Abstract

Monoallelic or biallelic RAD51C germline mutations results in chromosome instability disorders such as Fanconi anemia and cancers. The bona fide function of RAD51C is to assist RAD51 nucleoprotein filament onto single-strand DNA to complete homologous recombination (HR) repair. In addition to HR repair, the role of RAD51C in DNA replication is emerging when replication forks are transiently or irreversibly stalled. We identified novel RAD51C variants of uncertain significance (VUS) from breast, ovarian, pancreatic and gastric cancer patients and functionally characterized the effect of these variants in replication fork protection and double-strand breaks (DSB’s) repair. In RAD51C-deficient Chinese hamster CL-V4B cells, expression of RAD51C F164S, A87E, L134S and E49K variants heightened sensitivity to mitomycin C (MMC), etoposide and PARP inhibition. Differently, expression of subset of RAD51C variants R24L, R24W and R212H displayed mild sensitivity to MMC, etoposide and PARP inhibition. Further functional characterization of a subset of variants revealed that Rad51C F164S, A87E, L134S and E49K variants displayed reduced RAD51 foci formation and increased overall nuclear single strand DNA levels in the presence of replication stress. Additionally, DNA fiber assay revealed that RAD51C F164S, A87E, L134S and E49K variants displayed defective replication fork protection upon prolonged fork stalling. Investigations using patient-derived lymphoblastoid cell line carrying heterozygous RAD51C L134S variant showed an impairment in RAD51 chromatin association and replication fork protection, suggestive of deleteriousness of this VUS variant. Overall, our findings provide more insights into molecular roles of RAD51C in replication fork integrity maintenance and in DSB repair.

Introduction

RAD51C is one among the essential homologous recombination (HR) genes that includes BRCA1, BRCA2, PALB2, XRCC3, RAD51D and BRIP1 whose germline mutation carriers are at high risk for breast and ovarian cancers (1). Germline pathogenic or likely pathogenic variants in known cancer-predisposing genes have implications for heritability. Nonetheless, the impact of variants of uncertain significance (VUS) on the etiology of disease and cancer predisposition has not been fully appreciated. In recent years, RAD51C novel VUS have been encountered in the clinics (2) and some variants are functionally characterized in vitro (3–5). RAD51C is a part of two protein complexes named BCDX2 (RAD51B, RAD51C, RAD51D and XRCC2) and CX3 (RAD51C and XRCC3) whose primary role is to facilitate HR by assisting RAD51 nucleofilament formation at double-strand breaks (DSBs) (6). Rad51C gene encodes a ~40 kDa protein, consisting of 376 amino acids. Rad51C contains a Holliday junction resolution domain (1–126 amino acids), a nucleotide binding region such as walker A motif (125–132 amino acids) and walker B motif (238–242 amino acids), a region for interaction with Rad51B, Rad51D and XRCC3 (79–136 amino acids) and the C-terminal nuclear localization signal (366–370 amino acids) (7,8) (Fig. 1A). Previous functional characterization of RAD51C germline variants revealed that RAD51C G125V and L138F are highly sensitive to mitomycin C (MMC) induced inter-strand crosslinks (ICL) in both DT-40 chicken cells and in Chinese hamster cells (3–5). Similarly, RAD51C D159N, G264S, T287A, R366Q and R258H hypomorphs displayed partial sensitivity to MMC in both chicken DT-40 and Chinese hamster cells (3,4). The sensitivity to DNA-damaging agents is attributed to the ability of these variants to repair DSBs through HR. In addition to human cell lines reports, identification and characterization of Rad51 paralog complex, from Caenorhabditis elegans, revealed that RFS-1/RIP-1 is essential for HR and Rad51 foci formation in vivo (9). Furthermore, several biochemical and biophysical studies demonstrated that RFS-1/RIP-1 complex structurally remodels Rad51 filaments to an open conformation and this remodeling activity facilitates strand exchange with the template DNA (9). Mechanistically, RFS-1/RIP-1 limits the Rad51-ssDNA turnover by binding to the 5′-end of individual Rad51-ssDNA filaments and mediate remodeling in a 5′–3′ direction (10).

Identification of Rad51C germline variants. (A) Schematic representation of the identified variants at the protein level, positioned on the basis of the known functional domains in Rad51C. (B) Human Rad51C protein has been modeled using Swiss-model server (https://swissmodel.expasy.org) on the basis of the archaeal RadA as template PDB ID: 2ZUB. The residues are indicated in yellow.
Figure 1

Identification of Rad51C germline variants. (A) Schematic representation of the identified variants at the protein level, positioned on the basis of the known functional domains in Rad51C. (B) Human Rad51C protein has been modeled using Swiss-model server (https://swissmodel.expasy.org) on the basis of the archaeal RadA as template PDB ID: 2ZUB. The residues are indicated in yellow.

In addition to their classical HR-mediated DNA DSB’s repair, several studies uncovered single strand DNA gap prevention, replication fork protection and restart roles for HR and Fanconi anemia genes such as BRCA1, BRCA2, RAD51 and FANCD2 (11–15). However, the clear role of RAD51 paralogues in replication fork protection and stability is poorly understood. RAD51C, also a member of RAD51 paralog, has emerging roles in replication fork protection (5), and it promotes replication fork restart (16) in mammalian cell lines in the presence of hydroxyurea (HU)-induced fork stalling. Indeed, RAD51C G125V and L138F variants that lack functional RAD51C exhibited deficits in replication fork protection and restart (5). Hence, broader implementation of in vitro functional characterization including different chemotherapeutic drug sensitivity assay, RAD51 nucleation assay and DNA replication forks protection assay is essential for the pathogenicity assessment of the VUS variants.

In this study, we identified RAD51C VUS in patients diagnosed with breast, ovarian, pancreatic or gastric cancers and had familial history of cancers. Using multiple approaches such as drug sensitivity assay, RAD51 foci formation, replication fork protection and chromosome aberration assays, we report that a subset of RAD51C such as F164S, A87E, L134S and E49K are likely deleterious variants. The study has provided a weight of evidence that RAD51C functions in stalled replication fork protection beyond its key roles in DSB repair. Our results provide additional evidence suggesting the need for the functional characterization of missense RAD51C variants encountered in the clinic.

Results

Identification of RAD51C germline VUS

Demographic and clinical data of all 12 patients carrying RAD51C VUS mutation is summarized in Table 1. The list of the co-occurring mutations is summarized in Supplementary Table S1. Of all heterozygous RAD51C VUS carriers identified in our cohort, seven had breast cancer, three had ovarian cancer, one had pancreatic cancer and one had multiple cancers (gastric cancer, endometrial cancer, kidney cancer and squamous cell carcinoma). Among the seven breast cancer patients, two had a second breast cancer after their first diagnosis of breast cancer. The age at diagnosis of breast and ovarian cancer in the patients is between 30 and 49 years old. Overall, 9 of the 12 RAD51C carriers demonstrated family history of cancers. In summary, a total of 10 RAD51C missense variants was identified and are classified as VUS. Up to date, except that RAD51C c.635G > A (R212H) variant has been reported before yet functionally characterized (16–20), the rest RAD51C missense variants: c.70C > T (R24W), c.71G > T (R24L), c.145G > A (E49K), c.233C > T (T78I), c.253 T > G (C85G), c.260C > A (A87E), c.401 T > C (L134S), c.491 T > C (F164S) and c.589G > A (E197K) are novel. These missense variants are distributed around Walker A and Walker B motif on RAD51C genes (Fig. 1A). Rad51C 3D structure was homology modeled on the basis of archaeal RadA crystal structure and the variants discovered in this study are highlighted (Fig. 1B) The deleterious effects of RAD51C missense variants predicted by in silico tools are variable and inconsistent (Table 2); therefore, this has prompted us to functionally evaluate the significance of these germline missense variants identified in our patients.

