Clinical samples and public datasets
Breast cancer tissues and adjacent normal tissues were procured from Nanjing Medical University First Affiliated Hospital. The inclusion criteria for patient samples included those who had not received neoadjuvant therapy. Following resection, the collected samples were promptly frozen in liquid nitrogen to preserve their molecular integrity. Ethical considerations were given utmost importance, and all patients involved in this study provided written informed consent. The study protocol was approved by the Ethics Committee of Nanjing Medical University First Affiliated Hospital. Public datasets were obtained from Oncomine (https://www.oncomine.org/) [30], UALCAN (https://ualcan.path.uab.edu/index.html) [31], bc-GenExMiner 4.9 (http://bcgenex.ico.unicancer.fr/BC-GEM/GEM-Accueil.php?js=1) [32], Kaplan–Meier plotter (http://kmplot.com/analysis/) [33, 34], Search Tool for the Retrieval of Interacting Genes (STRING, https://string-db.org) [35], Comparative Toxicogenomics Database (CTD, http://ctdbase.org) [36], Genomics of Drug Sensitivity in Cancer (GDSC, https://www.cancerrxgene.org/) [37], SWISS-MODEL (https://swissmodel.expasy.org/) [38] and The Cancer Genome Atlas (TCGA) [39] for analysis.
Cell culture
Human breast cancer cell lines (MCF-7, ZR-75-1, BT474, MDA-MB-231, MDA-MB-453, and T-47D) and a normal mammary epithelial cell line (MCF-10A) were procured from the American Type Culture Collection (ATCC) (Manassas, VA, USA). The SUM-1315 cell line was generously provided by Stephen Ethier (University of Michigan, Ann Arbor, MI, USA). The HEK293T cell line was maintained and preserved by our laboratory. MCF-7, ZR-75-1, BT474, MDA-MB-231, MDA-MB-453, SUM1315, and HEK293T cells were cultured in DMEM (Gibco, USA), while T-47D cells were cultured in RPMI 1640 (Gibco, USA). The culture media for all cell lines contained 10% fetal bovine serum, 100 U/mL penicillin, and 100 mg/mL streptomycin. The cells were incubated at 37°C in a humidified atmosphere with 5% CO2.
Cell viability assay and determination of drug synergy
Cell viability assays were performed using the Cell Counting Kit-8 (CCK-8) (Vazyme, China) following the manufacturer’s instructions. Briefly, 2000 cells were seeded into each well of a 96-well plate, with 100 µL of DMEM supplemented with 10% FBS. At the specified time points, the medium was replaced with 100 µL of DMEM containing CCK-8 (90 µL DMEM and 10 µL CCK-8), and the cells were incubated for 2 h. The absorbance was measured spectrophotometrically at 450 nm.
To evaluate the synergistic effect of the two drugs, the combination index (CI) was determined using the app SiCoDEA [40]. The CI provides a quantitative measure of drug synergy, and it is calculated using Loewe additivity model and the Log-logistic [01] method as the drug-response curve model [41].
RNA extraction and quantitative real-time polymerase chain reaction (qRT-PCR)
Total RNA from cells or tissues was isolated using Trizol reagent (Takara, Japan). Complementary DNA (cDNA) was synthesized using the HiScript Q RT SuperMix (Vazyme, China). Subsequently, qRT-PCR was conducted using the AceQ qPCR SYBR Green Master Mix (Vazyme, China). β-actin were employed as endogenous controls for the relative expression of mRNA, respectively. The specific primer sequences utilized in this study are listed in Supplementary Table S1. The relative expression levels were determined using the 2−∆∆CT method, normalizing to the appropriate control and calculating the fold change.
Western blot analysis
The total protein from breast cancer cells was extracted using RIPA buffer and separated by 10% SDS-PAGE. Subsequently, the proteins were transferred onto a PVDF membrane (Bio-Rad, USA). The membranes were blocked using 5% skimmed milk powder and then incubated with primary antibodies against FANCI (1:2000, Proteintech, China, 20789-1-AP), PARP1 (1:2000, Proteintech, China, 13371-1-AP), Histone H3 (1:2000, Proteintech, China, 17168-1-AP), γH2AX (1:2000, Proteintech, China, 10856-1-AP), GAPDH (1:1000, Proteintech, China, 10856-1-AP), β-actin (1:1000, Proteintech, China, 20536-1-AP), pADPr (1:200, SANTA CRUZ, USA, sc-56198), and FLAG-Tag (1:1000, Beyotime, China, AF0036) at 4°C overnight. Subsequently, the membranes were incubated with secondary antibodies (1:5000) (Cell Signaling Technology, USA, 7074P2) at room temperature for 2 h. Finally, the protein bands were visualized using ImmobilobTM Western Chemiluminescent HRP Substrate (Millipore, USA).
