the undamaged sister chromatid to carry questions than answers. This Perspective of these facilitators is a predisposing factor
out high-fidelity repair of predominantly discusses how the BRCA1 and BRCA2 for HBOC syndrome when mutated, which
replication-associated DNA double- proteins function biochemically and how suggests that it is the BRCA1–BRCA2–HR
strand breaks (DSBs). HR appears to be this is related to their observed roles. We pathway that suppresses tumorigenesis.

68 | JANUARY 2012 | VOLUME 12 www.nature.com/reviews/cancer

© 2012 Macmillan Publishers Limited. All rights reserved


PERSPECTIVES

Table 1 | BRCA1 and BRCA2 functions: their domains and binding partners serine in SXXF motifs by ATM. The BRCA1‑
interacting proteins include abraxas, BRIP1
Function Domain Direct Indirect Refs and CtIP. The binding of these proteins
binding binding
make up separate BRCA1 macro-protein
BRCA1 complexes that have distinct and overlapping
Recruitment to BRCT Abraxas RAP80 19,20, functions in the DDR15 (FIG. 1; TABLE 1). A
DNA damage sites 108,109 fourth BRCA1‑containing complex medi-
DNA end BRCT and RING? CtIP MRN complex 13,14,22 ated through the BRCA1 coiled-coil domain
resection is composed of PALB2 and BRCA2 and is
G2/M checkpoint BRCT Abraxas RAP80 20,21 specifically involved in DSB repair by HR16–18.
How these multiple BRCA1 complexes work
BRCT CtIP MRN complex 13
in a coordinated manner is still unclear. It
SCD (S1423 and S1524 ATM MRN complex 111 will be interesting to uncover whether one
phosphorylation)
BRCA1‑containing complex is replaced with
S-phase SCD (S1387 phosphorylation) ATM MRN complex 112 a different BRCA1‑containing complex dur-
checkpoint
BRCT BRIP1 TOPBP1 40 ing the DDR, or whether BRCA1 functions
Repair during BRCT BRIP1 TOPBP1 110 as a platform on which initial DNA damage
DNA replication sensing proteins assemble and disassemble
HR Coiled-coil and S988 PALB2 BRCA2 16–18,25 and subsequent repair proteins associate.
phosphorylation As many of the current investigations have
been performed using global DNA damaging
BRCA2
agents, disentangling which BRCA1 complex
HR BRC RAD51 46,50,52 functions at different steps in the pathway
DBD DSS1 43–46 will be difficult. The study of a single, site-
N terminus PALB2 BRCA1 16–18,25 specific damage locus may help to shed light
on the crosstalk between the various BRCA1
C terminus RAD51 CDK2 53
macro-complexes and their assembly and
ATM, ataxia-telangiectasia mutated; BRIP1, BRCA1-interacting protein C‑terminal helicase 1; CDK2, disassembly.
cyclin-dependent kinase 2; CtIP, CtBP-interacting protein; DBD, DNA-binding domain; DSS1, deleted in
split-hand/split-foot syndrome; HR, homologous recombination; MRN, MRE11, RAD50 and Nijmegen
breakage syndrome protein 1 (NBS1); PALB2, partner and localizer of BRCA2; SCD, SQ/TQ cluster domain; BRCA1 and HR. BRCA1 is directly involved
TOPBP1, DNA topoisomerase 2-binding protein 1.
in HR‑mediated repair of DSBs19–21. BRCA1
binds to DSBs through its association with
BRCA1 functional domains and bind- unclear 10–13. The tumour suppressor func- the abraxas–RAP80 macro-complex, which
ing partners. BRCA1 is a versatile protein tion of the E3 ubiquitin ligase activity has associates with ubiquitylated histones
that links DNA damage sensing and DDR been questioned recently by the observation at DNA DSBs19 (FIG. 1). Next, BRCA1 is
effectors. BRCA1 interacts with tumour that in a knock-in mouse model expressing involved in processing DSBs through its
suppressors, DNA repair proteins and cell an E3‑ligase defective mutant of BRCA1, the interaction with CtIP and the MRN com-
cycle regulators through its various func- development of tumours was suppressed to plex (FIG. 1). The BRCA1–CtIP complex
tional domains and thereby has diverse the same extent as when wild-type BRCA1 promotes CtIP-mediated 5′-end resection
roles in multiple DNA repair pathways was expressed. BRCA1 ubiquitylation of of DSBs14, which is abrogated by three inde-
(particularly HR, NHEJ and single-strand CtBP-interacting protein (CtIP; also known pendent tumour-associated mutations in
annealing (SSA)) and in checkpoint regula- as RBBP8), a protein involved in DNA the BRCT domain of BRCA1 (REFS 22,23).
tion5,6. BRCA1 contains an amino‑terminal DSB resection through its association with BRCA1 is also required for RAD51 recruit-
RING domain that has E3 ubiquitin ligase the MRN complex (which is comprised of ment to the sites of DNA damage through its
activity (which catalyses protein ubiquityla- MRE11, RAD50 and Nijmegen breakage interactions with PALB2 and BRCA2 (FIG. 1).
tion) and a BRCT domain that facilitates syndrome protein 1 (NBS1; also known This interaction appears to be dependent
phospho-protein binding (FIG. 2a). Many as nibrin)), may have a role in DSB repair on CHK2‑mediated phos­phorylation
inherited cancer-associated BRCA1 muta- pathway choice, as CtIP-dependent resec- of S988 on BRCA1 (S.N.P., unpublished
tions have been found within the RING tion promotes HR and inhibits NHEJ14. observations). Importantly, knock-in mice
and BRCT domains, indicating that both Why the E3‑ligase function appears to be expressing an S971A mutant (in mice,
domains are involved in suppressing breast dispensable for tumour suppression has not S971 corresponds to human S988) develop
and ovarian cancer 7–9. BRCA1 E3 ubiquitin yet been satisfactorily answered, as many mammary and endometrial tumours after
ligase activity is enhanced when associ- tumour-producing mutations are located treatment with DNA damaging agents24.
ated with the RING domain of its partner in the RING domain, suggesting that there BRCA1‑deficient human cells express-
protein, BRCA1‑associated RING domain may be another function of the protein ing BRCA1‑S988A have defects in HR but
protein 1 (BARD1)10. The BRCA1–BARD1 associated with the RING domain that has retain normal checkpoint function and
heterodimer generates polyubiquitin chains yet to be defined. resistance to ionizing radiation, implying
at unconventional K6 linkages that do not The BRCT phosphopeptide-binding that the HR function of BRCA1 is distinct
appear to signal for protein degradation, but motif, which is conserved in multiple DDR from its other functions in the DDR and
may instead mediate downstream signalling proteins, is responsible for the association that this mutation causes a dissociation of
events through mechanisms that are still of BRCA1 with proteins phosphorylated on function25. Brca1‑null embryonic stem (ES)