Table 1

Clinicopathological characteristics of the RAD51C VUS variants

Patient no.RAD51C VUS variantsProtein changeGenderRaceCancer types and age of cancer diagnosisFamily history of cancers
1c.70C > TR24WFemaleChineseBreast cancer (30 years old) [invasive mucinous carcinoma]Breast cancer, kidney cancer, colorectal cancer
2c.71G > TR24LFemaleChineseGastric cancer (49 years old)
[Other poorly cohesive carcinoma],
Endometrial cancer (37)
[Endometroid],
Kidney cancer (39),
[Papillary urothelial carcinoma (TCC)],
Squamous cell carcinoma (51)
Colorectal cancer, breast cancer, lung cancer, prostate cancer, skin cancer, gastric cancer, endometrial cancer, pancreatic cancer
3c.145G > AE49KFemaleChineseOvarian cancer (37)
[Serous carcinoma]
Breast cancer, colorectal cancer, kidney cancer
4c.233C > TT78IFemaleChineseBreast cancer (42)Breast cancer
5c.233C > TT78IMaleChinesePancreatic cancer (55) [Adenocarcinoma]Nil
6c.253 T > GC85GFemaleChineseBreast cancer (39)
[Invasive ductal breast carcinoma],
Second breast cancer (59)
Nil
7c.260C > AA87EFemaleChineseBreast cancer (37)
[Invasive ductal breast carcinoma]
Nil
8c.401 T > CL134SFemaleChineseBreast cancer (49)
[Invasive ductal breast carcinoma]
Breast cancer, brain cancer
9c.491 T > CF164SFemaleChineseOvarian cancer (45)
[Serous carcinoma]
Kidney cancer
10c.589G > AE197KFemaleChineseOvarian cancer (48)
[Transitional cell carcinoma]
Breast cancer, gastric cancer
11c.635G > AR212HFemaleChineseBreast cancer (34)
[Invasive ductal breast carcinoma],
Second breast cancer (42)
Gastric cancer
12c.635G > AR212HFemaleChineseBreast cancer (44)
[Invasive lobular breast carcinoma],
Thyroid cancer
Lung cancer, breast cancer
Patient no.RAD51C VUS variantsProtein changeGenderRaceCancer types and age of cancer diagnosisFamily history of cancers
1c.70C > TR24WFemaleChineseBreast cancer (30 years old) [invasive mucinous carcinoma]Breast cancer, kidney cancer, colorectal cancer
2c.71G > TR24LFemaleChineseGastric cancer (49 years old)
[Other poorly cohesive carcinoma],
Endometrial cancer (37)
[Endometroid],
Kidney cancer (39),
[Papillary urothelial carcinoma (TCC)],
Squamous cell carcinoma (51)
Colorectal cancer, breast cancer, lung cancer, prostate cancer, skin cancer, gastric cancer, endometrial cancer, pancreatic cancer
3c.145G > AE49KFemaleChineseOvarian cancer (37)
[Serous carcinoma]
Breast cancer, colorectal cancer, kidney cancer
4c.233C > TT78IFemaleChineseBreast cancer (42)Breast cancer
5c.233C > TT78IMaleChinesePancreatic cancer (55) [Adenocarcinoma]Nil
6c.253 T > GC85GFemaleChineseBreast cancer (39)
[Invasive ductal breast carcinoma],
Second breast cancer (59)
Nil
7c.260C > AA87EFemaleChineseBreast cancer (37)
[Invasive ductal breast carcinoma]
Nil
8c.401 T > CL134SFemaleChineseBreast cancer (49)
[Invasive ductal breast carcinoma]
Breast cancer, brain cancer
9c.491 T > CF164SFemaleChineseOvarian cancer (45)
[Serous carcinoma]
Kidney cancer
10c.589G > AE197KFemaleChineseOvarian cancer (48)
[Transitional cell carcinoma]
Breast cancer, gastric cancer
11c.635G > AR212HFemaleChineseBreast cancer (34)
[Invasive ductal breast carcinoma],
Second breast cancer (42)
Gastric cancer
12c.635G > AR212HFemaleChineseBreast cancer (44)
[Invasive lobular breast carcinoma],
Thyroid cancer
Lung cancer, breast cancer

All the RAD51C variants identified in our study are listed in the table along with their clinicopathological characteristics.

Table 1

Clinicopathological characteristics of the RAD51C VUS variants

Patient no.RAD51C VUS variantsProtein changeGenderRaceCancer types and age of cancer diagnosisFamily history of cancers
1c.70C > TR24WFemaleChineseBreast cancer (30 years old) [invasive mucinous carcinoma]Breast cancer, kidney cancer, colorectal cancer
2c.71G > TR24LFemaleChineseGastric cancer (49 years old)
[Other poorly cohesive carcinoma],
Endometrial cancer (37)
[Endometroid],
Kidney cancer (39),
[Papillary urothelial carcinoma (TCC)],
Squamous cell carcinoma (51)
Colorectal cancer, breast cancer, lung cancer, prostate cancer, skin cancer, gastric cancer, endometrial cancer, pancreatic cancer
3c.145G > AE49KFemaleChineseOvarian cancer (37)
[Serous carcinoma]
Breast cancer, colorectal cancer, kidney cancer
4c.233C > TT78IFemaleChineseBreast cancer (42)Breast cancer
5c.233C > TT78IMaleChinesePancreatic cancer (55) [Adenocarcinoma]Nil
6c.253 T > GC85GFemaleChineseBreast cancer (39)
[Invasive ductal breast carcinoma],
Second breast cancer (59)
Nil
7c.260C > AA87EFemaleChineseBreast cancer (37)
[Invasive ductal breast carcinoma]
Nil
8c.401 T > CL134SFemaleChineseBreast cancer (49)
[Invasive ductal breast carcinoma]
Breast cancer, brain cancer
9c.491 T > CF164SFemaleChineseOvarian cancer (45)
[Serous carcinoma]
Kidney cancer
10c.589G > AE197KFemaleChineseOvarian cancer (48)
[Transitional cell carcinoma]
Breast cancer, gastric cancer
11c.635G > AR212HFemaleChineseBreast cancer (34)
[Invasive ductal breast carcinoma],
Second breast cancer (42)
Gastric cancer
12c.635G > AR212HFemaleChineseBreast cancer (44)
[Invasive lobular breast carcinoma],
Thyroid cancer
Lung cancer, breast cancer
Patient no.RAD51C VUS variantsProtein changeGenderRaceCancer types and age of cancer diagnosisFamily history of cancers
1c.70C > TR24WFemaleChineseBreast cancer (30 years old) [invasive mucinous carcinoma]Breast cancer, kidney cancer, colorectal cancer
2c.71G > TR24LFemaleChineseGastric cancer (49 years old)
[Other poorly cohesive carcinoma],
Endometrial cancer (37)
[Endometroid],
Kidney cancer (39),
[Papillary urothelial carcinoma (TCC)],
Squamous cell carcinoma (51)
Colorectal cancer, breast cancer, lung cancer, prostate cancer, skin cancer, gastric cancer, endometrial cancer, pancreatic cancer
3c.145G > AE49KFemaleChineseOvarian cancer (37)
[Serous carcinoma]
Breast cancer, colorectal cancer, kidney cancer
4c.233C > TT78IFemaleChineseBreast cancer (42)Breast cancer
5c.233C > TT78IMaleChinesePancreatic cancer (55) [Adenocarcinoma]Nil
6c.253 T > GC85GFemaleChineseBreast cancer (39)
[Invasive ductal breast carcinoma],
Second breast cancer (59)
Nil
7c.260C > AA87EFemaleChineseBreast cancer (37)
[Invasive ductal breast carcinoma]
Nil
8c.401 T > CL134SFemaleChineseBreast cancer (49)
[Invasive ductal breast carcinoma]
Breast cancer, brain cancer
9c.491 T > CF164SFemaleChineseOvarian cancer (45)
[Serous carcinoma]
Kidney cancer
10c.589G > AE197KFemaleChineseOvarian cancer (48)
[Transitional cell carcinoma]
Breast cancer, gastric cancer
11c.635G > AR212HFemaleChineseBreast cancer (34)
[Invasive ductal breast carcinoma],
Second breast cancer (42)
Gastric cancer
12c.635G > AR212HFemaleChineseBreast cancer (44)
[Invasive lobular breast carcinoma],
Thyroid cancer
Lung cancer, breast cancer

All the RAD51C variants identified in our study are listed in the table along with their clinicopathological characteristics.

