The BRCA1 tumor suppressor gene encodes a multidomain protein for which several functions have been described. These include a key role in homologous recombination repair (HRR) of DNA double-strand breaks, which is shared with two other high-risk hereditary breast cancer suppressors, BRCA2 and PALB2. Although both BRCA1 and BRCA2 interact with PALB2, BRCA1 missense variants affecting its PALB2-interacting coiled-coil domain are considered variants of uncertain clinical significance (VUS). Using genetically engineered mice, we show here that a BRCA1 coiled-coil domain VUS, Brca1 p.L1363P, disrupts the interaction with PALB2 and leads to embryonic lethality. Brca1 p.L1363P led to a similar acceleration in the development of Trp53-deficient mammary tumors as Brca1 loss, but the tumors showed distinct histopathologic features, with more stable DNA copy number profiles in Brca1 p.L1363P tumors. Nevertheless, Brca1 p.L1363P mammary tumors were HRR incompetent and responsive to cisplatin and PARP inhibition. Overall, these results provide the first direct evidence that a BRCA1 missense variant outside of the RING and BRCT domains increases the risk of breast cancer.

Significance:

These findings reveal the importance of a patient-derived BRCA1 coiled-coil domain sequence variant in embryonic development, mammary tumor suppression, and therapy response.

See related commentary by Mishra et al., p. 6080

BRCA1 and BRCA2 are the main high-risk hereditary breast cancer suppressor genes (1). The functions of BRCA1 and BRCA2 in homologous recombination repair (HRR) appear requisite for tumor suppression, because all known pathogenic variants analyzed to-date result in HRR defects (2–8). Besides BRCA1 and BRCA2, several other HRR genes are associated with a high risk for breast cancer, including PALB2 (9). PALB2 binds to BRCA2 via its WD40 domain and is important for BRCA2 localization to chromatin and sites of DNA damage (10). The coiled-coil domains of PALB2 and BRCA1 can also bind, and several groups have shown that disruption of this interaction affects HRR (11–13). Together this suggests that an intact BRCA1–PALB2–BRCA2 complex is a key requirement for hereditary breast cancer suppression. Although BRCA1 sequence variants disrupting the interaction with PALB2 have been identified in patients with suspected hereditary breast cancer (11), their impact on BRCA1-mediated tumor suppression is not clear. Thus far, all known pathogenic BRCA1 missense variants are restricted to the evolutionary conserved N-terminal RING and C-terminal BRCT domains. However, three missense variants within the PALB2-binding coiled-coil domain of BRCA1 (residues 1364–1437) are known to confer functional defects in HRR: p.M1400V, p.L1407P, and p.M1411T (2, 3, 11). Of these VUS, p.L1407P appears to have the most adverse effect on BRCA1 function. BRCA1 p.L1407P severely compromises the interaction of BRCA1 with PALB2, impairs HRR (2, 11), and leads to increased sensitivity to cisplatin and PARP1 inhibition (2, 3). More recently, a mouse model of Fanconi anemia harboring the BRCA1 p.L1407P murine equivalent, Brca1 p.L1363P, exemplified the importance of BRCA1–PALB2 interaction in vivo (14). Constitutive expression of this missense mutation recapitulated an array of Fanconi anemia–associated clinical phenotypes like growth retardation, infertility, and bone marrow failure, and highlighted defects in HRR and interstrand cross-link repair pathways, which rendered hypersensitivity to DNA-damaging agents. However, the impact of this clinically relevant BRCA1-PALB2 coiled-coil domain on breast cancer development and therapy response remains to be determined. Therefore, we set out to model the BRCA1 coiled-coil VUS p.L1407P in mice using CRISPR/Cas9 germline modification. Our Brca1 p.L1363P mouse model was analyzed for HRR defects and crossed into a K14cre conditional Brca1;Trp53 knockout model of BRCA1-associated breast cancer (15–17) to investigate the importance of BRCA1–PALB2 interaction for breast cancer suppression and treatment response.

Generation of Brca1L1363P mice

Brca1 p.L1363P mice were generated following a CRISPR/Cas9-mediated gene editing protocol (18). Zygotes isolated from FVB mice were coinjected with in vitro-transcribed Cas9 mRNA and Brca1 exon 13-targeting guide RNA (gRNA) from pX330 plasmid, along with oligonucleotide repair templates for the introduction of the p.L1363P variant (Supplementary Table S1). The Brca1 gRNA was designed using https://zlab.bio/guide-design-resources (19). Following implantation into pseudopregnant female FVB mice, offspring were screened for the desired missense mutation by Sanger sequencing, backcrossed once onto FVB mice, and maintained for the desired experimental genotype. To distinguish wild-type, heterozygous, and homozygous mutants, we established a three-primer PCR strategy (Supplementary Table S2). Brca1L1363P/F;Trp53F/F mice were crossed with a transgenic β-actin-cre germline deleter strain (FVB background; kindly provided by M. Vooijs and A. Berns, The Netherlands Cancer Institute, Amsterdam, the Netherlands) to obtain Brca1L1363P/+;Trp53Δ/+ mice. The β-actin-cre transgene was crossed out before compound mutant mice were used for experiments. Brca1L1363P/+ mice were bred with K14cre;Brca1F/F;Trp53F/F (KB1P) animals (FVB background; ref. 17) to generate K14cre;Brca1L1363P/F;Trp53F/F (KB1(L1363P)P) mice. All mouse experiments were approved by an independent animal ethics committee of The Netherlands Cancer Institute and executed according to Dutch and European guidelines.

Embryo isolations

Timed matings were established and the presence of a copulation plug demarcated embryonic day 0.5 (E0.5). Uteri from pregnant females were isolated at various days after coitum and kept on ice in PBS. Individual embryos were then isolated from the maternal decidua, and yolk sac tissue was collected for genotyping. Images were collected using Olympus SC30 digital camera and cellSens software (Olympus).

Mouse embryonic fibroblast isolation and culture

Brca1L1363P/+;Trp53Δ/+ mice carrying Brca1L1363P and Trp53Δ alleles in cis were intercrossed for the isolation of mouse embryonic fibroblasts (MEF). Heads and organs of E13.5 embryos were removed, and the remaining tissue was minced, rinsed in PBS, and incubated for 30 minutes in 0.5 mL 0.05% (w/v) trypsin-EDTA (GIBCO, 25300054) at 37°C. Cell aggregates were dissociated in DMEM (GIBCO, 11880028) supplemented with 10% volume for volume (v/v) FBS (Serana, S-FBSP-EU-015) and penicillin-streptomycin (P/S; GIBCO, 15070063). Cells were plated on 10 cm dishes and cultured at 3% O2.

Tumor-derived organoid and cell lines

Tumor-derived organoids were established following an adapted protocol (20). Cryopreserved tumor pieces were digested in Advanced DMEM/Ham's F-12 (AdDMEM/F12; GIBCO, 12634028) with 2 mg/mL collagenase type IV (GIBCO, 17104–019) for 30 minutes at 37°C while gently shaking. Suspensions were centrifuged at 1,500 rpm for 10 minutes and pellets were resuspended in ADDF+++ [AdDMEM/F12, 100× GlutaMAX (GIBCO, 35050061), 1 mol/L HEPES (Sigma-Aldrich, H4034), 100× P/S]. After centrifugation at 1,500 rpm for 10 minutes, pellets were resuspended in ADDF+++ and filtered (70 μm). Filtered organoids were pelleted (1,500 rpm, 10 minutes) and suspended in ENR growth medium [ADDF+++, 50x B-27 supplement (GIBCO, 17504044), 20 μg/mL EGF (Sigma-Aldrich, E4127), 500 mmol/L N-acetyl-L-cysteine (Sigma-Aldrich, A9165)] mixed 1:1 with Cultrex Reduced Growth Factor Basement Membrane Extract Type 2 (BME; Trevigen, 3533-001-02). Embedded organoids were seeded, incubated for 30 minutes at 37°C, and cultured in ENR medium supplemented with 10 μmol/L Nutlin-3a (Sigma-Aldrich, SML0580) under standard conditions (37°C, 5% CO2, 21% O2).

Mouse tumor cell lines were subsequently derived from respective early-passage organoid cultures. Organoids were dissociated by triturating in TrypLE (GIBCO, 12605010) and centrifuged at 800 rpm for 5 minutes. Pellets were suspended in DMEM/Nutrient Mixture F-12 media (DMEM/F12; GIBCO, 11320033) supplemented with 10% FBS, 100× P/S, 5 μg/mL insulin (Sigma-Aldrich, I0516), 5 ng/mL EGF, and 5 ng/mL cholera toxin (Gentaur, INABA 569B). Cell lines were cultured under low oxygen conditions (3% O2).

