The DNA damage response (DDR) is essential for the maintenance of genomic stability. Protein posttranslational modifications play pivotal roles in regulating the DDR process. Here, we found that SUMOylated RNF168 undergoes liquid–liquid phase separation (LLPS), which restricts the recruitment of RNF168 to DNA damage sites, reduces RNF168-catalyzed H2A ubiquitination, restrains 53BP1 in nuclear condensates, and ultimately impairs nonhomologous DNA end joining repair efficiency. Sentrin/SUMO-specific protease 1 (SENP1) was identified as a specific deSUMOylase of RNF168, and it was highly expressed in colorectal adenocarcinoma. In response to DNA damage, SENP1 decreased RNF168 SUMOylation and prevented RNF168 from forming nuclear condensates, thus promoting damage repair efficiency and cancer cell resistance to DNA damaging agents. Moreover, high SENP1 expression correlated with poor prognosis in patients with cancer, and SENP1 depletion sensitized cancer cells to chemotherapy. In summary, these findings reveal DDR is suppressed by SUMOylation-induced LLPS of RNF168 and suggest that SENP1 is a potential target for cancer therapy.

Significance:

Sentrin/SUMO-specific protease 1 decreases RNF168 SUMOylation and liquid–liquid phase separation to promote DNA damage repair, safeguarding genomic integrity and driving chemotherapy resistance.

In response to various types of DNA damage, cells use a set of signaling cascades, known as the DNA damage response (DDR), to sense and repair lesions to maintain genomic integrity and cellular homeostasis. A DNA double-strand break (DSB) is considered the most harmful DNA lesion (1). Two dedicated pathways, namely nonhomologous end joining (NHEJ) and homologous recombination (HR), are responsible for DSB repair (2–4). Following the occurrence of a DSB, a cascade of proteins is recruited to the damaged sites, among which, the RING finger protein 168 (RNF168), an E3 ubiquitin ligase of H2A (5, 6), has been identified as a necessary factor for the recruitment of repair proteins to damaged chromosomes, including 53BP1, the most critical effector in the NHEJ repair pathway (7).

Mounting evidence indicates that protein posttranslational modifications (PTM) play important roles in the DDR (8, 9). Similar to phosphorylation and ubiquitination, reversible SUMOylation, modulated by E1 activating enzyme, E2 conjugating enzyme, E3 ligating enzyme, and sentrin/SUMO-specific proteases (SENP), fine-tunes the recruitment of repair proteins to the damaged sites (10–12). For example, the SUMOylation E3 ligases PIAS1 and PIAS4 have been shown to promote DSB repair (10). SUMOylation of MDC1 is required for its degradation and removal from damaged sites (13). TIP60 SUMOylation attenuates its interaction with DNA-PKcs, thus facilitating HR (14). SENP2 regulates NEMO and NF-κB activation induced by DNA damage (15). However, whether SUMOylation/deSUMOylation regulates the function of RNF168 in the DDR remains unknown.

Recent studies have revealed that liquid–liquid phase separation (LLPS) acts as a governing mechanism for biomolecular condensation and membraneless organelle formation, which serves to regulate multiple biological processes including transcription, autophagy, intracellular signaling, and genomic integrity (16–20). For example, poly-ubiquitin chain-induced phase separation of adaptor protein p62 regulates protein quality by segregating the autophagosome marker LC3 (21). Pol II–mediated transcription takes place in nuclear condensates that assemble at both the promoter and enhancer loci (22–24). PTMs serve critical functions in governing LLPS (25–27); for example, SUMOylation regulates the disassembly and formation of stress granules (18), while ubiquitin binding disrupts UBQLN2 LLPS (28). PARylation has also been reported to promote the phase separation of hnRNP A1 (29). In addition, DDR proteins have been shown to undergo LLPS; for instance, DNA damage-induced 53BP1 phase separation coordinates damage detection with gene expression and checkpoint activation (30, 31). Moreover, LLPS regulates cellular processes involved in cancer cell pathology, and the dysregulation of LLPS is increasingly implicated as a previously hidden driver of oncogenic activity (19, 32). Therefore, it is of great importance to identify new PTM regulators that impact cancer progression through LLPS.

Here, we demonstrate that SUMOylated RNF168 induces LLPS, which sequesters 53BP1 into RNF168-formed nuclear puncta, subsequently reducing the recruitment of RNF168 to the DNA damage site and ultimately impairing the efficiency of the NHEJ repair pathway. In response to DSB, SENP1 specifically deSUMOylates RNF168 and disrupts the formation of nuclear condensates, which promotes NHEJ and cell survival. Moreover, SENP1 is expressed at a higher level in colon adenocarcinoma (COAD) and is related to a poor prognosis, whereas disrupting SENP1 leads to genomic instability and increases the sensitivity of colon cancer cells to chemotherapeutic drugs. Our findings provide novel insight into the molecular mechanism underlying the safeguarding of genomic integrity and the regulation of drug resistance by the SENP1-RNF168–53BP1 axis.

Antibodies, reagents, and plasmids

Rabbit anti-SUMO3 mAbs were purchased from Proteintech (catalog no. 11251–1-AP; RRID: AB_2198405) and Abcam (catalog no. 2970–1; RRID: AB_2198534). Mouse anti-Flag (catalog no. F3165; RRID: AB_259529) and anti-HA (catalog no. H9658; RRID: AB_260092) mAbs were bought from Sigma-Aldrich. Mouse anti-Myc (catalog no. M047–3; RRID: AB_591112), anti-His (catalog no. D291–3; RRID: AB_10597733), anti-α-tubulin (catalog no. M175–3; RRID: AB_10697817) mAbs and rabbit anti-GFP mAb (catalog no. 598; RRID: AB_591819) were procured from MBL. Rabbit anti-RNF168 and anti-SENP1 mAbs were purchased from Proteintech (catalog no. 21393–1-AP; RRID: AB_10733883) and Abcam (catalog no. ab108981; RRID: AB_10862449), respectively. Mouse anti-γH2AX (catalog no. DR1016–100UG; RRID: AB_437862) mAb was purchased from Millipore. Rabbit anti-H2A mAb was bought from Cell Signaling Technology (catalog no. 12349; RRID: AB_2687875). Rabbit anti-β-actin mAb was procured from Abclonal (catalog no. AC004, RRID: AB_2737399). Mouse anti-53BP1 mAb (catalog no. sc-517281; RRID: AB_2921289), rabbit anti-53BP1 mAb (catalog no. 88439; RRID: AB_10694558), and rabbit anti-BRCA1 pAb (catalog no. BS1036; RRID: AB_1662959) were purchased from Santa Cruz, Cell Signaling Technology, and Bioworld, respectively.

Plasmids expressing human SENP1, His-SUMO1, and His-SUMO3 were kindly provided by Prof. Jinke Cheng, Shanghai Jiao Tong University School of Medicine. Human wild-type RNF168 and the K210R mutant were inserted into the pcDNA-3Myc, pCMV-HA, pcDNA-3Flag, pEGFP-C1, and pET-28a vectors. Human RNF168 K173R and K203R mutants in the pcDNA-3Myc vector were generated by site-directed mutagenesis. The cDNAs of PIAS family genes were cloned into the pCMV-HA vector. All short hairpin RNA (shRNA) sequences were cloned into the pLKO.1 vector. SENP1 was inserted into the pEGFP-N3 vector. H2A was inserted into the pcDNA-3Flag vector. With a view to mimicking constitutive SUMOylation of RNF168, RNF168 was fused to SUMO3 in tandem by insertion mutagenesis, and then cloned into the pcDNA-3Myc and pEGFP-C1 vectors. All constructs were confirmed by DNA sequencing.

Cell culture

HEK293T, SW480, HeLa, MEF, NCM460, HT29, FHC, LOVO, and RKO cells were cultured in DMEM supplemented with 10% FBS (Gibco). HCT116 cells were maintained in RPMI1640 (Gibco). MEF SENP1 knockout cells and controlled wild-type cells were kind gifts from Jinke Cheng (Shanghai Jiao Tong University, Shanghai, China). The identities of all cell lines were authenticated by short tandem repeat analysis and verified by PCR to ensure the absence of Mycoplasma contamination.

Animal models

Experiments using mice were performed following the ‘Principles for the Utilization and Care of Vertebrate Animals’ and the ‘Guide for the Care and Use of Laboratory Animals’. Animal studies were approved by the Institutional Animal Care and Use Committee (IACUC) of the Center for Experimental Animal Research (China) and Peking University Laboratory Animal Center (IACUC no. LSC-ZhengX-2–1). Fifteen female BALB/c nude mice (4 weeks of age) were bought from Beijing Vital River Laboratory Animal Technology and randomly grouped into five categories. A total of 4 × 106 HCT116 cells were injected subcutaneously into the armpits, with 2 × 106 cells on each side. Tumor size was measured every 2 days from day 7 using a caliper, and the volume of each tumor was calculated using the following formula: volume = length × width2/2. Mice were all sacrificed on day 12 postinjection.

His-SUMO pulldown

HEK293T cells were transfected with His-SUMO and the indicated plasmids for 48 hours, then harvested and lysed in His-pulldown lysis buffer. The lysate was incubated with 40 μL Ni2+ beads for 4 hours at room temperature. After washing 4 times with different wash buffers, the beads were denatured in 2× SDS loading buffer, and the level of SUMOylation was assessed using the indicated antibodies.

Chromatin extraction

The chromatin extraction assays were conducted as previously described (33). Cells were collected and lysed in chromatin extraction buffer A (10 mmol/L PIPES pH 6.8, 100 mmol/L NaCl, 300 mmol/L sucrose, 3 mmol/L MgCl2, 1 mmol/L EGTA, 0.2% Triton X-100) on ice for 30 minutes and then centrifuged at 3,000 × g, 4°C for 5 minutes. The supernatant was collected as soluble element, and cell pellets were lysed in chromatin extraction buffer B (3 mmol/L EDTA, 0.2 mmol/L EGTA, 1 mmol/L DTT) and centrifuged at 3,000 × g, 4°C for 5 minutes. The supernatant was completely removed, and the sediment was resuspended in buffer C (50 mmol/L Tris pH 8.0, 150 mmol/L NaCl, 1 mmol/L EDTA, 0.1% SDS, 1% Triton X-100) as the chromatin element. Both elements were denatured in 2× SDS loading buffer and tested by immunoblotting.

Immunofluorescence microscopy

Cells were transfected with the indicated plasmids using PEI reagent. At 48 hours post-transfection, cells were collected and fixed in precooled methanol for 10 minutes at −20°C according to a previously described procedure (33). Nuclear DNA was stained with 4′,6-diamidino-2-phenylindole (DAPI; 1 g/mL). Images were obtained with a confocal microscope (Zeiss LSM-710 NLO & DuoScan, Germany; Zeiss LSM-800 NLO & DuoScan; Zeiss LSM-880 NLO & DuoScan; DeltaVision OMX SR) using the 63× oil objective lens. Quantitation was performed using the Imaris software (Bitplane).

Laser microirradiation

Laser microirradiation was carried out following previously described procedures (34). HeLa cells were cultured in glass-bottomed Petri dishes and irradiated with an ultraviolet laser (365-nm pulsed nitrogen laser, 16 Hz pulse, 60% output). Images were taken using a Nikon A1 confocal imaging system, and time-lapse images were captured at the indicated times. ImageJ software were used to calculate the signal intensity of each cell.

Protein purification

Expression of recombinant His-tagged proteins in E. coli strain BL21 or Rosetta was induced with 0.5 mmol/L isopropyl-β-D-thiogalactopyranoside when the bacterial cultures reached an OD600 value of 0.6. After expressing at 16°C for 20 hours, fusion proteins were purified on an Ni-Sepharose column according to the manufacturer's instructions (GE HealthCare).

