DHX9 cKO mice generation
DHX9loxp/loxp mice were generated at BIOCYTOGEN company by injecting Cas9 mRNA, sgRNAs, and pUC57 donor plasmid directly into zygotes. AicdaCre+/- mice were kindly provided by Dr. Baidong Hou at the Institute of Biophysics, CAS. DHX9loxp/loxp mice and AicdaCre+/- mice were maintained in C57BL/6J genetic background and intercrossed to generate the mice with conditional deletion of DHX9 in the activated B cells. Genotyping was performed by PCR with loxP-specific primer pairs and confirmed by Sanger sequencing (see Supplementary Table 3). The PCR products of the WT allele were 3,126 bp, while the DHX9-ablation allele showed a 403 bp fragment. Six- to ten-week-old mice were used for splenic B cell isolation and antibody titer measurement. The DHX9loxp/loxp, Aicda-Cre mice were used for subsequent analysis, and their littermates with genome type of DHX9loxp/loxp or DHX9loxp/+ were used as controls. All the surgical procedures were approved and performed under the guidelines of the IBP Animal Care and Use Committee.
RT-qPCR
Total RNA was extracted from WT or DHX9 cKO B cells with TRIzol reagent (Thermo Fisher, 15596018) and converted into cDNA with M-MLV Reverse Transcriptase (Promega, M1708) after depleting genomic DNA by RQ1 DNase I digestion. Quantitative PCR was performed on a StepOne Plus Real-Time PCR System (Applied Biosystems) with Hieff® qPCR SYBR Green Master mix (YEASEN, 11203ES08). GAPDH served as an internal control. All primers used in this study are shown in Supplementary Table 3.
Cell culture, immunization, and CSR
Splenic B cells were isolated from WT and DHX9 cKO mice by negative selection using the EasyStep mouse B cell isolation kit (Stem cell, 19854). The B220+ populations typically accounted for over 95% of the isolated cells. Primary B cells and CH12F3 cells were cultured in RPMI 1640 medium supplemented with 10% fetal bovine serum (FBS, Gibco, 16000044) and 50 µM 2-mecaptoethanol in 5% CO2 at 37 °C. LPS (25 mg/ml) was used to activate ex vivo cultured WT or DHX9 cKO B cells. DHX9 cKO mice and their WT littermates were intraperitoneally injected with 5 mg LPS per mouse (Sigma-Aldrich, L3024). After immunization, spleens or Peyers’patches were collected on day 10 for FACS analysis. Memory B cells (B220+/CD38+/GL7-/IgD-/IgM-) and plasma cells (B220-/CD138+) were gated and sorted out using different antibodies. The IgA+ or IgG3+ B cells from non-immunized or LPS-immunized mice were quantified by FACS using specific antibodies. The combination of CD40L (0.5 mg/ml, Peprotech, 315-15), IL-4 (20 ng/ml), and TGF-β (20 ng/ml, Peprotech, 100-21C) was directly added to the CH12F3 culture medium and incubated for additional 72 h to induce CSR from IgM to IgA. CSR was examined as we previously described30. The following antibodies were used for flow cytometry analysis: anti-B220 (Biolegend, 103224/103205; Invitrogen, 4339662), anti-CD19 (Biolegend, 115519), anti-GL7 (Biolegend, 144603) and anti-Fas (Biolegend, 152603), anti-IgG3 (Biolegend, 406803), anti-CD38 (Invitrogen, 2312470; Biolegend, 102711), anti-CD138 (Biolegend, 142505), anti-IgD (Biolegend, 405740/405716/405711; Invitrogen, 4346705), anti-IgM (Biolegend, 406505/406511), anti-IgA (Biolegend, 407003), anti-streptavidin PE (eBioscience, 12-4317-87), anti- streptavidin FITC (eBioscience, 11-4317-87) and anti-streptavidin e450 (Invitrogen, 2110549). All the FACS analyses were performed on Fortessa (BD Biosciences) and analyzed by FlowJo software (Tree Star).