NATURE REVIEWS | CANCER VOLUME 12 | JANUARY 2012 | 69

© 2012 Macmillan Publishers Limited. All rights reserved


PERSPECTIVES

Sensor Mediator Effector BRCA1 is to remove NHEJ proteins such as


DSB P p53‑binding protein 1 (53BP1) from DSBs35
H2AX to prevent aberrant end-joining and to regu-
RNF8
late the choice between HR and NHEJ. A
MDC1 RAP80
BRCA1 exon 11 deletion mutant exhibited
Abraxas BRCA1 decreased SSA in addition to decreased HR,
providing further evidence that BRCA1
Resection functions in both SSA and HR36. The rele-
P
MRE11–RAD50–NBS1 vance of SSA in genome stability and tumour
P suppression is not known, but is likely to be
CtIP
BRCA1 limited. Like NHEJ, SSA is also mutagenic,
BRCA1
producing deletions and insertions at sites
CHK2 P
ATM ATR P BRIP1 of long repeat sequences.
BRCA1 CHK1
Strand invasion TOPBP1
PALB2 ATR BRCA1 and checkpoint activation. The
BRCA1–BARD1 complex is involved in
BRCA2 the activation of G1/S, S-phase and G2/M
RAD51 checkpoints (TABLE 1). The G1/S-checkpoint
requires phosphorylation of BRCA1 by
RAD52 ATM or ATR, which facilitates phosphory­
HR
lation of p53 on S15. p53‑S15 phospho-
rylation is necessary for transcriptional
Figure 1 | Molecular mechanisms of the DNA damage response. In response to DNA| Cancer
Nature Reviews double-
induction of the cyclin dependent kinase
strand breaks (DSBs) or replication fork collapse (not shown), sensors (light blue) detect the damage,
and signalling mediators recruit or activate effectors that repair the damage and activate cell cycle
(CDK) inhibitor p21 and ionizing radiation-
checkpoints. BRCA1‑containing macro-complexes (dark blue) are crucial mediators of the DNA dam- induced G1/S checkpoint activation37.
age response. The BRCA1–abraxas–RAP80 complex associates with ubiquitylated histones near the BRCA1–BARD1 depletion compromises the
sites of DNA damage; this is dependent on phosphorylation of histone H2AX (γH2AX), mediator of induction of p21 and activation of the G1/S
DNA damage checkpoint protein 1 (MDC1) and RING finger protein 8 (RNF8). The BRCA1–CtBP- checkpoint in response to ionizing radia-
interacting protein (CtIP) complex associates with the MRN complex (which is comprised of MRE11, tion38. The exact mechanism of BRCA1–
RAD50 and Nijmegen breakage syndrome protein 1 (NBS1)), which senses DSBs and is responsible for BARD1 control of the S-phase and G2/M
DSB resection. The BRCA1–partner and localizer of BRCA2 (PALB2)–BRCA2 complex is important in checkpoints is not well characterized39.
mediating RAD51‑dependent homologous recombination (HR). CHK2‑dependent phosphorylation The BRCA1–BRIP1–DNA topoisomer-
of S988 in BRCA1 appears to be required for the BRCA1–PALB2–BRCA2 effector complex, which is
ase 2‑binding protein 1 (TOPBP1) macro-
important in RAD51‑mediated HR. The BRCA1–BRCA1‑interacting protein C-terminal helicase 1
(BRIP1)–DNA topoisomerase 2‑binding protein 1 (TOPBP1) complex is associated with DNA repair
complex appears to be necessary for the
during replication and may help mediate ataxia telangiectasia and Rad3‑related (ATR)–CHK1 signal- S‑phase checkpoint in response to stalled
ling, but its precise function is unknown. DNA damage is also recognized by ataxia-telangiectasia or collapsed replication forks40, whereas the
mutated (ATM) and ATR kinases, which phosphorylate BRCA1, BRCA1‑associated proteins and p53 BRCA1–abraxas–RAP80 macro-complex
and mediate signalling to form macro-complexes and activate cell cycle checkpoints. appears to be involved in the G2/M check-
point in response to ionizing radiation-
induced DNA damage (TABLE 1).
cells from mice expressing the human BRCA1 and other DNA repair pathways. The G2/M checkpoint is defective in cells
BRCA1‑S988A mutant also displayed no BRCA1 may also function in other DNA lacking functional BRCA1, BARD1, RAP80
difference in cell cycle profiles or sensitiv- repair pathways, including NHEJ and SSA. or abraxas20,21 (TABLE 1). Interestingly, partial
ity to DNA damaging agents compared to The role of BRCA1 in NHEJ is somewhat defects in G2/M checkpoint activation and
those expressing wild-type human BRCA1 controversial, as BRCA1 has been observed p53 stabilization were observed in mouse
(REF. 26). In this study, HR function was not to facilitate27,28, suppress29,30 or have no effect embryonic fibroblasts (MEFs) from knock-in
analysed, and sensitivity to DNA damaging on NHEJ31. These wide-ranging observa- mice expressing the cancer-associated
agents was measured by growth assay rather tions may be attributed to the variety of BRCA1‑S971A mutation (which is the
than clonogenic survival. The extent to assays used to measure NHEJ and the pos- equivalent to human BRCA1‑S988A)26.
which deficiency in HR contributes to cell sibility that BRCA1 has different roles in The checkpoint defect was mild and disap-
survival after ionizing radiation, even for the various subtypes of NHEJ. BRCA1 may peared 4 hours after treatment with ionizing
cells in S phase, remains a point of debate be involved in HR, NHEJ and SSA through radiation. Furthermore, after treatment
— HR is likely to be more important for its interaction with the MRN complex, with an alkylating agent, but not ionizing
surviving replication errors than for surviv- which is required for DNA end resection radiation, the levels of p53 were reduced
ing exogenous DNA damage resulting in before all three repair processes32,33. BRCA1 in MEFs expressing BRCA1‑S971A24,
DSBs, in which case other repair pathways recruitment to DSBs is facilitated by a DNA whereas in human HCC1937 cells express-
can be used. Similarly, tumour suppression damage-induced interaction of the BRCA1 ing BRCA1‑S988A, no detectable defect in
is likely to be related to fixing replication N terminus with the NHEJ protein KU80, the G2/M checkpoint was observed (S.N.P.,
errors rather than repairing DSBs, which thereby providing another mechanism for unpublished observations). The stability of
is why HR and tumour suppression are BRCA1 accumulation at DSBs34. A recent p53 has not been tested in BRCA1‑deficient
closely linked. study has proposed that a critical function of human cells and may need to be measured

70 | JANUARY 2012 | VOLUME 12 www.nature.com/reviews/cancer

© 2012 Macmillan Publishers Limited. All rights reserved


PERSPECTIVES

to verify whether, in human cells, BRCA1‑ a Human BRCA1 CHK2 ATM


S988A is a true separation-of-function muta-
Coiled
tion. These additional functions of BRCA1 RING NLS coil BRCT
P P P P
in checkpoints and in stabilizing p53 may S988 S1387, S1423,
contribute to genomic stability, which is pri- 1
S1524
1863
marily determined by the role of BRCA1 in
HR. Likewise, this broader range of BRCA1 1280 1524
SCD
functions may account for the severity BARD1 PALB2 • Abraxas
and pattern of genomic instability found • CtIP
in BRCA1‑deficient tumours relative to • BRIP1
BRCA2‑deficient tumours.
b Human BRCA2 CDK2
BRCA2 functional domains and binding
partners. In contrast to the multifunctional BRC repeats H OB OB OB NLS P
activities of BRCA1, the primary function S3291
1 T 3418
of BRCA2 is in HR (TABLE 1). BRCA2 medi-
ates the recruitment of the recombinase
21 39 1009 2083 DNA binding
RAD51 to DSBs; RAD51 recruitment is not
PALB2 RAD51 DSS1 RAD51
only essential for HR but is also responsible
for the tumour-suppressive function of this
Figure 2 | BRCA1 and BRCA2 functional domains. a | The BRCA1 amino terminus contains a RING
repair process41 (FIG. 1). BRCA2 contains a Natureand
Reviews | Cancer
domain that associates with BRCA1‑associated RING domain protein 1 (BARD1) a nuclear locali-
DNA-binding domain (DBD) that binds zation sequence (NLS). The central region of BRCA1 contains a CHK2 phosphorylation site on S988
single-stranded DNA (ssDNA) and double- (REF. 25). The carboxyl terminus of BRCA1 contains: a coiled-coil domain that associates with partner
stranded DNA (dsDNA) and eight BRC and localizer of BRCA2 (PALB2); a SQ/TQ cluster domain (SCD) that contains approximately ten poten-
repeats that bind RAD51 (FIG. 2b). The DBD tial ataxia-telangiectasia mutated (ATM) phosphorylation sites and spans amino acid residues
contains five components: a 190‑amino-acid 1280–1524; and a BRCT domain that binds ATM-phosphorylated abraxas, CtBP-interacting protein
α-helical domain, three oligonucleotide (CtIP) and BRCA1‑interacting protein C‑terminal helicase 1 (BRIP1). The BRCA1–abraxas complex is
binding (OB) folds that are ssDNA-binding associated with BRCA1 recruitment to sites of DNA damage19,20,108,109. The BRCA1–BRIP1 complex,
modules, and a tower domain (TD) that which also contains DNA topoisomerase 2-binding protein 1 (TOPBP1), is associated with DNA repair
protrudes from OB2 and binds dsDNA42. during replication110. The BRCA1–CtIP complex promotes ataxia-telangiectasia and Rad3‑related
(ATR) activation and homologous recombination (HR) by associating with the MRN complex (which is
The helical domain, OB1 and OB2 also asso-
comprised of MRE11, RAD50 and Nijmegen breakage syndrome protein 1 (NBS1)) and facilitating DNA
ciate with deleted in split-hand/split-foot double-strand break resection22. The central region of BRCA1, which contains the SCD, is phosphoryl-
syndrome (DSS1), which has been linked ated by ATM. This phosphorylation is important for BRCA1‑mediated G2/M and S‑phase checkpoint
to BRCA2 protein stabilization42–45. Ustilago activation, as expression of a BRCA1 mutant that lacks three of the phosphorylation sites (S1387,
maydis Brh2, the BRCA2 homologue in corn S1423 and S1524) fails to rescue defective checkpoint activation and ionizing radiation hypersensitiv-
smut, binds to DNA at the resected ends ity in a BRCA1‑deficient cell line111,112. b | The N terminus of BRCA2 binds PALB2 at amino acids 21–39
of a DSB, where both dsDNA and ssDNA (REF. 68). BRCA2 contains eight BRC repeats between amino acid residues 1009 and 2083 that bind
exist 46, presumably facilitating RAD51 fila- RAD51. The BRCA2 DNA-binding domain contains a helical domain (H), three oligonucleotide binding
ment formation at that site47. This implies (OB) folds and a tower domain (T), which may facilitate BRCA2 binding to both single-stranded DNA
that BRCA2 mediates RAD51 filament and double-stranded DNA46. This region also associates with deleted in split-hand/split-foot syndrome
(DSS1)42,44,45. The C terminus of BRCA2 contains an NLS and a cyclin-dependent kinase (CDK)
formation at the appropriate sites of ssDNA
phosphorylation site at S3291 that also binds RAD51 (REF. 53).
and prevents it from binding to dsDNA, as
supported by recent biochemical data using
purified BRCA2 (REF. 48). appears to be dispensable for HR in vivo (REFS 48,58,59). These studies showed that
Point mutations within BRC repeats and may be important for the disassembly the stoichiometry of RAD51 binding was
that compromise interactions with RAD51 of RAD51 complexes to facilitate mitotic 6–7 to 1, and that the protein could indeed
are found in individuals with HBOC syn- entry 2. Whether this association is impor- catalyse many steps of the RPA to RAD51
drome49. The BRC repeats have subtle dif- tant for suppression of breast and ovarian transition, as had been predicted by the
ferences in sequence and bind RAD51 with tumorigenesis is unknown. genetic studies. Future studies using the full-
varying affinity by mimicking the structure Much of our knowledge of BRCA2 length BRCA2 protein will help to further
of RAD51 monomers50. In addition to facili- comes from studying portions of the our understanding of the structure and
tating the recruitment of RAD51 to ssDNA, BRCA2 protein or investigating BRCA2 function of BRCA2.
the BRC repeats accelerate replication pro- orthologues, such as Brh2 in U. maydis and
tein A (RPA)-displacement from ssDNA by BRC‑2 in Caenorhabditis elegans 46,52,55–57. BRCA2, HR and replication fidelity.
RAD51 (REF. 51), block RAD51 nucleation at Insight into the details of mammalian Overwhelming evidence suggests that the
dsDNA and facilitate RAD51 filament for- BRCA2 function had been hampered by the primary function of BRCA2 is to facili-
mation on ssDNA by maintaining the active inability to isolate the full-length protein, tate HR (TABLE 1). BRCA2‑deficient cells
ATP-bound form of RAD51 on ssDNA52. which is 3,418 amino acids in humans. are defective in recruiting RAD51 to sites
The binding of RAD51 by the C terminus of Recently, however, three independent of DSBs and in repairing DSBs by HR60.
BRCA2 has been shown to be dependent on groups have successfully purified and func- Although human and mouse cells express-
CDK activity 53,54. This association, however, tionally validated full-length human BRCA2 ing a BRCA2 loss-of-function truncation