Table 2

RAD51C missense variants and in silico predictions

No.Nucleotide changeProtein changeProtein changedbSNPClinVarREVELSIFTMutation TastergnomADReported in study
1c.70C > Tp.Arg24TrpR24Wrs878855180VCV0002417760.258DamagingDisease causingNilNil
2c.71G > Tp.Arg24LeuR24Lrs777554369VCV0004219820.242DamagingPolymorphismNilNil
3c.145G > Ap.Glu49LysE49Krs753709131VCV0004235720.216DamagingDisease causing3.98E-06Nil
4c.233C > Tp.Thr78IleT78Irs112832782VCV0002417670.063ToleratedPolymorphism6.36E-05Nil
5c.253 T > Gp.Cys85GlyC85Grs1060502595VCV0004098530.156ToleratedDisease causing3.98E-06Nil
6c.260C > Ap.Ala87GluA87Ers1567786127VCV0006312800.562DamagingDisease causingNilNil
7c.401 T > Cp.Leu134SerL134Srs1233795152VCV0005387720.703DamagingDisease causing4.05E-06Nil
8c.491 T > Cp.Phe164SerF164Srs1060502589VCV0004098450.757DamagingDisease causingNilNil
9c.589G > Ap.Glu197LysE197Krs1555597094VCV0006608340.093ToleratedDisease causingNilNil
10c.635G > Ap.Arg212HisR212Hrs200857129VCV0001876330.639DamagingDisease causing8.36E-05PMID: 29263802, 21 597 919, 25 338 684, 25 980 754, 32 566 746
No.Nucleotide changeProtein changeProtein changedbSNPClinVarREVELSIFTMutation TastergnomADReported in study
1c.70C > Tp.Arg24TrpR24Wrs878855180VCV0002417760.258DamagingDisease causingNilNil
2c.71G > Tp.Arg24LeuR24Lrs777554369VCV0004219820.242DamagingPolymorphismNilNil
3c.145G > Ap.Glu49LysE49Krs753709131VCV0004235720.216DamagingDisease causing3.98E-06Nil
4c.233C > Tp.Thr78IleT78Irs112832782VCV0002417670.063ToleratedPolymorphism6.36E-05Nil
5c.253 T > Gp.Cys85GlyC85Grs1060502595VCV0004098530.156ToleratedDisease causing3.98E-06Nil
6c.260C > Ap.Ala87GluA87Ers1567786127VCV0006312800.562DamagingDisease causingNilNil
7c.401 T > Cp.Leu134SerL134Srs1233795152VCV0005387720.703DamagingDisease causing4.05E-06Nil
8c.491 T > Cp.Phe164SerF164Srs1060502589VCV0004098450.757DamagingDisease causingNilNil
9c.589G > Ap.Glu197LysE197Krs1555597094VCV0006608340.093ToleratedDisease causingNilNil
10c.635G > Ap.Arg212HisR212Hrs200857129VCV0001876330.639DamagingDisease causing8.36E-05PMID: 29263802, 21 597 919, 25 338 684, 25 980 754, 32 566 746

For the identified Rad51C variants, in silico predictions were performed using REVEL, SIFT and Mutation Taster online tools to determine the pathogenicity of the variants.

Table 2

RAD51C missense variants and in silico predictions

No.Nucleotide changeProtein changeProtein changedbSNPClinVarREVELSIFTMutation TastergnomADReported in study
1c.70C > Tp.Arg24TrpR24Wrs878855180VCV0002417760.258DamagingDisease causingNilNil
2c.71G > Tp.Arg24LeuR24Lrs777554369VCV0004219820.242DamagingPolymorphismNilNil
3c.145G > Ap.Glu49LysE49Krs753709131VCV0004235720.216DamagingDisease causing3.98E-06Nil
4c.233C > Tp.Thr78IleT78Irs112832782VCV0002417670.063ToleratedPolymorphism6.36E-05Nil
5c.253 T > Gp.Cys85GlyC85Grs1060502595VCV0004098530.156ToleratedDisease causing3.98E-06Nil
6c.260C > Ap.Ala87GluA87Ers1567786127VCV0006312800.562DamagingDisease causingNilNil
7c.401 T > Cp.Leu134SerL134Srs1233795152VCV0005387720.703DamagingDisease causing4.05E-06Nil
8c.491 T > Cp.Phe164SerF164Srs1060502589VCV0004098450.757DamagingDisease causingNilNil
9c.589G > Ap.Glu197LysE197Krs1555597094VCV0006608340.093ToleratedDisease causingNilNil
10c.635G > Ap.Arg212HisR212Hrs200857129VCV0001876330.639DamagingDisease causing8.36E-05PMID: 29263802, 21 597 919, 25 338 684, 25 980 754, 32 566 746
No.Nucleotide changeProtein changeProtein changedbSNPClinVarREVELSIFTMutation TastergnomADReported in study
1c.70C > Tp.Arg24TrpR24Wrs878855180VCV0002417760.258DamagingDisease causingNilNil
2c.71G > Tp.Arg24LeuR24Lrs777554369VCV0004219820.242DamagingPolymorphismNilNil
3c.145G > Ap.Glu49LysE49Krs753709131VCV0004235720.216DamagingDisease causing3.98E-06Nil
4c.233C > Tp.Thr78IleT78Irs112832782VCV0002417670.063ToleratedPolymorphism6.36E-05Nil
5c.253 T > Gp.Cys85GlyC85Grs1060502595VCV0004098530.156ToleratedDisease causing3.98E-06Nil
6c.260C > Ap.Ala87GluA87Ers1567786127VCV0006312800.562DamagingDisease causingNilNil
7c.401 T > Cp.Leu134SerL134Srs1233795152VCV0005387720.703DamagingDisease causing4.05E-06Nil
8c.491 T > Cp.Phe164SerF164Srs1060502589VCV0004098450.757DamagingDisease causingNilNil
9c.589G > Ap.Glu197LysE197Krs1555597094VCV0006608340.093ToleratedDisease causingNilNil
10c.635G > Ap.Arg212HisR212Hrs200857129VCV0001876330.639DamagingDisease causing8.36E-05PMID: 29263802, 21 597 919, 25 338 684, 25 980 754, 32 566 746

For the identified Rad51C variants, in silico predictions were performed using REVEL, SIFT and Mutation Taster online tools to determine the pathogenicity of the variants.

RAD51C F164S, A87E, L134S and E49K variants are sensitive to MMC, etoposide and PARP inhibition

To evaluate the functional impacts of the RAD51C missense VUS variants, we took advantage of the established RAD51C-deficient (CL-V4B) Chinese hamster cell line model system together with their RAD51C proficient parental cell lines (V79B) (17), previously used to demonstrate the role of RAD51C in DSB repair (4,18–20). CL-V4B cells displayed remarkable sensitivity to MMC. For complementation assay, we stably overexpressed flag-tagged RAD51C wild-type, empty vector (EV) and various RAD51C mutants (R24L, R24W, E49K, T78I, C85G, A87E, L134S, F164S, E197K, R212H) in CL-V4B cells. The protein expression of various mutants in CL-V4B cells are shown in Figure 2A. In agreement to previous reports (4,5), our experiment showed that CL-V4B cells stably expressed EV are dramatically sensitive to MMC and etoposide compared with the parental V79B cells. The introduction of RAD51C wild-type plasmid was able to restore survival upon MMC and etoposide to a level comparable to the V79B cells (Fig. 2B and C). Compared with wildtype RAD51C (WT), CL-V4B cells expressing the RAD51C F164S, A87E, L134S and E49K variants were significantly more sensitive to MMC-induced ICLs and etoposide-induced DSBs (Fig. 2B and C). The rest of the RAD51C mutants exhibited very mild to no sensitivity to MMC and etoposide (Fig. 2B and C). Because error-free HR repair is required to resolve ICL-induced DNA breaks (21), we questioned whether poor RAD51 localization and foci formation to the sites of breaks might contribute to this heightened sensitivity. Consistently, RAD51 foci formation was severely impaired in F164S and A87E mutants (Fig. 2D). However, RAD51 foci formation was moderately but significantly affected in L134S and E49K mutants. (Fig. 2D). We next investigated whether RAD51C mutants that display reduced RAD51 foci formation can confer lethality to PARP inhibition because of poor HR function, as reported in BRCA1/2-deficient cells (22). As speculated, RAD51C F164S, A87E, E49K and L134S variants were significantly sensitivity to PARP inhibition in a decreased order of magnitude (Fig. 2E). Overall, these results suggest that these subset of RAD51C missense variants phenocopy Rad51C-deficient scenario evidenced by reduced RAD51 foci formation and displaying increased sensitivity different DSB’s-inducing agents such as to MMC, etoposide and PARP inhibition.