Antibody quantification by ELISA
Mouse serum was collected from the orbital venous plexus of unchallenged WT and DHX9 cKO mice. The concentrations of Ig (IgA, IgM, IgG1, and IgG3) were measured using mouse Ig ELISA quantitation sets according to the manufacturer’s protocol (Bethyl Laboratories, E90-103, E90-101, E90-105, E90-111). The culture medium from DHX9WT, DHX9306-308del, and DHX9Y1189C mutant CH12F3 cells was collected for quantifying the IgA titers with mouse IgA ELISA quantitation set (E90-103). For patient serum, the concentration of IgG3 and IgE were measured by unlabeled mouse anti-human IgG3 hinge (SouthernBiotech, HP6050) and human IgE ELISA quantification kits (Bethyl Laboratories, A80-108), respectively. The absorbance was measured on a microplate spectrophotometer (Thermo Fisher Scientific, 1510).
Lentiviral shRNA packaging and transduction
The pLKO.1 lentiviral shRNA clones (TRCN0000335501, TRCN0000071058, TRCN0000071019, TRCN0000086693, TRCN0000312950) were applied to knockdown Wrn, Pif1, Fancj, Blm, and DHX36, respectively. The sequences of DHX9 shRNA used in CH12F3 cells are shown in Supplementary Table 3. To produce lentiviral particles, 6 mg of shRNA plasmid and packaging plasmids were co-transfected into 293T cells with Lipofectamine 2000 reagent as we previously described. The lentiviral particles in the culture medium were collected twice after 48 h and 72 h of transfection. For transduction, 5 ml of lentiviral supernatant and 5 ml of fresh medium were gently mixed with 8 mg/ml of polybrene (Sigma-Aldrich, H9268) and then applied to CH12F3 cells for 24 h. The stably transduced CH12F3 cells were selected by puromycin (8 mg/ml) for an additional 3 days.
GFP and mScarlet knock-in and mutant cell lines
To generate GFP- and mScarlet-tagged DHX9 cell lines, the homology repair template consisted of GFP or mScarlet cDNA sequence flanked on either side by 1200 bp homology arms was amplified from genomic DNA using KOD Xtreme Hot Start polymerase (Novagen, 71975-3). The repair templates were subsequently cloned into pEGFP-N1 using the Not1-HF restriction enzyme (NEB, R3189L), while the sgRNA was inserted into PX330-mcherry using the Bbs1 enzyme (Thermo Fisher Scientific, FD1014). The 5 mg of PX330-mcherry-sgRNA plasmid and 5 mg of homology repair template pEGFP-N1 were transfected into 106 CH12F3 cells using Electrotachometer (Lonza). The mCherry and GFP double-positive cells were sorted out after 24 h of transfection and expanded for one additional week. GFP- or mScarlet-positive cells were further sorted out and plated into a 96-well plate. Genotyping was performed using 2 × M5 Hiper plus Taq HiFi PCR mix (Mei5 Biotechnology, MF002-plus-01), and the PCR products were gel purified for Sanger sequencing. To further generate c.306-308del or p.Y1189C mutations, 106 GFP-knockin CH12F3 cells were transfected with 5 mg PX330-mcherry-sgRNA and 5 mg repair template containing the relevant mutation. The correct knock-in clones were individually cultured and confirmed by sanger sequencing. The sequences of sgRNA and primers used for genotyping are shown in Supplementary Table 3.
Plasmid construction
The pFastBac-hDHX9 plasmid was kindly provided by Dr. Frank Grosse, while the pCMV6-mDHX9 plasmid was purchased from Origene (MR226503). The human DHX9 fragment was inserted into the pMamBac-GFP vector between XhoI and EcoRI restriction sites for protein purification. The pMamBac-DHX9Y1189C-GFP, pMamBac-DHX9K417R-GFP, and pMamBac-DHX9ΔIDR3-GFP plasmids were generated by site-directed mutagenesis. The IDR1, IDR2, and IDR3 of DHX9 were inserted into the pSumo backbone or the pET-3C-GFP backbone between XhoI and EcoRI restriction sites. The pSANG10-3F-BG4 plasmid used for expressing BG4 scFv anti-G-quadruplex antibody was a gift from Shankar Balasubramanian (Addgene plasmid # 55756).