NATURE REVIEWS | CANCER VOLUME 12 | JANUARY 2012 | 71

© 2012 Macmillan Publishers Limited. All rights reserved


PERSPECTIVES

a Repair of a DSB b Repair at a fork c Repair behind a fork


Exposure to Blocking lesion at replication fork
Stalled replication fork
DSB-inducing agents

Or

Polymerase restart puts lesion


behind the fork, creating a DSG
Two-ended DSB One-ended DSB

Sister chromatid
BRCA1 and No BRCA1 or BRCA2,
BRCA2 BER creates DSB
BRCA1 and No BRCA1 BRCA1 and No BRCA1
BRCA2 or BRCA2 BRCA2 or BRCA2 HR to repair DSG BER to remove lesion?

HR (in S/G2) RAD52-mediated HR (in S/G2) RAD52-mediated


or NHEJ HR or NHEJ or BER HR (or BER)

Chromosome
aberrations

Figure 3 | Homologous recombination at different types of DNA dam- repair (BER) because an undamaged template is needed for repair;
Nature Reviews there-
| Cancer
age. Exogenous agents produce DNA double-strand breaks (DSBs) with fore, HR is the only available pathway for the repair of DSGs. Cells lacking
two ends (a), whereas during replication, blocking lesions on the template functional BRCA1 or BRCA2 exhibit an abundance of chromatid breaks,
strand can produce either one-ended DSBs (b) or daughter-strand gaps which indicates an attempt to repair DSGs in the absence of a functional
(DSGs) (c), both of which are preferentially repaired in the S and G2 phases BRCA-mediated HR pathway. DSBs that remain unrepaired behind a repli-
of the cell cycle by the BRCA1–BRCA2‑mediated homologous recombina- cation fork can also produce chromatid breaks or aberrant junctions or
tion (HR) pathway. During replication, template strand lesions may be exchanges. Hence, BRCA1 and BRCA2 have crucial roles in the repair of
repaired either behind the fork or at the fork. Gaps associated with lesions replication-associated lesions at or behind the replication fork. NHEJ,
on the parental strand behind the fork cannot be removed by base excision non-homologous end-joining.

mutant display some defects in replication created behind the replication fork (in addi- heterozygous cells must be haplo-insufficient,
and checkpoint control, BRCA2 is not tion to replication fork collapse) as a con- but this functional defect has so far been
essential for these processes61,62. Recently, sequence of gaps on the nascent strand that difficult to identify or investigate at the
BRCA2‑deficient hamster cells treated with are created by single-strand lesions on the cellular level. Defective HR can also be
hydroxyurea (which causes replication fork parental strand (FIG. 3). found in sporadic breast cancers despite
stalling and collapse) were shown to have the absence of a germline mutation in one
defects in maintaining the length of the Links between BRCA1 and BRCA2 of the crucial members of the HR pathway,
nascent strand of DNA, perhaps because Although germline Brca1 or Brca2 hetero­ as shown by array-comparative genomic
BRCA2 protects the nascent strand from zygous mutations in mice do not have a hybridization (aCGH) studies66 and func-
degradation at stalled replication forks2. strong phenotype, most homozygous muta- tional evaluations of human breast cancer
Thus, in addition to its role in repair by HR, tions are embryonically lethal63,64. Both Brca1 cells67. However, the nature of how these
these results imply a second role for BRCA2 and Brca2 homozygous mouse embryos are functional defects in HR occur is not yet
in protecting the replication fork. The criti- hypersensitive to ionizing radiation and have clear. Nevertheless, the common defect
cal evidence supporting these two functions widespread chromosome and chromatid in HR indicates that the function of the
of BRCA2 was a dissociation-of-function aberrations, which indicates error-prone BRCA1–BRCA2 pathway in mediating HR
mutant, S3291A, the expression of which repair of chromatid breaks. Interestingly, is important for tumour suppression in
allows normal DSB-induced HR but results the phenotype of BRCA-deficient mouse inherited and sporadic breast cancer.
in defective protection of replication forks. embryos mimics the phenotype of mice with
Without the dissociation-of-function inactivating mutations of Rad51 (REF. 65). PALB2 connects BRCA1 and BRCA2.
mutant, the reduced length of the nascent Therefore, these mouse models were the PALB2 binds directly to both BRCA1 and
strand after replication fork stalling could be initial evidence to indicate that BRCA1 and BRCA2 and thereby provides a physical link
secondary to deletions in the sister chroma- BRCA2 function in a common pathway of between the two proteins16–18 (FIG. 1). The
tid arising either from defective HR repair RAD51‑mediated HR63,64. N‑terminal coiled-coil domain of PALB2
of the collapsed replication fork or from In humans, the tumours that develop interacts with the coiled-coil domain of
defective daughter-strand gap (DSG) repair in patients with germline heterozygous BRCA1, and the C terminus of PALB2
by HR. We favour the view that the activity mutations in BRCA1 or BRCA2 are defec- interacts with the N terminus of BRCA2
of BRCA1 and BRCA2 can occur at DSBs tive in HR‑mediated repair. The germline (REF. 68) (FIG. 2b). The interaction of PALB2