RAD51C F164S, A87E, L134S and E49K variants are sensitive to MMC, etoposide and PARP inhibition. (A) Overexpression of Flag-RAD51C mutants in CL-V4B cells (Rad51C−/−). WT indicates Flag-tagged wildtype RAD51C. (B) MMC was treated for 5 days and percentage cellular viability was calculated for all the variants identified. (C) The variants expressing cells were treated with etoposide and recovered in fresh media for 4 days. Then, the percentage cell viability was calculated. (D) MMC-treated RAD51C variants and WT overexpressing cells were subjected RAD51 staining and cells with >5 foci were counted using confocal microscopy. (E) In total, 1 μM talozoparib was treated for 5 days and percentage cellular viability was calculated. Note: For B, C, D and E, unpaired t-test was performed in comparison with WT-RAD51C expressing cells. Each experiment was performed in triplicates.
Figure 2

RAD51C F164S, A87E, L134S and E49K variants are sensitive to MMC, etoposide and PARP inhibition. (A) Overexpression of Flag-RAD51C mutants in CL-V4B cells (Rad51C−/−). WT indicates Flag-tagged wildtype RAD51C. (B) MMC was treated for 5 days and percentage cellular viability was calculated for all the variants identified. (C) The variants expressing cells were treated with etoposide and recovered in fresh media for 4 days. Then, the percentage cell viability was calculated. (D) MMC-treated RAD51C variants and WT overexpressing cells were subjected RAD51 staining and cells with >5 foci were counted using confocal microscopy. (E) In total, 1 μM talozoparib was treated for 5 days and percentage cellular viability was calculated. Note: For B, C, D and E, unpaired t-test was performed in comparison with WT-RAD51C expressing cells. Each experiment was performed in triplicates.

A subset of RAD51C variants exhibit replication fork deprotection and chromosome instability in the presence of replication stress

Because F164S, A87E, L134S and E49K exhibited sensitivity to chemotherapeutics that induce DSBs accompanied with reduced RAD51 foci formation, we next focused our further functional analyses on these four potentially damaging variants. We first tested the effect of these variants in unperturbed DNA replication. Single molecule DNA fiber assay revealed that the expression of F164S, A87E, L134S and E49K variants did not affect replication fork speed in unperturbed S phase (Supplementary Fig. S1A). To address the effect of RAD51C variants in response to replication stress, we treated F164S, A87E, L134S and E49K expressing cells with HU that induces replication fork stalling, collapse and DSB’s induction upon prolonged treatment. Consistent to previous observations, Rad51C-deficient CL-V4B cells displayed dramatic sensitivity to HU treatment and expression of WT Rad51C rescued sensitivity in these cells (4,5). Interestingly, F164S and A87E displayed heightened sensitivity, whereas L134S and E49K showed moderate sensitivity to HU treatment (Fig. 3A). Replication fork stalling induced by HU remodels replication forks into a four-way junction called reversed forks whose controlled resection is required for prompt fork restart and stability (23). Hence, to this end, we next adopted DNA fiber nascent degradation assay as shown in Figure 3B to monitor the replication fork stability in Rad51C-deficient cells. In agreement with previous reports, Rad51C-deficient CL-V4B displayed reduced nascent DNA stability/protection in the presence of HU as measured by the degradation of nascent DNA tracks labeled with nucleotide analogs, IdU and CIdU (5) (Fig. 3B and C). Furthermore, expression of wild-type RAD51C or optional addition of MRE11 exonuclease inhibitor mirin both rescued nascent DNA degradation in CL-V4B cells (4) (Fig. 3C). This result further settles the role of RAD51C in protecting nascent DNA against MRE11 nuclease upon HU-induced replication fork remodeling in CL-V4B cell lines. Therefore, we next tested the replication fork stability of the four probably damaging RAD51C variants such as F164S, A87E, L134S and E49K in the presence of HU. Interestingly, Rad51C F164S, A87E, L134S and E49K variants expressing cells displayed compromised nascent DNA protection mirroring Rad51C-deficient state in CL-V4B cells (Fig. 3E). Unscheduled resection of the nascent DNA in the presence of replication stress leads to single-strand gaps accumulation, replication fork collapse and DSBs generation (12). Along these lines, we next monitored the overall nuclear ssDNA levels by incorporating BrdU into cells followed by HU treatment. RAD51C F164S, A87E, L134S and E49K mutants displayed increased overall nuclear ssDNA levels in the presence of replication stress induced by HU and these nuclear ssDNA accumulations were readily rescued by expression of wild-type RAD51C (Fig. 3E). RAD51 nucleoprotein filament formation is essential to prevent resection of nascent DNA by MRE11-mediated resection (13,24). Therefore, the ssDNA regions observed in the presence of HU in RAD51C mutants might be because of reduced RAD51 nucleoprotein filament formation, and this, in part, could have contributed to poor nascent DNA protection. In different mammalian cell lines, Rad51C loss has been shown to increase spontaneous sister chromatid gaps and chromosome breaks in the mitotic population (4,5). Hence, we next tested whether the unscheduled nascent DNA degradation observed in Rad51C variants in the presence of HU (Fig.3C and D), translates into chromosome abnormalities in metaphase spreads. Strikingly, in contrast to cells expressing WT Rad51C, expression of F164S, A87E, L134S and E49K Rad51C variants significantly increased chromosome breaks in the metaphase spreads (Fig.3F). Taken together, these data indicate that the identified Rad51C variants give rise to detectable impairment in replication fork stability, this further support the deleterious effects of these RAD51C missense variants.

A subset of RAD51C variants exhibit replication fork deprotection and chromosome instability in the presence of replication stress (A) Rad51C WT, EV, F164S, A87E, L134S and E49K variants expressing cells were treated with HU and the percentage cell viability was calculated. (B) Cartoon of the replication fork stability assay experimental setup and fiber interpretation is shown in the figure. Representative fiber image displaying fork degradation in CL-V4B-EV (Rad51C-deficient) cell lines is shown with respect to CL-V4B-WT (Rad51C-proficient). (C) CL-V4B-EV (Rad51C-deficient) is optionally treated with mirin and CL-V4B-WT (RAD51C-proficient) cells were labeled sequentially with nucleotide analog IdU and CldU for 30 min each followed by incubation with 4 mM HU for 5 h. At least 75 fibers were counted for each condition and Mann–Whitney test were performed. (D) RAD51C WT, EV, F164S, A87E, L134S and E49K variants expressing hamster cell lines were subjected to replication fork stability assay. At least 75 fibers were counted for each condition and Mann–Whitney test were performed. All the experiments were performed in triplicates. (E) RAD51C mutants expressing cells were treated with BrdU for 16 h followed by treatment with 2 mM HU for 4 h and stained for native BrdU signal omitting denaturing step and this represents the overall nuclear ssDNA intensity. At least 200 cells were counted for each condition and Mann–Whitney test were performed. The overall nuclear ssDNA signals were scored by measuring intensity of BrdU signal. (F) Chromosome breaks were detected by metaphase spread analysis. RAD51C WT, EV, F164S, A87E, L134S and E49K variants expressing cells were treated with 4 mM HU for 5 h. Left: Experimental plan and a representative metaphase spread with chromosome breaks indicated (Scalebar, 5 μm). Right: The graph bar depicts chromosome breaks per metaphase spreads. Unpaired t-test was performed in comparison with WT-RAD51C expressing cells. Each experiment was performed in triplicates. For each condition at least 50 metaphase spreads were analyzed.
Figure 3