Immunofluorescence
Splenic B cells were treated with PDS (2 mM; MCE, 1781882) for 72 h to stabilize G4 structures for examining the effect of G4 accumulation on DNA damage. Splenic B cells or CH12F3 cells were fixed with 4% paraformaldehyde (PFA, Sigma-Aldrich, F8775) in PBS and then permeabilized with 0.3% Triton X-100 for 15 min. For DNA G4 staining, cells were first treated with 50 µg/ml of RNase A to eliminate potential RNA G4s. After washing three times with PBS, splenic B cells were blocked with 10% normal donkey serum in PBS for 1 h. The primary antibodies against G4, R-loop, FLAG (Sigma-Aldrich, F1804; ABclonal, AE004), DHX9 (Invitrogen, PA5-19542; Abcam, ab26271), 53BP1 (Santa Cruz, sc-515841), Rad51 (Santa Cruz, sc-398587), and gH2AX (Millipore, 05-636) were diluted in PBST and incubated with cells overnight at 4 °C. Cells were washed with PBST for three times and then incubated with donkey anti-mouse 488 (Life technology, A21202), donkey anti-rabbit 488 (Life technology, A21206), donkey anti-mouse 594 (Life technology, A21203), donkey anti-rabbit 594 (Life technology, A21207), and goat anti-mouse 647 (Life technology, A28181) for 1 h at RT and counterstained with DAPI for 10 min. The cells were spread on cover slides and imaged with confocal FV1200. The BG4 and S9.6 antibodies were purified as previously described34,65.
DNA FISH and combined G4 staining
DNA FISH probes were prepared by PCR amplification of the genomic fragments covering the G4 peaks of IgH Sm, Myc, and Pim1. The purified PCR products were fluorescently labeled with Alexa Fluor 546 or 647 using a ULYSIS Nucleic Acid Labelling Kit (Thermo Fisher Scientific, U21652, U21660). The labeled DNA FISH probes need to be denatured at 95 °C for 10 min and snap-chill on ice before use. For DNA FISH, LPS-activated B cells were first fixed with 4% PFA for 15 min at RT and then permeabilized with 70% ethanol for 15 min. The permeabilized cells were subsequently denatured in 2 × SSC buffer containing 70% formamide for 10 min at 75 °C. The mixture of 1% salmon sperm DNA (Thermo Fisher Scientific, 15632011) and 1 mg of labeled probes were applied to the cells and hybridized overnight at 37 °C. On the next day, slides were washed 3 times with 2 × SSC buffer at 45 °C for 10 min and stained with DAPI (Sigma-Aldrich, F6057) for 10 min. After washing twice with 2 × SSC buffer, the slides were sealed with mounting medium (Vector Laboratories, H-1000) for visualization with the Olympus FV1200 microscope.
For combined DNA FISH and G4 immunofluorescent staining, the permeabilized cells were pre-treated with 50 µg/ml RNase A at 37 °C for 2 h and washed three times with PBS. After blocking with 10% normal donkey serum for 1 h, the BG4 antibody was 1:100 diluted with PBS buffer and directly added to the blocked cells overnight at 4 °C. Subsequently, the anti-flag and donkey anti-mouse 488 antibodies were applied to visualize G4. After washing three times with PBST, the cells were permeabilized with 0.7% Triton-X-100/0.1 M HCl for 10 min on ice and denatured with 1.9 M HCl for 30 min at RT. Cells were then hybridized with DNA probes overnight at 37 °C and washed three times with 2 × SSC for 20 min. The nuclei were finally counterstained with DAPI.