Lentiviral infection and plasmid transfection
Commercially available lentiviral vectors containing FANCI-coding and short hairpin RNA (shRNA)-targeting FANCI sequence (FANCI, FANCI-sh1, and FANCI-sh2) were obtained from NOVOBIO (Shanghai, China) to achieve up- or down-regulation of FANCI in breast cancer cells. Scrambled lentiviral constructs were employed as negative controls (shNC and vector) to assess the baseline response. All vectors were verified through DNA sequencing. When the breast cancer cell lines SUM-1315 and ZR-75-1 reached approximately 30 to 40% confluence, they were infected with FANCI-sh1, FANCI-sh2, shNC, FANCI, and the vector at an appropriate multiplicity of infection. Stable cell lines were established by treating them with 5 µg/ml Blasticidin (InvivoGen, USA, ant-bl-1) for 1 week. The expression of FANCI in the cells was evaluated by qRT-PCR and Western blot analysis. shRNA sequences used in this study are listed in Supplementary Table S2.
The FLAG-FANCI fragments vectors were obtained from Yormbio (Shanghai, China). Based on the protein structure provided by SWISS-MODEL (https://swissmodel.expasy.org/), we generated seven fragmented DNA sequences encoding truncated domains of FANCI, derived from its full-length sequence (NM 001113378.2). These truncated DNA fragments, tagged with the FLAG epitope, were then subcloned into vectors to create the FLAG-FANCI fragments expression plasmids. Supplementary Table S3 provides details about the domain and length of each truncated fragment.
Cell proliferation assay
Cell proliferation assays were performed using the Cell Counting Kit-8 (CCK-8) (Vazyme, China) according to the manufacturer’s instructions. Briefly, 2000 cells were seeded into each well of a 96-well plate, with 100 µL of DMEM supplemented with 10% FBS. At the designated time points, the medium was replaced with 100 µL of DMEM containing CCK-8 (90 µL DMEM and 10 µL CCK-8), and the cells were incubated for 2 h. The absorbance was measured spectrophotometrically at 450 nm.
Colony formation assay
For the colony formation assay, 500 cells were seeded in a six-well plate and cultured in DMEM medium supplemented with 10% FBS for approximately 2 weeks. Proliferating colonies were fixed with methanol and stained with 1% crystal violet (Beyotime, China). The number of colonies was quantified by visual observation of single-cell proliferation.
EdU incorporation assay
Cell proliferation was assessed using the 5-ethynyl-2’-deoxyuridine (EdU) assay (Beyotime, China). The incorporation of EdU into newly synthesized DNA provides a specific and quantitative measurement of cell proliferation. Breast cancer cells were seeded in 96-well plates at a density of 2 × 104 cells per well and cultured in DMEM medium supplemented with 10% FBS for 24 h. Following that, the cells were incubated with 50 µM EdU at 37°C for 2 h. Subsequently, the cells were fixed with 4% paraformaldehyde, permeabilized with 0.5% Triton X-100, and stained with 1× Apollo® reaction cocktail for 30 min. Finally, the cell nuclei were counterstained with DAPI, and the cells were visualized under a fluorescence microscope.
Transwell assay
For the Transwell assay, a total of 3 × 104 cells were suspended in 100 µL of serum-free medium and added to the upper compartment of each chamber. The lower chamber was filled with 600 µL of medium containing 20% fetal bovine serum. Transwell plates (Corning, USA) were placed in a humidified incubator with 95% air and 5% CO2 at 37°C for 48 h to allow cell migration. After incubation, the chambers were carefully washed with PBS, fixed with formaldehyde, and stained with 0.5% crystal violet (Beyotime, China). The migrated cells were then imaged and quantified using ImageJ software (National Institutes of Health, Bethesda, MD, USA) [42].
Cell wound-healing assay
The cell wound-healing assay was employed to assess cell migration. Breast cancer cells were seeded in 6-well plates and cultured until they reached confluence. Subsequently, linear scratch wounds were generated using a 200 µL sterile pipette tip. After washing the cells three times with PBS, images were acquired using an inverted fluorescence microscope at 0 and 48 h (Olympus, Japan).