A subset of RAD51C variants exhibit replication fork deprotection and chromosome instability in the presence of replication stress (A) Rad51C WT, EV, F164S, A87E, L134S and E49K variants expressing cells were treated with HU and the percentage cell viability was calculated. (B) Cartoon of the replication fork stability assay experimental setup and fiber interpretation is shown in the figure. Representative fiber image displaying fork degradation in CL-V4B-EV (Rad51C-deficient) cell lines is shown with respect to CL-V4B-WT (Rad51C-proficient). (C) CL-V4B-EV (Rad51C-deficient) is optionally treated with mirin and CL-V4B-WT (RAD51C-proficient) cells were labeled sequentially with nucleotide analog IdU and CldU for 30 min each followed by incubation with 4 mM HU for 5 h. At least 75 fibers were counted for each condition and Mann–Whitney test were performed. (D) RAD51C WT, EV, F164S, A87E, L134S and E49K variants expressing hamster cell lines were subjected to replication fork stability assay. At least 75 fibers were counted for each condition and Mann–Whitney test were performed. All the experiments were performed in triplicates. (E) RAD51C mutants expressing cells were treated with BrdU for 16 h followed by treatment with 2 mM HU for 4 h and stained for native BrdU signal omitting denaturing step and this represents the overall nuclear ssDNA intensity. At least 200 cells were counted for each condition and Mann–Whitney test were performed. The overall nuclear ssDNA signals were scored by measuring intensity of BrdU signal. (F) Chromosome breaks were detected by metaphase spread analysis. RAD51C WT, EV, F164S, A87E, L134S and E49K variants expressing cells were treated with 4 mM HU for 5 h. Left: Experimental plan and a representative metaphase spread with chromosome breaks indicated (Scalebar, 5 μm). Right: The graph bar depicts chromosome breaks per metaphase spreads. Unpaired t-test was performed in comparison with WT-RAD51C expressing cells. Each experiment was performed in triplicates. For each condition at least 50 metaphase spreads were analyzed.

Patient-derived cell line harboring RAD51C c.401 T > C (L134S) displayed compromised genome maintenance function

To further confirm the observed phenotype in patient samples, we next attempted to test the effect of these variants in genome stability maintenance. Because of limited biospecimen from patients, we were able to obtain only one EBV-immortalized B lymphoblastoid cell line (LCL) derived from a breast cancer patient harboring heterozygous RAD51C c.401 T > C (L134S) mutation. The patient presented with invasive ductal breast carcinoma at the age of 49 and has family history of breast and brain cancers (Fig. 3A). Because the RAD51C L134S variant expression in CL-V4B cells displayed moderate sensitivity to MMC, etoposide, PARP inhibition and exhibited poor fork protection function, we next questioned whether heterozygous RAD51C L134S LCL display these features of genome instability. At the basal level, the patient LCL showed mild reduction in endogenous RAD51C protein levels compared with healthy controls (Fig. 3B), whereas the total Rad51 levels and replication fork speed remained unaltered (Fig. 3C; Supplementary Fig. S1B). To access the HR function in LCLs, we induced DSB’s with etoposide and investigated the RAD51 chromatin association. Interestingly, treatment with etoposide increased the amount of chromatin bound phospho-RPA in both RAD51C L134S patient and healthy LCLs but only the patient cells showed impaired RAD51 chromatin association, suggesting the HR function is compromised (Fig. 3D). Like the observations from CL-V4B expressing Rad51C L134S variant (Fig.2C and3A), Rad51C L134S LCLs displayed mild sensitivity to etoposide and HU (Fig. 4D). Interestingly, replication fork stability assay revealed that RAD51C L134S carrier cells displayed reduced fork protection in comparison to healthy control (Fig.4F) that is consistent to results obtained from hamster cells from Figure 3D. Similarly, Rad51C L134S carrier treated with etoposide or HU displayed increased chromosome breaks in the metaphase spreads (Fig.4G), suggesting that these chromosome anomalies might be because of compromised HR and replication fork protection.

Patient-derived cell line harboring RAD51C c.401 T > C (L134S) displayed compromised genome maintenance function. (A) Pedigree of the affected family showed the proband presented with invasive ductal breast carcinoma at age of 49 and has family history of breast and brain cancers. (B) RAD51C total protein comparison between patient and two healthy controls level. (C) Rad51 total protein comparison between healthy and patient-derived LCLs. (D) Chromatin fractionation was performed after treating LCLs with 10 μM etoposide for 16 h and for RAD51, pRPA and MCM7. (E) Healthy and patient-derived Rad51C L134S LCLs were treated with 10 nM etoposide or 0.5 mM HU for 72 h and the percentage cellular viability was calculated. (F) Replication fork stability assay was performed as described previously in Figure 3B, at least 100 fibers were counted for each condition. The ratio of CIdU and IdU lengths were plotted and Mann–Whitney tests were performed. (G) Healthy and Rad51C L134S patient-derived cell lines were treated with 10 nM etoposide or 0.2 mM HU for 6 h and metaphase spreads were performed for chromosome breaks analyses. At least 50 metaphase spreads were analyzed for each condition.
Figure 4

Patient-derived cell line harboring RAD51C c.401 T > C (L134S) displayed compromised genome maintenance function. (A) Pedigree of the affected family showed the proband presented with invasive ductal breast carcinoma at age of 49 and has family history of breast and brain cancers. (B) RAD51C total protein comparison between patient and two healthy controls level. (C) Rad51 total protein comparison between healthy and patient-derived LCLs. (D) Chromatin fractionation was performed after treating LCLs with 10 μM etoposide for 16 h and for RAD51, pRPA and MCM7. (E) Healthy and patient-derived Rad51C L134S LCLs were treated with 10 nM etoposide or 0.5 mM HU for 72 h and the percentage cellular viability was calculated. (F) Replication fork stability assay was performed as described previously in Figure 3B, at least 100 fibers were counted for each condition. The ratio of CIdU and IdU lengths were plotted and Mann–Whitney tests were performed. (G) Healthy and Rad51C L134S patient-derived cell lines were treated with 10 nM etoposide or 0.2 mM HU for 6 h and metaphase spreads were performed for chromosome breaks analyses. At least 50 metaphase spreads were analyzed for each condition.

Discussion

Monoallelic germline alteration in RAD51C confer susceptibility to breast and ovarian cancer was first reported by Meindl et al. (3) and the predominance of RAD51C missense variants in breast and ovarian cancer is further highlighted by Osorio et al. (1), together suggested that RAD51C missense variants that are usually clinically classified as VUS should be further assessed for pathogenicity.

Here, our study demonstrates roles for novel Rad51C variants in the control of DNA replication fork integrity. In our cohort, altogether, we identified ten RAD51C germline VUS variants from 12 patients that have not yet been functionally characterized. Importantly, our functional characterization identified four missense Rad51C variants, E49K, A87E, L134S and F164S that displayed features of genome instability. E49K is located at domain required for Holliday junction resolution (25); A87E is located at domain required for its interaction with other paralogs such as RAD51B, RAD51D and XRCC3 (25,26). Interestingly, L134S is located at a 4-amino acid distance from the walker–ATP motif. L138F in walker–ATP motif has been reported to compromise replication fork protection and restart activity (5). Finally, F164S variant is located at the center of the protein between walker A and B motifs.

Remarkably, A87E and F164S variants displayed strong defect in Rad51 foci formation (Fig. 2D), with heightened sensitivity to MMC, etoposide and PARP inhibition (Fig. 2B, C and E). Differently, Rad51C E49K and L134S displayed moderate defect in Rad51 foci formation (Fig. 2D), with modest sensitivity to MMC and etoposide (Fig. 2B and C). These results suggest that the heightened cellular sensitivity to chemotherapeutics is because of severe defect in HR in Rad51C A87E and F164S variants in presence of DSB’s-inducing agents.

