Academic Editor

Article Metrics

  • Fig. 1.

    View in Article
    Full Image
  • Fig. 2.

    View in Article
    Full Image
  • Fig. 3.

    View in Article
    Full Image
  • Fig. 4.

    View in Article
    Full Image
  • Fig. 5.

    View in Article
    Full Image
  • Information

  • Download

  • Contents

Abstract

Background: The switching/sucrose non-fermentable (SWI/SNF) Related, Matrix Associated, Actin Dependent Regulator Of Chromatin, Subfamily A (SMARCA) member 2 and member 4 (SMARCA2/4) are paralogs and act as the key enzymatic subunits in the SWI/SNF complex for chromatin remodeling. However, the role of SMARCA2/4 in DNA damage response remains unclear. Methods: Laser microirradiation assays were performed to examine the key domains of SMARCA2/4 for the relocation of the SWI/SNF complex to DNA lesions. To examine the key factors that mediate the recruitment of SMARCA2/4, the relocation of SMARCA2/4 to DNA lesions was examined in HeLa cells treated with inhibitors of Ataxia-telangiectasia-mutated (ATM), Ataxia telangiectasia and Rad3-related protein (ATR), CREB-binding protein (CBP) and its homologue p300 (p300/CBP), or Poly (ADP-ribose) polymerase (PARP) 1/2 as well as in H2AX-deficient HeLa cells. Moreover, by concomitantly suppressing SMARCA2/4 with the small molecule inhibitor FHD286 or Compound 14, the function of SMARCA2/4 in Radiation sensitive 51 (RAD51) foci formation and homologous recombination repair was examined. Finally, using a colony formation assay, the synergistic effect of PARP inhibitors and SMARCA2/4 inhibitors on the suppression of tumor cell growth was examined. Results: We show that SMARCA2/4 relocate to DNA lesions in response to DNA damage, which requires their ATPase activities. Moreover, these ATPase activities are also required for the relocation of other subunits in the SWI/SNF complex to DNA lesions. Interestingly, the relocation of SMARCA2/4 is independent of γH2AX, ATM, ATR, p300/CBP, or PARP1/2, indicating that it may directly recognize DNA lesions as a DNA damage sensor. Lacking SMARCA2/4 prolongs the retention of γH2AX, Ring Finger Protein 8 (RNF8) and Breast cancer susceptibility gene 1 (BRCA1) at DNA lesions and impairs RAD51-dependent homologous recombination repair. Furthermore, the treatment of an SMARCA2/4 inhibitor sensitizes tumor cells to PARP inhibitor treatment. Conclusions: This study reveals SMARCA2/4 as a DNA damage repair factor for double-strand break repair.

1. Introduction

Genomic DNA constantly encounters genotoxic stresses that induce DNA damage. In response to DNA damage, the surveillance system of cells is quickly activated [1]. DNA lesions are detected by DNA damage sensors, which are associated with chromatin relaxation for DNA damage repair [2]. Several important chromatin remodeling complexes have been reported to participate in DNA damage response, one of which is the switching/sucrose non-fermentable (SWI/SNF) complex [3].

The SWI/SNF complex is evolutionarily conserved in eukaryotes and plays a key role in regulating the position of nucleosomes on genomic DNA [3, 4, 5, 6]. This complex is an ATP-dependent chromatin remodeling complex with multiple subunits. The key subunit in the SWI/SNF complex is subfamily A (SMARCA) member 2 (SMARCA2)/Brahma (BRM), an ATPase with the Asp-Glu-Gly-His (DEGH) helicase fold [7, 8]. It hydrolyzes ATP and provides the energy for the whole complex to disrupt the interaction between histones and DNA, thereby removing nucleosome barriers and exposing naked DNA for numerous biological activities, such as transcription and replication [9, 10]. SMARCA4/Brahma-related gene 1 (BRG1) is a paralog of SMARCA2, which contains a similar domain architecture and has a redundant function to SMARCA2 [2, 11, 12, 13, 14]. In addition to SMARCA2/4, the SWI/SNF complex also has up to 28 other subunits depending on different biological contexts [7]. However, nine of them are known as core subunits that are involved in all the known functions of the SWI/SNF complex [15].