Coimmunoprecipitation

Cultured cells were trypsinized, counted, washed twice with PBS, and lysed in NETNG-300 lysis buffer (300 mmol/L NaCl, 20 mmol/L Tris-HCl pH 7.5, 1 mmol/L EDTA, 0.5% NP40, 10% glycerol) complemented with 1 mg/mL Pefabloc (Roche, 11429868001) and cOmplete Mini Protease Inhibitor Cocktail (Roche, 11836153001). Protein concentrations were determined with Pierce BCA Protein Assay Kit (Thermo Fisher Scientific, 23225). For BRCA1 coimmunoprecipitation, lysates were incubated overnight with mouse anti-BRCA1 antibody (21) at 4°C. Subsequently, lysates were incubated with Pierce agarose beads (Thermo Fisher Scientific, 20423) for 2 hours at 4°C and spin column-purified complexes were resolved on NuPAGE 4%–12% Bis-Tris SDS-MOPS polyacrylamide gels (Invitrogen, NP0321). Western blot protein analysis of input and immunoprecipitated samples used the above mouse anti-BRCA1 (1:10) and rabbit polyclonal anti-PALB2 (1:2,000) primary antibodies and rabbit anti-mouse IgG/HRP (1:2,000; Dako, P0260, RID: AB_2636929) and goat anti-rabbit IgG/HRP (1:2,000; Dako, P0448, RID: AB_2617138) secondary antibodies. Protein was visualized using ECL (Bio-Rad, 1705060) and imaged with Fusion FX7 imaging system (Vilber).

Alpha track assay

Experiments were performed as described previously (22) with small modifications. MEFs were seeded on coverslips overnight, washed with PBS, and covered with a mylar foil (Birkelbach Kondensatortechnik), allowing α-particle irradiation from below by turning over the coverslip. Irradiation was done using a 241 Am point source by moving the source underneath three areas of the coverslip for 30 seconds. Cells were incubated for 1 hour at 37°C and washed with ice-cold PBS. Subsequently, cells were extracted with cold CSK buffer [10 mmol/L HEPES-KOH pH 7.9, 100 mmol/L NaCl, 300 mmol/L sucrose, 3 mmol/L MgCl2, 1 mmol/L EGTA, 0.5% (v/v) Triton X-100] and cold CSS buffer [10 mmol/L Tris pH 7.4, 10 mmol/L NaCl, 3 mmol/L MgCl2, 1% (v/v) Tween 20, 0.5% (w/v) sodium deoxycholate] for 5 minutes each before fixation with 4% paraformaldehyde for 30 minutes at room temperature. Fixed cells were washed twice for 10 minutes (0.1% Triton X-100 in PBS) and washed for 30 minutes in blocking solution (0.5% BSA and 0.15% glycine in PBS). Primary antibodies were diluted in blocking buffer and incubated overnight at 4°C. Cells were washed twice for 10 minutes (0.1% Triton X-100 in PBS) and once quickly in blocking buffer. Secondary antibodies were diluted in blocking buffer and cells were incubated for at least 1 hour at room temperature in the dark. Finally, cells were washed twice with PBS and coverslips were mounted using Vectashield with DAPI. Quantification was done as described previously. Primary antibodies include: rabbit anti-53BP1 (1:1,000; Novus Biologicals, NB100-304, RID: AB_10003037) and mouse anti-RPA34 (1:1,000; Abcam, Ab2175, RID: AB_302873). Secondary antibodies include: Alexa Fluor 594 goat anti-rabbit IgG (Invitrogen, A-31631) and Alexa Fluor 488 goat anti-mouse IgG (1:1,000; Invitrogen, A-11001, RID: AB_2534069).

RAD51 foci formation

Cells were grown overnight on 12 mm glass coverslips and γ-irradiated (Best Theratronics) the following day with 10 Gy. EdU was added to refreshed medium after 2 hours of recovery, and 1 hour later cells were washed with PBS++ (PBS with 0.5 mmol/L MgCl2 and 1 mmol/L CaCl2). Ice-cold 0.5% Triton X-100 (Sigma-Aldrich, X100) in PBS was used for pre-extraction, followed by a PBS++ wash and fixation with 4% formaldehyde solution for 10 minutes at room temperature. Fixed cells were washed once with PBS and permeabilized in ice-cold 1:1 methanol:acetone for 20 minutes. Click-iT EdU Alexa Fluor 647 imaging kit (Invitrogen, C10340) was used for EdU detection. Staining buffer [SB; 1% BSA (Sigma-Aldrich, A8022), 0.15% glycine (Sigma-Aldrich, 15527), 0.1% Triton X-100, 0.5% FBS, in PBS] was used for blocking and antibody dilutions. Cells were incubated in SB for 15 minutes prior to the primary antibody incubation of 2 hours at room temperature. Secondary antibody incubation was for 1 hour at room temperature. Coverslips were mounted with VECTASHIELD Hardset Antifade Mounting Medium with DAPI (Vector Laboratories, H-1500). Antibodies included recombinant rabbit monoclonal anti-RAD51 primary antibody (1:2,000; Abcam, ab133534, RID: AB_2722613) and goat anti-rabbit Alexa Fluor 488 secondary antibody (1:400; Invitrogen, A-11008, RID: AB_143165). Stacked images (∼5 μm Z-volume) were taken with Leica TCS SP5 confocal microscope at 63× oil objective, and a minimum of 50 EdU+ cells per line was analyzed in ImageJ using an in-house macro. Automatic segmentation of nuclei based on DAPI signal thresholds was used to detect nuclei. Foci were detected using maximum intensity Z-stack projections with background-subtraction correction, in addition to filtering parameters for discriminating foci diameter size and adjusting signal to noise. The mean intensity EdU signal was used to select S phase nuclei.

Colony formation assay

MEFs were plated in triplicate at a density of 1,000 cells per well in a 12-well plate. The following day, cisplatin (in saline-mannitol, Mayne Pharma) or AZD2461 PARP inhibitor (in DMSO, Syncom) was added to refreshed medium at varying concentrations. After 6 days, MEFs were fixed with 4% formaldehyde for 15 minutes, stained with crystal violet (Supelco, 101408) for 30 minutes, washed three times with demineralized water, and dried overnight. Plate images were taken with Gel Count (Oxford Optronix). For quantification, crystal violet was dissolved in 10% acetic acid for 1 hour while shaking, and absorbance was measured at 560 nm using Infinite 200 microplate reader (Tecan).

DNA fiber analysis

DNA fiber analysis was carried out according to the previously defined standard protocol (23, 24). Briefly, cells were sequentially pulse labeled with 30 μmol/L 5-Chloro-2′-deoxyuridine (CldU; Sigma-Aldrich, C6891) and 250 μmol/L idoxuridine (IdU; European Pharmacopoeia, I0050000) for 15 minutes each. For assessing fork degradation, cells were treated with 4 mmol/L hydroxyurea (Sigma-Aldrich, H8627) for 3 hours. After incubation, cells were collected and resuspended in PBS at 2.5 × 105 cells/mL. Labeled cells were mixed 1:1 with unlabeled cells, and 2.5 μL of cells were added to 7.5 μL of lysis buffer [200 mmol/L Tris-HCl pH 7.5, 50 mmol/L EDTA, 0.5% (w/v) SDS] on a glass slide. After 8 minutes, the slides were tilted at 15°–45° and the resulting DNA spreads were air dried and fixed in 3:1 methanol:acetic acid overnight at 4°C. The fibers were denatured with 2.5 mol/L HCl for 1 hour, washed with PBS, and blocked with 0.2% Tween 20 in 1% BSA/PBS for 40 minutes. The newly replicated CldU and IdU tracks were incubated for 2.5 hours in the dark at room temperature with anti-BrdU antibodies recognizing CldU (1:500; Abcam, ab6326, RID: AB_305426) and IdU (1:100; B44; BD Biosciences, 347580, RID: AB_400326), followed by a 1-hour incubation with secondary antibodies at room temperature in the dark: Alexa Fluor 488 goat anti-mouse IgG (1:300; Invitrogen, A11001, RID: AB_2534069) and Cy3 donkey anti-rat IgG (1:150; Jackson Immuno-Research Laboratories, 712-166-153, RID: AB_2340669). Fibers were visualized and imaged with Carl Zeiss Axio Imager D2 microscope using 63× Plan Apo 1.4 NA oil immersion objective. Data analysis was carried out with ImageJ software64 and statistical analysis using GraphPad Prism.

Metaphase spreads and chromosomal aberrations

Metaphase spreads were carried out according to the standard protocol described previously (24). Briefly, cells were treated with 500 nmol/L olaparib (AZD2281; Selleck Chemicals, S1060) for 24 hours before preparing metaphase spreads (final confluence of 50%–80%). Cells were arrested in metaphase by incubating with colcemid (N-methyl-N-deacetyl-colchicine; Roche, 10295892001) for the last 16 hours before harvesting the cells. Collected cells were then treated with hypotonic solution (0.075 mol/L KCl) for 30 minutes at 37 °C and fixed with 3:1 methanol:acetic acid. Telomere-FISH was further carried out to study chromosomal aberration. Metaphases were hybridized with telomere repeat-specific peptide nucleic acid probes (PNA Bio, F1002) as described previously (25) to label the telomeres. A minimum of 60 metaphase images were obtained using Carl Zeiss Axio Imager D2 microscope with 63x Plan Apo 1.4 NA oil immersion objective and analyzed with ImageJ software64.