Replication fork stalling in the presence of HU induces fork remodeling by SMARCAL1, ZRANB3, HLTF and Rad51 (12,27). The remodeled reversed forks are protected by BRCA2 that facilitate RAD51 nucleoprotein filament formation to counteract nucleases-mediated degradation of nascent DNA (11,13). In the process of fork remodeling and subsequent fork protection, lower amount of Rad51 is enough to promote replication fork reversal (23,28), whereas, higher levels of Rad51 is required for replication fork protection (29,30). This suggests that Rad51 levels at the stalled replication forks determine the outcome of the stalled DNA replication fork metabolism. In addition to BRCA2, Rad51 levels at the replication forks are regulated by Rad51 paralogs when challenged with replication stress-inducing agents such as CPT and HU (5,16). Intriguingly, it has been shown that loss of Rad51 paralogs such as Rad51C, XRCC2 and XRCC3 trigger MRE11-mediated nascent strand degradation in hamster cell lines (5). Furthermore, Rad51C K131A variant residing within the walker A motif and L138F variant identified in breast/ovarian cancers has also been shown to increase nascent DNA degradation (5).

Along these lines, our data identify novel Rad51C variants E49K, A87E, L134S and F164S, whose expression results in replication fork deprotection (Fig. 3D), as a result, there is an increase in the overall ssDNA accumulation upon HU-induced fork stalling (Fig. 3E). Although all the four variants displayed similar levels of replication fork deprotection, there were differences in the extent of cellular sensitivity and chromosome breaks between the variants under HU challenged conditions (Fig. 3A and F). A87E and F164S were extremely sensitive to HU mirroring Rad51C null background with increased chromosome breaks, whereas E49K and L134S displayed moderate sensitivity to HU with moderate chromosome breaks in the metaphase spreads (Fig. 3A and F). In A87E and F164S variant expressing cells, the unscheduled nascent DNA degradation could have been converted into DSB’s. Because of severe HR defects (Fig. 3D), the A87E and F164S expressing mutants could have triggered apoptosis with increased chromosome breaks (Fig. 3F) in the presence of HU.

In mammalian cells, it has been shown that the HR function of BRCA1/2 is essential for embryonic cell viability (31). But more recent studies suggest that in the absence of HR, preserving fork protection is sufficient to sustain cell viability in BRCA2-deficient mouse embryonic stem cells and tumor cells (32,33). In addition, another study showed that, preventing nascent strand degradation by suppressing fork reversal mechanism improved cellular fitness in BRCA1/2-deficient cancer cells (34). Further, different separation of function mutant studies in non-transformed human mammary cell lines revealed that HR-directed repair but not fork protection is critical for cellular survival (35). In this context, our data suggest that the variants that displayed strong HR defect showed heightened sensitivity for HU (e.g. A87E and F164S variants) in comparison with other two variants (e.g. E49K and L134S). Further, our data from patient-derived cell lines suggest that heterozygous missense Rad51C L134S mutation was sufficient to trigger nascent DNA degradation (Fig. 4E) but displayed only mild sensitivity to HU with slight increase in chromosome breaks. Taken together, our study hints that HR mechanism contributes to cellular fitness, whereas fork protection appears to contribute minimally for cell survival when challenged with HU in these cell lines. Future studies are required to understand the role of Rad51C pathogenic variants in replication fork protection, replication fork restart/HR and their relationship with cancer cell survival. Further biochemical reconstitution studies for these variants are essential to dissect their interaction ability with other RAD51 paralogues such as RAD51B, XRCC2, XRCC3 and RAD51D. Hence in vitro reconstitution of these variants with the DNA substrates will be highly informative in understanding the sequential role of Rad51 paralogs in replication fork remodeling and protection. The roles of RAD51C in cancer predisposition are not well understood, although several studies correlate RAD51C mutation with cancer susceptibility (36). Because PARP inhibitors are highly efficient in BRCA1/2-deficient cancers, narrowing down RAD51C variants that display HR and fork protection defect can help stratify patients beneficial for PARP inhibitors treatment. Therefore, evidence-based classification of RAD51C VUS variants is essentially needed for accurate molecular diagnosis and gene-informed genetic counseling.

Materials and Methods

Patient data and specimens

Patients consulted at the Cancer Genetics Service of National Cancer Centre Singapore with RAD51C germline variants identified through multi-gene panel testing were enrolled to the study with written informed consent. The genes included in the panel are listed in Supplementary Table S1 in detail. Testing was performed by commercial laboratories accredited by College of American Pathologists/Clinical Laboratory Improvement Amendments (CAP/CLIA). Patient demographics, personal medical information and family history were retrieved from electronic medical records. This study is approved by SingHealth Centralized Institution Review Board (CIRB 2011/826/B).

Cell cultures

Chinese hamster lung fibroblasts V79B (RAD51C-wild-type) and CL-V4B (RAD51C-deficient) (19,37) were cultured in Dulbecco's Modified Eagle Medium supplemented with 10% fetal bovine serum (FBS) (Life Technologies, Carlsbad, CA), 2 mM L-glutamine (Life Technologies) and 100 U/ml penicillin/streptomycin (Life Technologies). EBV-immortalized B LCLs were derived from a breast cancer patient harboring RAD51C c.401 T > C (L134S) and from healthy donors. LCLs were maintained in RPMI1640 supplemented with 10% FBS. All cells were maintained at 37°C in a 5% CO2–95% air atmosphere.

Plasmid constructs and establishment of stable cell lines

The cDNA encoding full length human RAD51C was PCR-amplified and cloned in pcDNA3.1(+) expression vector using Gibson assembly method. RAD51C mutant plasmids were generated by introducing the mutation using site-directed mutagenesis. EV, wild-type and mutant plasmids were transfected into V79B or CL-V4B cells using Lipofectamine 3000(Life Technologies). Stable cell lines were created by selection with 800-1000ug/ml G418(Life Technologies) for 2 weeks.

Western blot

The expression of RAD51C protein was confirmed by western blot. Cells were lysed in radioimmunoprecipitation assay lysis buffer (Thermo Fisher Scientific, Waltham, MA) supplemented with Halt Protease and Phosphatase Inhibitor cocktail (Thermo Fisher Scientific) and lysates were clarified by centrifugation at 16000 g for 20 min at 4°C. In total, 20 μg of protein was separated by SDS gel and transferred to polyvinylidene fluoride membranes. Membranes were incubated in 5% non-fat milk in Tris-buffered saline consisting of 0.1% Tween20 for 1 h at room temperature. The membrane was incubated with primary antibody diluted in blocking buffer overnight at 4°C. After three washings in Tris-buffered saline consisting of 0.1% Tween20, the membrane was incubated with horseradish peroxidase secondary antibody (Dako, Santa Clara, CA) for 1 h at room temperature. The bands were visualized with a Western Lightning chemiluminescent kit (PerkinElmer) and ChemiDoc Imaging system (Bio-Rad Laboratories, Inc., Hercules, CA). Pan-actin served as internal loading control.

Cell viability assay

The parental V79B and CL-V4B stably expressed RAD51C mutants were treated with ICLs agent MMC, etoposide, PARP inhibitor and HU to assess cellular viability. Cells seeded onto 96-well plate at a density of5000 cells per well were treated with MMC (100 nM, 5 days), etoposide (10 nM for 6 h and recovered with fresh media for 4 days), talazoparib (1 μM, 5 Days) and HU (0.4 mM for 12 h and recovered with fresh media for 4 days). Cell viability was determined via the cell counting kit CCK-8 assay (Dojindo Laboratories, Kumamoto, Japan) at an optical density of 450 nm on the Victor spectrophotometer (PerkinElmer Life Sciences, Waltham, MA).

RAD51 foci formation and metaphase spreads

Cells were treated with 10 ng MMC/ml for 6 h and then observed for RAD51 foci formation by immunofluorescence staining. Cells containing five or more foci were considered positive, with 200 cells scored for each condition over triplicate experiments. For the chromosome breaks assay, CL-V4B cells (Rad51C−/−) with the expression of WT and different Rad51C variants were treated with 4 mM HU for 5 h. The compound was washed of 3 times with 1× PBS and incubated with the fresh media for 12 h. Later the cells were incubated with colcemid (1 μg/ml) for 4 h and the metaphase spreads were performed as described previously (4,5).