It has been shown that the SWI/SNF complex participates in DNA damage repair. The complex is recruited to DNA double-strand breaks (DSBs) and participates in both non-homologous end joining and homologous recombination repair via sliding nucleosome barriers and exposing naked DNA ends to facilitate the loading of repair machinery [5, 6]. However, the recruitment mechanism of the SWI/SNF complex seems unclear. It has been reported that poly(ADP-ribosyl)ation (aka PARylation) signals or histone acetylation events mediate the recruitment of the SWI/SNF complex to DNA lesions [16, 17, 18, 19]. However, PARylation is mainly induced by DNA single-strand breaks [20], whereas the SWI/SNF complex is known to participate in DSB repair [5, 6]. It is also unclear if any specific histone acetylation events are significantly enriched at DNA lesions. Moreover, earlier studies have shown that loss of SMARCA2 is sufficient for impairing DSB repair [5, 6]. However, it was recently identified that SMARCA4, the paralog of SMARCA2, plays a redundant role in the SWI/SNF complex [21]. Since both SMARCA2 and SMARCA4 are ATPase in the SWI/SNF complex, only lacking both of them, but not either of them, can suppress the function of the SWI/SNF complex [21, 22]. The DSB repair relies on the chromatin remodeling activity of the SWI/SNF complex, during which the energy is derived from the ATPase of SMARCA2 or SMARCA4 in the SWI/SNF complex. With these unsolved questions, we have carefully examined the recruitment of SMARCA2 and SMARCA4 to DNA lesions as well as their functions in homologous recombination (HR) repair. We demonstrate that lacking both SMARCA2 and SMARCA4 inhibits HR repair. Moreover, the recruitment of SMARCA2 and SMARCA4 to DNA lesions is independent of PARylation and histone acetylation but requires the ATPase activities of SMARCA2 and SMARCA4.

2. Experimental Procedures
2.1 Plasmids

To express green fluorescent protein (GFP)-tagged SMARCA2, SMARCA4, SMARACC1 and SMARCD1, the cDNAs of SMARCA2, SMARCA4, SMARACC1 and SMARCD1 were synthesized by Tsingke Biotechnology (Beijing, China) based on the sequences from NCBI (NM_003070 for SMARCA2, NM_001128844 for SMARCA4, NM_003074 for SMARCC1, and NM_003076 for SMARCD1). The complementary DNAs (cDNAs) were then cloned into pEGFP-C1 (Clontech, Beijing, China) to generate GFP-SMARCA2, GFP-SMARCA4, GFP-SMARCC1, and GFP-SMARCD1. For the expression of truncation mutants of SMARCA2, N-terminal (N-Ter, amino acids 1-653), helicase domain (Hel, amino acids 654-1383), and bromo domain (BRD, amino acids 1384-1590) were cloned into pEGFP-C1 to generate GFP-N-Ter, GFP-Hel and GFP-BRD. The K755A mutant of SMARCA2 and the K785A mutant of SMARCA4 were generated by site-directed mutagenesis from the wild type SMARCA2 and SMARCA4, respectively [21, 22].

2.2 Cell Culture and Transfection

HeLa, HCC1187, MDA-MB-468 and MDA-MB-231 cells were purchased from American Type Culture Collection. HeLa cells were cultured in Dulbecco’s Modified Eagle Medium (DMEM) (Catalog number: 11965092; Thermo Fisher Scientific, Waltham, MA, USA), HCC1187 cells were cultured in Roswell Park Memorial Institute (RPMI)-1640 medium (Catalog number: 11875168; Thermo Fisher Scientific, Waltham, MA, USA), and MDA-MB-468 and MDA-MB-231 cells were cultured in Leibovitz’s L-15 Medium (Catalog number: 21083027; Thermo Fisher Scientific, Waltham, MA, USA), containing 10% fetal bovine serum and 1% penicillin-streptomycin (Catalog number: 15140122; Thermo Fisher Scientific, Waltham, MA, USA) at 37 °C with 5% CO2. All cell lines were validated by Short Tandem Repeat (STR) profiling and tested negative for mycoplasma.

To ectopically express GFP-tagged proteins, transfection of the cells with all pEGFP constructs was carried out using Lipofectamine 2000 (Catalog number: 11668500; Thermo Fisher Scientific, Waltham, MA, USA) according to the manufacturer’s protocol.