Copy number variation sequencing

llumina HiSeq 2500 using V4 chemistry (Illumina Inc.) was performed as previously described with 65 base single reads (26), and aligned and summarized as done before (27). The entire analysis was implemented by Julian de Ruiter using Snakemake (Snakemake version 5.19.2; wrapper version 0.60.0; ref. 28) and is freely available on GitHub (https://github.com/jrderuiter/snakemake-cnvseq). An unsupervised clustering was performed on the segmented copy number data. Copy number instability was scored by calculating the fraction of bins with copy number values above or below a threshold of 2.5 and 1.5, respectively, in the segmented copy number data. Copy number variation sequencing (CNV-seq) data generated in this study have been deposited in the NCBI's Gene Expression Omnibus (GEO) database (GEO GSE182449).

RNA sequencing

llumina TruSeq mRNA libraries were generated and sequenced with 65 base single reads on HiSeq 2500 using V4 chemistry (Illumina Inc.) as described previously (26), and reads were aligned and expression counts generated as before (27). The analysis was implemented by Julian de Ruiter using Snakemake (Snakemake version 5.19.2; wrapper version 0.63.0; ref. 28) and is freely available on GitHub (https://github.com/jrderuiter/snakemake-rnaseq). Singscore package (29) was used to determine epithelial–mesenchymal transition (EMT) scores based on the EMT signature in Supplementary Table S2B of Huang and colleagues (30). RNA sequencing data generated in this study have been deposited in the NCBI's GEO database (GEO GSE182448).

Orthotopic transplantation and drug interventions

Cryopreserved tumor pieces (1–2 mm diameter) from KP, KB1P, and KB1(L1363P)P spontaneous tumor donors were transplanted orthotopically into the fourth mammary gland of recipient female FVB mice, ages 8–12 weeks. Once outgrown tumors reached 200 mm3 (day 0), animals were randomly apportioned among three treatment arms (cisplatin, PARP inhibitor AZD2461, or no treatment). Cisplatin (6 mg/kg) was administered intravenously into the tail on day 0. Repeat doses were given after 14 days to unresponsive tumors (>100 mm3); responsive tumors (<100 mm3) received a repeat dose once returning to 200 mm3. A maximum of four doses was administered. AZD2461 (100 mg/kg) was administered by oral gavage for 29 consecutive days (days 0–28). Animals of all treatment arms were sacrificed when tumor burden reached 1,200 mm3 or if they showed signs of distress outlined in the Code of Practice for Animal Experimentation in Cancer Research.

Statistical analysis

Details for statistical methods are explained in each figure legend. All multiple comparison follow-up tests (Figs. 2B, 2C, 2F, 2G, and 4I) were performed relative to the indicated wild-type Brca1 MEF and tumor cell line controls. Statistical analysis for RAD51 IRIF (Figs. 2C and 4I) was performed on only the γ-irradiated groups, non-irradiated lines were excluded from comparison. Log-rank (Mantel–Cox) was used for Kaplan–Meier survival curves while applying Bonferroni multiple comparisons correction with a significance threshold of P = 0.05. All statistical significance was determined using GraphPad Prism.

Generation of Brca1 p.L1363P (FVB) mice

We used CRISPR/Cas9-mediated genome editing in FVB mouse zygotes to model the BRCA1 coiled-coil domain VUS c.4220T>C p.L1407P, which disrupts the interaction of BRCA1 with PALB2. The BRCA1 coiled-coil domain is well conserved between humans and mice (Fig. 1A), where the murine equivalent of human p.L1407P is p.L1363P. As expected, the COILS algorithm (31) predicts that both human BRCA1 p.L1407P and mouse BRCA1 p.L1363P disable the alpha-helical structure of the coiled-coil domain (Fig. 1B). Because CRISPR/Cas9 often modifies both alleles of a target gene and Brca1 is essential for embryonic development (32, 33), we combined the homology-dependent recombination template for introduction of the p.L1363P variant with a second template that only disrupts gRNA recognition (Supplementary Fig. S1A). Through this approach, we obtained a Brca1 p.L1363P (Brca1LP; Supplementary Fig. S1A and S1B) founder, which was crossed with wild-type FVB mice for subsequent experiments. Heterozygous Brca1LP mice are viable without any apparent aberrant phenotypes.

Figure 1.

Brca1L1363P/L1363P (FVB) mice are not viable. A, DNA and protein changes for human BRCA1 p.L1407P and mouse Brca1 p.L1363P. The missense mutations and leucine to proline amino acid change are indicated in red. Additional silent mutations for the mouse p.L1363P variant are indicated in green. B, Probability of coiled-coil formation for human BRCA1 p.L1407P and mouse Brca1 p.L1363P as determined by the COILS prediction algorithm. C, E10.5 Brca1+/+, Brca1L1363P/+, and Brca1L1363P/L1363P mouse embryo littermates and E10.5 Brca1L1363P/L1363P;Trp53Δ/+ and Brca1+/+;Trp53Δ/+ embryo littermates. Scale bar, 1 mm. D, Representative images of P21 IHC staining in E13.5 Brca1L1363P/+;Trp53Δ/+ (LP/+;Δ/+) and Brca1L1363P/L1363P;Trp53Δ/+ (LP/LP;Δ/+) mouse fetal heart and liver tissue sections. Scale bar, 50 μm. E, Quantification of P21 IHC expression. Error bars, SD (n = 4 per group). *, P = 0.0286 (Mann–Whitney U test).

Figure 1.

Brca1L1363P/L1363P (FVB) mice are not viable. A, DNA and protein changes for human BRCA1 p.L1407P and mouse Brca1 p.L1363P. The missense mutations and leucine to proline amino acid change are indicated in red. Additional silent mutations for the mouse p.L1363P variant are indicated in green. B, Probability of coiled-coil formation for human BRCA1 p.L1407P and mouse Brca1 p.L1363P as determined by the COILS prediction algorithm. C, E10.5 Brca1+/+, Brca1L1363P/+, and Brca1L1363P/L1363P mouse embryo littermates and E10.5 Brca1L1363P/L1363P;Trp53Δ/+ and Brca1+/+;Trp53Δ/+ embryo littermates. Scale bar, 1 mm. D, Representative images of P21 IHC staining in E13.5 Brca1L1363P/+;Trp53Δ/+ (LP/+;Δ/+) and Brca1L1363P/L1363P;Trp53Δ/+ (LP/LP;Δ/+) mouse fetal heart and liver tissue sections. Scale bar, 50 μm. E, Quantification of P21 IHC expression. Error bars, SD (n = 4 per group). *, P = 0.0286 (Mann–Whitney U test).

Close modal

Homozygous Brca1 p.L1363P (FVB) mice die during embryonic development

For a first functional analysis of Brca1 p.L1363P in vivo, heterozygous Brca1LP mice were intercrossed and their offspring was genotyped. No Brca1LP/LP mice were born; therefore, embryos were analyzed at different stages of embryonic development. Like Brca1-null mice, Brca1LP/LP mice showed a general growth defect at mid-gestation (Fig. 1C). Although the phenotype was less severe than those of Brca1-null mice (32, 33) or mice carrying pathogenic mutations in the Brca1 RING or BRCT domain-encoding sequences (15, 16), no living embryos were observed after E11.5 (Table 1).

Table 1.

Genotype distribution of Brca1L1363P/+ x Brca1L1363P/+ offspring.

AgeTotalBrca1+/+Brca1LP/+Brca1LP/LPBrca1 n.d.P
E10.5 20 4 (5) 9 (10) 7 (5)  0.5769 
E11.5 30 7 (8) 12 (15) 8 (8) 3a 0.8157 
E13.5 30 5 (8) 22 (15) 2a (8) 1a 0.0152 
Postnatal 162 63 (41) 99 (81) 0 (41)  <0.0001 
AgeTotalBrca1+/+Brca1LP/+Brca1LP/LPBrca1 n.d.P
E10.5 20 4 (5) 9 (10) 7 (5)  0.5769 
E11.5 30 7 (8) 12 (15) 8 (8) 3a 0.8157 
E13.5 30 5 (8) 22 (15) 2a (8) 1a 0.0152 
Postnatal 162 63 (41) 99 (81) 0 (41)  <0.0001 

Note: Expected numbers of mice are indicated in parentheses.

Abbreviation: n.d., not determined.

aResorption. P values (two-tailed) show the χ2 test for goodness of fit.

Loss of Trp53 is known to delay embryonic lethality in Brca1- and Palb2-null mice (33–35), so we questioned whether this phenotype would likewise occur in Brca1LP/LP mice on a Trp53-deficient background. Brca1 and Trp53 are both located on chromosome 11 (at 65.18 and 42.83 cM, respectively), so we bred male mice with Brca1LP and Trp53Δ2–10 (Trp53Δ; ref. 36) in cis (as determined from genotyped offspring) and crossed these with Brca1LP/+ or Brca1LP/+;Trp53Δ/+ female mice. While heterozygous inactivation of Trp53 delayed embryonic lethality in Brca1LP/LP mice, it did not result in viable offspring (Table 2), Brca1LP/LP;Trp53Δ/+ embryos were smaller than Brca1-proficient littermates (Fig. 1C) but continued to develop until at least E13.5. For a molecular explanation to the growth defect of Brca1LP/LP embryos, we checked markers of proliferation, apoptosis, and cell-cycle regulation (Fig. 1D and E; Supplementary Fig. S1C). In line with the partial rescue of embryonic development, heterozygous deletion of Trp53 allowed most E13.5 Brca1LP/LP cells to proliferate, as indicated by the expression of Ki-67, and we observed only few cleaved caspase 3–positive apoptotic cells (Supplementary Fig. S1C). However, similar to Brca1-knockout embryos (32), E13.5 Brca1LP/LP;Trp53Δ/+ embryos showed increased expression of the TP53 effector protein Cyclin-dependent kinase inhibitor 1A (P21) in several tissues including the embryonic heart and liver (Fig. 1D and E). P21-mediated G1 phase cell cycle arrest can explain the marked growth suppression of critical organs such as heart and liver and likely contributes to the embryonic lethality of Brca1LP/LP and Brca1LP/LP;Trp53Δ/+ mice.