COMET assay
Splenic B cells from WT and DHX9 cKO mice were ex vivo cultured and stimulated with LPS for 72 h. DNA damage was assessed with a Comet Assay Kit (R&D, 4250-050-K). Briefly, 10 µl of cell suspension was mixed with 50 µl of 0.5% low-melting agarose and spread onto a CometSlide (Trevigen) pre-coated with 100 µl of 1% normal-melting agarose. Cells were lysed by immersing the slides in a freshly prepared lysis solution (2.5 M NaCl, 100 mM ethylenediaminetetraacetic acid (EDTA), 10 mM Tris, 1% Triton X-100, 10% DMSO, pH 10.0) at 4 °C for 2 h. Subsequently, slides were washed three times in 1 × PBS and placed in a gel electrophoresis apparatus with freshly prepared electrophoresis buffer (1 mM EDTA, 300 mM NaOH, pH 13.0) for 25 min to allow DNA unwinding. Electrophoresis was performed at 20 V with a starting current of 300 mA for 20 min. Lastly, slides were neutralized with 0.4 M Tris (pH 7.5) three times and stained with 1 × SYBR Gold solution for visualization under a fluorescence microscope (Life Technologies). Splenic B cells isolated from WT mice were treated with 100 mM hydrogen peroxide (H2O2) for 20 min at 4 °C and served as a positive control. The tail moment was calculated by the length between the center of the head and the tail.
Western blot
Western blot was performed as previously described30. The following antibodies were used: anti-DHX9 (Invitrogen, PA5-19542), anti-GAPDH (Cell Signaling Technology, 2118L), anti-Actin (Invitrogen, MA5-15739), anti-Wrn (Immunoway, YT7780), anti-Pif1 (Proteintech, 19006-1-AP), anti-Fancj (Proteintech, 24436-1-AP), anti-Blm (Bioss, bs-12872R), anti-DHX36 (Proteintech, 13159-1-AP), anti-ROD1 (Abnova, H00009991-M01), anti-AID (Life Technologies, 392500), anti-53BP1 (Santa Cruz, sc-515841), anti-Rad51 (Santa Cruz, sc-398587), anti-gH2AX (Millipore, 05-636), and anti-H2AX (Abcam, ab20669). Horseradish peroxidase (HRP)-coupled secondary antibodies (1:5,000) were purchased from Pierce (31460, 31430), and mouse anti-rabbit IgG mAb (Conformation Specific, HRP Conjugate, 5127S) was purchased from Cell Signaling Technology.
Protein purification
DHX9 and mutant proteins were expressed using an insect and mammalian expression system. Briefly, 1 mg of pMamBac-hDHX9, hDHX9WT-GFP, hDHX9Y1189C-GFP, hDHX9ΔIDR3-GFP, or hDHX9K417R-GFP plasmids were individually transfected into 106 Sf9 cells and incubated at 27 °C for 72 h. The P1 baculovirus supernatant was collected by centrifugation at 500 g for 5 min, and 1-2% of the P1 baculovirus was directly added to 2 × 106/ml Sf9 suspension cells and incubated at 27 °C on a shaker at 130 rpm for an additional 72 h. The supernatant was centrifuged at 500 g for 5 min to collect P2 baculovirus, which was used to generate high-titer P3 baculovirus. To express the recombinant proteins, 3 × 106/ml HEK293F (Gibco, R79007) suspension cells were infected with 10-20% of P3 baculovirus for 12 h. The infected cells were treated with 10 mM sodium butyrate and incubated for an additional 60 h at 30 °C on a shaker at 130 rpm. The cells were collected by centrifugation at 4000 rpm with a Superspeed centrifuge (Thermo Fisher Scientific, LYNX6000) for 10 min at 4 °C. After resuspending the cell pellets in binding buffer (20 mM Tris-Cl pH 8.0, 500 mM NaCl, 25 mM imidazole) and lysed with an ultrasonic disruptor (Ningbo Scientz Biotechnology, SB-80). The lysates were centrifuged at 18000 rpm for 1 h at 4 °C, and the supernatant was used for purifying recombinant proteins with StrepTrapTM HP (Cytiva, 10278182). After eliminating nucleic acids with a buffer (20 mM Tris-Cl pH 8.0, 2 M NaCl, 25 mM imidazole), the proteins were eluted with an elution buffer (20 mM Tris-Cl pH 8.0, 500 mM NaCl, 25 mM imidazole, and 10mM desulfurizing biotin), and purified with a HiloadTM 16/600 SuperdexTM 200pg column (Cytiva, 28-9893-35) on an ÄKTA Pure (Cytiva). The tagged proteins were eluted with a buffer containing 20 mM Tris-Cl pH 8.0, 300 mM KCl, and 1 mM DTT. The PreScission protease was added to the eluted protein solution to remove his tag from purified proteins and incubated overnight at 4 °C. The His-Strep-GFP tag and His-tag were eliminated by HisTrapTM HP (Cytiva, 10251115). Untagged proteins were acquired in flowthrough and purified with a HiloadTM 16/600 SuperdexTM 200pg column on an ÄKTA Pure (Cytiva) using buffer (20 mM Tris-Cl pH 8.0, 500 mM NaCl, and 1 mM DTT).