To generate HeLa SMARCA2 or HeLa SMARCA4 knockout cells, CRISPR/Cas9-mediated gene editing was performed. Cells were transfected with single guide RNA (sgRNA) against SMARCA2, constructed in pSpCas9(BB)-2A-Puro (PX459 V2.0) vector (Addgene plasmid ID: 62988). The sgRNA sequence targeting SMARCA2 was 5-TCCCATCCTATGCCGACGAT-3. To generate HeLa SMARCA4 knockout cells, the sgRNA sequence for SMARCA4 was 5-CCGGCGAGGGACCCGGGCTA-3. Transfection of the cells was carried out using Lipofectamine 2000 (Thermo Fisher Scientific) according to the manufacturer’s protocol. The transfected cells were selected in medium containing 2 µg/mL puromycin and then sub-cloned into 96-well plates 24 h post transfection. The monoclonal cell lines were identified. For radiation sensitive 51 (RAD51) gene knockdown, the sequence of si-RAD51 was 5-CUAAUCAGGUGGUAGCUCAUU-3. The sgRNAs and small interfering RNA (si-RNA) were synthesized by Tsingke Biotechnology company. Si-RNA transfections were carried out using Lipofectamine RNAiMAX (Catalog number: 13778075; Thermo Fisher Scientific, Waltham, MA, USA), according to the manufacturer’s protocol.

2.3 Antibodies

The following antibodies were purchased from their respective companies: anti-γH2AX (Novus, NB100-384), anti-RAD51 (Catalog Number: MA1-23271; Invitrogen, Carlsbad, CA, USA), anti-ring finger protein 8 (RNF8) (Catalog number: 14112-1-AP, Proteintech, Wuhan, Hubei, China), anti-breast cancer susceptibility gene 1 (BRCA1) (Catalog Number: NB100-404, Novus, Littleton, CO, USA), anti-SMARCA2 (Catalog number: HPA029981; Sigma, Louis, MO, USA).

2.4 Laser Microirradiation Assays

Laser microirradiation assays were performed according to a previous study [23]. GFP-tagged constructs were transfected into the specified cells and plated on 35-mm glass-bottom dishes. Cellular DNA damage in the nuclei of cultured cells was induced using microirradiation with a pulsed nitrogen laser (Spectra-Physics; 365 nm, 10 Hz pulse). This laser system was directly integrated into the epifluorescence microscope’s imaging pathway, focusing through a Plan-Apochromat 63×/NA 1.40 oil immersion objective. The laser power was adjusted to 50–70% of its maximum capacity. The resulting green fluorescence stripes were captured, analyzed using ImageJ software (National Institutes of Health, Bethesda, MD, USA) and the results presented represent images from 20 cells across three independent experiments. For inhibitor treatment, cells were treated with different inhibitors at 1 µM for 2 hours followed by laser microirradiation. The SMARCA2/4 inhibitor (FHD286, HY-144835), Ataxia-telangiectasia-mutated (ATM) inhibitor (KU55933, HY-12016), Ataxia telangiectasia and Rad3-related protein (ATR) inhibitor (VE-821, HY-14731), CREB-binding protein (CBP)/p300 inhibitor (CBP/p300-IN-21, HY-155229), and Poly (ADP-ribose) polymerase (PARP) 1/2 inhibitor (Olaparib, HY-10162) were purchased from MedChemExpress (Shanghai, China).

2.5 Immunofluorescence (IF) Staining

Cells growing on glass coverslips were irradiated with 5 Gy and allowed to recover for 12 h before immunofluorescence. Then, cells were treated with fixation buffer (4% PFA, 0.1% Triton X-100). The slides were incubated with different primary antibodies in 1% BSA at room temperature for 2 h or 4 °C overnight. Subsequently, the slides were washed with bovine serum albumin (PBS) and incubated with a secondary antibody. DNA was stained, and coverslips were mounted with 4,6-diamidino-2-phenylindole (DAPI) Fluoromount-G™ (Catalog Number: 36308ES20; Yeason, Shanghai, China). Cells were treated with FHD286 or Compound 14 at 1 µM for 2 hours followed by ionizing irradiation. Compound 14 was another specific SMARCA2/4 ATPase dual inhibitor, which was synthesized according to the compound structure [24]. SMARCA2, γH2AX, BRCA1, RNF8 and RAD51 foci were stained with the antibodies list in section 2.3.