Coimmunoprecipitation

Cells were transfected with the indicated plasmids using PEI reagent. At 48 hours post-transfection, cells were collected and washed with PBS buffer, lysed in modified RIPA buffer (50 mmol/L Tris-HCl pH 7.4, 150 mmol/L NaCl, 1 mmol/L EDTA, protease inhibitors), and sonicated using a JY92-II sonicator (on for 5 seconds, off for 5 seconds; 10 cycles). After pre-clearing with Protein G-Sepharose beads for 1 hour, the supernatant was incubated with the indicated antibodies or control IgG antibodies overnight, and then gently mixed with 50 μL Protein G Sepharose beads for a further 4 hours. Finally, the samples were denatured, separated by SDS-PAGE, and transferred to nitrocellulose membranes (GE HealthCare). Membranes were blocked by 5% skim milk and then probed with specific primary and secondary antibodies. The protein signals were detected and quantitated using the Odyssey Infrared Imaging System and Odyssey V3.0 software (LI-COR Bio-sciences).

Immunoprecipitation mass spectrometry

HEK293T cells were transfected with Flag-RNF168 and Flag-pcDNA plasmids for 48 hours, and then lysed in RIPA buffer, sonicated using a JY92-II sonicator, and pre-cleared with Protein G Sepharose beads. The supernatants were first incubated overnight with an anti-Flag antibody and then with 50 μL Protein G Sepharose beads for a further 4 hours. Following centrifugation at 3,000 rpm, 4°C for 5 minutes, the supernatants were completely removed and Flag-peptide was added to competitively bind to the Protein G Sepharose beads. RNF168 and its associated proteins were released, precipitated using TCA, and subjected to mass spectrometry (MS) analysis. The proteomics data have been deposited in the ProteomeXchange Consortium via the PRIDE partner repository, with the dataset identifier PXD036815.

Generation of RNF168−/− and SENP1−/− cells

Two guide RNAs were designed against sequences conserved across the SENP1 or RNF168 locus using the Brunello human CRISPR-Cas9 screening library. The following guides were selected:

SENP1-gRNA1: aagatagggaatatacactgtgg

SENP1-gRNA2: gtgtaaaggaaaatgtgtggtgg

RNF168-gRNA1: cagcgtgtggttacacgggaggg

RNF168-gRNA2: agaaattctctcgtcaacgtgg

In brief, lentiviral vector particles were generated by transient transfection of HEK293T cells with gRNA (cloned into the LentiCRISPR V2 vector) and the packaging plasmids (PMD2.G and PSPAX2) using Lipofectamine 3000 (Invitrogen). Lentiviral vector supernatants were collected on days 2 and 3 post-transfection and used for knockout in HEK293T/HCT116 cells. SENP1- and RNF168-knockout clones were grown from single cells obtained by flow cytometry, and the knockout efficiency of each positive clone was verified by immunoblotting.

Generation of SENP1- and RNF168-knockdown cells

Seven shRNA sequences were synthesized and cloned into the pLKO.1 vector:

#1: ccggccgaaagacctcaagtggattctcgagaatccacttgaggtctttcggtttttg

#2: ccggtactggaactaagacatcgagctcgagctcgatgtcttagttccagtatttttg

#3: ccggaagacatcgagacaggtgtaaaggactcgagtcctttacacctgtctcgatgtctttttttg

#4: ccggctcgatgtcttagttccagtactcgagtactggaactaagacatcgagttttt

#5: ccggcgagaaagattgcgccagattctcgagaatctggcgcaatctttctcgttttt

#6: ccggtgaccattacacgcaaagatactcgagtatctttgcgtgtaatggtcattttt

shRNA168: ccggcgacactttctccacagatatctcgagatatctgtggagaaagtgtcgtttttg

Lentiviral vector particles were generated by transient transfection of HEK293T cells with the lentiviral packaging components (pLP1, pLP2, pVSVG). Supernatants were collected on days 2 and 3 and used to infect target cells. Knockdown cells were screened using puromycin.

Neutral comet assay

Neutral comet assays were performed to detect cell genome stability using the CometAssay Kit (Trevigen). Cells were harvested and resuspended in ice-cold PBS buffer. The neutral single-cell agarose gel was placed in specific running buffer to let DNA helix uncoiling and the level of DNA damage was visualized by electrophoresis under alkaline conditions. Images were obtained using Olympus Ix73 fluorescence microscope with a 10× objective lens. Quantitation was conducted using the Casp Lab software v1.2.2 (University of Wroclaw, Wroclaw, Poland). Approximately 80 cells were analyzed in each group using the Comet Assay IV software (Perceptive Instruments).

NHEJ and HR assays

The DNA repair efficiency assays were conducted as previously described (34). For the NHEJ assays, cells were transfected with the linearized pEGFP-Pem1-Ad2 plasmids and DsRed plasmid for 48 hours, and then harvested and washed with 1× PBS buffer. For the HR assays, cells were cotransfected with DR-GFP, an I-SceI expression vector, and a DsRed plasmid, and then harvested at 48 hours post-transfection and washed with 1× PBS buffer.

The intensity of green (EGFP) and red (DsRed) fluorescence signals was measured by FACS using a LSRFortessa or FACSVerse instrument (BD Biosciences). The percentage of EGFP/DsRed double-positive cells to DsRed-positive cells was taken as the NHEJ or HR repair efficiency. The results were normalized to those of the RNF168 wild-type cells. All the samples were analyzed using the FlowJo software to determine the number of GFP-positive cells relative to that of cells expressing DsRed.

IHC

Paraffin-embedded human colon normal and cancer samples were kindly provided by Xiuqin Zhang (Peking University). Slides were stained with rabbit anti-SENP1 (1:100) at 4°C overnight and then with secondary antibodies. Visualization was carried out using DAB (3′3′-diaminobenzidine tetrahydrochloride) as the substrate. The intensity of SENP1 was classified as follows: 1, no staining; 2, weak reactivity; 3, moderate reactivity; 4, strong reactivity; 5, very strong reactivity. A Leica DM IRE2 microscope was used to obtain the images.

Apoptosis assay

The apoptosis assay was performed using the Annexin V-FITC Apoptosis Detection kit (Beyotime Biotechnology). Cells were collected and stained with Annexin V and propidium iodide (PI), and the apoptosis rate was detected using a LSRFortessa or FACSVerse instrument (BD Biosciences). The Q3 quadrant represented Annexin V–positive cells, which are in the early stage of apoptosis. The Q2 quadrant showed cells positive for both Annexin V and PI, which are in the late stage of apoptosis. A total of 1 × 104 cells were counted for each group.

Cell invasion assay

Cells were seeded onto Transwell inserts (Corning) containing Matrigel-coated (BD Biosciences) porous membranes and incubated for 24 hours. Using cotton swabs to remove the cells remaining on the insert, while using methanol to fix cells adhering to the lower side of the membrane. Then crystal violet staining was conducted, and observed by microscopy.

Duolink proximity ligation assay

A Duolink In Situ PLA Kit (DUO92101, Sigma-Aldrich) was used to perform the proximity ligation assay (PLA) assay according to the manufacturer's instructions as previously described (34). In brief, cells were fixed using 4% paraformaldehyde for 20 minutes at room temperature, permeabilized with 0.5% Triton X-100, and blocked with 1× blocking solution buffer. Subsequently, cells were incubated at 4°C overnight with primary antibodies targeting RNF168 and SUMO3 and then with PLA PLUS and MINUS PLA probes at 37°C for 1 hour. After washing three times, the ligation-ligase solution was added, and cells were incubated at 37°C for 30 minutes, followed by incubating with amplification polymerase solution at 37°C in the dark for 100 minutes, after which, cells were stained with Mounting Medium containing DAPI. Fluorescence images were obtained under a confocal laser scanning microscope (Zeiss LSM 880) using a 63× oil objective lens.

In vitro SUMOylation assay

HEK293T cells were transfected with HA-tagged SUMOylation E3 ligase PIAS1, which was subsequently enriched using an anti-HA antibody. The in vitro SUMOylation assay was conducted following previously described procedures (34). Briefly, His-tagged wild-type or mutant RNF168 proteins were diluted in in vitro SUMOylation buffer, immunoprecipitated with Mg-ATP, SUMO E1, SUMO E2, SUMO3, and Protein G Sepharose bound to E3 ligase. The mixture was incubated for 2 hours at 37°C following the manufacturer's manual from Abcam.

Fluorescence recovery after photobleaching assay

Fluorescence recovery after photobleaching (FRAP) experiments were performed using a Zeiss LSM 880 microscope with a 63× oil immersion objective. EGFP-RNF168-SUMO3 bodies (in vivo) were bleached until the fluorescence intensity was reduced to 30% of the initial value. After photobleaching, recovery time-lapse images were captured for the indicated times. Following a previously described method (29), the pre-bleaching fluorescence intensity was set to 100% and the fluorescence intensity at each time point was normalized (It) and used to calculate the fluorescence recovery according to the following formula: FR(t) = It/Ipre-bleaching. Image J was used for quantitation, and GraphPad Prism was used to plot and analyze FRAP data.

In vitro droplet formation assay

Proteins at the indicated concentration were incubated in droplet formation buffer (150 mmol/L NaCl, 50 mmol/L Tris-HCl pH 7.5, 10% glycerol, 1 mmol/L DTT, 10% PEG8000) for 2 minutes as reported previously (16). The in vitro droplet formation assay was carried out on glass-bottomed Petri dishes and observed using a Zeiss LSM 880 microscope with a 63× oil immersion objective.

Three-dimensional rendering and sphericity measurement

Z-Stack scanning images of HeLa cells transiently expressing GFP-RNF168-SUMO3 were acquired using a confocal microscope (Zeiss LSM 880) with a 63× oil immersion lens. Three-dimensional (3D) rendering was conducted using the Imaris software and calibrated with spherical beads. Sphericity was calculated using the Imaris software according to the following formula:

Acid chromatin fractionation

Cells transiently expressing the indicated plasmids were lysed in NP-40 lysis buffer, incubated at 4°C for 30 minutes, and centrifuged at 18,000 × g for a further 30 minutes. The supernatant was removed and neutralized with 1 mol/L Tris-HCl (pH 8.0) as the soluble fraction. The pellet was washed with ddH2O and resuspended in 0.2 mol/L HCl as the nuclear fraction. Samples were denatured in 6× SDS loading buffer, subjected to SDS-PAGE, and detected by immunoblotting.

Fission and fusion live-cell imaging

HeLa cells were cultured on glass-bottomed Petri dishes and transfected with the indicated plasmids. The Micropoint system (Andor) was directly coupled to the epifluorescence path of the Nikon A1 confocal imaging system, and time-lapse images were captured at the indicated times.

Metaphase spread assay

MEF cells were treated with 1 μmol/L nocodazole for 12 hours to arrest mitosis at G2–M phase. Cells were collected and resuspended in 0.06 mol/L KCl, incubated at 37°C for 30 minutes, and then centrifuged at 1,000 rpm for 5 minutes. The supernatant was removed and the pellet was fixed in freshly prepared fixation buffer (methyl alcohol: glacial acetic acid = 3:1) for 10 minutes. The cell suspension was extracted, dropped onto a slide, and dried at 60°C. Images were taken using a Zeiss LSM 880 microscope with a 63× oil immersion objective.

Cell proliferation assay

A CCK-8 kit (Dojindo Molecular Technologies Inc) was used to perform the cell proliferation assay according to the manufacturer's protocol. Cell suspensions (100 μL/well) were seeded onto 96-well plates and allowed to adhere. During the process of cell growth, 10 μL CCK-8 solution was added to each well at different time points and the absorbance was measured at 450 nm using a microplate reader.

Wound healing assay

Cells were cultured to 100% confluence and a physical gap was then created using a p10 tip. The process of cell migration into the gap was monitored by photography at the indicated times.

Drug resistance assay

Cells were cultured on 6-well plates at the indicated density and treated with different concentrations of chemotherapeutic drugs. After culture for 14 days, colony formation was observed by crystal violet staining and quantitated using the Image-J software.