For expressing IDR1, IDR2, and IDR3 truncated proteins, pSUMO-IDR1, pSUMO-IDR2, and pSUMO-IDR3 plasmids were individually transformed into BL21 (DE3) strain. The proteins were expressed and purified as we previously described30. To remove the his-sumo tag on protein, Ulp1 protease was added to the eluted product and dialyzed with buffer (20 mM Tris-HCl pH 8.0, 500 mM NaCl) overnight at 4 °C. The dialyzed protein samples were purified using another Ni-NTA resin to remove the protease, His-SUMO tag, or uncleaved proteins. The proteins were concentrated by centrifugation using an ultrafiltration tube and purified with a pre-equilibrated HiLoadTM 16/60 SuperdexTM 75pg column (Cytiva, 17-1068-01). Finally, the IDR1, IDR2, and IDR3 proteins were eluted using a buffer containing 20 mM Tris-HCl, pH 8.0, 300 mM KCl, and 1 mM DTT. The purity of the proteins was confirmed by fractionating on 4-15% SDS-PAGE and stained with Coomassie blue G-250 (Urchem, 71011381).
In vitro phase separation assay
To monitor temperature-dependent DHX9 phase separation, 1.6 mM DHX9 proteins in 300 ml buffer P (20 mM Tris-HCl pH 8.0, 150 mM NaCl, 1 mM DTT) were first incubated at 4 °C for 20 min and then transferred to 37 °C for additional 10 min. Subsequently, the mixture was incubated at 4 °C for 60 min. Images were collected at different time points by the FV3000 microscope. The sample without DHX9 proteins served as a negative control. To examine the dynamics of liquid droplets, GFP-tagged DHX9, the truncates, or the mutant proteins were dissolved in buffer P (20 mM Tris-HCl pH 8.0, 150 mM NaCl, 1 mM DTT) at a final concentration of 40 mM and transferred to a glass slide for monitoring the phase behaviors at indicated time points. For examining the wetting properties of liquid droplets on the surface of a glass slide, GFP-tagged DHX9 dissolved in phase separation buffer was transferred to the glass slide and incubated at RT for 0.5 h. Then the images were collected at indicated time points. To evaluate the influence of salt ions on DHX9 droplets, different amounts of DHX9 proteins were added to the buffer containing 20 mM Tris-HCl pH 8.0, 1 mM DTT, and varying concentrations of NaCl. The tubes were incubated at RT for 10 min and then loaded onto the glass slide for monitoring droplet formation under different conditions.