Table 2.

Genotype distribution of Brca1L1363P/+;Trp53Δ/+ x Brca1L1363P/+ offspring.

AgeTotalBrca1+/+ Trp53+/+Brca1+/+ Trp53Δ/+Brca1LP/+ Trp53+/+Brca1LP/+ Trp53Δ/+Brca1LP/LP Trp53Δ/+P
E13.5 26 1 (7) 0 (0) 10 (7) 9 (7) 6 (7) 0.0566 
Postnatal 78 19 (20) 1 (0) 33 (20) 25 (20) 0 (20) <0.0001 
AgeTotalBrca1+/+ Trp53+/+Brca1+/+ Trp53Δ/+Brca1LP/+ Trp53+/+Brca1LP/+ Trp53Δ/+Brca1LP/LP Trp53Δ/+P
E13.5 26 1 (7) 0 (0) 10 (7) 9 (7) 6 (7) 0.0566 
Postnatal 78 19 (20) 1 (0) 33 (20) 25 (20) 0 (20) <0.0001 

Note: Expected numbers of mice are indicated in parentheses and are based on crosses with Brca1LP and Trp53Δ mutations in cis. P values (two-tailed) show the χ2 test for goodness of fit.

In the complete absence of TP53, Brca1LP/LP mice developed apparently normal until at least E13.5, although no postnatal survival was observed upon compound heterozygous intercrosses (Table 3). This allowed us to isolate E13.5 MEFs from Brca1LP/LP;Trp53Δ/Δ embryos and Brca1LP/+;Trp53Δ/Δ littermate controls for further evaluation of the functional consequences of Brca1 p.L1363P.

Table 3.

Genotype distribution of Brca1L1363P/+;Trp53Δ/+ x Brca1L1363P/+;Trp53Δ/+ offspring.

AgeTotalBrca1+/+ Trp53+/+Brca1+/+ Trp53Δ/+Brca1+/+ Trp53Δ/ΔBrca1LP/+ Trp53+/+Brca1LP/+ Trp53Δ/+Brca1LP/+ Trp53Δ/ΔBrca1LP/LP Trp53+/+Brca1LP/LP Trp53Δ/+Brca1LP/LP Trp53Δ/Δ
Postnatal 118 25 16 22 46 
AgeTotalBrca1+/+ Trp53+/+Brca1+/+ Trp53Δ/+Brca1+/+ Trp53Δ/ΔBrca1LP/+ Trp53+/+Brca1LP/+ Trp53Δ/+Brca1LP/+ Trp53Δ/ΔBrca1LP/LP Trp53+/+Brca1LP/LP Trp53Δ/+Brca1LP/LP Trp53Δ/Δ
Postnatal 118 25 16 22 46 

Note: Offspring from compound heterozygous parents with Brca1LP and Trp53Δ in cis or trans.

BRCA1 p.L1363P is unable to bind PALB2 and shows hypomorphic activity in HRR

To verify whether mouse Brca1 p.L1363P phenocopies human BRCA1 p.L1407P, we analyzed Brca1LP/LP;Trp53Δ/Δ (LP/LP) mutant and Brca1LP/+;Trp53Δ/Δ (LP/+) control MEFs for BRCA1–PALB2 interaction and HRR defects. As expected, based on sequence conservation and the predicted effect of leucine to proline substitution on the coiled-coil structure (Fig. 1A and B), Brca1 p.L1363P severely attenuated BRCA1-PALB2 binding. While PALB2 readily coimmunoprecipitated with BRCA1 in whole-cell extracts of Brca1LP/+;Trp53Δ/Δ MEFs, this was not possible for Brca1LP/LP;Trp53Δ/Δ MEFs (Fig. 2A). Although the abrogation of BRCA1-PALB2 binding did not affect the initial DNA end-resection step of HRR, as measured by recruitment of RPA to 53BP1-positive alpha tracks (Fig. 2B), it still led to HRR defects. Ionizing radiation–induced foci (IRIF) of RAD51, a marker for HRR proficiency, were significantly decreased in Brca1LP/LP;Trp53Δ/Δ MEFs (Fig. 2C; Supplementary Fig. S2). This correlated with an increased sensitivity to cisplatin and PARP1 inhibition (Fig. 2D and E; Supplementary Fig. S3A–S3D) and accumulation of DNA chromosomal abnormalities upon PARP1 inhibitor treatment (Fig. 2F). Abrogation of BRCA1-PALB2 binding may affect functions of both BRCA1 and BRCA2, including those outside of HRR such as their shared role in protecting stalled replication forks (37, 38). Nevertheless, and in line with previously published work (39, 40), DNA fiber analysis of Brca1LP/LP;Trp53Δ/Δ MEFs did not reveal defects in replication fork protection (Fig. 2G). Taken together, these data show that the abrogation of BRCA1–PALB2 interaction by Brca1 p.L1363P impairs HRR, without affecting the roles of BRCA1 in DNA end-resection and replication fork protection.

Figure 2.

Brca1 p.L1363P MEFs show impaired BRCA1–PALB2 interaction and HRR deficiency. A, Coimmunoprecipitation of BRCA1 with PALB2 in Brca1L1363P/+;Trp53Δ/Δ (LP/+) and Brca1L1363P/L1363P;Trp53Δ/Δ (LP/LP) MEFs and a Brca1-deficient KB1P mammary tumor cell line control. B, RPA alpha track analysis of DSB end-resection in LP/LP MEFs and wild-type Brca1+/+ (+/+), hypomorphic Brca1Δ11/Δ11 (Δ11/Δ11), and LP/+ control MEFs. The graph depicts the mean and SEM. P value summaries are relative to +/+. *, P = 0.0174 (one-way ANOVA with Dunn multiple comparison test). C, RAD51 foci formation following γ-irradiation–induced DNA damage in +/+, Δ11/Δ11, LP/+, and LP/LP MEFs. RAD51 foci were detected by immunofluorescence 4 hours after cultures received 10 Gy irradiation (+IR) or were left as non-irradiated treatment controls (−IR). Error bars, mean ± SD. Statistical analysis compares the mean ranks of +IR Δ11/Δ11, LP/+, and LP/LP MEFs with +IR +/+ MEFs. P value summaries are relative to γ-irradiated +/+ control MEFs. ***, P = 0.0001; ****, P < 0.0001 (Kruskal–Wallis one-way ANOVA with Dunn multiple comparison test). D and E, Dose–response curves of +/+, Δ11/Δ11, LP/+, and LP/LP MEFs to cisplatin (D) and AZD2461 PARP inhibitor (E). Graphs depict the relative viability as determined by absorbance of crystal violet at 560 nm. Data represent the mean ± SD. F, Metaphase spread analysis of genomic instability upon olaparib treatment in LP/LP MEFs and +/+, Δ11/Δ11, and LP/+ control MEFs. The graph depicts the mean and SD. P value summaries indicate the statistical comparison with +/+ MEFs. ****, P < 0.0001 (one-way ANOVA with Dunn multiple comparison test). G, DNA fiber analysis of replication fork degradation in +/+, Δ11/Δ11, LP/+, and LP/LP MEFs. Top, schematic of degradation assay labeling. MEFs were sequentially labeled with CldU and IdU, followed by hydroxyurea (HU) treatment to stall DNA replication forks. Bottom, fork degradation measured by the relative shortening of IdU tracts after hydroxyurea treatment is plotted as the ratio of IdU to CldU tract length. P value summaries represent each mean rank comparison with +/+ MEF controls. ****, P < 0.0001 (Kruskal–Wallis one-way ANOVA with Dunn multiple comparison test). ns, not significant.

Figure 2.