Bioinformatics analysis

Expression variance and survival rate curves were analyzed and constructed using the UALCAN portal and GEPIA2 website, respectively, with COAD-related data screened out from the database. SUMOylation sites were predicted by the SUMOplot Analysis Program, GPS-SUMO, JASSA v4, and the Ron Hay lab group websites. Sequence alignment was performed using the ClustalW website and depicted using the ENDscript/ESPript website.

Statistical analysis

Statistical results were acquired from at least three independent biological replicates. Figure legends explained each detailed n values. Almost all data included are presented as the mean ± SEM, unless otherwise specified. GraphPad Prism 8.0 software was employed to obtain P values using Student t test (two-tailed), Mann–Whitney test (for two group comparisons), or one-way ANOVA. *, P < 0.05; **, P < 0.01; ***, P < 0.001.

Data and materials availability

The data supporting this study are available from the corresponding author upon reasonable request. The MS proteomics data have been deposited in the ProteomeXchange Consortium, with the dataset identifier PXD036815. Source data are provided with this paper. The data analyzed in this study were obtained from The Cancer Genome Atlas (TCGA) database (Colon Adenocarcinoma TCGA data) at https://ualcan.path.uab.edu/index.html.

RNF168 is predominantly SUMOylated at Lys210

SUMOylation plays important roles in the regulation of MDC1, 53BP1, and BRCA1 functions in the DDR pathway (10, 13). A previous study suggested that RNF168 is a substrate of SUMOylation (35); however, the modification site and functional significance of RNF168 SUMOylation remain to be elucidated. To further characterize RNF168 SUMOylation, a His-SUMO pulldown assay was performed to evaluate the type of SUMOylation, which revealed that RNF168 was modified by both SUMO1 and SUMO3 (Fig. 1A). Because SUMO2/3 proteins are more abundant and responsible for environmental stress than SUMO1 (36, 37), we chose SUMO3 for further study. The SUMOylation of RNF168 by SUMO3 was also confirmed using denaturing immunoprecipitation (IP) assays (Supplementary Fig. S1A). More importantly, the SUMOylation of endogenous RNF168 was validated in HEK293T, HCT116, and SW480 cells using IP assays (Fig. 1B). Using His-SUMO pulldown, we further confirmed that RNF168 is mono-SUMOylated, with Myc-tagged chimeric plasmids containing one SUMO3, a homodimer of SUMO3, or a heterodimer of SUMO3 and SUMO1, and endogenous RNF168 as the controls (Supplementary Fig. S1B).

Figure 1.

RNF168 is predominantly SUMOylated at Lys210. A, RNF168 was SUMOylated by both SUMO1 and SUMO3. HEK293T cells coexpressing HA-RNF168 and His-tagged SUMO1 or SUMO3 were collected at 48 hours post-transfection and incubated with Ni-NTA to determine the SUMOylation type using a His-SUMO pulldown assay. B, Endogenous RNF168 SUMOylation was confirmed using IP assays in HEK293T, HCT116, and SW480 cells with the indicated antibodies. C, The effect of the PIAS family on RNF168 SUMOylation in HEK293T cells was determined using His-SUMO pulldown assay. D, Co-IP assays were performed to examine the interaction between RNF168 and PIAS family members including PIAS1, PIAS3, and PIASy in HEK293T cells transfected with the indicated plasmids. E, Lys210 was the major SUMOylation site for RNF168. HEK293T cells transfected with Myc-RNF168 wild-type (WT) and its K-to-R mutants were subjected to IP assays using the indicated antibodies. F,In vitro SUMOylation assay was performed using RNF168 WT and RNF168 K210R proteins purified from E. coli, and HA-tagged PIAS1 proteins were immunoprecipitated from HA-PIAS1-transfected HEK293T cells.

Figure 1.

RNF168 is predominantly SUMOylated at Lys210. A, RNF168 was SUMOylated by both SUMO1 and SUMO3. HEK293T cells coexpressing HA-RNF168 and His-tagged SUMO1 or SUMO3 were collected at 48 hours post-transfection and incubated with Ni-NTA to determine the SUMOylation type using a His-SUMO pulldown assay. B, Endogenous RNF168 SUMOylation was confirmed using IP assays in HEK293T, HCT116, and SW480 cells with the indicated antibodies. C, The effect of the PIAS family on RNF168 SUMOylation in HEK293T cells was determined using His-SUMO pulldown assay. D, Co-IP assays were performed to examine the interaction between RNF168 and PIAS family members including PIAS1, PIAS3, and PIASy in HEK293T cells transfected with the indicated plasmids. E, Lys210 was the major SUMOylation site for RNF168. HEK293T cells transfected with Myc-RNF168 wild-type (WT) and its K-to-R mutants were subjected to IP assays using the indicated antibodies. F,In vitro SUMOylation assay was performed using RNF168 WT and RNF168 K210R proteins purified from E. coli, and HA-tagged PIAS1 proteins were immunoprecipitated from HA-PIAS1-transfected HEK293T cells.

Close modal

His-SUMO pulldown screening of the SUMO E3 ligases of RNF168 showed that PIAS1, PIAS3, and PIASy promoted RNF168 SUMOylation (Fig. 1C), and coimmunoprecipitation (co-IP) confirmed that these E3 ligases interacted with RNF168 (Fig. 1D). Among them, PIAS1 displayed the strongest effect; therefore, we used this E3 ligase in subsequent experiments. Furthermore, the in vitro SUMOylation assay demonstrated that RNF168 was SUMOylated by SUMO3 in the presence of PIAS1 (Supplementary Fig. S1C–S1E).

Lysine residues undergoing SUMOylation are typically found within the SUMO modification consensus motif ψKXE, in which ψ is a large hydrophobic residue and X is any residue. Prediction using SUMOplot (https://www.abcepta.com/sumoplot) and GPS-SUMO (http://sumosp.biocuckoo.org/) revealed three lysine residues of RNF168, K173, K203, and K210, as potential SUMOylation sites. Accordingly, we constructed three RNF168 mutants, K173R, K203R, and K210R, and examined their effects on RNF168 SUMOylation using IP assays. In comparison with wild-type RNF168, only the K210R mutation notably reduced the SUMOylation level of RNF168 (Fig. 1E). Similarly, an in vitro SUMOylation assay using purified proteins (Supplementary Fig. S1E) showed that the SUMOylation level of the RNF168 K210R mutant is markedly decreased as compared with that of wild-type RNF168 (Fig. 1F). Furthermore, sequence alignment revealed that K210 is conserved among diverse species (Supplementary Fig. S1F). Collectively, these results demonstrate that RNF168 is mono-SUMOylated predominantly at K210.

SUMOylated RNF168 undergoes LLPS

To explore the impact of SUMOylation on RNF168, following previous studies that have used a chimera to simulate stable modifications (38–40), we constructed a GFP-tagged chimera RNF168-SUMO3 to mimic the SUMOylated form of RNF168 and conducted immunofluorescence (IF) assays using HeLa cells expressing different plasmids (Fig. 2A). Strikingly, microscopic visualization of fluorescently tagged RNF168-SUMO3 chimeric proteins revealed that SUMOylation induced RNF168 condensation, while the SUMOylation-defective K210R mutant did not yield puncta (Fig. 2A; Supplementary Fig. S2A). Because intermolecular interactions can be weakened or enhanced by the modification of key residues (41), we hypothesized that SUMOylation may play a key role in regulating the formation of RNF168 nuclear condensates through LLPS. To test this hypothesis, we characterized the puncta using 3D Z-stack scanning and 3D shape rendering and calculated the volume and surface area using the sphericity formula. The results showed that the puncta of SUMOylated RNF168 had a spherical shape, with a diameter of approximately 1.86 μm (Fig. 2B). Moreover, we conducted a FRAP assay in RNF168-SUMO3-expressing HeLa cells to investigate whether SUMOylation also affects the dynamics of RNF168 puncta in vivo. The fluorescence intensity of the SUMOylation-mimetic RNF168-SUMO3 protein was almost completely recovered within 1 minute post-bleaching, reflecting that SUMOylated RNF168 underwent rapid exchange with the surrounding environment (Fig. 2C and D). Subsequent tracing of the fusion and fission processes by live cell imaging revealed the occurrence of liquid-like properties (Fig. 2E; Supplementary Video 1, 2).

Figure 2.

SUMOylated RNF168 undergoes LLPS. A, SUMOylation-mimetic GFP-RNF168-SUMO plasmid was constructed (top), transfected into HeLa cells, and subjected to IF analyses. Representative images are shown. Scale bar, 2 μm. B, GFP-RNF168-SUMO3 formed puncta in HeLa cells. Three-dimensional Z-stack scanning of HeLa cells transfected with GFP-RNF168-SUMO3. An enlargement of the boxed punctum is shown in the inset. Rendered 3D shapes of a punctum with panels showing the XY, XZ, and YZ planes. A plot showing the sphericity of puncta (n = 169) calculated using the sphericity formula. Error bar, ± SD. Scale bar, 2 μm. C, Fluorescence intensity recovery after photobleaching (FRAP) of a GFP-RNF168-SUMO3 punctum. Representative images of the FRAP analysis of GFP-RNF168-SUMO3 are shown. The indicated region was chosen randomly under live-cell observation and photobleached to 30% of the original intensity. D, Quantitation of FRAP results of GFP-RNF168-SUMO3 puncta (n = 15). The FRAP recovery curve generated by averaging the signals of 15 droplets per group with similar sizes after photobleaching. The relative fluorescence intensity of each droplet prior to photobleaching was set to 100%. Time 0 refers to the time point right at the start of photobleaching. Data are presented as the mean ± SEM. E, GFP-RNF168-SUMO3 puncta underwent fusion and fission. Representative images of the fission and fusion observations of GFP-RNF168-SUMO3 puncta executed by live-cell tracking at the indicated time. F, Workflow of the in vitro droplet formation assay used for G and H. G and H, SUMOylation induced RNF168 phase separation in vitro. In vitro LLPS assays were performed using the indicated proteins following the procedure in F. The protein concentration used was 400 nmol/L. Scale bar, 2 μm.

Figure 2.

SUMOylated RNF168 undergoes LLPS. A, SUMOylation-mimetic GFP-RNF168-SUMO plasmid was constructed (top), transfected into HeLa cells, and subjected to IF analyses. Representative images are shown. Scale bar, 2 μm. B, GFP-RNF168-SUMO3 formed puncta in HeLa cells. Three-dimensional Z-stack scanning of HeLa cells transfected with GFP-RNF168-SUMO3. An enlargement of the boxed punctum is shown in the inset. Rendered 3D shapes of a punctum with panels showing the XY, XZ, and YZ planes. A plot showing the sphericity of puncta (n = 169) calculated using the sphericity formula. Error bar, ± SD. Scale bar, 2 μm. C, Fluorescence intensity recovery after photobleaching (FRAP) of a GFP-RNF168-SUMO3 punctum. Representative images of the FRAP analysis of GFP-RNF168-SUMO3 are shown. The indicated region was chosen randomly under live-cell observation and photobleached to 30% of the original intensity. D, Quantitation of FRAP results of GFP-RNF168-SUMO3 puncta (n = 15). The FRAP recovery curve generated by averaging the signals of 15 droplets per group with similar sizes after photobleaching. The relative fluorescence intensity of each droplet prior to photobleaching was set to 100%. Time 0 refers to the time point right at the start of photobleaching. Data are presented as the mean ± SEM. E, GFP-RNF168-SUMO3 puncta underwent fusion and fission. Representative images of the fission and fusion observations of GFP-RNF168-SUMO3 puncta executed by live-cell tracking at the indicated time. F, Workflow of the in vitro droplet formation assay used for G and H. G and H, SUMOylation induced RNF168 phase separation in vitro. In vitro LLPS assays were performed using the indicated proteins following the procedure in F. The protein concentration used was 400 nmol/L. Scale bar, 2 μm.