72 | JANUARY 2012 | VOLUME 12 www.nature.com/reviews/cancer

© 2012 Macmillan Publishers Limited. All rights reserved


PERSPECTIVES

with BRCA2 was shown to be essential for anaemia pathway — which includes BRCA2 BRCA1 and BRCA2 in tumorigenesis
loading RAD51 onto RPA-bound ssDNA69. — has some functional overlap with the Common genetic alterations are associated
Furthermore, the BRCA1–PALB2 interac- BRCA1–BRCA2 pathway, but the human with heterozygous BRCA1 or BRCA2 muta-
tion is a prerequisite for the recruitment syndrome Fanconi anaemia (which is caused tions, and these include loss of the wild-type
of BRCA2 and RAD51 to the site of DNA by defects in members of the Fanconi anae- BRCA1 or BRCA2 allele (LOH), loss of TP53
damage and for HR, but had no impact on mia pathway) is markedly different to HBOC (which encodes p53), and loss of ATM or
BRCA1‑mediated S‑phase checkpoint acti- syndrome and is characterized by anaemia, CHK2 function. These additional alterations
vation17,18. Depletion of PALB2 expression skeletal abnormalities and predisposition to may allow cells to bypass checkpoint con-
in cells phenocopied BRCA2 deficiency and squamous cell carcinomas. In addition, the trols and evade apoptosis, and thereby initi-
abrogated the interaction between BRCA1 inheritance of Fanconi anaemia is autosomal ate tumorigenesis. The fact that both BRCA1
and BRCA2. Additionally, BRCA1 phos­ recessive (owing to the inheritance of two and BRCA2 mutation carriers display these
phorylation on S988 by CHK2 promotes hypo­morphic alleles), whereas HBOC syn- similar somatic alterations further confirms
formation of the BRCA1–PALB2–BRCA2 drome shows autosomal-dominant inherit- that their role in HR‑mediated repair is
complex (FIG. 1), which may explain why ance, with loss of the second allele (loss of important for tumour suppression.
mutating this site abrogates HR25. It is hetero­zygosity (LOH)) occurring in the can-
unknown whether there are other regulators cers that arise in mutation carriers. The effect Loss of the wild-type BRCA allele. When
of the BRCA1–PALB2–BRCA2 complex. of a defective BRCA1 or BRCA2 allele in the a tumour develops in patients with HBOC
germ line must cause haploinsufficiency of syndrome caused by BRCA mutation, loss
BRCA1 and BRCA2 function in a common HR to trigger the subsequent genetic altera- of the wild-type BRCA allele (LOH) was ini-
pathway. Repair by HR can be triggered at tions that result in cancer. Presumably, haplo­ tially always reported, as would be expected
ionizing radiation-induced two-ended DSBs insufficiency from a single defective allele of a tumour suppressor. However, more
or at one-ended DSBs that are generated by has different biological impacts compared recently this dogma has been questioned.
the cleavage of replication forks that have with biallelic inheritance of hypomorphic Initial studies of Brca1+/– and Brca2+/– mice
been stalled secondary to a blocking lesion. alleles. However, the total number of fami- showed no increase in tumour formation
In some instances, single-strand lesions that lies in the world that have been diagnosed compared with wild-type mice76, but sub-
do not produce a strand break are bypassed with Fanconi anaemia caused by defects in sequent studies using Brca1+/–Trp53+/– mice
by the replication machinery and replica- BRCA1–BRCA2 pathway genes (BRCA2 showed a slight increase in mammary car-
tion is restarted downstream of the lesion, (also known as FANCD1), BRIP1 (also known cinoma incidence compared with Trp53+/–
leaving behind a region of ssDNA or a DSG as FANCJ and BACH1), PALB2 (also known as mice77. All tumours in this study retained
in which no DSB end is present 70 (FIG. 3). FANCN), RAD51C (also known as FANCO) BRCA1 protein expression, which rules out
Replication-associated one-ended DSBs or SLX4 (also known as FANCP)) is small, epigenetic silencing. Similar results were
or DSGs recruit BRCA1, PALB2, BRCA2 and whether these patients show all the char- also observed in Brca2+/– mice: mammary
and RAD51. Brca1‑mutant mice exhibit acteristic features of Fanconi anaemia has tumours developed in a p53‑deficient back-
telomere dysfunction, chromosome trans- been debated73. ground78; however, in this study, epigenetic
locations and chromatid aberrations71, and In the BRCA1–BRCA2‑mediated HR silencing cannot be ruled out because levels
Brca2‑mutant mice accumulate chromatid pathway, BRCA1 functions upstream of of BRCA2 protein were not measured. A
breaks and aberrant chromatid exchanges61, BRCA2, the function of which is dependent study of breast cancer tissue samples from
providing further evidence in support of on BRCA1. In mammalian cells, HR can patients with HBOC syndrome caused by
a BRCA1–BRCA2‑mediated HR response also occur through an alternative, BRCA1– BRCA mutation showed that out of 18 cases
to replication-associated DNA damage. BRCA2‑independent, RAD52‑dependent in which LOH was observed, 11 patients
Similarly, Brca1- and Brca2‑mutant mice pathway. When BRCA2 function is disrupted showed loss of the mutant rather than the
develop thymic lymphomas61,71, which is a in a tumour cell, RAD52 helps the cell to stay wild-type allele, suggesting that loss of
common tumour that arises in mice owing viable. Indeed, cells that are simultaneously the wild-type BRCA allele is not required
to defective DSB repair. Moreover, condi- depleted of RAD52 and BRCA1, RAD52 and for tumorigenesis79. By contrast, all ovarian
tional expression of homozygous Brca1 or PALB2 (S.N.P., unpublished observations) or cancer tissue samples tested showed LOH of
Brca2 mutants in mammary epithelium was RAD52 and BRCA2 (REF. 74) exhibit synthetic the wild-type BRCA allele79. LOH was also
sufficient to generate mammary cancers72. lethality. Taken together, these results sup- not observed in a small study of pancreatic
BRCA1 is involved in DDR signalling, port the hypothesis that BRCA1 and BRCA2 cancer tissue samples from carriers of the
checkpoint activation and HR and may are connected in a common HR pathway Icelandic founder mutation BRCA2‑999del5,
also play a role in other DNA repair pro- that functions to repair DSBs, collapsed which produces a truncated form of BRCA2
cesses, such as NHEJ and SSA. Conversely, DNA replication forks or DSGs. Germline (REFS 80,81). However, it is important to
BRCA2 is primarily involved in HR. The mutations in genes involving this common keep in mind the methodological issues
human syndromes associated with BRCA1 HR pathway are all associated with HBOC about measuring LOH in microdissected
or BRCA2 germline mutations are almost syndrome, suggesting that this pathway is the tumour samples by PCR, because a small
identical, and the only common functional crucial tumour suppressor activity. Mutations amount of contaminating normal tissue
link between the two BRCA proteins is the in BRCA1 and BRCA2 are the predominant could produce a wild-type allele. In addi-
HR pathway. Therefore, it seems reason- cause of HBOC syndrome, with ATM tion, epigenetic silencing of the wild-type
able to conclude that the HR pathway is and CHK2 mutations being less common. allele, which cannot be detected by PCR,
crucial for protecting the genome and that Mutations in PALB2 that lead to HBOC syn- must also be considered. In the absence of
this pathway is disrupted in tumours aris- drome are very rare compared to mutations LOH, haploinsufficiency of BRCA activity
ing in these mutation carriers. The Fanconi in BRCA1 and BRCA2 (REF. 75). may cause enough genomic instability to

NATURE REVIEWS | CANCER VOLUME 12 | JANUARY 2012 | 73

© 2012 Macmillan Publishers Limited. All rights reserved


PERSPECTIVES

promote tumorigenesis. Although some fail to suppress transformation and exhibit it vulnerable to genetic instability in the
data suggest that loss of the wild-type allele gain of function transforming activity in context of BRCA1 or BRCA2 deficiency? A
may not be required for all BRCA-associated rat embryo fibroblasts91,92. Future studies number of theories have surfaced to explain
tumorigenesis, in most cases LOH does may reveal p53 functions that are uniquely the tissue specificity of HBOC syndrome,
occur. However, whether BRCA heterozygo- impaired in BRCA-deficient cells. The rarity but as yet none is definitive.
sity promotes loss of the wild-type allele or of these mutants in human cancer and their One hypothesis relates to the connection
whether loss occurs randomly is still unclear. multiple occurrences in BRCA-associated between BRCA1 function and the regulation
BRCA LOH is thought to occur by either breast tumours suggests that these novel p53 of ER signalling 96, whereby BRCA1 represses
deletion or gene conversion. When LOH mutants are selected for during malignant the transcription of hormone-mediated
does occur, a germline mutation in BRCA1 progression in the unique genetic back- signalling factors and therefore functions in
results in loss of the wild-type BRCA1 allele ground of BRCA1- or BRCA2‑mutation- growth control. Early in BRCA1 research,
but not the wild-type BRCA2 allele, and vice associated tumours. Therefore, the many transcriptional effects of BRCA1
versa. The similar pattern of LOH observed common HR defect in both BRCA1- and were described, including co-activation
in patients carrying a BRCA1 or a BRCA2 BRCA2‑deficient cells may be responsible for and co-repression of target genes, but the
mutation and the fact that only one of the the selection of these specific p53 mutants. importance of these effects has lessened
BRCA genes is affected in an individual (and over time, suggesting that some of the initial
not both genes in one individual) suggests Loss of ATM or CHK2 function. Consistent observations were a product of overexpres-
that tumorigenesis in BRCA1 and BRCA2 with being in the same signalling pathway sion studies97. A recent report has suggested
mutation carriers is primarily caused by as p53, loss of Atm or Chek2 also rescues the that the effect of BRCA1 is to maintain het-
functional inactivation of either BRCA pro- embryonic lethality of Brca1 mutant mice erochromatin, and the loss of Brca1 in mice
tein, and we suggest that it is the deficiency and leads to the development of multiple could be reversed by expressing histone 2A
in HR that leads to tumorigenesis (as tumours, although at a lower frequency fused to ubiquitin98. Some of the reported
discussed above). compared to mice with Brca1 and Trp53 effects of BRCA1 on transcription may actu-
mutations93. In addition, ATM expression ally be due to effects on chromatin structure.
Loss of p53 expression. p53 plays a vital part can be aberrantly reduced or lost in tumours However, these findings do not explain why
in maintaining genomic integrity by regulat- expressing BRCA1 or BRCA2 mutants com- germline BRCA2 mutations have the same
ing the transcription of target genes that are pared with sporadic tumours without BRCA1 tissue predisposition.
involved in cell cycle arrest, apoptosis and or BRCA2 mutations94. These data suggest Our preferred theory, which is still specu-
DNA repair 82. Multiple studies suggest that that the genomic instability caused by hetero­ lative but reflects ongoing work in our labora-
the loss of p53 cooperates with the loss of zygous BRCA mutations may lead to the tory, is that hormonally driven growth during
BRCA1 or BRCA2 in tumorigenesis78,83–86. selection of ATM-deficient cells. Although each menstrual cycle produces reactive oxy-
TP53 mutations are present in 30–50% of ATM, BRCA1 and BRCA2 are in a common gen species, which cause measurable oxida-
human cancers, and they occur in tumours signalling pathway, additional loss of ATM tive DNA damage99–102. The consequence of
with BRCA1 or BRCA2 mutations with activity may contribute to a selective growth oxidative DNA damage is the production of a
greater frequency than in sporadic tumours advantage95. This apparently paradoxical subset of lesions that cause DNA replication
with wild-type BRCA1 and BRCA2 (REFS finding makes sense when one considers that stress and result in one-ended DSBs or DSGs.
83,87). A susceptibility to develop breast ATM functions in multiple DDR signalling In other words, oxidative DNA damage can
carcinomas is also a clinical feature of Li– pathways. Alternatively, in genetically unsta- produce replication stress that demands the
Fraumeni syndrome, which is caused by ble BRCA-deficient tumours, random gains use of the BRCA1–BRCA2–HR pathway. This
germline mutation of TP53 and is associ- and losses occur across the genome — sec- explanation would account for the common
ated with predisposition to develop several ondary to genetic instability — and ATM loss features of BRCA1 and BRCA2 predisposing
types of cancer. Loss of Trp53 delays embry- could occur secondary to global instability. to breast and ovarian cancer.
onic lethality by 2 to 3 days in Brca1–/– or
Brca2–/– mice88. Loss of Trp53 also partially Breast and ovarian tissue tropism Subtypes of breast cancer. An additional
rescues the embryonic lethality of Palb2–/– Why breast and ovarian cancer? Given that unsolved mystery is why BRCA1 mutation
mice16,17,69,89. Presumably, chromosome BRCA1 and BRCA2 protect the genome carriers develop predominantly (but not
breaks caused by loss of BRCA function from errors that arise during DNA replica- exclusively) ER‑negative tumours, whereas
activate p53‑dependent checkpoint controls tion, it is logical that cells driven to replicate BRCA2 mutation does not favour the devel-
and/or apoptosis to prevent tumour forma- would develop potentially oncogenic genetic opment of any particular subtype of breast
tion. Selective pressures then favour the pro- alterations in the absence of BRCA1 or cancer 103 (TABLE 2). However, the breast can-
liferation of cells with loss of p53 function. BRCA2 function. However, most cancers cers that arose in Ashkenazi Jewish women
In support of this idea, T cells from T cell are driven to grow and divide, so this feature with founder mutations in BRCA1 or BRCA2
lineage-specific BRCA2‑deficient mice have alone does not determine why there is a were mostly triple-negative (ER-negative,
an accumulation of chromosome aberra- major predisposition to breast and ovarian progesterone receptor (PR)-negative and
tions that result in increased p53‑mediated cancer in individuals who lack functional ERBB2‑non-amplified)104, so the association
apoptosis90. However, p53 mutants (such as BRCA1 or BRCA2. One common feature is with ER expression and BRCA genes is not
T150I, G199R and R202S) that were identi- that breast and ovarian epithelial cells are rigid. These observations pose the question
fied specifically in tumours from BRCA1 subject to strong growth signals by hormo- of why there should be a difference in the
or BRCA2 mutation carriers retain the nal stimulation during the normal menstrual biological subtypes of breast cancer when
transactivation, checkpoint and apoptotic cycle. The question then becomes: what fea- the two proteins appear to be working in a
activities of wild-type p53, but they still tures of hormonally triggered growth make common pathway of DNA repair (TABLE 3).