Replication fork stability assay

We adopted single molecule DNA fiber assay to measure the replication fork stability upon HU-induced nucleotide depletion. We sequentially labelled with 50 μM IdU and 250 μM CIdU nucleotide analogs for 30 min each and treated with 4 mM HU for 5 h to stall replication forks optionally supplemented with 50 μM mirin. The labelled cells were harvested with ice–cold 1× phosphate-buffered saline (PBS) and mixed gently. In total, 4 μl of cellular suspension were mixed with 6 μl of lysis buffer (200 mM Tris pH 7.5, 50 mM EDTA and 0.5% SDS). This mixture was layered on the super frost slides, after 3-min incubation the slides were slowly tilted at 45 angle to allow cell suspension to flow to the end of the slide. Later the slides were dried and fixed with 3:1 methanol: acetic acid for overnight at 4°C. Dried slides were rehydrated and denatured with 2.5 M HCl for 60 min. The denatured slides were washed thrice with 1× PBS and incubated with blocking buffer (1% bovine serum albumin, 1× PBS and 0.1% Triton-X). IdU and CIdU were stained using rat-BrdU antibody (Abcam #ab6326) and mouse-BrdU antibody (Becton Dickinson, 347580) with their respective 488 and 594 conjugated fluorophores. At least 75 fibers were counted for each condition over triplicate experiments.

Acknowledgements

We are grateful to Professor. Ganesh Nagaraju, Indian Institute of Science for kindly providing us the V79B, Rad51C proficient and CL-V4B Rad51C-deficient Chinese Hamster Cell Lines. We thank Mr Gopishankar Thirumoorthy, CCBT for technical support.

Conflict of Interest statement. All authors have no conflict of interest to declare.

Funding

Singapore Ministry of Education (MOE) under its Academic Research Fund (AcRF) Tier 1 [2019-T1-001-018]; National Cancer Centre Research Fund Terry Fox Grant [NCCRF-YR2018-NOV-1]; National Medical Research Council (NMRC) Clinician Scientist Award [MOH-000654].

Data Availability Statement

All the data related to this article will be made available by Joanne Ngeow ([email protected]) and Arun Mouli Kolinjivadi ([email protected]) upon reasonable request.

References

1

Osorio
,
A.
,
Endt
,
D.
,
Fernandez
,
F.
,
Eirich
,
K.
,
de la
 
Hoya
,
M.
,
Schmutzler
,
R.
,
Caldes
,
T.
,
Meindl
,
A.
,
Schindler
,
D.
and
Benitez
,
J.
(
2012
)
Predominance of pathogenic missense variants in the RAD51C gene occurring in breast and ovarian cancer families
.
Hum. Mol. Genet.
,
21
,
2889
2898
.

2

Federici
,
G.
and
Soddu
,
S.
(
2020
)
Variants of uncertain significance in the era of high-throughput genome sequencing: a lesson from breast and ovary cancers
.
J. Exp. Clin. Cancer Res.
,
39
,
46
.

3

Meindl
,
A.
,
Hellebrand
,
H.
,
Wiek
,
C.
,
Erven
,
V.
,
Wappenschmidt
,
B.
,
Niederacher
,
D.
,
Freund
,
M.
,
Lichtner
,
P.
,
Hartmann
,
L.
,
Schaal
,
H.
 et al. (
2010
)
Germline mutations in breast and ovarian cancer pedigrees establish RAD51C as a human cancer susceptibility gene
.
Nat. Genet.
,
42
,
410
414
.

4

Somyajit
,
K.
,
Subramanya
,
S.
and
Nagaraju
,
G.
(
2012
)
Distinct roles of FANCO/RAD51C protein in DNA damage signaling and repair: implications for Fanconi anemia and breast cancer susceptibility
.
J. Biolumin. Chemilumin.
,
287
,
3366
3380
.

5

Somyajit
,
K.
,
Saxena
,
S.
,
Babu
,
S.
,
Mishra
,
A.
and
Nagaraju
,
G.
(
2015
)
Mammalian RAD51 paralogs protect nascent DNA at stalled forks and mediate replication restart
.
Nucleic Acids Res.
,
43
,
9835
9855
.

6

Masson
,
J.Y.
,
Tarsounas
,
M.C.
,
Stasiak
,
A.Z.
,
Stasiak
,
A.
,
Shah
,
R.
,
McIlwraith
,
M.J.
,
Benson
,
F.E.
and
West
,
S.C.
(
2001
)
Identification and purification of two distinct complexes containing the five RAD51 paralogs
.
Genes Dev.
,
15
,
3296
3307
.

7

The UniProt
,
C.
(
2017
)
UniProt: the universal protein knowledgebase
.
Nucleic Acids Res.
,
45
,
D158
D169
.

8

Miller
,
K.A.
,
Sawicka
,
D.
,
Barsky
,
D.
and
Albala
,
J.S.
(
2004
)
Domain mapping of the Rad51 paralog protein complexes
.
Nucleic Acids Res.
,
32
,
169
178
.

9

Taylor
,
M.R.G.
,
Spirek
,
M.
,
Chaurasiya
,
K.R.
,
Ward
,
J.D.
,
Carzaniga
,
R.
,
Yu
,
X.
,
Egelman
,
E.H.
,
Collinson
,
L.M.
,
Rueda
,
D.
,
Krejci
,
L.
 et al. (
2015
)
Rad51 paralogs remodel pre-synaptic Rad51 filaments to stimulate homologous recombination
.
Cell
,
162
,
271
286
.

10

Taylor
,
M.R.G.
,
Spirek
,
M.
,
Jian Ma
,
C.
,
Carzaniga
,
R.
,
Takaki
,
T.
,
Collinson
,
L.M.
,
Greene
,
E.C.
,
Krejci
,
L.
and
Boulton
,
S.J.
(
2016
)
A polar and nucleotide-dependent mechanism of action for RAD51 paralogs in RAD51 filament remodeling
.
Mol. Cell
,
64
,
926
939
.

11

Kolinjivadi
,
A.M.
,
Crismani
,
W.
and
Ngeow
,
J.
(
2020
)
Emerging functions of Fanconi anemia genes in replication fork protection pathways
.
Hum. Mol. Genet.
,
29
,
R158
R164
.

12

Kolinjivadi
,
A.M.
,
Sannino
,
V.
,
de
 
Antoni
,
A.
,
Techer
,
H.
,
Baldi
,
G.
and
Costanzo
,
V.
(
2017
)
Moonlighting at replication forks - a new life for homologous recombination proteins BRCA1, BRCA2 and RAD51
.
FEBS Lett.
,
591
,
1083
1100
.

13

Kolinjivadi
,
A.M.
,
Sannino
,
V.
,
De Antoni
,
A.
,
Zadorozhny
,
K.
,
Kilkenny
,
M.
,
Techer
,
H.
,
Baldi
,
G.
,
Shen
,
R.
,
Ciccia
,
A.
,
Pellegrini
,
L.
 et al. (
2017
)
Smarcal1-mediated fork reversal triggers Mre11-dependent degradation of nascent DNA in the absence of Brca2 and stable Rad51 nucleofilaments
.
Mol. Cell
,
67
(
867–881
), e867.

14

Panzarino
,
N.J.
,
Krais
,
J.J.
,
Cong
,
K.
,
Peng
,
M.
,
Mosqueda
,
M.
,
Nayak
,
S.U.
,
Bond
,
S.M.
,
Calvo
,
J.A.
,
Doshi
,
M.B.
,
Bere
,
M.
 et al. (
2021
)
Replication gaps underlie BRCA deficiency and therapy response
.
Cancer Res.
,
81
,
1388
1397
.

15

Schlacher
,
K.
,
Christ
,
N.
,
Siaud
,
N.
,
Egashira
,
A.
,
Wu
,
H.
and
Jasin
,
M.
(
2011
)
Double-strand break repair-independent role for BRCA2 in blocking stalled replication fork degradation by MRE11
.
Cell
,
145
,
529
542
.