Brca1 p.L1363P MEFs show impaired BRCA1–PALB2 interaction and HRR deficiency. A, Coimmunoprecipitation of BRCA1 with PALB2 in Brca1L1363P/+;Trp53Δ/Δ (LP/+) and Brca1L1363P/L1363P;Trp53Δ/Δ (LP/LP) MEFs and a Brca1-deficient KB1P mammary tumor cell line control. B, RPA alpha track analysis of DSB end-resection in LP/LP MEFs and wild-type Brca1+/+ (+/+), hypomorphic Brca1Δ11/Δ11 (Δ11/Δ11), and LP/+ control MEFs. The graph depicts the mean and SEM. P value summaries are relative to +/+. *, P = 0.0174 (one-way ANOVA with Dunn multiple comparison test). C, RAD51 foci formation following γ-irradiation–induced DNA damage in +/+, Δ11/Δ11, LP/+, and LP/LP MEFs. RAD51 foci were detected by immunofluorescence 4 hours after cultures received 10 Gy irradiation (+IR) or were left as non-irradiated treatment controls (−IR). Error bars, mean ± SD. Statistical analysis compares the mean ranks of +IR Δ11/Δ11, LP/+, and LP/LP MEFs with +IR +/+ MEFs. P value summaries are relative to γ-irradiated +/+ control MEFs. ***, P = 0.0001; ****, P < 0.0001 (Kruskal–Wallis one-way ANOVA with Dunn multiple comparison test). D and E, Dose–response curves of +/+, Δ11/Δ11, LP/+, and LP/LP MEFs to cisplatin (D) and AZD2461 PARP inhibitor (E). Graphs depict the relative viability as determined by absorbance of crystal violet at 560 nm. Data represent the mean ± SD. F, Metaphase spread analysis of genomic instability upon olaparib treatment in LP/LP MEFs and +/+, Δ11/Δ11, and LP/+ control MEFs. The graph depicts the mean and SD. P value summaries indicate the statistical comparison with +/+ MEFs. ****, P < 0.0001 (one-way ANOVA with Dunn multiple comparison test). G, DNA fiber analysis of replication fork degradation in +/+, Δ11/Δ11, LP/+, and LP/LP MEFs. Top, schematic of degradation assay labeling. MEFs were sequentially labeled with CldU and IdU, followed by hydroxyurea (HU) treatment to stall DNA replication forks. Bottom, fork degradation measured by the relative shortening of IdU tracts after hydroxyurea treatment is plotted as the ratio of IdU to CldU tract length. P value summaries represent each mean rank comparison with +/+ MEF controls. ****, P < 0.0001 (Kruskal–Wallis one-way ANOVA with Dunn multiple comparison test). ns, not significant.

Close modal

Brca1 p.L1363P shows a defect in mammary tumor suppression

The embryonic lethality of Brca1LP/LP mice indicates that an intact BRCA1 coiled-coil domain is functionally important in vivo, in line with its requirement for BRCA1-mediated HRR (2, 11). To analyze whether the functional defect of Brca1 p.L1363P also compromises mammary tumor suppression, the Brca1LP allele was crossed into a K14cre-driven (FVB) mouse model for conditional inactivation of Brca1 and Trp53 in epithelial cells (17). Resulting K14cre;Brca1LP/F;Trp53F/F [KB1(L1363P)P] female mice were monitored for tumor formation and compared with K14cre;Brca1F/F;Trp53F/F (KB1P), K14cre;Brca1F/+;Trp53F/F (KB1hetP), and K14cre;Brca1LP/+;Trp53F/F [KB1(L1363P)hetP] control cohorts. KB1(L1363P)P and KB1P mice developed tumors at similar median latencies of 166.5 and 173 days, respectively (Fig. 3A; Supplementary Fig. S4A). These median latencies are significantly shorter than the 203.5 days for KB1hetP controls. Thus, in our model, Brca1 p.L1363P causes a similar acceleration of Trp53-deficient tumor formation as complete loss of Brca1. The median tumor-free latency of 228 days for KB1(L1363P)hetP mice did not differ from that of KB1hetP mice, showing that Brca1 p.L1363P does not have any dominant negative effects on tumor suppression in this mouse model. Although K14cre-mediated inactivation of Trp53 results in a range of epithelial tumors, including skin and mammary tumors, concomitant loss of Brca1 is known to skew the tumor type spectrum toward mammary tumors (17). Strikingly, the percentage of mammary tumors is even higher in KB1(L1363P)P mice than in KB1P mice, coinciding with a moderate but significantly shorter median mammary tumor-free survival (Fig. 3B and C). If and how this skew toward mammary tumor formation is caused by hypomorphic Brca1 p.L1363P activity remains to be determined. Regardless, the combined data from our tumor observation cohorts clearly show that Brca1 p.L1363P abolishes BRCA1-mediated mammary tumor suppression.

Figure 3.

Tumor development in female KB1(L1363P)P mice. A, Spontaneous tumor-free survival curves for KB1(L1363P)P mice compared with KB1P, KB1hetP, and KB1(L1363P)hetP. KB1(L1363P)P tumors formed with a median tumor-free latency (T50) of 166.5 days, with no significant difference compared with KB1P (T50 = 173 days; P = 0.5395) but developed tumors significantly faster than KB1hetP (T50 = 203.5 days; P < 0.0001) and KB1(L1363P)hetP [T50 = 228 days; P < 0.0001; log-rank (Mantel–Cox) test]. B, Distribution of KB1P, KB1(L1363P)P, KB1hetP, and KB1(L1363P)hetP tumor types. Numbers indicate individual mice. C, Mammary tumor-free survival curves for KB1(L1363P)P (T50 = 167 days) mice versus KB1P (T50 = 184 days; P = 0.0102), KB1hetP (T50 = 203.5 days; P = 0.0006), and KB1(L1363P)hetP (T50 = 228 days; P < 0.0001) mice [log-rank (Mantel–Cox) test]. D, Distribution of KB1P, KB1(L1363P)P, KB1hetP, and KB1(L1363P)hetP mammary tumor histological types. Tumors are characterized as adenocarcinomas (adeno), carcinosarcomas (c/s), and sarcomatoids (sarc). All available mammary tumors were scored; numbers indicate individual tumors. E, Level of genomic instability according to histological type in KB1(L1363P)P mammary tumors compared with KB1P, KB2P, and KP controls. The graph displays the percentage of discrete copy number aberrations identified by segmentation of CNV-seq profiles. The median level of genomic instability is indicated by the inner line, while the box represents the interquartile range, with whiskers extending to the highest and lowest values. F, Principal component analysis of mRNA expression in KB1(L1363P)P mammary tumors compared with KB1P, KB2P, and KP mammary tumors. Tumor genotype is indicated by color and tumor histological type by point shape.

Figure 3.

Tumor development in female KB1(L1363P)P mice. A, Spontaneous tumor-free survival curves for KB1(L1363P)P mice compared with KB1P, KB1hetP, and KB1(L1363P)hetP. KB1(L1363P)P tumors formed with a median tumor-free latency (T50) of 166.5 days, with no significant difference compared with KB1P (T50 = 173 days; P = 0.5395) but developed tumors significantly faster than KB1hetP (T50 = 203.5 days; P < 0.0001) and KB1(L1363P)hetP [T50 = 228 days; P < 0.0001; log-rank (Mantel–Cox) test]. B, Distribution of KB1P, KB1(L1363P)P, KB1hetP, and KB1(L1363P)hetP tumor types. Numbers indicate individual mice. C, Mammary tumor-free survival curves for KB1(L1363P)P (T50 = 167 days) mice versus KB1P (T50 = 184 days; P = 0.0102), KB1hetP (T50 = 203.5 days; P = 0.0006), and KB1(L1363P)hetP (T50 = 228 days; P < 0.0001) mice [log-rank (Mantel–Cox) test]. D, Distribution of KB1P, KB1(L1363P)P, KB1hetP, and KB1(L1363P)hetP mammary tumor histological types. Tumors are characterized as adenocarcinomas (adeno), carcinosarcomas (c/s), and sarcomatoids (sarc). All available mammary tumors were scored; numbers indicate individual tumors. E, Level of genomic instability according to histological type in KB1(L1363P)P mammary tumors compared with KB1P, KB2P, and KP controls. The graph displays the percentage of discrete copy number aberrations identified by segmentation of CNV-seq profiles. The median level of genomic instability is indicated by the inner line, while the box represents the interquartile range, with whiskers extending to the highest and lowest values. F, Principal component analysis of mRNA expression in KB1(L1363P)P mammary tumors compared with KB1P, KB2P, and KP mammary tumors. Tumor genotype is indicated by color and tumor histological type by point shape.

Close modal

KB1(L1363P)P mammary tumors show EMT-like phenotypes and limited genomic instability

KB1P mammary tumors are mainly adenocarcinomas, defined by their epithelial nature and solid growth pattern (Fig. 3D; Supplementary Fig. S4B; refs. 15–17). In contrast, KB1(L1363P)P mammary tumors are predominantly carcinosarcomas with both epithelial and mesenchymal components or wholly mesenchymal sarcomatoid tumors (Fig. 3D; Supplementary Fig. S4B–S4D). As previously observed for K14cre;Trp53F/F (KP), KB1P, and K14cre;Brca2F/F;Trp53F/F (KB2P) mammary tumors, tumors with mesenchymal phenotypes show more stable DNA copy number profiles than adenocarcinomas (Fig. 3E; ref. 41). This dampening can partially be explained by a larger contribution of stromal cells in which Brca1 and Trp53 are not switched off (Supplementary Figs. S4E-S4G and S5A). The three KB1(L1363P)P adenocarcinomas available for sequencing showed profound copy number changes, but this sample size is too small to draw robust conclusions on the effect of Brca1 p.L1363P on mammary tumor genome stability. Averaged over all histology types, KB1(L1363P)P mammary tumors do not show more copy number gains or losses than KP tumors (Fig. 3E; Supplementary Fig. S5B), which seems largely dependent on histology type. The same applies to differences in gene expression, which mainly confirm the histological classifications of adenocarcinoma, carcinosarcoma, and sarcomatoid tumors, and show that most KB1(L1363P)P mammary tumors have an EMT-like phenotype (Fig. 3F; Supplementary Fig. S5C and S5D).