2.6 HR Efficiency Assays

The DR-GFP reporter assay system was provided by Dr. Xiaochen Gai [25] at Westlake University. The assay was performed as described in a previous study [25]. The DR-GFP reporter system was specifically designed to quantify homologous recombination repair, where the iGFP gene fragment functions as a template for the HR process following an I-SceI-induced DSB in the upstream SceGFP cassette. To initiate this process, the HA-I-SceI-GR pCW tet-on plasmid was transfected into the cells. Doxycycline is then administered to trigger the expression of I-SceI for 18 hours. Acetonide treatment facilitates the translocation of I-SceI from the cytoplasm to the nucleus. Subsequently, I-SceI cuts the targeting site, generating a DSB, which upon repair, leads to the expression of GFP. Flow cytometry was performed to examine the efficiency of homologous recombination repair by calculating the GFP positive populations. For RAD51 knockdown, cells with DR-GFP insertion were transfected with negative control siRNA (siRNA-NC) or siRNA-RAD51 for 24 hours following HA-I-SceI-GR pCW tet-on plasmid transfection. For inhibitor treatment, cells with DR-GFP insertion were pretreated with or without FHD286 (1 µM) or Compound 14 (1 µM) for 2 hours before acetonide treatment.

2.7 Knockdown and Re-Expression Studies

SMARCA4 and SMARCA2 double knockdown was carried out by siRNA transfection directed against SMARCA2 (siRNA-SMARCA2): 5-UCGUCGAGCAAUCAUUUGGUU-3 and (siRNA-SMARCA4) SMARCA4: 5- UAGCAUUGAGGGCUGUCUCCA-3. A nontargeting control siRNA (siRNA-NC): 5-AACCACGUGAGGCAUCCAGGC-3 was used as a negative control. For double knockdown, individual siRNAs of SMARCA2 and SMARCA4 were co-transfected into cells with DR-GFP insertion 24 hours before HA-I-SceI-GR pCW tet-on plasmid transfection. The siRNA transfections were carried out using Lipofectamine RNAiMAX (Invitrogen). The full-length siRNA-resistant SMARCA2 was cloned into S protein-FLAG-Streptavidin binding peptide (SFB) vector. The K755A mutant of SMARCA2 was generated by site-directed mutagenesis from the siRNA-resistant full-length SMARCA2. Re-expression of full-length SMARCA2 or the ATPase inactive mutant (K755A) was performed by plasmid transfection using Lipofectamine 2000 (Thermo Fisher Scientific) 20 hours before HA-I-SceI-GR pCW tet-on plasmid transfection. Knockdown and/or re-expression were confirmed by Western blotting using the following primary antibodies: SMARCA4 (Catalog number: sc-17796; Santa Cruz, Shanghai, China), SMARCA2 (Catalog number: sc-166579; Santa Cruz, Shanghai, China), Vinculin (Catalog number: A14193; Abclonal, Wuhan, Hubei, China).

2.8 Clonogenic Assays

For the clonogenic cell proliferation assay, cells were seeded in 6-well plate (1000 cells/well). The cells were then treated with either a vehicle control or drugs with the indicated concentration on the next day, and culture medium was refreshed every 3 days for a total of 14 days in total. At the endpoints of the colony formation assays, the colonies were washed with PBS and fixed with 70% methanol for 10 minutes. This was followed by staining with 0.1% crystal violet. The number of colonies was counted by ImageJ software, and survival graphs were generated from three replicate wells, with colony numbers normalized to untreated controls.

2.9 Statistical Analysis

Statistical analyses were performed using GraphPad Prism7 (GraphPad Software, Inc., San Diego, CA, USA). The statistical significance was assessed using a two-tailed unpaired Student’s t-test. All of the data were presented as mean ± SD. The significance levels are annotated as follows: *, p 0.05. **, p 0.01. ***, p 0.001.

3. Results
3.1 The Enzymatic Activities of SMARCA2/4 are Essential for the Relocation of SMARCA2/4 to DNA Lesions

To study the molecular mechanism by which the SWI/SNF complex is recruited to DNA lesions, we examined SMARCA2/4, the key enzymatic subunits in the SWI/SNF complex. Using laser microirradiation assay and observation by fluorescence microscopy, we found that both GFP-tagged SMARCA2/4 relocated to DNA lesions (Fig. 1A). We also measured the recruitment kinetics and found that within five seconds, SMARCA2/4 was recruited to DNA lesions (Supplementary Fig. 1).

Fig. 1.