Close modal

To confirm the LLPS of SUMOylated RNF168, we performed an in vitro droplet formation assay using purified SUMOylated and unSUMOylated RNF168 proteins according to the procedure shown in Fig. 2F. The droplets were only observed in the presence of SUMOylated RNF168 and not within the RNF168 and SUMO3 protein mixture (Fig. 2G). Consistent with the observations in Fig. 2A, the droplet number of the RNF168 K210R SUMOylation-deficient mutant was largely decreased relative to that of the SUMOylated wild-type RNF168 (Fig. 2H), suggesting that SUMOylation of RNF168 is essential for LLPS formation. In addition, because proteins that undergo LLPS frequently contain intrinsically disordered regions (IDR; ref. 27), we employed AlphaFold and the IUPred2 website to identify a long IDR sequence in RNF168 that includes the SUMOylation site Lys210 (Supplementary Fig. S2B and S2C). Taken together, these results indicate that SUMOylated RNF168 undergoes LLPS.

SUMOylation-induced LLPS inhibits the function of RNF168 in the DDR pathway

RNF168 is an early response protein in the DSB repair pathway, and RNF168-catalyzed H2A K13/15 ubiquitination (6) is critical for 53BP1 accumulation (7). To evaluate the effect of SUMOylation on RNF168 function, laser microirradiation was used to generate localized DNA damage in HeLa cells expressing GFP-RNF168 or GFP-RNF168-SUMO3, and the recruitment of RNF168 to the damaged site was monitored. Notably, wild-type RNF168 was quickly recruited to the damaged site, whereas the SUMOylated form was not (Fig. 3A). Next, we constructed RNF168−/− HEK293T and HCT116 cells using the CRISPR/Cas9 system (Supplementary Fig. S3A and S3B) and re-expressed wild-type RNF168, RNF168-SUMO3, and RNF168 K210R to evaluate their interactions with H2A in response to etoposide treatment-induced DNA damage. As expected, relative to wild-type RNF168, the SUMOylated RNF168 showed a weaker interaction with H2A, while the SUMOylation-deficient mutant K210R showed a stronger H2A interaction (Fig. 3B and C; Supplementary Fig. S3C and S3D). Consistently, the ubiquitin pulldown and acid chromatin fractionation assays detected no H2A ubiquitination in cells without etoposide treatment, and the level of H2A ubiquitination decreased in cells expressing RNF168-SUMO3 and increased in those expressing the K210R mutant in response to DNA damage (Fig. 3D and E). These results indicate that SUMOylation inhibits RNF168 function in the DDR by reducing its recruitment to the damaged site and attenuating its function as an E3 ubiquitin ligase of H2A.

Figure 3.

SUMOylation-induced LLPS inhibits the function of RNF168 in the DDR pathway. A, RNF168-SUMO3 was not recruited to the DNA damage site. HeLa cells transfected with GFP-RNF168 or GFP-RNF168-SUMO3 were subjected to laser microirradiation, and the dynamic accumulation of GFP-tagged protein at DSB sites was monitored using fluorescent microscopy at different time points. White line, positions for laser microirradiation. B, The SUMOylation deficiency mutant K210R promotes the interaction between RNF168 and H2A in response to DNA damage. HCT116 RNF168−/− cells expressing the indicated plasmids were treated with or without 40 μmol/L etoposide for 2 hours and then subjected to a co-IP assay using the indicated antibodies. The RNF168-H2A interaction was quantitated using ImageJ software. C, The interaction between RNF168 and H2A is enhanced in the RNF168 K210R mutant following etoposide treatment. HCT116 RNF168 knockout (KO) cells were transfected with HA-RNF168 or HA-RNF168 K210R in the presence or absence of Myc-H2A. Following treatment with or without 40 μmol/L etoposide for 2 hours, cells were subjected to a co-IP assay using the indicated antibodies. D, SUMOylation of RNF168 attenuated its effect on H2A K15 ubiquitination. HEK293T RNF168 KO cells expressing Myc-RNF168 or Myc-RNF168-SUMO3 were subjected to a ubiquitin pulldown assay to evaluate H2A K15 ubiquitination using a specific antibody with or without etoposide treatment. E, H2A K15 ubiquitination was promoted in the RNF168 K210R mutant following DNA damage. HCT116 RNF168 KO cells expressing HA-RNF168 or HA-RNF168 K210R were treated with or without 40 μmol/L etoposide for 2 hours and then subjected to an acid chromatin fractionation assay. The level of endogenous H2A ubiquitination at lysine 15 was detected using anti-H2AK15ub antibody. F, Colocalization of BRCA1 and 53BP1 with the SUMOylated RNF168-induced condensates was assessed using IF in HeLa cells transfected with GFP-RNF168-SUMO3 using antibodies against 53BP1 or BRCA1 (red). Nuclei were stained with DAPI (blue). Fields were observed using confocal microscopy. Scale bar, 2 μm. G, An IF assay was performed in EGFP-RNF168-SUMO3-expressed HeLa cells using antibodies against 53BP1. Fields were observed by OMX SR SIM microscopy. Scale bar, 1 μm. H, The effect of RNF168 SUMOylation on NHEJ repair efficiency. HCT116 RNF168 knockdown cells were transfected with Myc-RNF168 wild-type (WT), Myc-RNF168 K210R or Myc-RNF168-SUMO3 plasmids; NHEJ efficiency was determined using the Pem1-Ad2-EGFP reporter assay. WT HCT116 cells were used as control and set to 1.0. Data are presented as the mean ± SD from three independent experiments. Statistical analysis was performed using a Student t test. I, SUMOylation inhibited the recruitment of 53BP1 to damaged site on chromatin. HEK293T RNF168 KO cells transfected with Myc-RNF168 or Myc-RNF168-SUMO3 were treated with or without 40 μmol/L etoposide for 2 hours, after which, chromatin was isolated and the recruitment of 53BP1 to chromatin was analyzed using immunoblotting. J, HeLa cells transfected with EGFP-RNF168 or EGFP-RNF168-SUMO3 and treated with 40 μmol/L etoposide for 2 hours. An IF assay was performed using antibodies against 53BP1 (red) and γH2AX (CY5). Nuclei were stained with DAPI (blue). Scale bar, 2 μm. Approximately 12 cells were selected in each group, and the colocalization Pearson correlation coefficient was calculated using Zen. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001; ns, nonsignificant.

Figure 3.

SUMOylation-induced LLPS inhibits the function of RNF168 in the DDR pathway. A, RNF168-SUMO3 was not recruited to the DNA damage site. HeLa cells transfected with GFP-RNF168 or GFP-RNF168-SUMO3 were subjected to laser microirradiation, and the dynamic accumulation of GFP-tagged protein at DSB sites was monitored using fluorescent microscopy at different time points. White line, positions for laser microirradiation. B, The SUMOylation deficiency mutant K210R promotes the interaction between RNF168 and H2A in response to DNA damage. HCT116 RNF168−/− cells expressing the indicated plasmids were treated with or without 40 μmol/L etoposide for 2 hours and then subjected to a co-IP assay using the indicated antibodies. The RNF168-H2A interaction was quantitated using ImageJ software. C, The interaction between RNF168 and H2A is enhanced in the RNF168 K210R mutant following etoposide treatment. HCT116 RNF168 knockout (KO) cells were transfected with HA-RNF168 or HA-RNF168 K210R in the presence or absence of Myc-H2A. Following treatment with or without 40 μmol/L etoposide for 2 hours, cells were subjected to a co-IP assay using the indicated antibodies. D, SUMOylation of RNF168 attenuated its effect on H2A K15 ubiquitination. HEK293T RNF168 KO cells expressing Myc-RNF168 or Myc-RNF168-SUMO3 were subjected to a ubiquitin pulldown assay to evaluate H2A K15 ubiquitination using a specific antibody with or without etoposide treatment. E, H2A K15 ubiquitination was promoted in the RNF168 K210R mutant following DNA damage. HCT116 RNF168 KO cells expressing HA-RNF168 or HA-RNF168 K210R were treated with or without 40 μmol/L etoposide for 2 hours and then subjected to an acid chromatin fractionation assay. The level of endogenous H2A ubiquitination at lysine 15 was detected using anti-H2AK15ub antibody. F, Colocalization of BRCA1 and 53BP1 with the SUMOylated RNF168-induced condensates was assessed using IF in HeLa cells transfected with GFP-RNF168-SUMO3 using antibodies against 53BP1 or BRCA1 (red). Nuclei were stained with DAPI (blue). Fields were observed using confocal microscopy. Scale bar, 2 μm. G, An IF assay was performed in EGFP-RNF168-SUMO3-expressed HeLa cells using antibodies against 53BP1. Fields were observed by OMX SR SIM microscopy. Scale bar, 1 μm. H, The effect of RNF168 SUMOylation on NHEJ repair efficiency. HCT116 RNF168 knockdown cells were transfected with Myc-RNF168 wild-type (WT), Myc-RNF168 K210R or Myc-RNF168-SUMO3 plasmids; NHEJ efficiency was determined using the Pem1-Ad2-EGFP reporter assay. WT HCT116 cells were used as control and set to 1.0. Data are presented as the mean ± SD from three independent experiments. Statistical analysis was performed using a Student t test. I, SUMOylation inhibited the recruitment of 53BP1 to damaged site on chromatin. HEK293T RNF168 KO cells transfected with Myc-RNF168 or Myc-RNF168-SUMO3 were treated with or without 40 μmol/L etoposide for 2 hours, after which, chromatin was isolated and the recruitment of 53BP1 to chromatin was analyzed using immunoblotting. J, HeLa cells transfected with EGFP-RNF168 or EGFP-RNF168-SUMO3 and treated with 40 μmol/L etoposide for 2 hours. An IF assay was performed using antibodies against 53BP1 (red) and γH2AX (CY5). Nuclei were stained with DAPI (blue). Scale bar, 2 μm. Approximately 12 cells were selected in each group, and the colocalization Pearson correlation coefficient was calculated using Zen. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001; ns, nonsignificant.

Close modal

It is well known that 53BP1 and BRCA1 play pivotal roles in NHEJ and HR repair; therefore, we used IF to elucidate whether SUMOylation of RNF168 further affects the downstream repair proteins 53BP1 and BRCA1. Notably, endogenous 53BP1, but not BRCA1, colocalized with the RNF168-SUMO3 condensates (Fig. 3F; Supplementary Fig. S3E). In addition, super-resolution microscopy confirmed the colocalization of RNF168-SUMO3 with 53BP1 (Fig. 3G; Supplementary Fig. S3F). These observations suggest that SUMOylated RNF168 restrains 53BP1 within the phase-separated puncta. Moreover, we found that RNF168 knockdown HCT116 cells have a significantly reduced repair efficiency of NHEJ and HR. The NHEJ repair efficiency could be rescued by the reintroduction of wild-type RNF168 and was even promoted by the K210R mutant but not by RNF168-SUMO3 (Fig. 3H; Supplementary Fig. S3G), while the HR repair efficiency was similarly recovered by wild-type RNF168, the K210R mutant, and RNF168-SUMO3 (Supplementary Fig. S3H), suggesting that RNF168 SUMOylation has no significant impact on HR repair. In accordance with these data, the recruitment of 53BP1 to chromatin was notably reduced in etoposide-treated RNF168−/− HEK293T cells expressing the RNF168-SUMO3 chimera (Fig. 3I). Furthermore, we examined whether RNF168 SUMOylation affected the colocalization of 53BP1 with γH2AX (a damage marker) using IF assays, which revealed that SUMOylated RNF168-induced puncta are larger than the DNA damage-induced foci containing wild-type RNF168 (Supplementary Fig. S3I). In etoposide-treated RNF168-SUMO3-expressing cells, SUMOylation-induced nuclear condensates of RNF168 compartmentalized 53BP1, resulting in reduced colocalization of 53BP1 with γH2AX; however, in cells harboring RNF168, 53BP1 was not restrained in the RNF168 puncta and colocalized with γH2AX (Fig. 3J). Taken together, these data demonstrate that SUMOylation drives RNF168 phase separation, which reduces RNF168 recruitment to the damaged site, inhibits H2A ubiquitination, and sequesters 53BP1 in the droplets, ultimately downregulating NHEJ repair efficiency.