KB1(L1363P)P mammary tumors respond to cisplatin and PARP inhibition

To analyze the response of KB1(L1363P)P mammary tumors to HRR deficiency–targeted therapy, we performed orthotopic transplantations with spontaneous donor tumors as described previously (15, 16, 42). To capture the heterogeneity of KB1(L1363P)P tumors, we transplanted five different donor tumors: one adenocarcinoma, three carcinosarcomas, and one sarcomatoid (Supplementary Fig. S6A and S6B). For comparison of treatment responses, we transplanted three KB1P adenocarcinomas, two KP adenocarcinomas, and one KP carcinosarcoma. Once outgrowths of transplanted tumor pieces reached a size of 200 mm3, mice were treated with cisplatin, the PARP inhibitor AZD2461, or left untreated (Fig. 4A). KB1(L1363P)P mammary tumors responded significantly better than KP tumors to both cisplatin and AZD2461 and showed an overall intermediate response to both DNA repair-targeting drugs compared with Brca1-proficient and Brca1-deficient controls (Fig. 4B4E; Supplementary Fig. S6C). While a faster growth rate may partially explain the decreased sensitivity of KB1(L1363P)P tumors to these targeted therapies (Fig. 4C and 4FH; Supplementary Fig. S6D–S6I), it seems likely that the HRR defect of tumor cells expressing hypomorphic BRCA1 p.L1363P—with intact RING and BRCT domains—is less severe than that of Brca1-null tumor cells. To establish whether this intermediate response is explained by a partial HRR defect in tumor cells expressing hypomorphic BRCA1 p.L1363P, we performed RAD51 IRIF analysis of KB1(L1363P)P mammary tumor-derived cell cultures. In contrast to KB1P, KB1(L1363P)P tumor cells were able to induce RAD51 foci in response to γ-radiation; however, RAD51 formation was clearly compromised compared with KP (Fig. 4I and J). Collectively, the results of our study suggest that BRCA1 coiled-coil variants that attenuate BRCA1–PALB2 interaction increase the risk of developing breast cancer but leave sufficient functionality to partially withstand HRR deficiency–targeted therapy.

Figure 4.

Treatment response of KB1(L1363P)P mammary tumors. A, Schematic overview of the orthotopic transplantations of spontaneous donor tumors and treatment regimens for intervention studies. Tx denotes orthotopic transplantation and T0 (day 0) marks the start of each respective treatment group once outgrown tumor volume reaches ± 200 mm3. B, Cause of death of KP (n = 13), KB1P (n = 15), and KB1(L1363P)P (n = 23) cisplatin-treated mice. Mice that were sensitive to treatment completed the four-cycle treatment regimen and did not reach endpoint tumor volume of 1,200 mm3 within 14 days of the final treatment. Resistant mice were sacrificed if tumor volume reached 1,200 mm3 within the 14-day window following treatment. Mice sacrificed due to nontumor burden-related causes were considered drug toxicity–associated events. C, Survival of untreated mice. Median survival (T50): KP T50 = 13 days, n = 13 mice; KB1P T50 = 12 days, n = 15 mice; KB1(L1363P)P T50 = 9 days, n = 21 mice. D, Survival of cisplatin-treated mice. Median survival (T50): KP T50 = 34.5 days, n = 13 mice; KB1P T50 = 111 days, n = 15 mice; KB1(L1363P)P T50 = 86 days, n = 23 mice. Mice sacrificed because of cisplatin side effects are censored. E, Survival of AZD2461-treated mice. Median survival T50: KP T50 = 14 days, n = 13 mice; KB1P T50 = 41 days, n = 14 mice; KB1(L1363P)P T50 = 18 days, n = 21 mice. One KB1(L1363P)P mouse died from an unknown cause and was censored. F–H, Comparison of treatment survival curves (C–E) per genotype for KP (F), KB1P (G), and KB1(L1363P)P (H) mice. I, Ionizing radiation-induced RAD51 foci in KP, KB1P, and KB1(L1363P)P tumor-derived cell lines. Short-term tumor cultures were derived from untreated transplanted tumors and either received (+IR) or did not receive (−IR) 10 Gy γ-irradiation. The scatter plot graphs the mean ± SD. P value summaries are relative to γ-irradiated KP 293. **, P = 0.0030; ***, P = 0.0001; ****, P < 0.0001 (Kruskal–Wallis one-way ANOVA with Dunn multiple comparison test). J, Representative immunofluorescence images of RAD51 foci in γ-irradiated KP, KB1P, and KB1(L1363P)P untreated tumor-derived cell lines. Scale bar, 10 μm.

Figure 4.

Treatment response of KB1(L1363P)P mammary tumors. A, Schematic overview of the orthotopic transplantations of spontaneous donor tumors and treatment regimens for intervention studies. Tx denotes orthotopic transplantation and T0 (day 0) marks the start of each respective treatment group once outgrown tumor volume reaches ± 200 mm3. B, Cause of death of KP (n = 13), KB1P (n = 15), and KB1(L1363P)P (n = 23) cisplatin-treated mice. Mice that were sensitive to treatment completed the four-cycle treatment regimen and did not reach endpoint tumor volume of 1,200 mm3 within 14 days of the final treatment. Resistant mice were sacrificed if tumor volume reached 1,200 mm3 within the 14-day window following treatment. Mice sacrificed due to nontumor burden-related causes were considered drug toxicity–associated events. C, Survival of untreated mice. Median survival (T50): KP T50 = 13 days, n = 13 mice; KB1P T50 = 12 days, n = 15 mice; KB1(L1363P)P T50 = 9 days, n = 21 mice. D, Survival of cisplatin-treated mice. Median survival (T50): KP T50 = 34.5 days, n = 13 mice; KB1P T50 = 111 days, n = 15 mice; KB1(L1363P)P T50 = 86 days, n = 23 mice. Mice sacrificed because of cisplatin side effects are censored. E, Survival of AZD2461-treated mice. Median survival T50: KP T50 = 14 days, n = 13 mice; KB1P T50 = 41 days, n = 14 mice; KB1(L1363P)P T50 = 18 days, n = 21 mice. One KB1(L1363P)P mouse died from an unknown cause and was censored. F–H, Comparison of treatment survival curves (C–E) per genotype for KP (F), KB1P (G), and KB1(L1363P)P (H) mice. I, Ionizing radiation-induced RAD51 foci in KP, KB1P, and KB1(L1363P)P tumor-derived cell lines. Short-term tumor cultures were derived from untreated transplanted tumors and either received (+IR) or did not receive (−IR) 10 Gy γ-irradiation. The scatter plot graphs the mean ± SD. P value summaries are relative to γ-irradiated KP 293. **, P = 0.0030; ***, P = 0.0001; ****, P < 0.0001 (Kruskal–Wallis one-way ANOVA with Dunn multiple comparison test). J, Representative immunofluorescence images of RAD51 foci in γ-irradiated KP, KB1P, and KB1(L1363P)P untreated tumor-derived cell lines. Scale bar, 10 μm.

Close modal

In this study, we present the first direct evidence that a BRCA1 missense variant outside of the RING and BRCT domains increases the risk to develop breast cancer. In line with previous observations (9, 11), we show that mouse Brca1 p.L1363P coiled-coil domain variant (equivalent to human BRCA1 p.L1407P) disrupts the interaction of BRCA1 with PALB2 and causes a defect in RAD51 recruitment to DNA double-strand breaks (DSB). Although this correlates with increased sensitivity to cisplatin and PARP inhibition, the functions of BRCA1 p.L1363P in DNA DSB end-resection and replication fork protection remain unaffected. Thus, the BRCA1 p.L1363P HRR defect differs from those caused by BRCA1 loss-of-function mutations in the RING and BRCT domains. In addition, we make a number of in vivo observations that clearly separate the defects caused by this coiled-coil domain mutation from those of previously generated Brca1-mutant mouse models.

Brca1 p.L1363P homozygous embryos show a milder embryonic phenotype than homozygous Brca1-null (17, 32), RING mutant (15, 16), or BRCT mutant (16) mice, although they still show a general growth retardation and die before day E13.5 of embryonic development. The observed embryonic lethality of homozygous Brca1 p.L1363P mice seems at odds with the viability of the reciprocal Palb2 coiled-coil domain mouse mutant. The Palb2 p.(L24_K26delinsAAA) mutation of Palb2CC6 mice also disrupts the interaction of PALB2 with BRCA1, yet homozygous Palb2CC6/CC6 mice are viable and only show a defect in male fertility that is linked to impaired meiosis (43). Palb2CC6/CC6 MEFs, however, have a growth defect and their premature senescence and mitomycin C sensitivity (43) are in line with the phenotypes we observed in Brca1LP/LP embryos and fibroblasts. It cannot be excluded that the consequences of disrupting BRCA1–PALB2 interaction are more or less severe depending on genetic background (mixed for Palb2CC6 mice vs. FVB for Brca1LP mice). Genetic background effects on the embryonic phenotypes of Brca1-mutant mice have been observed before (44) and are in fact supported by the prolonged survival of Brca1LP/LP embryos upon reduction of Trp53 gene dosage. Alternatively, inactivation of the BRCA1 coiled-coil domain may affect other interactions or processes, or PALB2 p.(L24_K26delinsAAA) may be less disruptive for BRCA1–PALB2 interaction than BRCA1 p.L1363P. Of note, a recently published C57BL/6J x C3H/HeJ mouse model carrying an in-frame deletion in the BRCA1 coiled-coil region also shows embryonic lethality, albeit not with complete penetrance (40), while Brca1 p.L1363P homozygosity in mice with a mixed C57BL/6J x 129/Sv genetic background showed growth defects at E13.5 but were nonetheless born at expected Mendelian frequencies (14).