The relocation of the switching/sucrose non-fermentable (SWI/SNF) Related, Matrix Associated, Actin Dependent Regulator Of Chromatin, Subfamily A (SMARCA) member 2 and member 4 (SMARCA2/4) to DNA lesions. (A) pEGFP-C1-SMARCA2, pEGFP-C1-SMARCA4, or pEGFP-C1 was transfected into HeLa cells for 24 hours. Following laser microirradiation, the laser stripes of green fluorescent protein (GFP)-SMARCA2/4 were observed using fluorescence microscopy. The laser stripe is indicated with white arrows. The table on the right presents the cellular status, specifically indicating the presence or absence of stripes. Scale bar: 10 μm. (B) Schematic of the deletion mutants of SMARCA2. (C) GFP-tagged truncation mutants of SMARCA2 were transiently expressed in HeLa cells. Following laser microirradiation, the laser stripes of GFP-N-Ter, GFP-Hel and GFP-bromo domain (BRD) were examined using fluorescence microscopy. The laser stripe is indicated with white arrows. The table on the right presents the cellular status, specifically indicating the presence or absence of stripes. Scale bar: 10 μm. (D) GFP-SMARCA2-K755A or GFP-SMARCA4-K785A were transiently expressed in HeLa cells. Following laser microirradiation, the laser stripes of these enzymatically dead mutants were examined. Scale bar: 10 μm.

SMARCA2/4 has several distinct domains, including the N-terminal QLQ motif and HAS domain, which mediate the interaction with other subunits of the SWI/SNF complex; the ATPase/helicase domain in the middle, which is the solo enzymatic domain in the whole complex; the C-terminal bromodomain that is known to recognize acetylated histones [10, 22] (Fig. 1B). Since SMARCA2/4 shares an identical domain architecture, we generated deletion mutants of SMARCA2 and found that only the ATPase/helicase domain (indicated as GFP-Hel), but not other domains (indicated as GFP-N-Ter or GFP-BRD), was able to relocate to DNA lesions (Fig. 1C). Moreover, we established the ATPase-dead constructs which mutated the key lysine residue into alanine in ATPase/helicase domains of SMARCA2 (K755A) and SMARCA4 (K785A) to abolish their ATP-binding activities [22, 26], and found that these ATPase activities were required to target SMARCA2/4 to DNA lesions (Fig. 1D).

3.2 Relocation of the SWI/SNF Complex to DNA Lesions is Dependent on the ATPase Activities of SMARCA2/4

Next, we examined other core subunits of the SWI/SNF complex, including SMARCC1 and SMARCD1. We show the first evidence that, similar to SMARCA2/4, both SMARCC1 and SMARCD1 relocated to DNA lesions when cells were treated with laser microirradiation (Fig. 2A). Interestingly, when we pre-treated cells with FHD286, a specific dual inhibitor to kill both ATPase activities of SMARCA2/4 [27], SMARCC1 and SMARCD1 were no longer recruited to DNA lesions in the laser microirradiation assays (Fig. 2B). This suggests that the ATPase activity of SMARCA2/4 is also required for targeting other subunits of the SWI/SNF complex to the sites of DNA damage.

Fig. 2.

Relocation of the switching/sucrose non-fermentable (SWI/SNF) to DNA lesions. (A) pEGFP-C1-SMARCC1, pEGFP-C1-SMARCD1, or pEGFP-C1 was transfected into HeLa cells for 24 hours. Following laser microirradiation, the laser stripes of GFP-SMARCC1 or GFP-SMARCD1 were observed using fluorescence microscopy. The laser stripe is indicated with white arrows. The right table presents the cellular status, specifically indicating the presence or absence of stripes. Scale bar: 10 μm. (B) GFP-SMARCA2, GFP-SMARCC1 or GFP-SMARCD1 was transiently expressed in HeLa cells. The cells were pre-treated with or without 1 µM FHD286 for two hours followed by laser microirradiation. The localization of SMARCA2, SMARCC1, and SMARCD1 was examined. The laser stripe is indicated with white arrows. The right table presents the cellular status, specifically indicating the presence or absence of stripes. Scale bar: 10 μm. (C) GFP-SMARCA2, GFP-SMARCC1 or GFP-SMARCD1 was transfected into HeLa cells for 24 hours. Cells were treated with 1 µM Ataxia-telangiectasia-mutated (ATM) inhibitor (ku55933), 1 µM Ataxia telangiectasia and Rad3-related protein (ATR) inhibitor (VE-821), 1 µM CREB-binding protein (CBP) and its homologue p300 (CBP/p300) inhibitor (CBP/p300-IN-21) or 1 µM Poly (ADP-ribose) polymerase (PARP) inhibitor (olaparib) for two hours followed by laser microirradiation. The localization of SMARCA2, SMARCC1 and SMARCD1 was examined. The localization of the subunits of the SWI/SNF complex was also examined in the H2AX KO HeLa cells. The laser stripe is indicated with white arrows. The right table presents the cellular status, specifically indicating the presence or absence of stripes. Scale bar: 10 μm.