SENP1 is the deSUMOylating enzyme that regulates RNF168 SUMO modification

Because our data indicate that SUMOylation negatively regulates the function of RNF168 in the DDR, we subsequently explored the regulatory mechanism underlying RNF168 SUMOylation. Considering that SENPs are deSUMOylating enzymes that reverse SUMOylation and are critical for the regulation of many protein functions, we sought to identify the specific SENP responsible for the regulation of RNF168 SUMOylation. IP and subsequent MS were conducted, which identified SENP1 and SENP3 as potential partners of RNF168 (Fig. 4A). Co-IP assays demonstrated an association between endogenous and exogenous RNF168 and SENP1 (Fig. 4B; Supplementary Fig. S4A), while an interaction between RNF168 and SENP3 was not detected even following overexpression (Supplementary Fig. S4B). Furthermore, the His-SUMO pulldown assay showed that SENP1 clearly reduced RNF168 SUMOylation (Fig. 4C), while SENP1 knockdown in HEK293T, HCT116, and SW480 cells increased RNF168 SUMOylation (Fig. 4DF; Supplementary Fig. S4C and S4D). To clarify whether SENP1 decreases RNF168 SUMOylation in a deSUMOylase activity-dependent manner, we constructed a SENP1 C603A mutant in which the catalytic activity was abolished. Indeed, the level of RNF168 SUMOylation was substantially reduced by wild-type SENP1; however, the C603A mutant had significantly less effect (Fig. 4D). In addition, SENP1 attenuated the interaction between RNF168 and its E3 ligase PIAS1 (Fig. 4G). These findings indicate that the ability of SENP1 to reduce RNF168 SUMOylation relies not only on its catalytic activity but also on its ability to impede the interaction between RNF168 and PIAS1. Taken together, these data suggest that SENP1 is the specific enzyme responsible for RNF168 deSUMOylation (Fig. 4H).

Figure 4.

SENP1 is the deSUMOylation enzyme that regulates RNF168 SUMO modification. A, The interaction proteins of RNF168 detected by IP assays, followed by MS. B, Association between endogenous RNF168 and SENP1 in HEK293T cells was detected using co-IP assays with anti-IgG, anti-SENP1 antibody. C, SENP1 deSUMOylated RNF168. The effect of SENP1 on RNF168 SUMOylation in HEK293T cells was determined using His-SUMO pulldown assay. D, SENP1 downregulation of RNF168 SUMOylation partially depends on its catalytic activity. HEK293T cells were transfected with the indicated plasmids and then subjected to His-SUMO pulldown assay. E and F, Depletion of SENP1 enhanced RNF168 SUMOylation in HCT116 (E) and SW480 (F) cells. Cells transfected with the indicated plasmids were subjected to IP assays. G, SENP1 inhibited the interaction between RNF168 and its E3 ligase PIAS1. HEK293T cells transfected with the indicated plasmids were subjected to co-IP to detect the effects of SENP1 on the interaction between RNF168 and PIAS1. H, Regulation of RNF168 SUMOylation by the PIAS family and SENP1.

Figure 4.

SENP1 is the deSUMOylation enzyme that regulates RNF168 SUMO modification. A, The interaction proteins of RNF168 detected by IP assays, followed by MS. B, Association between endogenous RNF168 and SENP1 in HEK293T cells was detected using co-IP assays with anti-IgG, anti-SENP1 antibody. C, SENP1 deSUMOylated RNF168. The effect of SENP1 on RNF168 SUMOylation in HEK293T cells was determined using His-SUMO pulldown assay. D, SENP1 downregulation of RNF168 SUMOylation partially depends on its catalytic activity. HEK293T cells were transfected with the indicated plasmids and then subjected to His-SUMO pulldown assay. E and F, Depletion of SENP1 enhanced RNF168 SUMOylation in HCT116 (E) and SW480 (F) cells. Cells transfected with the indicated plasmids were subjected to IP assays. G, SENP1 inhibited the interaction between RNF168 and its E3 ligase PIAS1. HEK293T cells transfected with the indicated plasmids were subjected to co-IP to detect the effects of SENP1 on the interaction between RNF168 and PIAS1. H, Regulation of RNF168 SUMOylation by the PIAS family and SENP1.

Close modal

SENP1 is involved in the DDR by downregulating RNF168 SUMOylation and is essential for the maintenance of genomic integrity

SUMOylation is a reversible and dynamic PTM that plays critical roles in cellular processes. Considering that SENP1 specifically deSUMOylates RNF168 and that SUMOylation negatively regulates the damage repair function of RNF168, we investigated whether SENP1 responds to DNA damage to dynamically regulate the function of RNF168 in the DDR. We performed a laser microirradiation assay using EGFP-SENP1-expressing cells to examine whether SENP1 is recruited to the damaged site. Notably, SENP1 was rapidly recruited to the damaged sites (within 10 sec after laser treatment; Fig. 5A), and IF assays revealed that SENP1 formed foci in the nucleus in response to bleomycin, ionizing radiation, or etoposide treatment (Supplementary Fig. S5A).

Figure 5.

SENP1 is involved in the DDR by downregulating RNF168 SUMOylation and is essential for maintaining genomic integrity. A, SENP1 was recruited to the DNA damage site. HeLa cells transfected with EGFP-SENP1 were subjected to laser microirradiation, and the dynamic accumulation of EGFP-tagged protein at DSB sites was monitored using fluorescent microscopy at different time points. B, SENP1 was recruited to DNA damage sites in an ATM-dependent manner. HeLa cells expressing EGFP-SENP1 were treated with 10 μmol/L ATM inhibitor KU55933, 10 μmol/L DNA-PK inhibitor NU7026, 5 μmol/L ATR inhibitor VE821, or 10 μmol/L PARP inhibitor olaparib for 2 hours and then subjected to laser microirradiation assays. C, Association between endogenous RNF168 and SENP1 in HEK293T cells treated with or without 40 μmol/L etoposide was detected using co-IP assays with anti-IgG or anti-SENP1 antibody. D, Endogenous SUMOylated RNF168 decreased in HEK293T cells treated with 40 μmol/L etoposide. E and F, The LLPS of endogenous RNF168 in the nucleus of the wild-type (WT) and SENP1−/− HEK293T cells (E) and HCT116 cells (F) was tested by IF assay using specific antibodies against RNF168. Scale bar, 2 μm. G, The effect of SENP1 depletion on NHEJ repair efficiency was assessed in SENP1 knockdown HCT116 cells using the Pem1-Ad2-EGFP reporter NHEJ repair efficiency assay. WT HCT116 cells were used as a control. Data are expressed as the mean ± SD from three independent experiments. H, SENP1 knockdown impaired genomic stability. SENP1 WT and SENP1 knockdown HEK293T cells were subjected to the neutral comet assay. Approximately 80 cells were counted in each group. Graphs show the mean ± SEM of comet tails. I, The effect of SENP1 depletion on chromosome aberrations was visualized by performing metaphase spread assays. SENP1 WT and SENP1 knockout (KO) MEF cells were treated with nocodazole (1 μmol/L) for 12 hours and subjected to the metaphase spread assay. Chromosome aberrations such as breakage and fusion were analyzed in approximately 30 metaphase spreads for each genotype. Representative aberrations and the aberration percentage of SENP1 WT and SENP1 KO are shown. Scale bar, 5 μm. Graphs show the mean ± SEM of chromosomal breakage and fusion events. Statistical analysis was performed using a Student t test. **, P < 0.01; ***, P < 0.001; ****, P < 0.0001.

Figure 5.

SENP1 is involved in the DDR by downregulating RNF168 SUMOylation and is essential for maintaining genomic integrity. A, SENP1 was recruited to the DNA damage site. HeLa cells transfected with EGFP-SENP1 were subjected to laser microirradiation, and the dynamic accumulation of EGFP-tagged protein at DSB sites was monitored using fluorescent microscopy at different time points. B, SENP1 was recruited to DNA damage sites in an ATM-dependent manner. HeLa cells expressing EGFP-SENP1 were treated with 10 μmol/L ATM inhibitor KU55933, 10 μmol/L DNA-PK inhibitor NU7026, 5 μmol/L ATR inhibitor VE821, or 10 μmol/L PARP inhibitor olaparib for 2 hours and then subjected to laser microirradiation assays. C, Association between endogenous RNF168 and SENP1 in HEK293T cells treated with or without 40 μmol/L etoposide was detected using co-IP assays with anti-IgG or anti-SENP1 antibody. D, Endogenous SUMOylated RNF168 decreased in HEK293T cells treated with 40 μmol/L etoposide. E and F, The LLPS of endogenous RNF168 in the nucleus of the wild-type (WT) and SENP1−/− HEK293T cells (E) and HCT116 cells (F) was tested by IF assay using specific antibodies against RNF168. Scale bar, 2 μm. G, The effect of SENP1 depletion on NHEJ repair efficiency was assessed in SENP1 knockdown HCT116 cells using the Pem1-Ad2-EGFP reporter NHEJ repair efficiency assay. WT HCT116 cells were used as a control. Data are expressed as the mean ± SD from three independent experiments. H, SENP1 knockdown impaired genomic stability. SENP1 WT and SENP1 knockdown HEK293T cells were subjected to the neutral comet assay. Approximately 80 cells were counted in each group. Graphs show the mean ± SEM of comet tails. I, The effect of SENP1 depletion on chromosome aberrations was visualized by performing metaphase spread assays. SENP1 WT and SENP1 knockout (KO) MEF cells were treated with nocodazole (1 μmol/L) for 12 hours and subjected to the metaphase spread assay. Chromosome aberrations such as breakage and fusion were analyzed in approximately 30 metaphase spreads for each genotype. Representative aberrations and the aberration percentage of SENP1 WT and SENP1 KO are shown. Scale bar, 5 μm. Graphs show the mean ± SEM of chromosomal breakage and fusion events. Statistical analysis was performed using a Student t test. **, P < 0.01; ***, P < 0.001; ****, P < 0.0001.

Close modal

To identify the upstream regulator of SENP1 recruitment, we treated cells with inhibitors of ATM (KU55933), DNA-PK (NU7026), ATR (VE821), and PARP (olaparib) and detected SENP1 recruitment by laser microirradiation. The results showed that KU55933 treatment blocked the accumulation of SENP1, which indicates that ATM plays a critical role in the regulation of SENP1 recruitment (Fig. 5B). Because ATM is a kinase, we next explored whether SENP1 interacts with ATM and undergoes phosphorylation. Indeed, both the interaction between SENP1 and ATM and the phosphorylation level of SENP1 were markedly increased in cells treated with etoposide in comparison with untreated cells (Supplementary Fig. S5B), and the level of SENP1 phosphorylation reduced in cells treated with ATM inhibitor (KU55933; Supplementary Fig. S5C), suggesting that SENP1 is rapidly recruited to DNA damage sites in an ATM-dependent manner. Consistently, we found that etoposide treatment promoted the association between SENP1 and RNF168 (Fig. 5C), and the level of endogenous RNF168 SUMOylation decreased after etoposide treatment (Fig. 5D).

Because our results showed that SUMOylation promotes the LLPS of RNF168 and that SENP1 deSUMOylates RNF168, we compared the puncta of endogenous RNF168 in wild-type cells with those in SENP1−/− cells using IF to further characterize the physiological effect of SENP1 on the LLPS formation of endogenous RNF168. Significant endogenous RNF168 LLPS was observed in both HEK293T and HCT116 SENP1 knocked out but not wild-type cells (Fig. 5E and F). Consistently, knockdown of SENP1 reduced the NHEJ repair efficiency (Fig. 5G). Moreover, we assessed the effects of SENP1 depletion on genomic integrity by performing neutral comet and metaphase spread assays. Relative to wild-type SENP1, SENP1 knockdown or knockout induced longer comet tails and a higher frequency of chromosomal breakage and fusion events (Fig. 5H and I). These data suggest that SENP1 is involved in the DNA damage repair pathway by interacting with RNF168 and downregulating RNF168 SUMOylation, thereby playing an essential role in the maintenance of genomic integrity.