In the context of K14cre-driven conditional Brca1;Trp53 knockout mouse breast cancer models, the Brca1LP allele has a similar effect on overall tumor latency as other Brca1 loss-of-function alleles (15–17); however, the tumor spectrum of KB1(L1363P)P mice differs from these other models by several aspects. K14cre;Trp53F/F-based models can develop skin tumors, yet KB1(L1363P)P mice show a remarkable skew toward mammary tumor formation. In addition, whereas Brca1-null, RING mutant, or BRCT-mutant mammary tumors are mainly basal-like adenocarcinomas, KB1(L1363P)P mammary tumors show EMT-like characteristics that also associate with relatively low genomic instability, as assessed by copy number aberrations. Despite these phenotypes, mammary tumors of KB1(L1363P)P mice are HRR-deficient and show an intermediate response to cisplatin and PARP inhibition compared with Brca1 wild-type KP and Brca1-null KB1P mammary tumors.

Taken together, our data suggest that BRCA1 coiled-coil mutations disabling PALB2 binding confer a similarly increased risk to develop breast cancer as known pathogenic BRCA1 RING, BRCT, and frameshift mutations. Similar to what we observed in our mouse model, human BRCA1 coiled-coil mutant breast tumors may show a difference in histology compared with tumors lacking a functional RING or BRCT domain. BRCA1-associated breast cancers are frequently of the basal-like subtype, in contrast to BRCA2- or PALB2-associated breast cancers, which show similar histology types as sporadic cases (45, 46). Because BRCA1 coiled-coil mutations affect the interaction with PALB2 but leave other BRCA1 domain functions intact, BRCA1 coiled-coil mutant tumors may well lack a bias toward the basal-like subtype. In addition, human BRCA1 coiled-coil mutant breast tumors are expected to be sensitive to HRR deficiency–targeted treatment, although response and resistance mechanisms may differ from other BRCA1-deficient tumors.

No disclosures were reported.

E.M. Pulver: Conceptualization, data curation, formal analysis, validation, investigation, visualization, methodology, writing–original draft, project administration, writing–review and editing. C. Mukherjee: Data curation, formal analysis, validation, investigation, methodology. G. van de Kamp: Data curation, formal analysis, validation, investigation, methodology. S.J. Roobol: Data curation, formal analysis, validation, investigation, methodology. M.B. Rother: Data curation, formal analysis, validation, investigation, methodology. H. van der Gulden: Data curation, investigation, writing–review and editing. R. de Bruijn: Data curation, software, formal analysis, investigation, methodology, writing–review and editing. M.V. Lattanzio: Investigation. E. van der Burg: Data curation, investigation. A.P. Drenth: Data curation, investigation. N.S. Verkaik: Investigation. K. Hahn: Funding acquisition, investigation, methodology. S. Klarenbeek: Investigation, methodology, writing–review and editing. R. de Korte–Grimmerink: Data curation, investigation. M. van de Ven: Supervision. C.E.J. Pritchard: Data curation, validation, investigation, methodology. I.J. Huijbers: Supervision. B. Xia: Resources, writing–review and editing. D.C. van Gent: Supervision, funding acquisition, validation. J. Essers: Supervision, funding acquisition, validation, writing–review and editing. H. van Attikum: Supervision, funding acquisition, validation. A. Ray Chaudhuri: Resources, supervision, funding acquisition, validation. P. Bouwman: Conceptualization, data curation, formal analysis, supervision, funding acquisition, validation, investigation, visualization, methodology, writing–original draft, project administration, writing–review and editing. J. Jonkers: Conceptualization, supervision, funding acquisition, validation, visualization, writing–original draft, project administration, writing–review and editing.