It has been reported that several DNA repair factors may mediate the recruitment of the SWI/SNF complex to DNA lesions, such as γH2AX, p300/CBP and PARylation [15, 19, 28]. Here, we examined H2AX-deficient cells as well as cells treated with inhibitors of ATM, ATR, p300/CBP, or PARP1/2. To our surprise, the SWI/SNF complex was still recruited to DNA lesions under these conditions (Fig. 2C). Only suppression of the ATPase/Helicase domain of SMARCA2/4 abolishes the recruitment of the SWI/SNF complex.

3.3 The Enzymatic Activity of SMARCA2/4 Suppresses the Dissociation of DNA Damage Response Factors from DSBs

Earlier studies have shown that the enrichment of DNA damage response factors, such as γH2AX, requires SMARCA2 [18, 19]. However, different results have shown that the SWI/SNF complex is the downstream effector of γH2AX at the beginning of the DSB response [29]. To further elucidate the role of the SWI/SNF complex in DNA damage response, we used CRISPR-Cas9 technology to knockout either SMARCA2 or SMARCA4 and treated cells with ionizing radiation (IR) to induce DSBs. However, without either SMARCA2 or SMARCA4, the enrichment of γH2AX at DSBs was not affected (Supplementary Fig. 2). Since lacking both SMARCA2 and SMARCA4 causes synthetic lethality for cells, we cannot use CRISPR-Cas9 technology to obtain SMARCA2/4 double knockout cells. Thus, we had to pre-treated cells with FHD286 for two hours to transiently suppress the enzymatic activity of SMARCA2/4 before the induction of DSBs by IR. Interestingly, the foci of γH2AX were not suppressed by the treatment of FHD286. Instead, we found that the foci of γH2AX existed for a prolonged period following IR treatment (Fig. 3). As a control, without IR treatment, neither FHD286 nor Compound 14 induced foci formation of γH2AX, suggesting that these chemical compound per se do not induce DSBs (Supplementary Fig. 3). We also examined the downstream factors of γH2AX, including RNF8 and BRCA1. Again, both foci of RNF8 and BRCA1 stayed for an extended period (Fig. 3). Moreover, these results were further validated when we treated cells with another potent selective SMARCA2/4 dual inhibitor, Compound 14 [24] (Fig. 3). Collectively, these results demonstrate that the suppression of the enzymatic activity of SMARCA2/4 impairs the dissociation of some DNA damage response factors from DSBs.

Fig. 3.

Lacking the enzymatic activities of SMARCA2/4 extends the accumulation of γH2AX, Ring Finger Protein 8 (RNF8) and Breast cancer susceptibility gene 1 (BRCA1). HeLa cells were pre-treated with or without FHD286 (1 µM) or Compound 14 (1 µM) for two hours, followed by 5 Gy of IR. Following the indicated recovery time, the foci of γH2AX (Top panel), RNF8 and BRCA1 were examined by immunofluorescence staining with anti-γH2AX, anti-RNF8 or anti-BRCA1 antibodies, respectively. The cells were also counterstained by 4,6-diamidino-2-phenylindole (DAPI). Foci number per cell was analyzed (Bottom panel). p values were calculated using Student’s t-test. “ns” indicates nonsignificant, *p < 0.05, ** p < 0.01, ***p < 0.001. Scale bars: 10 µm.

3.4 The Enzymatic Activities of SMARCA2/4 Facilitate RAD51-Mediated HR Repair

Upon SMARCA2/4 inhibition, the prolonged accumulation of DNA damage response factors, such as γH2AX, indicates the defects in DSB repair. It has been shown that the SWI/SNF complex is involved in HR repair. Here, we found that following IR treatment, SMARCA2 formed nuclear foci and colocalized with RAD51, a key enzyme in HR repair (Fig. 4A). Since SMARCA2/4 are key enzymatic subunits in the SWI/SNF complex, we asked if the ATPase activity of SMARCA2/4 is required for HR repair. In the process of HR repair, RAD51 recognizes ssDNA overhangs at DSB ends and motorizes ssDNA overhangs to invade into sister chromatids. Thus, RAD51 plays a key role during HR repair. Interestingly, when we treated cells with either FHD286 or Compound 14 to suppress both SMARCA2 and SMARCA4, we found that the IR-induced foci of RAD51 were suppressed (Fig. 4A). However, lacking either SMARCA2 or SMARCA4 did not affect the foci formation of RAD51 (Fig. 4B). Moreover, we performed a well-established GFP reporter assay to measure the efficacy of HR [25]. Following the treatment of FHD286 or Compound 14, HR repair was impaired (Fig. 4C). Moreover, we knocked down SMARCA2/4 by siRNA, which also impaired HR repair. However, when we reintroduced siRNA-resistant full-length SMARCA2 but not the enzymatically dead mutant K755A, we could rescue the HR defects (Fig. 4D).