To examine the influence of G4 DNA on DHX9 droplet, 40 mM of GFP-tagged DHX9 in buffer D (20 mM HEPES pH 7.5, 150 mM KCl, 1 mM DTT) were mixed with 10 mM of Cy5-labeled Sm-G4. After transferring to the glass slide and incubating at RT for 1 h, the phase behavior was recorded using the FV3000 microscope. To study the ability of DHX9 or Y1189C mutant on unwinding G4, 10 mM GFP-tagged DHX9 or Y1189C mutant were first mixed with 2.5 mM Cy5-labeled Sm-G4 in buffer D (20 mM HEPES, pH 7.5, 150 mM KCl, 1 mM DTT) containing 3 mM MgCl2, followed by adding Ficoll-400 to a final concentration of 150 mg/ml (Solarbio, F8150), and ATP (Meilunbio, MB3157) or AMP-PNP (Sigma-Aldrich, A2647) to a final concentration of 3 mM. The mixtures were transferred to a glass slide and imaged at different time points by FV3000 microscopy.
In vitro DNA phase separation assay
Single-stranded Sm DNA fragments, including 1 × Sm, 2 × Sm, and 4 × Sm, were synthesized by BGI genomics. To generate longer G4-containing ssDNA fragments, we developed a PCR-based lambda exonuclease digestion approach. The forward and reverse primers were synthesized and 5’ end-labeled with biotin or phosphorylation, respectively. The G4-containing Sm (1000 and 1900 bp) and Myc (202 bp) regions were PCR amplified with the primer pairs, 2 × M-PCR OPTI Mix (Biotool, B45012), and 50 ng murine genomic DNA. A 1000 bp non-G repeat fragment from IgH was amplified to serve as a negative control. The PCR fragments were gel purified, and 5 mg of them were treated with 1 ml of lambda exonuclease (NEB, M0262S) for 3 h at 37 °C to digest the phosphorylated strand. The biotinylated forward strand DNA was purified by phenol: chloroform: isoamyl alcohol (25: 24: 1) extraction and ethanol precipitation. The precipitated ssDNA fragments were resuspended in HEPES buffer (20 mM HEPES pH 7.5, 100 mM KCl, 1 mM DTT) and folded into G4 structures. Circular Dichroism (CD) was used to determine the conformation of the G4 structures.
To test the effect of G4 length on droplet formation, 300 ng Sm-G4 with varying lengths were added to the buffer G (20 mM HEPES pH 7.5, 100 mM KCl, 1 mM DTT) containing 10% PEG8000 and incubated for 1 h at RT. The solution was transferred onto the glass slide, and images were collected by an FV3000 microscope. For monitoring the internal dynamics of G4 condensation, the 1900 nt Sm-G4 and 202 nt Myc-G4 fragments were labeled with streptavidin-647 (Invitrogen, S21374) or streptavidin-488 (Invitrogen, S11223), respectively. 300 ng Sm-G4-647 or Myc-G4-488 was individually added to the buffer G containing 10% PEG8000, and images were collected at indicated time points by the FV3000 microscope. To examine the fusion ability between different G4 molecules, 300 ng Sm-G4-647 and Myc-G4-488 were added to the buffer G containing 10% PEG8000 and incubated for 1 h at RT. Images were collected by an FV3000 microscope.
G4 and linear DNA purification
To generate G4 substrate, Sm and Myc ssDNA fragments were first dissolved in HEPES buffer (20 mM HEPES pH 7.5, 100 mM KCl, 1 mM DTT) and then denatured at 95 °C for 5 min, followed by slowly cooling down to RT. To make linear DNA, we denature both fragments at 95 °C for 10 min and immediately put the tube on ice to prevent G4 structure formation. The size exclusion column Superdex75 or 200 (GE Healthcare) was used to purify G4 and linear DNA fractions in an AKTA purifier. Circular dichroism (CD) spectra for the Sm-G4 and Sm-linear were recorded at RT on a Chirascan Plus at the wavelength range of 220-350 nm.
EMSA
25 nM of Sm-G4 labeled with ROX dye was mixed with increasing amounts of full-length DHX9 or Y1189C mutant proteins in binding buffer (20 mM HEPES pH 7.5, 100 mM KCl, 1 mM DTT, 5% glycerol) and incubated on ice for 30 min. The samples were then fractionated on a 5% polyacrylamide gel at 4 °C in TBE buffer (40 mM Tris-HCl, PH 8.3, 45 mM boric acid, 1 mM EDTA). The shift bands were visualized at the ROX channel using Typhoon 7000. Bound and free DNA were quantified using ImageJ software, and the binding curves were fitted by GraphPad Prism 8.0 software.