SENP1 depletion inhibits cell proliferation, migration, and invasion

Because our results indicate that SENP1 is critical for genomic stability maintenance, we next explored the regulatory functions of SENP1 in cancer cells. We examined the effect of SENP1 on cell growth by generating both SENP1 knockout and knockdown HCT116 cells and verifying SENP1 depletion (Fig. 6A). SENP1 knockdown HCT116 cells showed slower growth rates than those of wild-type cells (Fig. 6B). Furthermore, we investigated the effect of SENP1 depletion on the sensitivity of cancer cells to chemotherapy drugs using SENP1 wild-type and SENP1−/− HCT116 cells treated with etoposide. Knockout of SENP1 resulted in a higher apoptosis rate than that of wild-type cells (Fig. 6C). Consistent with these observations, relative to wild-type HCT116 cells, SENP1 knockdown inhibited the migration and invasion abilities of cancer cells (Fig. 6D and E). Together, these results indicate that SENP1 plays important roles in regulating cell proliferation and migration, and dysfunction of SENP1 renders cancer cells sensitive to DNA damaging agents.

Figure 6.

SENP1 depletion inhibits cell proliferation, migration, and invasion. A, Knockdown and knockout (KO) of SENP1 were confirmed by immunoblotting. B, Cell Counting Kit-8 (CCK8) cell proliferation assay was used to detect cell proliferation in SENP1 wild-type (WT) and SENP1 knockdown HCT116 cells. C, SENP1 depletion increased the apoptosis rate of HCT116 cells after 40 μmol/L etoposide treatment. Apoptosis was assessed with FACS analysis using control or SENP1 KO HCT116 cells. WT cells shown in the Q4 quadrant are negative for both stains. D, The wound closure assay was performed to measure the effect of SENP1 deficiency on cell migration. Representative images of the wound healing assay at different time points indicating attenuated migratory abilities of HCT116 cells with SENP1 knockdown. E, The Transwell assay was performed to explore the effect of SENP1 knockdown on cell invasion. Representative light photomicrographs of migrated HCT116 cells through the Transwell at 6 hours (three replicates), stained with crystal violet. Student t test; *, P < 0.05; ***, P < 0.001.

Figure 6.

SENP1 depletion inhibits cell proliferation, migration, and invasion. A, Knockdown and knockout (KO) of SENP1 were confirmed by immunoblotting. B, Cell Counting Kit-8 (CCK8) cell proliferation assay was used to detect cell proliferation in SENP1 wild-type (WT) and SENP1 knockdown HCT116 cells. C, SENP1 depletion increased the apoptosis rate of HCT116 cells after 40 μmol/L etoposide treatment. Apoptosis was assessed with FACS analysis using control or SENP1 KO HCT116 cells. WT cells shown in the Q4 quadrant are negative for both stains. D, The wound closure assay was performed to measure the effect of SENP1 deficiency on cell migration. Representative images of the wound healing assay at different time points indicating attenuated migratory abilities of HCT116 cells with SENP1 knockdown. E, The Transwell assay was performed to explore the effect of SENP1 knockdown on cell invasion. Representative light photomicrographs of migrated HCT116 cells through the Transwell at 6 hours (three replicates), stained with crystal violet. Student t test; *, P < 0.05; ***, P < 0.001.

Close modal

SENP1 is upregulated in COAD and negatively correlated with cancer prognosis through the downregulation of RNF168 SUMOylation

SENP1 plays important roles in maintaining genomic integrity and regulating cell status when exposed to chemotherapeutic drugs. To explore the biological significance of SENP1, we first investigated the relationship between SENP1 and human cancers. Bioinformatics analyses revealed that SENP1 is highly expressed in tumor tissue of COAD patients (Fig. 7A). Consistent with these analyses, IHC staining confirmed the higher abundance of SENP1 in COAD tissues than that in adjacent normal tissues (Fig. 7B). And higher expression of SENP1 were observed in the human COAD cell lines HCT116, SW480, LOVO, HT29, and RKO, than in the normal colon cell lines NCM460 and FHC (Fig. 7C). We further validated the effect of SENP1 level on phase separation of endogenous RNF168 by IF assays using cancer cell HCT116 and normal cell FHC that displayed different expression level of endogenous SENP1. As expected, condensation of endogenous RNF168 was observed obviously in FHC cells with very low level of SENP1 and HCT116 cells with knocked out SENP1, but not in wild-type HCT116 that expressing high level of SENP1 (Fig. 7D), suggesting that the level of endogenous SENP1 plays a critical role in regulation of the formation of RNF168 LLPS under physiological condition. In addition, 53BP1 was also colocalized with endogenous RNF168 condensates in FHC cells (Supplementary Fig. S6A). Moreover, PLA experiments were performed to compare the levels of endogenous RNF168 SUMOylation in cancer tissues and adjacent normal tissues. As expected, COAD tissues exhibited a weaker association between RNF168 and SUMO3 relative to adjacent normal tissues (Fig. 7E), indicating that the overall RNF168 SUMOylation level decreased in colon cancer. In addition, in situ PLA assays showed that SENP1 knockdown resulted in condensation of SUMOylated RNF168 (Fig. 7F), demonstrating that SENP1 knockdown increased SUMOylation of RNF168.

Figure 7.

SENP1 is upregulated in COAD and negatively correlated to cancer prognosis through downregulation of RNF168 SUMOylation. A, SENP1 was upregulated in COAD. Expression variance of SENP1 in COAD patients using UALCAN portal analysis. B, SENP1 was upregulated in COAD tumor tissues. Left, representative images of IHC staining of SENP1 in human COAD specimens and adjacent tissue specimens. Scale bar, 50 μm. Right, images of IHC staining were quantitated (n = 4). Statistical analysis was performed using a Student t test. Data are expressed as the mean ± SEM. C, Immunoblotting of SENP1 expression level in normal FHC and NCM460 cells and human colon cancer cell lines HCT116, SW480, HT29, LOVO, and RKO. D, LLPS of endogenous RNF168 was verified using IF assays in FHC cells, HCT116 cells, and SENP1−/− HCT116 cells. Scale bar, 5 μm. E, Endogenous SUMOylation of RNF168 decreased in COAD tumor tissues. Left, representative images of in situ interaction between RNF168 and SUMO3 was detected using Duolink PLA IF in COAD tumor tissue specimens and adjacent normal tissue specimens. Scale bar, 10 μm. Right, quantitation of the PLA dots indicating RNF168-SUMO3 interactions is shown as mean ± SEM, with approximately 10 cells selected in each group. F, Knockdown of SENP1 promoted endogenous RNF168 SUMOylation. SW480 cells were transfected with SENP1 shRNA #5 or #6 and subjected to PLA analyses using the indicated antibodies. Scale bar, 5 μm. Right, immunoblotting of SENP1 expression level was confirmed in the indicated control and SENP1 knockdown SW480 cells, with approximately 10 cells selected in each group. G, RNF168-K210R SUMOylation deficiency caused drug resistance to oxaliplatin in HCT116 cells. HCT116 RNF168 knockout (KO) cells transfected with HA-RNF168 or HA-RNF168-K210R were treated with 50 μmol/L oxaliplatin for 48 hours, and apoptosis rates were subsequently assessed using Annexin V and PI staining. HCT116 wild-type (WT) cells and HCT116 RNF168 KO cells were used as controls. H, Reintroduction of WT RNF168 or the K210R mutant to RNF168−/− HCT116 cells was confirmed by immunoblotting. I, Crystal violet staining analysis was conducted using RNF168−/− HCT116 cells reintroduced by WT or K210R RNF168. Cells were treated with different concentrations of SN38 for 2 hours and drug-free medium was applied to the samples for 14 days, colonies were fixed in methanol, stained with crystal violet, washed with water, and photographed. J, Experimental design for constructing CDX nude mice models. Mice injected with indicated cells were sacrificed on the 12th day after injection; photographs of mice tumors in each group (n = 5) are shown. Tumor weight was measured using a caliper, and tumor volume was calculated using the following formula: volume = (length × width2)/2. Data are expressed as the mean ± SEM. Statistical analysis was performed using a Student t test. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001. The expression level of γH2AX in tumor tissues was assessed by immunoblotting.

Figure 7.

SENP1 is upregulated in COAD and negatively correlated to cancer prognosis through downregulation of RNF168 SUMOylation. A, SENP1 was upregulated in COAD. Expression variance of SENP1 in COAD patients using UALCAN portal analysis. B, SENP1 was upregulated in COAD tumor tissues. Left, representative images of IHC staining of SENP1 in human COAD specimens and adjacent tissue specimens. Scale bar, 50 μm. Right, images of IHC staining were quantitated (n = 4). Statistical analysis was performed using a Student t test. Data are expressed as the mean ± SEM. C, Immunoblotting of SENP1 expression level in normal FHC and NCM460 cells and human colon cancer cell lines HCT116, SW480, HT29, LOVO, and RKO. D, LLPS of endogenous RNF168 was verified using IF assays in FHC cells, HCT116 cells, and SENP1−/− HCT116 cells. Scale bar, 5 μm. E, Endogenous SUMOylation of RNF168 decreased in COAD tumor tissues. Left, representative images of in situ interaction between RNF168 and SUMO3 was detected using Duolink PLA IF in COAD tumor tissue specimens and adjacent normal tissue specimens. Scale bar, 10 μm. Right, quantitation of the PLA dots indicating RNF168-SUMO3 interactions is shown as mean ± SEM, with approximately 10 cells selected in each group. F, Knockdown of SENP1 promoted endogenous RNF168 SUMOylation. SW480 cells were transfected with SENP1 shRNA #5 or #6 and subjected to PLA analyses using the indicated antibodies. Scale bar, 5 μm. Right, immunoblotting of SENP1 expression level was confirmed in the indicated control and SENP1 knockdown SW480 cells, with approximately 10 cells selected in each group. G, RNF168-K210R SUMOylation deficiency caused drug resistance to oxaliplatin in HCT116 cells. HCT116 RNF168 knockout (KO) cells transfected with HA-RNF168 or HA-RNF168-K210R were treated with 50 μmol/L oxaliplatin for 48 hours, and apoptosis rates were subsequently assessed using Annexin V and PI staining. HCT116 wild-type (WT) cells and HCT116 RNF168 KO cells were used as controls. H, Reintroduction of WT RNF168 or the K210R mutant to RNF168−/− HCT116 cells was confirmed by immunoblotting. I, Crystal violet staining analysis was conducted using RNF168−/− HCT116 cells reintroduced by WT or K210R RNF168. Cells were treated with different concentrations of SN38 for 2 hours and drug-free medium was applied to the samples for 14 days, colonies were fixed in methanol, stained with crystal violet, washed with water, and photographed. J, Experimental design for constructing CDX nude mice models. Mice injected with indicated cells were sacrificed on the 12th day after injection; photographs of mice tumors in each group (n = 5) are shown. Tumor weight was measured using a caliper, and tumor volume was calculated using the following formula: volume = (length × width2)/2. Data are expressed as the mean ± SEM. Statistical analysis was performed using a Student t test. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001. The expression level of γH2AX in tumor tissues was assessed by immunoblotting.