Fig. 4.

Suppression of the enzymatic activities of SMARCA2/4 impairs RAD51-dependent homologous recombination (HR) repair. (A) HeLa cells were pre-treated with or without FHD286 (1 µM) or Compound 14 (1 µM) for two hours, followed by 5 Gy of IR. Foci formation of RAD51 and SMARCA2 was examined by immunofluorescence staining with anti-RAD51 or anti-SMARCA2 antibodies. The cells were also counterstained by DAPI. The number of foci per cell was analyzed. p values were calculated using Student’s t-test. “ns” indicates nonsignificant, ***p < 0.001. Scale bars: 10 µm. (B) SMARCA2 or SMARCA4 was knocked out in HeLa cells. IR-induced foci of RAD51 were examined following 5 Gy of IR. The cells were also counterstained by DAPI. The number of foci per cell was analyzed. p values were calculated using Student’s t-test. “ns” indicates nonsignificant. Scale bars: 10 µm. (C) HR efficacy was examined by DR-GFP reporter assays. The Y axis SSC-A (side scattering intensity) indicates the cells of interest gated for GFP fluorescence analysis. For the RAD51 knockdown, the cells with DR-GFP insertion were transfected with negative control siRNA (siRNA-NC) or siRNA-RAD51 for 24 hours following HA-I-SceI-GR pCW tet-on plasmid transfection. For the inhibitor treatment, cells with DR-GFP insertion were pretreated with or without FHD286 (1 µM) or Compound 14 (1 µM) for 2 hours before the acetonide treatment. GFP-positive populations were calculated by flow cytometry. (D) The enzymatic activity of SMARCA2 facilitates HR repair. HR efficacy was examined by DR-GFP reporter assays. The Y axis SSC-A (side scattering intensity) indicates the cells of interest gated for GFP fluorescence analysis. GFP-positive populations were calculated by flow cytometry (bottom right panel). p values were calculated using Student’s t-test. “ns” indicates nonsignificant, and ***p < 0.001. For SMARCA2/4 double knockdown, the cells with DR-GFP insertion were transfected with negative control siRNA (siRNA-NC) or siRNA-SMARCA2/4 24 hours before HA-I-SceI-GR pCW tet-on plasmid transfection. For SMARCA2 re-expression, knockdown cells were transfected with full-length siRNA-resistant S protein-FLAG-Streptavidin binding peptide (SFB)-SMARCA2 or the K755A mutant 20 hours before HA-I-SceI-GR pCW tet-on plasmid transfection. Knockdown and/or re-expression were confirmed by Western using indicated antibodies (Top right panel). Vinculin was used as a protein loading control. Figure was created with BioRender.com.

HR-deficient cells are often hypersensitive to PARP inhibitor treatment. To date, PARP inhibitors have been used for the treatment of breast, ovarian, pancreatic, and prostate cancers with HR deficiency. Interestingly, FHD286 is currently in clinical trials for cancer treatment. Since FHD286 treatment impairs HR repair, we ask if FHD286 treatment can sensitize tumor cells to PARP inhibitor treatment. We selected three breast cancer cell lines (HCC1187, MDA-MB-468 and MDA-MB-231) that are not sensitive to PARP inhibitor olaparib. Interestingly, with 50 nM of FHD286, all these cells line were sensitive to 100 nM olaparib treatment (Fig. 5), indicating that FHD286 may have an additive effect with PARP inhibitors for cancer treatment in the future.

Fig. 5.

FHD286 sensitizes tumor cells to the Poly (ADP-ribose) polymerase (PARP) inhibitor treatment. A total of 1000 cells were seeded in each well and treated with indicated doses of FHD286 and/or PARP inhibitor olaparib. Colony formation was examined at Day 14 with 0.1% crystal violet staining. Statistical analyses are shown in the lower panel. p values were calculated using Student’s t-test. “ns” indicates nonsignificant, *p < 0.05, **p < 0.01, ***p < 0.001.