In vitro helicase assay
To examine the G4 unwinding activity of DHX9 and Y1189C/K417R mutants, 100 nM of the ROX-labeled Sm-G4 was dissolved in 10 ml of 20 mM Tris-HCl, pH 7.5, 3.5 mM MgCl2, 3.5 mM ATP, 0.1 mg/ml BSA, 5 mM DTT and 10% (v/v) glycerol. The Sm linear sequence was used as a negative control. After adding the different amounts of DHX9 or Y1189C mutant proteins and incubating at 37 °C for 30 min, the reaction was terminated by snap cooling on ice, followed by the addition of 3.5 ml stop solution (50 mM EDTA, 2% (w/v) SDS, 0.1% (w/v) xylene cyanol, 0.1% (w/v) bromophenol blue, 40% (w/v) glycerol). The samples were fractionated on 5% native polyacrylamide gels and visualized with a ROX channel using a Typhoon 7000. Sm-G4 or Sm-linear DNA was quantified using ImageJ and GraphPad Prism 8.0.
To test the G4 unwinding activity of DHX9 or different mutants in phase state, Sm-G4 was dissolved in phase buffer containing 20 mM HEPES, 150 mM KCl, 1 mM MgCl2, 1 mM DTT with 1 mM ATP or AMP/PNP. DHX9 or mutants were added to the mixture and incubated at 37 °C for 20 min. The G4 structure resolving ability of DHX9 or mutants was examined using CD spectroscopy.
G4 pulldown assay
The 110 nt 5’-biotinylated Sm linear sequence was folded into Sm-G4 in a folding buffer (20 mM HEPES pH 7.5, 100 mM KCl, 1 mM DTT) as previously described 29. The MyOne streptavidin C1 beads (Life Technology, 65002) were washed three times with binding buffer (20 mM Tris-HCl, pH8.0, 500 mM KCl) and then coupled with the biotin-labeled Sm-G4 or Sm-linear for 1 h at RT, respectively. Splenic B cells were ex vivo cultured and stimulated with LPS for 72 h. The cells were lysed in protein lysis buffer (50 mM Tri–HCl pH 7.5, 150 mM NaCl, 5 mM EDTA, 5 mM EGTA, 1 % NP-40, protease inhibitor), and the extract was pre-cleared at 4 °C for 1 h with C1 beads. The pre-cleared lysate was diluted with an equal volume of 2 × binding buffer (20 mM Tris-HCl pH7.5, 2 mM EDTA, 200 mM KCl, 0.2 mM DTT, 10% glycerol, 0.02 mg/ml BSA) and incubated with Sm-DNA-coupled C1 beads for 1 h at 4 °C. After thoroughly washing with lysis buffer three times, the G4-binding proteins were eluted into 1 × LDS sample buffer (Invitrogen, NP0008) for western blot analysis.