16

Berti
,
M.
,
Teloni
,
F.
,
Mijic
,
S.
,
Ursich
,
S.
,
Fuchs
,
J.
,
Palumbieri
,
M.D.
,
Krietsch
,
J.
,
Schmid
,
J.A.
,
Garcin
,
E.B.
,
Gon
,
S.
 et al. (
2020
)
Sequential role of RAD51 paralog complexes in replication fork remodeling and restart
.
Nat. Commun.
,
11
,
3531
.

17

Drexler
,
G.A.
,
Rogge
,
S.
,
Beisker
,
W.
,
Eckardt-Schupp
,
F.
,
Zdzienicka
,
M.Z.
and
Fritz
,
E.
(
2004
)
Spontaneous homologous recombination is decreased in Rad51C-deficient hamster cells
.
DNA Repair (Amst.)
,
3
,
1335
1343
.

18

Gayarre
,
J.
,
Martin-Gimeno
,
P.
,
Osorio
,
A.
,
Paumard
,
B.
,
Barroso
,
A.
,
Fernandez
,
V.
,
de la
 
Hoya
,
M.
,
Rojo
,
A.
,
Caldes
,
T.
,
Palacios
,
J.
 et al. (
2017
)
Characterisation of the novel deleterious RAD51C p.Arg312Trp variant and prioritisation criteria for functional analysis of RAD51C missense changes
.
Br. J. Cancer
,
117
,
1048
1062
.

19

Godthelp
,
B.C.
,
Wiegant
,
W.W.
,
van
 
Duijn-Goedhart
,
A.
,
Scharer
,
O.D.
,
van
 
Buul
,
P.P.
,
Kanaar
,
R.
and
Zdzienicka
,
M.Z.
(
2002
)
Mammalian Rad51C contributes to DNA cross-link resistance, sister chromatid cohesion and genomic stability
.
Nucleic Acids Res.
,
30
,
2172
2182
.

20

Loveday
,
C.
,
Turnbull
,
C.
,
Ruark
,
E.
,
Xicola
,
R.M.
,
Ramsay
,
E.
,
Hughes
,
D.
,
Warren-Perry
,
M.
,
Snape
,
K.
,
Breast Cancer Susceptibility
,
C.
,
Eccles
,
D.
 et al. (
2012
)
Germline RAD51C mutations confer susceptibility to ovarian cancer
.
Nat. Genet.
,
44
,
475
476
 
author reply 476
.

21

Long
,
D.T.
,
Raschle
,
M.
,
Joukov
,
V.
and
Walter
,
J.C.
(
2011
)
Mechanism of RAD51-dependent DNA interstrand cross-link repair
.
Science
,
333
,
84
87
.

22

Giovannini
,
S.
,
Weller
,
M.C.
,
Repmann
,
S.
,
Moch
,
H.
and
Jiricny
,
J.
(
2019
)
Synthetic lethality between BRCA1 deficiency and poly(ADP-ribose) polymerase inhibition is modulated by processing of endogenous oxidative DNA damage
.
Nucleic Acids Res.
,
47
,
9132
9143
.

23

Zellweger
,
R.
,
Dalcher
,
D.
,
Mutreja
,
K.
,
Berti
,
M.
,
Schmid
,
J.A.
,
Herrador
,
R.
,
Vindigni
,
A.
and
Lopes
,
M.
(
2015
)
Rad51-mediated replication fork reversal is a global response to genotoxic treatments in human cells
.
J. Cell Biol.
,
208
,
563
579
.

24

Zadorozhny
,
K.
,
Sannino
,
V.
,
Belan
,
O.
,
Mlcouskova
,
J.
,
Spirek
,
M.
,
Costanzo
,
V.
and
Krejci
,
L.
(
2017
)
Fanconi-anemia-associated mutations destabilize RAD51 filaments and impair replication fork protection
.
Cell Rep.
,
21
,
333
340
.

25

French
,
C.A.
,
Tambini
,
C.E.
and
Thacker
,
J.
(
2003
)
Identification of functional domains in the RAD51L2 (RAD51C) protein and its requirement for gene conversion
.
J. Biolumin. Chemilumin.
,
278
,
45445
45450
.

26

Lio
,
Y.C.
,
Mazin
,
A.V.
,
Kowalczykowski
,
S.C.
and
Chen
,
D.J.
(
2003
)
Complex formation by the human Rad51B and Rad51C DNA repair proteins and their activities in vitro
.
J. Biolumin. Chemilumin.
,
278
,
2469
2478
.

27

Thakar
,
T.
and
Moldovan
,
G.L.
(
2021
)
The emerging determinants of replication fork stability
.
Nucleic Acids Res.
,
49
,
7224
7238
.

28

Bhat
,
K.P.
,
Krishnamoorthy
,
A.
,
Dungrawala
,
H.
,
Garcin
,
E.B.
,
Modesti
,
M.
and
Cortez
,
D.
(
2018
)
RADX modulates RAD51 activity to control replication fork protection
.
Cell Rep.
,
24
,
538
545
.

29

Liu
,
W.
,
Krishnamoorthy
,
A.
,
Zhao
,
R.
and
Cortez
,
D.
(
2020
)
Two replication fork remodeling pathways generate nuclease substrates for distinct fork protection factors
.
Sci. Adv.
,
6
, 1–10.

30

Bhat
,
K.P.
and
Cortez
,
D.
(
2018
)
RPA and RAD51: fork reversal, fork protection, and genome stability
.
Nat. Struct. Mol. Biol.
,
25
,
446
453
.

31

Moynahan
,
M.E.
and
Jasin
,
M.
(
2010
)
Mitotic homologous recombination maintains genomic stability and suppresses tumorigenesis
.
Nat. Rev. Mol. Cell Biol.
,
11
,
196
207
.

32

Ding
,
X.
,
Ray Chaudhuri
,
A.
,
Callen
,
E.
,
Pang
,
Y.
,
Biswas
,
K.
,
Klarmann
,
K.D.
,
Martin
,
B.K.
,
Burkett
,
S.
,
Cleveland
,
L.
,
Stauffer
,
S.
 et al. (
2016
)
Synthetic viability by BRCA2 and PARP1/ARTD1 deficiencies
.
Nat. Commun.
,
7
,
12425
.

33

Ray Chaudhuri
,
A.
,
Callen
,
E.
,
Ding
,
X.
,
Gogola
,
E.
,
Duarte
,
A.A.
,
Lee
,
J.E.
,
Wong
,
N.
,
Lafarga
,
V.
,
Calvo
,
J.A.
,
Panzarino
,
N.J.
 et al. (
2016
)
Replication fork stability confers chemoresistance in BRCA-deficient cells
.
Nature
,
535
,
382
387
.

34

Taglialatela
,
A.
,
Alvarez
,
S.
,
Leuzzi
,
G.
,
Sannino
,
V.
,
Ranjha
,
L.
,
Huang
,
J.W.
,
Madubata
,
C.
,
Anand
,
R.
,
Levy
,
B.
,
Rabadan
,
R.
 et al. (
2017
)
Restoration of replication fork stability in BRCA1- and BRCA2-deficient cells by inactivation of SNF2-family fork remodelers
.
Mol. Cell
,
68
,
414
, e418–
430
.

35

Feng
,
W.
and
Jasin
,
M.
(
2017
)
Homologous recombination and replication fork protection: BRCA2 and more!
 
Cold Spring Harbor Symp. Quant. Biol.
,
82
,
329
338
.

36

Clague
,
J.
,
Wilhoite
,
G.
,
Adamson
,
A.
,
Bailis
,
A.
,
Weitzel
,
J.N.
and
Neuhausen
,
S.L.
(
2011
)
RAD51C germline mutations in breast and ovarian cancer cases from high-risk families
.
PLoS One
,
6
,
e25632
.

37

Zdzienicka
,
M.Z.
and
Simons
,
J.W.
(
1987
)
Mutagen-sensitive cell lines are obtained with a high frequency in V79 Chinese hamster cells
.
Mutat. Res.
,
178
,
235
244
.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://academic.oup.com/journals/pages/open_access/funder_policies/chorus/standard_publication_model)

Supplementary data