74 | JANUARY 2012 | VOLUME 12 www.nature.com/reviews/cancer

© 2012 Macmillan Publishers Limited. All rights reserved


PERSPECTIVES

Table 2 | Human cancers arising in BRCA1 or BRCA2 mutation carriers


Cancer BRCA1 mutations BRCA2 mutations Notes
type
Breast 70–80% lifetime risk 50–60% lifetime risk Breast and ovarian cancer is the dominant cancer predisposition in BRCA1 and
BRCA2 mutation carriers. BRCA1 mutation carriers develop breast and ovarian
cancer at a younger age than BRCA2 mutation carriers113
Ovarian 50% lifetime risk 30% lifetime risk Breast and ovarian cancer is the dominant cancer predisposition in BRCA1 and
BRCA2 mutation carriers. LOH of the wild-type BRCA allele is always found
Prostate Ashkenazi Jewish founder 20-fold increased risk <1% of BRCA2 mutation carriers have prostate cancer. Prostate cancer is even
mutations are associated rarer in BRCA1 mutation carriers, except in members of the Ashkenazi Jewish
with increased risk population with BRCA1 mutations
Pancreatic Anecdotal evidence and Tenfold increased risk <1% of BRCA2 mutation carriers have pancreatic cancer. No incidence has
case reports only been clearly documented in BRCA1 mutation carriers
Gastric None reported Limited reports It is unclear whether stomach cancer is associated with BRCA2 mutations
Others None reported Brain, medulloblastoma, Fanconi anaemia subtype D1 (caused by BRCA2 mutations) is associated with
pharyngeal, CLL and AML cancer of the central nervous system
Fallopian Observed, but rare Rare This cancer type is like ovarian cancer, but it is a rare cancer overall and is still
tube uncommon in BRCA mutation carriers
AML, acute myeloid leukaemia; CLL, chronic lymphocytic leukaemia; LOH, loss of heterozygosity.

One explanation is that there is no differ- the resulting cellular consequences is not All of these explanations need more sup-
ence in the predisposition to genetic altera- understood. A final explanation is that the portive evidence and should be the focus of
tions between carriers of BRCA1 or BRCA2 cell-of-origin of ER‑negative tumours is future studies into the molecular genetics
mutations, but that the true connection to more susceptible to alterations in BRCA1, of breast cancer.
the ER‑negative subtype is the occurrence of whereas BRCA2 haplo-insufficiency predis- One intriguing clue regarding BRCA
co-inherited mutations (or even polymor- poses to loss of the second allele in all cell mutations and tissue tropism is that male
phisms) with the BRCA1 mutant haplotype. lineages, which would therefore result in the breast cancer, which is almost always
This idea has been studied in some detail, same biomarker profile as sporadic cancers. ER-positive, is more strongly associated
and at present there is no candidate co-
inherited mutation or polymorphism to sup-
port this hypothesis. A second explanation Table 3 | Characteristics of BRCA1- and BRCA2-mutation-associated breast cancers
is that the role of BRCA1 in transcriptional Phenotype BRCA1 BRCA2 Notes
co-activation (or co-repression), which is a ER expression Negative in 80–90% Positive in 60–65% One of the major
function not shared by BRCA2, produces mysteries to be solved
changes in gene expression that are sufficient
PR expression Predominantly negative Positive in the majority Less complete
to change the expression of the ER bio- of cases data relative to ER
marker. ER‑negative tumours have a char- expression
acteristic gene expression profile105, and so ERBB2 Usually absent ~15% have ERBB2 amplification
it would be intriguing to determine whether amplification amplification can occur in BRCA
the profile of BRCA1‑associated ER‑negative mutation carriers
and sporadic ER‑negative tumours is simi- Early onset Highly prevalent between Less prevalent between
lar. However, for this hypothesis to be cor- 30 and 50 years of age 40 and 70 years of age
rect, a mechanistic connection between Lobular cancers Less likely As frequent as in
BRCA1‑dependent transcription and sporadic breast cancer
the transcriptional profile of ER‑negative (~15%)
tumours should be established. A third High grade Likely Common More common than
explanation is that a different mutational sporadic cancers
spectrum is induced by BRCA1 heterozygo- Basal markers Frequent Less common Tumours have
sity compared with BRCA2 heterozygosity, cytokeratin profile of
which could potentially be due to the dif- basal or myoepithelial
ferences in DNA repair found with BRCA1 markers
deficiency (which could include defects in HR function Defective Defective Some debate over the
SSA and NHEJ). Analyses using aCGH show frequency of LOH for
some similarities between BRCA1- and the wild-type allele
BRCA2‑associated cancers, including large Prognosis No difference overall. No difference
deletions and amplifications66. However, relative to Local recurrence in the
sporadic cancer breast is increased with
some differences are also detectable66, such at the same conservative surgery and
as the locus specificity of the alterations. stage radiation therapy
The importance of these alterations in ER, oestrogen receptor; HR, homologous recombination; LOH, loss of heterozygosity; PR, progesterone
specific loci in terms of how they arise or receptor.