Immunofluorescent staining
For immunofluorescent staining, adherent breast cancer cells were washed with PBS, fixed with 4% paraformaldehyde, permeabilized with 0.1% Triton X-100, and blocked with 1% bovine serum albumin (BSA) in PBS. The cells were then incubated with primary antibodies against γH2AX (2 µg, Proteintech, China, 10856-1-AP), FANCI (2 µg, Proteintech, China, 20789-1-AP), or PARP1 (2 µg, Proteintech, China, 13371-1-AP) in 1% BSA at 4°C overnight. Afterward, the cells were washed three times with PBS for 5 min each and incubated with the appropriate secondary antibody (Beyotime, China, A0516/A0468). DNA staining was performed using Gold Antifade Mountant with DAPI (Invitrogen, USA, P36931). Immunofluorescence imaging was carried out using a Leica Stellaris STED confocal laser scanning microscope (Leica Microsystems, Buffalo Grove, USA). The data was analyzed by Leica Application Suite X.
Animal experiment
All animal experiments were conducted in accordance with the guidelines of the Institutional Animal Care and Use Committee of Nanjing Medical University. In different stages of the study, we injected 1×106 wild-type SUM-1315 cells or cells expressing shNC or FANCI-sh1 vectors into the mammary fat pads of 4-week-old female BALB/c nude mice for the in vivo xenograft model. The mice were housed in cages with five animals each, maintained at a room temperature of 23°C ± 1°C, and provided with ad libitum access to standard rodent diet and water. Tumor volumes were monitored every 4 days by measuring the length and width of the tumors using calipers. The tumor volume was calculated using the formula: (length) × (width)2/2. A total of three animal experiments were conducted at different stages.
In the first animal experiment, fourteen female BALB/c nude mice were randomly divided into two groups (seven mice each group): shNC group and FANCI-sh1 group, without any further treatment. This experiment aimed to elucidate the impact of FANCI on the proliferation of breast cancer cells in in vivo animal models.
In the second animal experiment, fifty-six mice were divided into four treatment groups using random assignment: shNC (control group), shNC + talazoparib (talazoparib group), FANCI-sh1 + sterile PBS (FANCI knockdown group), and FANCI-sh1 + talazoparib(combination treatment group). Each group was further divided into two subgroups (seven mice each subgroup): one receiving immediate treatment after cell implantation and the other receiving delayed treatment initiated when the tumors reached an average volume of 15 mm3. Talazoparib was administered at a dosage of 0.33 mg/kg daily through intraperitoneal injection. This experiment aimed to investigate the effect of FANCI on the therapeutic efficacy of PARP inhibitors in in vivo animal models, as well as to compare the outcomes between early treatment and delayed treatment strategies.
In the third experiment, a total of twenty-eight mice were injected with SUM-1315 wild-type cells and subsequently divided into four groups (seven mice per group) using random assignment. The groups included: Control (treated with sterile PBS), talazoparib (0.33 mg/kg/day, administered intraperitoneally), palbociclib (100mg/kg/day, administered orally), and talazoparib + palbociclib (combination treatment). Each group received the respective treatment once daily at the indicated dosage. The experiment was ceased after twenty-seven days, and tumors were harvested and imaged. Euthanasia of the mice and tumor collection were performed upon reaching the humane endpoint.
Immunoprecipitation and co-immunoprecipitation assays
Breast cancer cells were washed three times with cold PBS and then homogenized in IP lysis buffer (Beyotime, China) supplemented with protease and phosphatase inhibitor cocktails (Beyotime, China). To remove non-specific binding, total protein lysates were precleared by adding 1.0 µg of the appropriate control IgG (normal mouse or rabbit IgG, depending on the host species of the primary antibody) along with 20 µl of resuspended Protein A/G agarose beads (SANTA CRUZ, USA, SC-2003). Discard the pellet after 30 min incubation at 4℃. The precleared lysate was then used for each immunoprecipitation (IP) using the specific antibody of interest, and Protein A/G agarose beads were added to the cells, followed by overnight incubation at 4℃. The precipitated proteins were subsequently washed and analyzed by western blot. The mass spectrometry analysis was conducted by BGI Genomics (Shenzhen, China). For FLAG-tag protein IP Assay, cell extracts were immunoprecipitated with Anti-FLAG Agarose Gel (Beyotime, China, P2202S-4) at 4℃ overnight. The immunoprecipitates were thoroughly washed with lysis buffer five times, eluted with SDS loading buffer, boiled at 99℃ for 5 min, and then immunoblotted with the indicated antibodies. The samples were incubated with primary antibodies against FANCI (2µg, Proteintech, China, 20789-1-AP), PARP1 (2µg, Proteintech, China, 13371-1-AP), normal rabbit IgG (2µg, Beyotime, China, A7016).
Statistical Analysis
Statistical analysis was conducted using SPSS 24.0 and GraphPad Prism 8.0. The experimental data are presented as mean ± standard deviation (SD). Group comparisons were assessed using Student’s t-test or ANOVA as appropriate. A significance level of P < 0.05 was considered statistically significant.