Close modal

The clinical effectiveness of chemotherapy is limited by drug resistance, which is the major cause of treatment failure and mortality in patients with colon cancer (42). Because deSUMOylation of RNF168 plays a protective role by promoting NHEJ repair in the cellular response to DNA damage, we therefore speculated that the decreased RNF168 SUMOylation level caused by high SENP1 levels in cancer tissues might contribute to drug resistance. To verify this hypothesis, HCT116 RNF168 knockout cells transfected with HA-RNF168 or HA-RNF168-K210R were treated with the clinical COAD medicine oxaliplatin and subjected to apoptosis analyses. RNF168 knockout promoted the rate of apoptosis; reintroduction of the SUMOylation-defective K210R mutant led to a lower rate of apoptosis relative to RNF168−/− cells rescued with RNF168 wild-type (Fig. 7G). In keeping with these results, we generated HCT116 RNF168−/− cells stably expressing the lentivirus transfer vector pLV04-RNF168 or pLV04-RNF168 K210R (Fig. 7H) and performed a colony formation assay. After treatment with different concentrations of SN38 (a metabolite of the COAD chemotherapeutic drug irinotecan) or oxaliplatin, HCT116 RNF168−/− cells expressing the SUMOylation-defective K210R mutant displayed a higher survival rate than those reintroduced with wild-type RNF168 (Fig. 7I; Supplementary Fig. S6B and S6C). These results suggest that cells with lower levels of RNF168 SUMOylation are more resistant to chemotherapy drugs.

To elucidate the physiological effect of RNF168 SUMOylation in mice, HCT116 wild-type cells, HCT116 RNF168−/−cells, HCT116 SENP1−/− cells, and RNF168−/− HCT116 cells with reintroduced SUMOylation-defective K210R or wild-type RNF168 were subcutaneously injected into the armpits of 4-week-old female BALB/c nude mice. After inoculation, we observed tumor volume every two days until the tumor sizes approached approximately 1,000 mm3. As expected, RNF168−/− and SENP1−/−, which strongly disturb genomic integrity, inhibited tumor growth as well; the tumor size in the SENP1−/− group was notably smaller than the wild-type group. Surprisingly, the RNF168−/− group failed to form tumors, but this failure was rescued by reintroduction of RNF168 wild-type and the K210R mutant. As expected, rescue with the SUMOylation-deficient mutant K210R, which promotes NHEJ repair, showed larger tumors than the wild-type RNF168 group. In addition, the levels of DNA damage marker γH2AX were assessed in the tumor tissues, which showed that smaller tumor correlated with more extensive damage (Fig. 7J).

Consequently, analyses of the correlation between SENP1 and COAD prognosis using TCGA databases revealed that COAD patients with higher SENP1 expression levels had lower survival (Supplementary Fig. S6D). Furthermore, other types of cancer such as mesothelioma (MESO), liver hepatocellular carcinoma (LIHC), kidney renal papillary cell carcinoma (KIRP), and adrenocortical carcinoma (ACC) showed a similar survival tendency as the COAD patients (Supplementary Fig. S6E). Collectively, these data suggest that a high level of SENP1 in cancer tissues affects chemotherapy drug efficacy and cancer prognosis through downregulation of RNF168 SUMOylation.

The DDR is fine-tuned by many types of posttranslational protein modifications. In the present study, we identified that PIAS1 promotes SUMOylation and SENP1 deSUMOylates RNF168 at Lys210. More importantly, SUMOylated RNF168 undergoes LLPS in the nucleus, which blocks the recruitment of RNF168 to the damaged site, restrains the RNF168-H2A interaction and H2A ubiquitination, compartmentalizes 53BP1 in the condensates, and ultimately downregulates NHEJ repair efficiency. In response to DNA damage, SENP1 is quickly recruited to the damaged sites where it decreases RNF168 SUMOylation, which disrupts RNF168 LLPS and facilitates damage repair. SENP1 is upregulated in COAD, which renders cancer cells insensitive to chemotherapy drugs through its inhibition of RNF168 SUMOylation-mediated LLPS, thus causing drug resistance.

Previous studies have reported the crucial role of RNF168 in the DSB repair pathway and in human diseases; dysfunction of RNF168 leads to radiosensitivity, immunodeficiency, dysmorphic features, and learning difficulties, known as the RIDDLE syndrome (1, 43). In the DDR pathway, RNF168 ubiquitinates histone H2A at K13/15 (6), and ubiquitinated H2A serve as a scaffold for recruitment of downstream repair proteins such as 53BP1 (7). In addition, RNF168 itself is a substrate of ubiquitination and SUMOylation (5, 8, 35). SUMO1 has been suggested to modify RNF168 (35), and RNF168 has been shown to associate with proteins modified by SUMO2 and/or SUMO3 (44). However, the functional effects of RNF168 SUMOylation remain largely unknown. Here, we confirmed that RNF168 is mono-SUMOylated and demonstrated that RNF168 SUMOylation had a significant function in the regulation of DNA damage repair by undergoing LLPS.

Because the germline P granule has been reported to be the first phase-separated condensate, many LLPS-formed membraneless organelles, including nucleoli, Cajal bodies, centromeres, promyelocytic leukemia nuclear bodies, and stress granules, have been reported to play important roles in regulating many biological processes (17, 21, 45). In the DDR pathway, many proteins undergo LLPS as well, which regulate the efficiency of DNA repair. For example, phase separation of the RNA binding protein FUS is sufficient for repair initiation (46), and TopBP1 assembles biomolecule condensates and triggers the checkpoint in response to DNA replication impediments (47). Many factors, especially PTMs, are involved in the regulation of LLPS under different conditions. Poly-ubiquitination-modulated p62 phase separation drives the segregation of autophagic cargo (21). Dephosphorylation and SUMOylation are required for the formation of stress-induced NELF condensates (48). In addition, SUMOylation also coordinates the disassembly of the stress granules that contributes to amyotrophic lateral sclerosis pathogenesis (18). Here, we demonstrate that RNF168 undergoes LLPS, which is upregulated by SUMOylation. More importantly, we identified 53BP1 as a component of RNF168 condensates (Fig. 3F and G, Supplementary Fig. S3E and S3F), which is consistent with previous observations that RNF168 is associated with 53BP1 (7) and that 53BP1 shows phase separation properties (30, 31). It is noteworthy that RNF168 LLPS downregulates DNA damage repair by blocking the recruitment of RNF168 to damaged sites and compartmentalizing the downstream key repair protein 53BP1 as well, elucidating a novel mechanism underlying PTM-mediated LLPS in the DDR pathway.

DeSUMOylation enzyme SENPs are well known to balance protein levels between their SUMOylated and un-SUMOylated forms. The aberrant expression of SENPs is correlated with cancer development and progression. In response to fasting, SENP1 regulates mitochondrial metabolism by deSUMOylating Sirt3 (49). SENP2 inhibits NF-κB activity by reducing NEMO SUMOylation (15). Furthermore, mitotic SENP3 activation promotes the innate immune response in tumor cells (50). In our study, we demonstrate that in response to DNA damage, SENP1 is recruited to DNA damage sites in an ATM-dependent manner to promote NHEJ by deSUMOylating RNF168; however, whether SENP1 is directly phosphorylated by ATM and the identity of the phosphorylation site require further investigation. More importantly, the level of endogenous SENP1 plays critical role in regulation of the LLPS of RNF168 and NHEJ. We found that SENP1 is highly expressed in COAD patients who showed a low level of RNF168 SUMOylation, reduced RNF168 LLPS, promoted NHEJ efficiency, and experienced chemotherapeutic drug resistance (Fig. 7). Consistently, we demonstrated that COAD cells with depleted SENP1 were more sensitive to chemotherapy drugs and showed decreased abilities of proliferation and migration, thus leading to effective treatment. Using cell-derived xenograft (CDX) nude mice models, we further proved that knockout of SENP1 and RNF168 significantly inhibited tumor growth, which could be rescued by introduction of the RNF168 SUMOylation-deficient mutant K210R. Meanwhile, analysis of the TCGA database revealed that the expression level of SENP1 is negatively correlated with survival rate of many types of cancers including MESO, LIHC, KIRP, and ACC, suggesting that SENP1 affects cancer treatment efficacy by modulating RNF168 SUMOylation.

In summary, we identified that RNF168 SUMOylation impeded RNF168 function in DNA damage repair through LLPS. The formation of reversible functional micrometer-sized RNF168 condensates by SENP1-modulated SUMOylation in response to DNA damage provides insight into a novel molecular mechanism underlying DDR regulation. Depletion of SENP1 renders cancer cells sensitive to DNA damaging drugs, suggesting a potential strategy for enhancing the efficacy of chemotherapy.

No disclosures were reported.

M. Wei: Conceptualization, data curation, formal analysis, investigation, visualization, methodology, writing–original draft. X. Huang: Investigation. L. Liao: Investigation. Y. Tian: Investigation. X. Zheng: Conceptualization, resources, supervision, funding acquisition, writing–original draft, project administration, writing–review and editing.

The authors sincerely thank Prof. Jinke Cheng for providing the human SUMO1, SUMO2/3, and SENP1 plasmids. The authors are grateful to the National Center for Protein Sciences at Peking University, particularly Guilan Li, Yinghua Guo, and Liying Du, for technical help. They also appreciate the assistance of Ye Liang from the Core Facility of the State Key Laboratory of Membrane Biology at Peking University and Siying Qin, Dong Liu, and Liqin Fu from the Core Facilities of Life Sciences at Peking University for their assistance with the protein MS analysis and microscopy imaging. This work was supported by the National Key R&D Program of China (2022YFA1302803), the National Natural Science Foundation of China (82130081, 32270756), and Natural Science Foundation of Beijing Municipality (5212008).

The publication costs of this article were defrayed in part by the payment of publication fees. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/).