NATURE REVIEWS | CANCER VOLUME 12 | JANUARY 2012 | 75

© 2012 Macmillan Publishers Limited. All rights reserved


PERSPECTIVES

with BRCA2 mutations106. A second clue is BRCA1- or BRCA2-deficient cancers produce DNA breaks
that the range of cancer types observed in
BRCA2 mutation carriers is broader (BRCA2
mutation carriers can develop prostate and
pancreatic cancer, among others) than the
range observed in BRCA1 mutation carri-
ers. The mechanistic explanation here could
be linked to the cellular stresses to which Accumulation of chromatid breaks
the epithelial cells are exposed, although
why this selects for differences in the repair
pathway response to stress is not clear. Radials Deletions and insertions
BRCA2‑deficient cells have been reported
to be capable of carrying out SSA, whereas
BRCA1‑deficient cells cannot 36. However, Dicentrics Translocations
the role of SSA in the maintenance of (cell death) (cell survival) aCGH
genome integrity is not clear. In addition, Centromere Centromere
recent work has suggested that 53BP1 medi-
Translocation produced by
ates the genetic and chromosome rearrange- exchange and cell division
ments specifically in BRCA1‑deficient cells,
as loss of 53BP1 lessens the severity of the Figure 4 | BRCA-deficient cells accumulate chromatid breaks and chromatid exchanges. In
Nature Reviews
the absence of BRCA1 or BRCA2 function, chromatid breaks accumulate, resulting | Cancer
in aberrant chro-
repair defect caused by loss of BRCA1 (but
not loss of BRCA2)35. In this study, 53BP1 matid exchanges or other processes involving illegitimate end-joining. If two chromatid breaks are
protected DSB ends from 5′-end resection, joined to produce a chromosome structure containing two centromeres, a dicentric quadri-radial
chromosome is formed, which leads to cell death at mitosis. If an exchange is made with a chromatid
which initiates HR. If this was the entire
fragment without a centromere, processing and cell division can produce a viable cell with a transloca-
explanation for the effect of 53BP1, this pro- tion. All of the hallmarks of BRCA-deficient cancers can be explained by the production of chromatid
tein should also protect against the defective breaks and illegitimate end-joining. Without exchange events between different chromosomes, inter-
HR seen with BRCA2 defects, but this does stitial deletions, terminal deletions and insertions of chromosome fragments can originate from the
not seem to be the case. Instead, the speci- chromatid break. In the absence of homologous recombination, the resulting phenotypes can be seen
ficity towards BRCA1 would suggest that either by spectral karyotyping or by array-comparative genomic hybridization (aCGH), which detects
53BP1 is protecting the genome from defec- large losses and gains across the genome.
tive SSA, which could be more important in
genome stability than currently considered.
In conclusion, although we have stressed the frequency and pathological associations of development. The genetic instability result-
role of BRCA1 and BRCA2 functioning in a HR pathway defects both in primary breast ing from these growth defects and the loss
common pathway of DNA repair, there may tumours and in established breast cancer cell of DDR mediators leads to multiple genetic
be subtle differences between BRCA1- and lines. We have found that there is a signifi- gains and losses, but understanding which
BRCA2‑deficient cells that account for this cant prevalence of HR defects in sporadic are the crucial secondary targets and
curious difference in the range of cancers breast cancer (S.N.P., unpublished obser- which are nonspecific changes is one of the
that develop. vations). In addition, we have observed key research challenges in understanding the
the same phenotype in breast cancer cell aetiology of cancers associated with BRCA
Sporadic breast and ovarian cancers lines, none of which has known BRCA1 or loss. The observations that defects in HR can
We and others have reported that HR BRCA2 mutations or changes in BRCA1 be acquired rather than inherited support
defects occur in sporadic breast cancers and BRCA2 expression. The sporadic nature the view that genetic instability provides a
as well as the cancers arising in carriers of of these tumours with BRCA-like features selective advantage to breast cancer cells.
BRCA1 or BRCA2 mutations66,67,107. The would make the explanation less likely to Both mechanisms result in genetic instabil-
extent of this type of DNA repair defect be an unknown genetic alteration and more ity, which is perhaps triggered by oxidative
in other types of cancer is not known, as likely to be an epigenetic acquired event in metabolism producing replication stress.
specific assays are needed to detect this tumorigenesis. However, the implications Therefore, the number of BRCA1–BRCA2–
phenotype. Our approach has been to test are profound — the number of patients who HR pathway-defective breast cancers may be
the functional integrity of the HR pathway might be suitable for therapeutic strategies to 4 to 5 times greater than originally thought,
directly in ex vivo human tumour samples target defects in HR would be substantially making the number of patients who are
by examining the formation of nuclear foci expanded. amenable to DNA repair targeting strategies
of RAD51 and BRCA1 induced by ionizing much higher than previously estimated.
radiation67 (such nuclear foci indicate the Conclusions and perspectives
recruitment of DDR proteins to a site of BRCA1 and BRCA2 function in the DDR Rohini Roy, Jarin Chun and Simon N. Powell are at the
Molecular Biology Program and Department of
DNA damage). Others have suggested that during S and G2 phase by mediating HR Radiation Oncology, Memorial Sloan-Kettering Cancer
a ‘BRCA-like’ phenotype exists if tumour to maintain replication fidelity. The loss of Center, 1275 York Avenue, New York,
cells show the characteristic large-region BRCA1 or BRCA2 function in normal cells New York 10065, USA.
gains and losses that are also seen in BRCA- results in growth defects, which are required, Correspondence to S.N.P. 
e‑mail: powells@mskcc.org
deficient tumours (FIG. 4). We have initiated in combination with the subsequent loss
a more comprehensive assessment of the of other DDR mediators, for tumour doi:10.1038/nrc3181