4. Discussion

In this study, we have demonstrated that the enzymatic activity of SMARCA2/4 plays a key role in the recruitment of the SWI/SNF complex to DNA lesions, facilitating DSB repair. Earlier studies have shown that other factors, such as PARylation, acetylation, or phosphorylation events, mediate the recruitment of the SWI/SNF complex [16, 17, 18, 19]. Here, we have shown that only the ATPase activities of SMARCA2/4 are essential for the recruitment of this chromatin remodeling complex. Notably, chromatin remodeling should be one of the early DNA damage response events. Only after the removal of the nucleosome barrier can the repair machinery access DNA lesions for repair [8, 30]. Since the ATPase domains of SMARCA2/4 directly interact with genomic DNA [10, 13], it is possible that the SWI/SNF complex constantly scans genomic DNA and is able to detect DSBs. It also allows for quick chromatin remodeling for damage repair in response to DSBs. Laser microirradiation induces the rapid relocation of SMARCA2/4 within five seconds, confirming their quick response to DNA damage. Thus, we favor the possibility that SMARCA2/4 are able to quickly and directly recognize DNA lesions as DNA damage sensors.

Earlier studies have shown that the lack of SMARCA2 is sufficient to impair HR repair [5, 6]. However, numerous analyses have suggested that SMARCA2 and SMARCA4 are paralogs and have redundant functions in the SMARCA2/4 [2, 3, 11, 22]. In our study, we have demonstrated that abolishing both enzymatic activities of SMARCA2/4 is necessary to suppress HR repair. Notably, the SMARCA4 gene is frequently deleted during tumorigenesis [21, 31]. Therefore, if the cells only contain SMARCA2, ablating SMARCA2’s enzymatic activity is sufficient to disrupt the function of the SWI/SNF complex and impair HR repair.

In our study, we have demonstrated that suppressing SMARCA2/4 did not hinder the foci formation of several upstream DNA damage response factors, such as γH2AX, RNF8 and BRCA1, but rather extended the duration of these foci. Conversely, suppression the enzymatic activity of SMARCA2/4 abolished the foci formation of downstream repair machinery, such as RAD51. In response to DSBs, the SWI/SNF complex’s biological function is to remove nucleosome barriers and expose naked DNA for DNA end processing [13, 15]. Typically, at DSB ends, a few hundred base pairs are processed into ssDNA and coated with RAD51, forming RAD51-ssDNA filaments for strand invasion [32, 33]. It is estimated that only a few nucleosomes at DSB ends are evicted by chromatin remodeling complexes. Therefore, without the enzymatic activities of SMARCA2/4, we could not detect RAD51 at DSBs. However, unlike bona fide repair machinery, γH2AX mediates DNA damage response signals extending up to megabases away from DSBs [25, 34, 35, 36]. Thus, the enrichment of γH2AX does not seem to be controlled by the SWI/SNF complex. Instead, due to the delayed DSB repair caused by the suppression of SMARCA2/4, these response factors, such as γH2AX, remain at DSBs for a prolonged time.

Collectively, our studies provide an in-depth understanding of the biological functions of SMARCA2/4 in the context of DNA damage repair.

5. Conclusions

The SMARCA2 and SMARCA4 proteins play a crucial role as DNA damage sensors. The ATPase activities are essential for recruiting the SWI/SNF complex to DNA lesions. Consequently, the loss of SMARCA2 and SMARCA4 impairs HR repair, a vital process for maintaining genome integrity. This finding suggests that treatment with SMARCA2 and SMARCA4 inhibitors may sensitize tumor cells to PARP inhibitors, a class of drugs used in cancer therapy.

Availability of Data and Materials

The datasets used and/or analysed during the current study are available from the corresponding author on reasonable request.

Author Contributions

LY and DW designed the research project, performed the experiments, analyzed the data, and wrote the manuscript. Both authors have thoroughly reviewed and approved the content of the final manuscript. Both authors have participated sufficiently in the work and agreed to be accountable for all aspects of the work.

Ethics Approval and Consent to Participate

Not applicable.

Acknowledgment

We apologize to all authors whose work could not be mentioned due to space limitations. Figures were created with BioRender.com.

Funding

This work was supported in part by grants from Hangzhou City Leading Innovation and Entrepreneurship Team (TD2020004), “Pioneer” and “Leading Goose” R&D Program of Zhejiang (2024SSYS0033), Westlake University Education Foundation, and Westlake Laboratory of Life Sciences and Biomedicine.

Conflict of Interest

The authors declare no conflict of interest. Duo Wu is from the SynRx Therapeutics.

References

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.