1.
Stewart
GS
,
Panier
S
,
Townsend
K
,
Al-Hakim
AK
,
Kolas
NK
,
Miller
ES
, et al
.
The RIDDLE syndrome protein mediates a ubiquitin-dependent signaling cascade at sites of DNA damage
.
Cell
2009
;
136
:
420
34
.
2.
Smeenk
G
,
Wiegant
WW
,
Marteijn
JA
,
Luijsterburg
MS
,
Sroczynski
N
,
Costelloe
T
, et al
.
Poly(ADP-ribosyl)ation links the chromatin remodeler SMARCA5/SNF2H to RNF168-dependent DNA damage signaling
.
J Cell Sci
2013
;
126
:
889
903
.
3.
Garvin
AJ
.
Beyond reversal: ubiquitin and ubiquitin-like proteases and the orchestration of the DNA double-strand break repair response
.
Biochem Soc Trans
2019
;
47
:
1881
93
.
4.
Xu
Y
,
Xu
D
.
Repair pathway choice for double-strand breaks
.
Essays Biochem
2020
;
64
:
765
77
.
5.
Pinato
S
,
Scandiuzzi
C
,
Arnaudo
N
,
Citterio
E
,
Gaudino
G
,
Penengo
L
.
RNF168, a new RING finger, MIU-containing protein that modifies chromatin by ubiquitination of histones H2A and H2AX
.
BMC Mol Biol
2009
;
10
:
1
14
.
6.
Mattiroli
F
,
Vissers
JH
,
van Dijk
WJ
,
Ikpa
P
,
Citterio
E
,
Vermeulen
W
, et al
.
RNF168 ubiquitinates K13–15 on H2A/H2AX to drive DNA damage signaling
.
Cell
2012
;
150
:
1182
95
.
7.
Doil
C
,
Mailand
N
,
Bekker-Jensen
S
,
Menard
P
,
Larsen
DH
,
Pepperkok
R
, et al
.
RNF168 binds and amplifies ubiquitin conjugates on damaged chromosomes to allow accumulation of repair proteins
.
Cell
2009
;
136
:
435
46
.
8.
Pinato
S
,
Gatti
M
,
Scandiuzzi
C
,
Confalonieri
S
,
Penengo
L
.
UMI, a novel RNF168 ubiquitin binding domain involved in the DNA damage signaling pathway
.
Mol Cell Biol
2011
;
31
:
118
26
.
9.
Han
ZJ
,
Feng
YH
,
Gu
BH
,
Li
YM
,
Chen
H
.
The posttranslational modification, SUMOylation, and cancer (Review)
.
Int J Oncol
2018
;
52
:
1081
94
.
10.
Galanty
Y
,
Belotserkovskaya
R
,
Coates
J
,
Polo
S
,
Miller
KM
,
Jackson
SP
.
Mammalian SUMO E3-ligases PIAS1 and PIAS4 promote responses to DNA double-strand breaks
.
Nature
2009
;
462
:
935
9
.
11.
Morris
JR
,
Boutell
C
,
Keppler
M
,
Densham
R
,
Weekes
D
,
Alamshah
A
, et al
.
The SUMO modification pathway is involved in the BRCA1 response to genotoxic stress
.
Nature
2009
;
462
:
886
90
.
12.
Wang
Y
,
Dasso
M
.
SUMOylation and deSUMOylation at a glance
.
J Cell Sci
2009
;
122
:
4249
52
.
13.
Luo
K
,
Zhang
H
,
Wang
L
,
Yuan
J
,
Lou
Z
.
Sumoylation of MDC1 is important for proper DNA damage response
.
EMBO J
2012
;
31
:
3008
19
.
14.
Gao
SS
,
Guan
H
,
Yan
S
,
Hu
S
,
Song
M
,
Guo
ZP
, et al
.
TIP60 K430 SUMOylation attenuates its interaction with DNA-PKcs in S-phase cells: Facilitating homologous recombination and emerging target for cancer therapy
.
Sci Adv
2020
;
6
:
eaba7822
.
15.
Lee
MH
,
Mabb
AM
,
Gill
GB
,
Yeh
ET
,
Miyamoto
S
.
NF-kappaB induction of the SUMO protease SENP2: a negative feedback loop to attenuate cell survival response to genotoxic stress
.
Mol Cell
2011
;
43
:
180
91
.
16.
Boija
A
,
Klein
IA
,
Sabari
BR
,
Dall'Agnese
A
,
Coffey
EL
,
Zamudio
AV
, et al
.
Transcription factors activate genes through the phase-separation capacity of their activation domains
.
Cell
2018
;
175
:
1842
55
.
17.
Verdile
V
,
De Paola
E
,
Paronetto
MP
.
Aberrant phase transitions: side effects and novel therapeutic strategies in human disease
.
Front Genet
2019
;
10
:
173
.
18.
Marmor-Kollet
H
,
Siany
A
,
Kedersha
N
,
Knafo
N
,
Rivkin
N
,
Danino
YM
, et al
.
Spatiotemporal proteomic analysis of stress granule disassembly using APEX reveals regulation by SUMOylation and links to ALS pathogenesis
.
Mol Cell
2020
;
80
:
876
91
.
19.
Mehta
S
,
Zhang
J
.
Liquid–liquid phase separation drives cellular function and dysfunction in cancer
.
Nat Rev Cancer
2022
;
22
:
239
52
.
20.
Zhang
L
,
Geng
X
,
Wang
F
,
Tang
J
,
Ichida
Y
,
Sharma
A
, et al
.
53BP1 regulates heterochromatin through liquid phase separation
.
Nat Commun
2022
;
13
:
360
.
21.
Sun
D
,
Wu
R
,
Zheng
J
,
Li
P
,
Yu
L
.
Polyubiquitin chain-induced p62 phase separation drives autophagic cargo segregation
.
Cell Res
2018
;
28
:
405
15
.
22.
Boehning
M
,
Dugast-Darzacq
C
,
Rankovic
M
,
Hansen
AS
,
Yu
T
,
Marie-Nelly
H
, et al
.
RNA polymerase II clustering through carboxy-terminal domain phase separation
.
Nat Struct Mol Biol
2018
;
25
:
833
40
.
23.
Chong
S
,
Dugast-Darzacq
C
,
Liu
Z
,
Dong
P
,
Dailey
GM
,
Cattoglio
C
, et al
.
Imaging dynamic and selective low-complexity domain interactions that control gene transcription
.
Science
2018
;
36
:
eaar25551
.
24.
Sabari
BR
,
Dall'Agnese
A
,
Boija
A
,
Klein
IA
,
Coffey
EL
,
Shrinivas
K
, et al
.
Coactivator condensation at super-enhancers links phase separation and gene control
.
Science
2018
;
361
:
eaar3958
.
25.
Boeynaems
S
,
Alberti
S
,
Fawzi
NL
,
Mittag
T
,
Polymenidou
M
,
Rousseau
F
, et al
.
Protein phase separation: a new phase in cell biology
.
Trends Cell Biol
2018
;
28
:
420
35
.
26.
Arva
A
,
Kasu
YAT
,
Duncan
J
,
Alkhatatbeh
MA
,
Brower
CS
.
The ligand of ate1 is intrinsically disordered and participates in nucleolar phase separation regulated by jumonji domain containing 6
.
Proc Natl Acad Sci USA
2021
;
118
:
e2015887118
.
27.
Owen
I
,
Shewmaker
F
.
The role of posttranslational modifications in the phase transitions of intrinsically disordered proteins
.
Int J Mol Sci
2019
;
20
:
5501
.
28.
Dao
TP
,
Kolaitis
RM
,
Kim
HJ
,
O'Donovan
K
,
Martyniak
B
,
Colicino
E
, et al
.
Ubiquitin modulates liquid–liquid phase separation of UBQLN2 via disruption of multivalent interactions
.
Mol Cell
2018
;
69
:
965
78
.
29.
Duan
Y
,
Du
A
,
Gu
J
,
Duan
G
,
Wang
C
,
Gui
X
, et al
.
PARylation regulates stress granule dynamics, phase separation, and neurotoxicity of disease-related RNA-binding proteins
.
Cell Res
2019
;
29
:
233
47
.
30.
Pessina
F
,
Giavazzi
F
,
Yin
Y
,
Gioia
U
,
Vitelli
V
,
Galbiati
A
, et al
.
Functional transcription promoters at DNA double-strand breaks mediate RNA-driven phase separation of damage-response factors
.
Nat Cell Biol
2019
;
21
:
1286
99
.
31.
Kilic
S
,
Lezaja
A
,
Gatti
M
,
Bianco
E
,
Michelena
J
,
Imhof
R
, et al
.
Phase separation of 53BP1 determines liquid-like behavior of DNA repair compartments
.
EMBO J
2019
;
38
:
e101379
.
32.
Wang
B
,
Zhang
L
,
Dai
T
,
Qin
Z
,
Lu
H
,
Zhang
L
, et al
.
Liquid–liquid phase separation in human health and diseases
.
Signal Transduct Target Ther
2021
;
6
:
290
.
33.
Yang
CZ
,
Zang
WC
,
Tang
ZF
,
Ji
YP
,
Xu
RD
,
Yang
YF
, et al
.
A20/TNFAIP3 regulates the DNA damage response and mediates tumor cell resistance to DNA-damaging therapy
.
Cancer Res
2018
;
78
:
1069
82
.
34.
Xu
RD
,
Yu
SY
,
Zhu
D
,
Huang
XP
,
Xu
YQ
,
Lao
YM
, et al
.
hCINAP regulates the DNA damage response and mediates the resistance of acute myelocytic leukemia cells to therapy
.
Nat Commun
2019
;
10
:
3812
.
35.
Danielsen
JR
,
Povlsen
LK
,
Villumsen
BH
,
Streicher
W
,
Nilsson
J
,
Wikstrom
M
, et al
.
DNA damage-inducible SUMOylation of HERC2 promotes RNF8 binding via a novel SUMO-binding Zinc finger
.
J Cell Biol
2012
;
197
:
179
87
.
36.
Saitoh
H
,
Hinchey
J
.
Functional heterogeneity of small ubiquitin-related protein modifiers SUMO-1 versus SUMO-2/3
.
J Biol Chem
2000
;
275
:
6252
8
.
37.
Tatham
MH
,
Jaffray
E
,
Vaughan
OA
,
Desterro
JM
,
Botting
CH
,
Naismith
JH
, et al
.
Polymeric chains of SUMO-2 and SUMO-3 are conjugated to protein substrates by SAE1/SAE2 and Ubc9
.
J Biol Chem
2001
;
276
:
35368
74
.
38.
Liu
YC
,
Lin
MC
,
Chen
HC
,
Tam
MF
,
Lin
LY
.
The role of small ubiquitin-like modifier-interacting motif in the assembly and regulation of metal-responsive transcription factor 1
.
J Biol Chem
2011
;
286
:
42818
29
.
39.
Zhang
Z
,
Du
J
,
Wang
S
,
Shao
L
,
Jin
K
,
Li
F
, et al
.
OTUB2 promotes cancer metastasis via hippo-independent activation of YAP and TAZ
.
Mol Cell
2019
;
73
:
7
21
.
40.
Waters
E
,
Wilkinson
KA
,
Harding
AL
,
Carmichael
RE
,
Robinson
D
,
Colley
HE
, et al
.
The SUMO protease SENP3 regulates mitochondrial autophagy mediated by Fis1
.
EMBO Rep
2022
;
23
:
1743
5
.
41.
Lv
P
,
Du
Y
,
He
C
,
Peng
L
,
Zhou
X
,
Wan
Y
, et al
.
O-GlcNAcylation modulates liquid–liquid phase separation of SynGAP/PSD-95
.
Nat Chem
2022
;
14
:
831
40
.
42.
Picco
G
,
Cattaneo
CM
,
van Vliet
EJ
,
Crisafulli
G
,
Rospo
G
,
Consonni
S
, et al
.
Werner helicase is a synthetic-lethal vulnerability in mismatch repair-deficient colorectal cancer refractory to targeted therapies, chemotherapy, and immunotherapy
.
Cancer Discov
2021
;
11
:
1923
37
.
43.
Stewart
GS
,
Stankovic
T
,
Byrd
PJ
,
Wechsler
T
,
Miller
ES
,
Huissoon
A
, et al
.
RIDDLE immunodeficiency syndrome is linked to defects in 53BP1-mediated DNA damage signaling
.
Proc Natl Acad Sci USA
2007
;
104
:
16910
5
.
44.
Shire
K
,
Wong
AI
,
Tatham
MH
,
Anderson
OF
,
Ripsman
D
,
Gulstene
S
, et al
.
Identification of RNF168 as a PML nuclear body regulator
.
J Cell Sci
2016
;
129
:
580
91
.
45.
Brangwynne
CP
,
Eckmann
CR
,
Courson
DS
,
Rybarska
A
,
Hoege
C
,
Gharakhani
J
, et al
.
Germline P granules are liquid droplets that localize by controlled dissolution/condensation
.
Science
2009
;
324
:
1729
32
.
46.
Levone
BR
,
Lenzken
SC
,
Antonaci
M
,
Maiser
A
,
Rapp
A
,
Conte
F
, et al
.
FUS-dependent liquid–liquid phase separation is important for DNA repair initiation
.
J Cell Biol
2021
;
220
:
e202008030
.
47.
Frattini
C
,
Promonet
A
,
Alghoul
E
,
Vidal-Eychenie
S
,
Lamarque
M
,
Blanchard
MP
, et al
.
TopBP1 assembles nuclear condensates to switch on ATR signaling
.
Mol Cell
2021
;
81
:
1231
45
.
48.
Rawat
P
,
Boehning
M
,
Hummel
B
,
Aprile-Garcia
F
,
Pandit
AS
,
Eisenhardt
N
, et al
.
Stress-induced nuclear condensation of NELF drives transcriptional downregulation
.
Mol Cell
2021
;
81
:
1013
26
.
49.
Wang
T
,
Cao
Y
,
Zheng
Q
,
Tu
J
,
Zhou
W
,
He
J
, et al
.
SENP1-Sirt3 signaling controls mitochondrial protein acetylation and metabolism
.
Mol Cell
2019
;
75
:
823
34
.
50.
Hu
G
,
Chen
Y
,
Yang
X
,
Wang
Y
,
He
J
,
Wang
T
, et al
.
Mitotic SENP3 activation couples with cGAS signaling in tumor cells to stimulate antitumor immunity
.
Cell Death Dis
2022
;
13
:
640
.