76 | JANUARY 2012 | VOLUME 12 www.nature.com/reviews/cancer

© 2012 Macmillan Publishers Limited. All rights reserved


PERSPECTIVES

1. Foulkes, W. D. Molecular origins of cancer: inherited 27. Zhong, Q., Boyer, T. G., Chen, P. L. & Lee, W. H. 52. Carreira, A. et al. The BRC repeats of BRCA2
susceptibility to common cancers. N. Engl. J. Med. Deficient nonhomologous end-joining activity in cell- modulate the DNA-binding selectivity of RAD51. Cell
359, 2143–2153 (2008). free extracts from Brca1‑null fibroblasts. Cancer Res. 136, 1032–1043 (2009).
2. Schlacher, K. et al. Double-strand break repair- 62, 3966–3970 (2002). 53. Esashi, F. et al. CDK-dependent phosphorylation of
independent role for BRCA2 in blocking stalled 28. Zhong, Q., Chen, C. F., Chen, P. L. & Lee, W. H. BRCA1 BRCA2 as a regulatory mechanism for
replication fork degradation by MRE11. Cell 145, facilitates microhomology-mediated end joining of recombinational repair. Nature 434, 598–604
529–542 (2011). DNA double strand breaks. J. Biol. Chem. 277, (2005).
3. Futreal, P. A. et al. A census of human cancer genes. 28641–28647 (2002). 54. Ayoub, N. et al. The carboxyl terminus of Brca2 links
Nature Rev. Cancer 4, 177–183 (2004). 29. Moynahan, M. E., Chiu, J. W., Koller, B. H. & Jasin, M. the disassembly of Rad51 complexes to mitotic entry.
4. Rahman, N. & Stratton, M. R. The genetics of breast Brca1 controls homology-directed DNA repair. Mol. Curr. Biol. 19, 1075–1085 (2009).
cancer susceptibility. Annu. Rev. Genet. 32, 95–121 Cell 4, 511–518 (1999). 55. Saeki, H. et al. Suppression of the DNA repair defects
(1998). 30. Snouwaert, J. N. et al. BRCA1 deficient embryonic of BRCA2‑deficient cells with heterologous protein
5. Huen, M. S., Sy, S. M. & Chen, J. BRCA1 and its stem cells display a decreased homologous fusions. Proc. Natl Acad. Sci. USA 103, 8768–8773
toolbox for the maintenance of genome integrity. recombination frequency and an increased frequency (2006).
Nature Rev. Mol. Cell Biol. 11, 138–148 (2010). of non-homologous recombination that is corrected by 56. Esashi, F., Galkin, V. E., Yu, X., Egelman, E. H. &
6. Deng, C. X. & Brodie, S. G. Roles of BRCA1 and its expression of a brca1 transgene. Oncogene 18, West, S. C. Stabilization of RAD51 nucleoprotein
interacting proteins. Bioessays 22, 728–737 (2000). 7900–7907 (1999). filaments by the C‑terminal region of BRCA2. Nature
7. Friedman, L. S. et al. Confirmation of BRCA1 by 31. Wang, H. et al. Nonhomologous end-joining of ionizing Struct. Mol. Biol. 14, 468–474 (2007).
analysis of germline mutations linked to breast and radiation-induced DNA double-stranded breaks in 57. Petalcorin, M. I., Sandall, J., Wigley, D. B. &
ovarian cancer in ten families. Nature Genet. 8, human tumor cells deficient in BRCA1 or BRCA2. Boulton, S. J. CeBRC‑2 stimulates D‑loop formation
399–404 (1994). Cancer Res. 61, 270–277 (2001). by RAD‑51 and promotes DNA single-strand
8. Couch, F. J. & Weber, B. L. Mutations and 32. Fu, Y. P. et al. Breast cancer risk associated with annealing. J. Mol. Biol. 361, 231–242 (2006).
polymorphisms in the familial early-onset breast genotypic polymorphism of the nonhomologous end- 58. Jensen, R. B., Carreira, A. & Kowalczykowski, S. C.
cancer (BRCA1) gene. Breast Cancer Information Core. joining genes: a multigenic study on cancer Purified human BRCA2 stimulates RAD51‑mediated
Hum. Mutat. 8, 8–18 (1996). susceptibility. Cancer Res. 63, 2440–2446 (2003). recombination. Nature 467, 678–683 (2010).
9. Shattuck-Eidens, D. et al. A collaborative survey of 33. Paull, T. T., Cortez, D., Bowers, B., Elledge, S. J. & 59. Liu, J., Doty, T., Gibson, B. & Heyer, W. D. Human
80 mutations in the BRCA1 breast and ovarian Gellert, M. Direct DNA binding by Brca1. Proc. Natl BRCA2 protein promotes RAD51 filament formation
cancer susceptibility gene. Implications for Acad. Sci. USA 98, 6086–6091 (2001). on RPA-covered single-stranded DNA. Nature Struct.
presymptomatic testing and screening. JAMA 273, 34. Wei, L. et al. Rapid recruitment of BRCA1 to DNA Mol. Biol. 17, 1260–1262 (2010).
535–541 (1995). double-strand breaks is dependent on its 60. Yuan, S. S. et al. BRCA2 is required for ionizing
10. Wu, L. C. et al. Identification of a RING protein that association with Ku80. Mol. Cell. Biol. 28, 7380– radiation-induced assembly of Rad51 complex in vivo.
can interact in vivo with the BRCA1 gene product. 7393 (2008). Cancer Res. 59, 3547–3551 (1999).
Nature Genet. 14, 430–440 (1996). 35. Bunting, S. F. et al. 53BP1 inhibits homologous 61. Patel, K. J. et al. Involvement of Brca2 in DNA repair.
11. Sato, K. et al. Nucleophosmin/B23 is a candidate recombination in Brca1‑deficient cells by blocking Mol. Cell 1, 347–357 (1998).
substrate for the BRCA1‑BARD1 ubiquitin ligase. resection of DNA breaks. Cell 141, 243–254 (2010). 62. Daniels, M. J., Wang, Y., Lee, M. & Venkitaraman,
J. Biol. Chem. 279, 30919–30922 (2004). 36. Stark, J. M., Pierce, A. J., Oh, J., Pastink, A. & Jasin, M. A. R. Abnormal cytokinesis in cells deficient in the
12. Nishikawa, H. et al. Mass spectrometric and Genetic steps of mammalian homologous repair with breast cancer susceptibility protein BRCA2. Science
mutational analyses reveal Lys‑6‑linked polyubiquitin distinct mutagenic consequences. Mol. Cell. Biol. 24, 306, 876–879 (2004).
chains catalyzed by BRCA1‑BARD1 ubiquitin ligase. 9305–9316 (2004). 63. Evers, B. & Jonkers, J. Mouse models of BRCA1 and
J. Biol. Chem. 279, 3916–3924 (2004). 37. Siliciano, J. D. et al. DNA damage induces BRCA2 deficiency: past lessons, current understanding
13. Yu, X., Fu, S., Lai, M., Baer, R. & Chen, J. BRCA1 phosphorylation of the amino terminus of p53. Genes and future prospects. Oncogene 25, 5885–5897
ubiquitinates its phosphorylation-dependent binding Dev. 11, 3471–3481 (1997). (2006).
partner CtIP. Genes Dev. 20, 1721–1726 (2006). 38. Fabbro, M. et al. BRCA1‑BARD1 complexes are 64. Deng, C. X. & Scott, F. Role of the tumor suppressor
14. Yun, M. H. & Hiom, K. CtIP‑BRCA1 modulates the required for p53Ser‑15 phosphorylation and a G1/S gene Brca1 in genetic stability and mammary gland
choice of DNA double‑strand‑break repair pathway arrest following ionizing radiation-induced DNA tumor formation. Oncogene 19, 1059–1064
throughout the cell cycle. Nature 459, 460–463 damage. J. Biol. Chem. 279, 31251–31258 (2004). (2000).
(2009). 39. Xu, B., Kim, S. & Kastan, M. B. Involvement of Brca1 65. Tsuzuki, T. et al. Targeted disruption of the Rad51
15. Mohammad, D. H. & Yaffe, M. B. 14‑3‑3 proteins, FHA in S‑phase and G2-phase checkpoints after ionizing gene leads to lethality in embryonic mice. Proc. Natl
domains and BRCT domains in the DNA damage irradiation. Mol. Cell. Biol. 21, 3445–3450 (2001). Acad. Sci. USA 93, 6236–6240 (1996).
response. DNA Repair 8, 1009–1017 (2009). 40. Greenberg, R. A. et al. Multifactorial contributions 66. Stefansson, O. A. et al. Genomic profiling of breast
16. Sy, S. M., Huen, M. S. & Chen, J. PALB2 is an integral to an acute DNA damage response by BRCA1/ tumours in relation to BRCA abnormalities and
component of the BRCA complex required for BARD1-containing complexes. Genes Dev. 20, phenotypes. Breast Cancer Res. 11, R47 (2009).
homologous recombination repair. Proc. Natl Acad. 34–46 (2006). 67. Willers, H. et al. Utility of DNA repair protein foci for
Sci. USA 106, 7155–7160 (2009). 41. Moynahan, M. E., Pierce, A. J. & Jasin, M. BRCA2 is the detection of putative BRCA1 pathway defects in
17. Zhang, F., Fan, Q., Ren, K. & Andreassen, P. R. PALB2 required for homology-directed repair of chromosomal breast cancer biopsies. Mol. Cancer Res. 7,
functionally connects the breast cancer susceptibility breaks. Mol. Cell 7, 263–272 (2001). 1304–1309 (2009).
proteins BRCA1 and BRCA2. Mol. Cancer Res. 7, 42. Yang, H. et al. BRCA2 function in DNA binding and 68. Oliver, A. W., Swift, S., Lord, C. J., Ashworth, A. &
1110–1118 (2009). recombination from a BRCA2‑DSS1‑ssDNA structure. Pearl, L. H. Structural basis for recruitment of BRCA2
18. Zhang, F. et al. PALB2 links BRCA1 and BRCA2 in the Science 297, 1837–1848 (2002). by PALB2. EMBO Rep. 10, 990–996 (2009).
DNA-damage response. Curr. Biol. 19, 524–529 43. Li, J. et al. DSS1 is required for the stability of BRCA2. 69. Xia, B. et al. Control of BRCA2 cellular and clinical
(2009). Oncogene 25, 1186–1194 (2006). functions by a nuclear partner, PALB2. Mol. Cell 22,
19. Wang, B. et al. Abraxas and RAP80 form a BRCA1 44. Kojic, M., Yang, H., Kostrub, C. F., Pavletich, N. P. & 719–729 (2006).
protein complex required for the DNA damage Holloman, W. K. The BRCA2‑interacting protein DSS1 70. Nagaraju, G. & Scully, R. Minding the gap: the
response. Science 316, 1194–1198 (2007). is vital for DNA repair, recombination, and genome underground functions of BRCA1 and BRCA2 at stalled
20. Kim, H., Chen, J. & Yu, X. Ubiquitin-binding protein stability in Ustilago maydis. Mol. Cell 12, 1043–1049 replication forks. DNA Repair 6, 1018–1031 (2007).
RAP80 mediates BRCA1‑dependent DNA damage (2003). 71. McPherson, J. P. et al. A role for Brca1 in chromosome
response. Science 316, 1202–1205 (2007). 45. Kristensen, C. N., Bystol, K. M., Li, B., Serrano, L. & end maintenance. Hum. Mol. Genet. 15, 831–838
21. Kim, H., Huang, J. & Chen, J. CCDC98 is a Brenneman, M. A. Depletion of DSS1 protein disables (2006).
BRCA1‑BRCT domain-binding protein involved in the homologous recombinational repair in human cells. 72. Deng, C. X. & Brodie, S. G. Knockout mouse models
DNA damage response. Nature Struct. Mol. Biol. 14, Mutat. Res. 694, 60–64 (2010). and mammary tumorigenesis. Semin. Cancer Biol. 11,
710–715 (2007). 46. Yang, H., Li, Q., Fan, J., Holloman, W. K. & Pavletich, 387–394 (2001).
22. Yu, X., Wu, L. C., Bowcock, A. M., Aronheim, A. & N. P. The BRCA2 homologue Brh2 nucleates RAD51 73. Alter, B. P. & Kupfer, G. Fanconi anemia. GeneReviews
Baer, R. The C‑terminal (BRCT) domains of BRCA1 filament formation at a dsDNA–ssDNA junction. [online], http://www.ncbi.nlm.nih.gov/books/
interact in vivo with CtIP, a protein implicated in the Nature 433, 653–657 (2005). NBK1401/ (2011).
CtBP pathway of transcriptional repression. J. Biol. 47. Powell, S. N., Willers, H. & Xia, F. BRCA2 keeps Rad51 74. Feng, Z. et al. Rad52 inactivation is synthetically lethal
Chem. 273, 25388–25392 (1998). in line. High-fidelity homologous recombination with BRCA2 deficiency. Proc. Natl Acad. Sci. USA 108,
23. Chen, L., Nievera, C. J., Lee, A. Y. & Wu, X. Cell cycle- prevents breast and ovarian cancer? Mol. Cell 10, 686–691 (2011).
dependent complex formation of BRCA1.CtIP.MRN is 1262–1263 (2002). 75. Hellebrand, H. et al. Germline mutations in the PALB2
important for DNA double-strand break repair. J. Biol. 48. Thorslund, T. et al. The breast cancer tumor gene are population specific and occur with low
Chem. 283, 7713–7720 (2008). suppressor BRCA2 promotes the specific targeting of frequencies in familial breast cancer. Hum. Mutat. 32,
24. Kim, S. S. et al. Uterus hyperplasia and increased RAD51 to single-stranded DNA. Nature Struct. Mol. e2176–e2188 (2011).
carcinogen-induced tumorigenesis in mice carrying a Biol. 17, 1263–1265 (2010). 76. Gowen, L. C., Johnson, B. L., Latour, A. M., Sulik, K. K.
targeted mutation of the Chk2 phosphorylation site in 49. Venkitaraman, A. R. Linking the cellular functions of & Koller, B. H. Brca1 deficiency results in early
Brca1. Mol. Cell. Biol. 24, 9498–9507 (2004). BRCA genes to cancer pathogenesis and treatment. embryonic lethality characterized by neuroepithelial
25. Zhang, J. et al. Chk2 phosphorylation of BRCA1 Annu. Rev. Pathol. 4, 461–487 (2009). abnormalities. Nature Genet. 12, 191–194 (1996).
regulates DNA double-strand break repair. Mol. Cell. 50. Pellegrini, L. et al. Insights into DNA recombination 77. Cressman, V. L. et al. Mammary tumor formation in
Biol. 24, 708–718 (2004). from the structure of a RAD51–BRCA2 complex. p53- and BRCA1‑deficient mice. Cell Growth Differ.
26. Chang, S., Biswas, K., Martin, B. K., Stauffer, S. & Nature 420, 287–293 (2002). 10, 1–10 (1999).
Sharan, S. K. Expression of human BRCA1 variants in 51. Wooster, R. et al. Identification of the breast cancer 78. Jonkers, J. et al. Synergistic tumor suppressor activity
mouse ES cells allows functional analysis of BRCA1 susceptibility gene BRCA2. Nature 378, 789–792 of BRCA2 and p53 in a conditional mouse model for
mutations. J. Clin. Invest. 119, 3160–3171 (2009). (1995). breast cancer. Nature Genet. 29, 418–425 (2001).