The authors thank Jinhyuk Bhin for his assistance with bioinformatics analyses. The authors thank the NKI animal laboratory facilities and pathology unit, the NKI genomics core facility, and the NKI bioimaging facility for expert technical support. This work was supported by grants from the Dutch Cancer Society (KWF; NKI 2015-7877 to P. Bouwman, J. Jonkers, and Maaike P.G. Vreeswijk; 11008/2017-1 to A. Ray Chaudhuri; and 12092/2018 to J. Essers); the Dutch Research Council (NWO; VIDI VI. 193.131 to A. Ray Chaudhuri; VICI 91814643 to J. Jonkers; and VICI VI.C.182.052 to H. van Attikum); Oncode Institute, which is partly financed by KWF, NWO research program Mosaic (grant 017.008.022 to J. Jonkers); the Lundbeck Foundation (grant R223-2016-956 to J. Jonkers, Claus Storgaard Sørensen, and Finn Cilius Nielsen); Technologie stichting STW (project number 13577 to D.C. van Gent and J. Essers); and the Swiss National Science Foundation to K. Hahn.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Nielsen
FC
,
van Overeem Hansen
T
,
Sørensen
CS
. 
Hereditary breast and ovarian cancer: new genes in confined pathways
.
Nat Rev Cancer
2016
;
16
:
599
612
.
2.
Anantha
RW
,
Simhadri
S
,
Foo
TK
,
Miao
S
,
Liu
J
,
Shen
Z
, et al
Functional and mutational landscapes of BRCA1 for homology-directed repair and therapy resistance
.
Elife
2017
;
6
:
e21350
.
3.
Bouwman
P
,
van der Gulden
H
,
van der Heijden
I
,
Drost
R
,
Klijn
CN
,
Prasetyanti
P
, et al
A high-throughput functional complementation assay for classification of BRCA1 missense variants
.
Cancer Discov
2013
;
3
:
1142
55
.
4.
Lu
C
,
Xie
M
,
Wendl
MC
,
Wang
J
,
McLellan
MD
,
Leiserson
MDM
, et al
Patterns and functional implications of rare germline variants across 12 cancer types
.
Nat Commun
2015
;
6
:
10086
.
5.
Millot
GA
,
Carvalho
MA
,
Caputo
SM
,
Vreeswijk
MPG
,
Brown
MA
,
Webb
M
, et al
A guide for functional analysis of BRCA1 variants of uncertain significance
.
Human Mutat
2012
;
33
:
1526
37
.
6.
Ransburgh
DJR
,
Chiba
N
,
Ishioka
C
,
Toland
AE
,
Parvin
JD
. 
Identification of breast tumor mutations in BRCA1 that abolish its function in homologous DNA recombination
.
Cancer Res
2010
;
70
:
988
95
.
7.
Starita
LM
,
Islam
MM
,
Banerjee
T
,
Adamovich
AI
,
Gullingsrud
J
,
Fields
S
, et al
A multiplex homology-directed DNA repair assay reveals the impact of more than 1,000 BRCA1 missense substitution variants on protein function
.
Am J Hum Genet
2018
;
103
:
498
508
.
8.
Towler
WI
,
Zhang
J
,
Ransburgh
DJR
,
Toland
AE
,
Ishioka
C
,
Chiba
N
, et al
Analysis of BRCA1 variants in double-strand break repair by homologous recombination and single-strand annealing
.
Hum Mutat
2013
;
34
:
439
45
.
9.
Antoniou
AC
,
Casadei
S
,
Heikkinen
T
,
Barrowdale
D
,
Pylkäs
K
,
Roberts
J
, et al
Breast-cancer risk in families with mutations in PALB2
.
N Engl J Med
2014
;
371
:
497
506
.
10.
Xia
B
,
Sheng
Q
,
Nakanishi
K
,
Ohashi
A
,
Wu
J
,
Christ
N
, et al
Control of BRCA2 cellular and clinical functions by a nuclear partner, PALB2
.
Mol Cell
2006
;
22
:
719
29
.
11.
Sy
SMH
,
Huen
MSY
,
Chen
J
. 
PALB2 is an integral component of the BRCA complex required for homologous recombination repair
.
Proc Natl Acad Sci U S A
2009
;
106
:
7155
60
.
12.
Zhang
F
,
Ma
J
,
Wu
J
,
Ye
L
,
Cai
H
,
Xia
B
, et al
PALB2 links BRCA1 and BRCA2 in the DNA-damage response
.
Curr Biol
2009
;
19
:
524
9
.
13.
Zhang
F
,
Fan
Q
,
Ren
K
,
Andreassen
PR
. 
PALB2 functionally connects the breast cancer susceptibility proteins BRCA1 and BRCA2
.
Mol Cancer Res
2009
;
7
:
1110
8
.
14.
Park
D
,
Bergin
SM
,
Jones
D
,
Ru
P
,
Koivisto
CS
,
Jeon
Y-J
, et al
Ablation of the Brca1-Palb2 interaction phenocopies Fanconi anemia in mice
.
Cancer Res
2020
;
80
:
4172
84
.
15.
Drost
R
,
Bouwman
P
,
Rottenberg
S
,
Boon
U
,
Schut
E
,
Klarenbeek
S
, et al
BRCA1 RING function is essential for tumor suppression but dispensable for therapy resistance
.
Cancer Cell
2011
;
20
:
797
809
.
16.
Drost
R
,
Dhillon
KK
,
van der Gulden
H
,
van der Heijden
I
,
Brandsma
I
,
Cruz
C
, et al
BRCA1185delAG tumors may acquire therapy resistance through expression of RING-less BRCA1
.
J Clin Invest
2016
;
126
:
2903
18
.
17.
Liu
X
,
Holstege
H
,
van der Gulden
H
,
Treur-Mulder
M
,
Zevenhoven
J
,
Velds
A
, et al
Somatic loss of BRCA1 and p53 in mice induces mammary tumors with features of human BRCA1-mutated basal-like breast cancer
.
Proc Natl Acad Sci U S A
2007
;
104
:
12111
6
.
18.
Yang
H
,
Wang
H
,
Jaenisch
R
. 
Generating genetically modified mice using CRISPR/Cas-mediated genome engineering
.
Nat Protoc
2014
;
9
:
1956
68
.
19.
Hsu
PD
,
Scott
DA
,
Weinstein
JA
,
Ran
FA
,
Konermann
S
,
Agarwala
V
, et al
DNA targeting specificity of RNA-guided Cas9 nucleases
.
Nat Biotechnol
2013
;
31
:
827
32
.
20.
Duarte
AA
,
Gogola
E
,
Sachs
N
,
Barazas
M
,
Annunziato
S
,
R de Ruiter
J
, et al
BRCA-deficient mouse mammary tumor organoids to study cancer-drug resistance
.
Nat Methods
2018
;
15
:
134
40
.
21.
Sotiropoulou
PA
,
Karambelas
AE
,
Debaugnies
M
,
Candi
A
,
Bouwman
P
,
Moers
V
, et al
BRCA1 deficiency in skin epidermis leads to selective loss of hair follicle stem cells and their progeny
.
Genes Dev
2013
;
27
:
39
51
.
22.
Xu
G
,
Chapman
JR
,
Brandsma
I
,
Yuan
J
,
Mistrik
M
,
Bouwman
P
, et al
REV7 counteracts DNA double-strand break resection and affects PARP inhibition
.
Nature
2015
;
521
:
541
4
.
23.
Cornacchia
D
,
Dileep
V
,
Quivy
J-P
,
Foti
R
,
Tili
F
,
Santarella-Mellwig
R
, et al
Mouse Rif1 is a key regulator of the replication-timing programme in mammalian cells: Mouse Rif1 controls replication timing
.
EMBO J
2012
;
31
:
3678
90
.
24.
Mukherjee
C
,
Tripathi
V
,
Manolika
EM
,
Heijink
AM
,
Ricci
G
,
Merzouk
S
, et al
RIF1 promotes replication fork protection and efficient restart to maintain genome stability
.
Nat Commun
2019
;
10
:
3287
.
25.
Difilippantonio
MJ
,
Petersen
S
,
Chen
HT
,
Johnson
R
,
Jasin
M
,
Kanaar
R
, et al
Evidence for replicative repair of DNA double-strand breaks leading to oncogenic translocation and gene amplification
.
J Exp Med
2002
;
196
:
469
80
.
26.
Henneman
L
,
van Miltenburg
MH
,
Michalak
EM
,
Braumuller
TM
,
Jaspers
JE
,
Drenth
AP
, et al
Selective resistance to the PARP inhibitor olaparib in a mouse model for BRCA1-deficient metaplastic breast cancer
.
Proc Natl Acad Sci U S A
2015
;
112
:
8409
14
.
27.
Gogola
E
,
Duarte
AA
,
de Ruiter
JR
,
Wiegant
WW
,
Schmid
JA
,
de Bruijn
R
, et al
Selective loss of PARG restores parylation and counteracts PARP inhibitor-mediated synthetic lethality
.
Cancer Cell
2018
;
33
:
1078
93
.
28.
Koster
J
,
Rahmann
S
. 
Snakemake–a scalable bioinformatics workflow engine
.
Bioinformatics
2012
;
28
:
2520
2
.
29.
Foroutan
M
,
Bhuva
DD
,
Lyu
R
,
Horan
K
,
Cursons
J
,
Davis
MJ
. 
Single sample scoring of molecular phenotypes
.
BMC Bioinformatics
2018
;
19
:
404
.
30.
Huang
S
,
Hölzel
M
,
Knijnenburg
T
,
Schlicker
A
,
Roepman
P
,
McDermott
U
, et al
MED12 controls the response to multiple cancer drugs through regulation of TGF-β receptor signaling
.
Cell
2012
;
151
:
937
50
.
31.
Lupas
A
,
Van Dyke
M
,
Stock
J
. 
Predicting coiled coils from protein sequences
.
Science
1991
;
252
:
1162
4
.
32.
Hakem
R
,
de la Pompa
JL
,
Sirard
C
,
Mo
R
,
Woo
M
,
Hakem
A
, et al
The tumor suppressor gene Brca1 is required for embryonic cellular proliferation in the mouse
.
Cell
1996
;
85
:
1009
23
.
33.
Ludwig
T
,
Chapman
DL
,
Papaioannou
VE
,
Efstratiadis
A
. 
Targeted mutations of breast cancer susceptibility gene homologs in mice: lethal phenotypes of Brca1, Brca2, Brca1/Brca2, Brca1/p53, and Brca2/p53 nullizygous embryos
.
Genes Dev
1997
;
11
:
1226
41
.
34.
Bouwman
P
,
Drost
R
,
Klijn
C
,
Pieterse
M
,
van der Gulden
H
,
Song
J-Y
, et al
Loss of p53 partially rescues embryonic development of Palb2 knockout mice but does not foster haploinsufficiency of Palb2 in tumour suppression
.
J Pathol
2011
;
224
:
10
21
.
35.
Hakem
R
,
de la Pompa
JL
,
Elia
A
,
Potter
J
,
Mak
TW
. 
Partial rescue of Brca1 (5–6) early embryonic lethality by p53 or p21 null mutation
.
Nat Genet
1997
;
16
:
298
302
.
36.
Jonkers
J
,
Meuwissen
R
,
van der Gulden
H
,
Peterse
H
,
van der Valk
M
,
Berns
A
. 
Synergistic tumor suppressor activity of BRCA2 and p53 in a conditional mouse model for breast cancer
.
Nat Genet
2001
;
29
:
418
25
.
37.
Schlacher
K
,
Christ
N
,
Siaud
N
,
Egashira
A
,
Wu
H
,
Jasin
M
. 
Double-strand break repair-independent role for BRCA2 in blocking stalled replication fork degradation by MRE11
.
Cell
2011
;
145
:
529
42
.
38.
Schlacher
K
,
Wu
H
,
Jasin
M
. 
A distinct replication fork protection pathway connects Fanconi anemia tumor suppressors to RAD51-BRCA1/2
.
Cancer Cell
2012
;
22
:
106
16
.
39.
Daza-Martin
M
,
Starowicz
K
,
Jamshad
M
,
Tye
S
,
Ronson
GE
,
MacKay
HL
, et al
Isomerization of BRCA1-BARD1 promotes replication fork protection
.
Nature
2019
;
571
:
521
7
.
40.
Nacson
J
,
Di Marcantonio
D
,
Wang
Y
,
Bernhardy
AJ
,
Clausen
E
,
Hua
X
, et al
BRCA1 mutational complementation induces synthetic viability
.
Mol Cell
2020
;
78
:
951
9
.e6.
41.
Holstege
H
,
van Beers
E
,
Velds
A
,
Liu
X
,
Joosse
SA
,
Klarenbeek
S
, et al
Cross-species comparison of aCGH data from mouse and human BRCA1- and BRCA2-mutated breast cancers
.
BMC Cancer
2010
;
10
:
455
.
42.
Rottenberg
S
,
Nygren
AOH
,
Pajic
M
,
van Leeuwen
FWB
,
van der Heijden
I
,
van de Wetering
K
, et al
Selective induction of chemotherapy resistance of mammary tumors in a conditional mouse model for hereditary breast cancer
.
Proc Natl Acad Sci U S A
2007
;
104
:
12117
22
.
43.
Simhadri
S
,
Peterson
S
,
Patel
DS
,
Huo
Y
,
Cai
H
,
Bowman-Colin
C
, et al
Male fertility defect associated with disrupted BRCA1-PALB2 interaction in mice
.
J Biol Chem
2014
;
289
:
24617
29
.
44.
Ludwig
T
,
Fisher
P
,
Ganesan
S
,
Efstratiadis
A
. 
Tumorigenesis in mice carrying a truncating Brca1 mutation
.
Genes Dev
2001
;
15
:
1188
93
.
45.
Honrado
E
,
Osorio
A
,
Palacios
J
,
Benitez
J
. 
Pathology and gene expression of hereditary breast tumors associated with BRCA1, BRCA2 and CHEK2 gene mutations
.
Oncogene
2006
;
25
:
5837
45
.
46.
Li
A
,
Geyer
FC
,
Blecua
P
,
Lee
JY
,
Selenica
P
,
Brown
DN
, et al
Homologous recombination DNA repair defects in PALB2-associated breast cancers
.
NPJ Breast Cancer
2019
;
5
:
23
.