NATURE REVIEWS | CANCER VOLUME 12 | JANUARY 2012 | 77

© 2012 Macmillan Publishers Limited. All rights reserved


PERSPECTIVES

79. King, T. A. et al. Heterogenic loss of the wild-type transformation suppression from other wild- 105. Sorlie, T. et al. Gene expression patterns of breast
BRCA allele in human breast tumorigenesis. Ann. type p53 functions. Oncogene 18, 2451–2459 carcinomas distinguish tumor subclasses with
Surg. Oncol. 14, 2510–2518 (2007). (1999). clinical implications. Proc. Natl Acad. Sci. USA 98,
80. Skoulidis, F. et al. Germline Brca2 heterozygosity 93. Cao, L. et al. ATM‑Chk2‑p53 activation prevents 10869–10874 (2001).
promotes KrasG12D-driven carcinogenesis in a murine tumorigenesis at an expense of organ homeostasis 106. Bane, A. L. et al. BRCA2 mutation-associated breast
model of familial pancreatic cancer. Cancer Cell 18, upon Brca1 deficiency. EMBO J. 25, 2167–2177 cancers exhibit a distinguishing phenotype based on
499–509 (2010). (2006). morphology and molecular profiles from tissue
81. Thorlacius, S. et al. A single BRCA2 mutation in male 94. Tommiska, J. et al. The DNA damage signalling microarrays. Am. J. Surg. Pathol. 31, 121–128
and female breast cancer families from Iceland with kinase ATM is aberrantly reduced or lost in (2007).
varied cancer phenotypes. Nature Genet. 13, BRCA1/BRCA2-deficient and ER/PR/ 107. Holstege, H. et al. BRCA1‑mutated and basal-like
117–119 (1996). ERBB2‑triple‑negative breast cancer. Oncogene 27, breast cancers have similar aCGH profiles and a high
82. Meek, D. W. Tumour suppression by p53: a role for 2501–2506 (2008). incidence of protein truncating TP53 mutations. BMC
the DNA damage response? Nature Rev. Cancer 9, 95. Sullivan, A. et al. Concomitant inactivation of p53 Cancer 10, 654 (2010).
714–723 (2009). and Chk2 in breast cancer. Oncogene 21, 1316–1324 108. Liu, Z., Wu, J. & Yu, X. CCDC98 targets BRCA1 to
83. Ramus, S. J. et al. Increased frequency of TP53 (2002). DNA damage sites. Nature Struct. Mol. Biol. 14,
mutations in BRCA1 and BRCA2 ovarian tumours. 96. Fan, S. et al. BRCA1 inhibition of estrogen receptor 716–720 (2007).
Genes Chromosomes Cancer 25, 91–96 (1999). signaling in transfected cells. Science 284, 109. Sobhian, B. et al. RAP80 targets BRCA1 to specific
84. Rowley, M. et al. Inactivation of Brca2 promotes 1354–1356 (1999). ubiquitin structures at DNA damage sites. Science
Trp53‑associated but inhibits KrasG12D‑dependent 97. Somasundaram, K. et al. Arrest of the cell cycle by the 316, 1198–1202 (2007).
pancreatic cancer development in mice. tumour-suppressor BRCA1 requires the CDK-inhibitor 110. Cantor, S. B. et al. BACH1, a novel helicase-like
Gastroenterology 140, 1303–1313.e3 (2011). p21WAF1/CiP1. Nature 389, 187–190 (1997). protein, interacts directly with BRCA1 and contributes
85. McAllister, K. A. et al. Spontaneous and irradiation- 98. Zhu, Q. et al. BRCA1 tumour suppression occurs via to its DNA repair function. Cell 105, 149–160
induced tumor susceptibility in BRCA2 germline heterochromatin-mediated silencing. Nature 477, (2001).
mutant mice and cooperative effects with a p53 179–184 (2011). 111. Cortez, D., Wang, Y., Qin, J. & Elledge, S. J.
germline mutation. Toxicol. Pathol. 34, 187–198 99. Sipe, H. J. Jr, Jordan, S. J., Hanna, P. M. & Mason, R. P. Requirement of ATM-dependent phosphorylation of
(2006). The metabolism of 17β‑estradiol by lactoperoxidase: a Brca1 in the DNA damage response to double-strand
86. Liu, X. et al. Somatic loss of BRCA1 and p53 in mice possible source of oxidative stress in breast cancer. breaks. Science 286, 1162–1166 (1999).
induces mammary tumors with features of human Carcinogenesis 15, 2637–2643 (1994). 112. Xu, B., O’Donnell, A. H., Kim, S. T. & Kastan, M. B.
BRCA1‑mutated basal-like breast cancer. Proc. Natl 100. Malins, D. C., Holmes, E. H., Polissar, N. L. & Phosphorylation of serine 1387 in Brca1 is
Acad. Sci. USA 104, 12111–12116 (2007). Gunselman, S. J. The etiology of breast cancer. specifically required for the Atm-mediated S‑phase
87. Schuyer, M. & Berns, E. M. Is TP53 dysfunction Characteristic alteration in hydroxyl radical-induced checkpoint after ionizing irradiation. Cancer Res. 62,
required for BRCA1‑associated carcinogenesis? Mol. DNA base lesions during oncogenesis with potential 4588–4591 (2002).
Cell. Endocrinol. 155, 143–152 (1999). for evaluating incidence risk. Cancer 71, 3036–3043 113. Krainer, M. et al. Differential contributions of BRCA1
88. Hakem, R., de la Pompa, J. L., Elia, A., Potter, J. & (1993). and BRCA2 to early-onset breast cancer. N. Engl.
Mak, T. W. Partial rescue of Brca15–6 early embryonic 101. Lavigne, J. A. et al. The effects of catechol- J. Med. 336, 1416–1421 (1997).
lethality by p53 or p21 null mutation. Nature Genet. O‑methyltransferase inhibition on estrogen
16, 298–302 (1997). metabolite and oxidative DNA damage levels in Acknowledgements
89. Bouwman, P. et al. Loss of p53 partially rescues estradiol-treated MCF‑7 cells. Cancer Res. 61, The authors are supported by grants from the US National
embryonic development of Palb2 knockout mice but 7488–7494 (2001). Cancer Institute and the Susan G. Komen For The Cure
does not foster haploinsufficiency of Palb2 in tumour 102. Hamada, J. et al. Increased oxidative DNA damage in foundation.
suppression. J. Pathol. 224, 10–21 (2011). mammary tumor cells by continuous epidermal growth
90. Cheung, A. M. et al. Loss of Brca2 and p53 factor stimulation. J. Natl Cancer Inst. 93, 214–219 Competing interests statement
synergistically promotes genomic instability and (2001). The authors declare no competing financial interests.
deregulation of T‑cell apoptosis. Cancer Res. 62, 103. Armes, J. E. et al. Distinct molecular pathogeneses of
6194–6204 (2002). early-onset breast cancers in BRCA1 and BRCA2
91. Crook, T. et al. p53 mutation with frequent novel mutation carriers: a population-based study. Cancer
DATABASES
National Cancer Institute Drug Dictionary: http://www.
codons but not a mutator phenotype in BRCA1- and Res. 59, 2011–2017 (1999).
cancer.gov/drugdictionary
BRCA2‑associated breast tumours. Oncogene 17, 104. Comen, E. et al. Relative contributions of BRCA1 and
Pathway Interaction Database: http://pid.nci.nih.gov
1681–1689 (1998). BRCA2 mutations to “triple-negative” breast cancer in
92. Smith, P. D. et al. Novel p53 mutants selected in Ashkenazi Women. Breast Cancer Res. Treat. 129, ALL LINKS ARE ACTIVE IN THE ONLINE PDF
BRCA-associated tumours which dissociate 185–190 (2011).

78 | JANUARY 2012 | VOLUME 12 www.nature.com/reviews/cancer

© 2012 Macmillan Publishers Limited. All rights reserved

You might also like