ssDRIP-seq and G4 ChIP-seq
ssDRIP-seq was performed as previously described34. G4 ChIP-seq was modified from a previously published protocol66. Briefly, splenic B cells were crosslinked with 1% formaldehyde for 10 min at RT and stopped by adding 125 mM glycine for 10 min. After washing twice with ice-cold PBS, the cells were pellet down at 300 g for 5 min at 4 °C. The cell pellets were resuspended in 1 ml of ice-cold cyto lysis buffer (10 mM Tris-Cl, pH 8.0, 10 mM NaCl, 0.5% NP-40, 1 × protease inhibitor cocktail) and incubated on ice for 10 min with occasional inversion every 2 min. Pellet down cells at 2,300 g for 5 min at 4 °C. The supernatant was discarded, and the remaining nuclear pellet was resuspended in 500 ml of nuclear lysis buffer (1% SDS, 10 mM EDTA, 50 mM Tris-HCl, pH 8.1, 1 × protease inhibitor cocktail). The nuclear lysate was then sonicated seven times at the maximum setting by BRANSON SLPe Sonifier (output setting 4, 10 seconds per cycle). The soluble chromatin was collected by centrifugation at 14,000 rpm for 10 min. After quantification, approximately 1 mg of chromatin (2.5 ml) was transferred to a 1.5 ml Eppendorf LoBind microcentrifuge tube, followed by adding 1 ml of RNase A and 43.5 ml of blocking buffer (1% BSA, 25 mM HEPES, pH 7.5, 10.5 mM NaCl, 110 mM KCl, 1 mM MgCl2) and incubated in a thermomixer at 37 °C for 20 min. Subsequently, 1 mg of BG4 antibody was added to the mixture and incubated by rotation at 1400 rpm in a thermomixer at 16 °C for 1 h. To pulldown BG4 bound chromatin fragments, 10 ml of pre-blocked anti-FLAG M2 magnetic beads (Sigma-Aldrich, M8823) was further applied to the tube and incubated at 16 °C for 1 h. The beads were washed three times with 200 ml of ice-cold wash buffer (100 mM KCl, 10 mM Tris-HCl, pH 7.4, 0.1% Tween-20), and then washed with the wash buffer for additional three times at 1400 rpm for 10 min at 37 °C. Placed the tube on a magnetic stand for 1 min and discarded the supernatant. Elute G4 DNA from the beads by adding 100 ml of TE buffer (10 mM Tris-HCl, pH 8.0, 1 mM EDTA) and 2 ml of proteinase K to the tube and incubated at 37 °C for 2 h at 1400 rpm, followed by de-crosslinking overnight at 65 °C. The G4 DNA in the supernatant was purified with a QIAquick PCR purification kit (Qiagen, 28104) and quantified by Qubit. The purified G4 DNA was converted into a library using the Accel-NGS 1S Plus DNA Library Kit (Swift Biosciences, 10024).
Imaging of DHX9 or G4 condensate in living cells
To monitor G4 condensate in vivo, CH12F3 cells were transfected with 10 pmol of Sm-G4 and Myc-G4 for 12 h, followed by counterstaining with Hoechst33342 for 10 min. The stained CH12F3 cells were transferred to a glass-bottom cell culture dish (NEST, 801002) for imaging by an FV1200 microscope. For examining the phase-separation ability of GFP- and mScarlet-tagged DHX9306-308del, DHX9Y1189C, or WT proteins in vivo, the corresponding CH12F3 cells were individually plated onto a confocal dish for 12 h, and the cells were stained with Hoechst33342 for imaging by FV1200 as described above.
FRAP
For in vitro FRAP experiments, DHX9 proteins or DNA G4 droplets were first formed at room temperature and transferred onto a glass slide. The selected regions were photobleached with 100% laser power by an FV3000 microscope at 488 nm and 647 nm. Images were acquired over a time course of 20 min. Fluorescence intensity in bleached regions was measured by Imaris and further analyzed by GraphPad Prism. For DHX9-related FRAP experiments within cells, the GFP/mScarlet-tagged or 306-308del/Y1189C mutant CH12F3 cells were cultured in a 96-well plate (PerkinElmer, 6055302) for 12 h. The CH12F3 cells were rinsed three times with PBS and stained with Hoechst33342 to visualize the nucleus. A single DHX9-GFP condensate was partially photobleached with 100% laser power by FV1200 on 488 nm and 594 nm. Live-cell images were recorded over a time course of 5 min. For G4 FRAP analysis, CH12F3 cells were transfected with Sm-G4 and cultured for 12 h in a 96-well plate. After counterstaining with Hoechst33342, G4 condensates were partially photobleached with 100% laser power by FV1200 on 594 nm. Cells were imaged over a time course of 5 min.
Patient samples collection
All the subjects enrolled for genetic analysis had written informed consent, and the protocol was approved by the Ethics Committee of Shanghai Jiao Tong University Affiliated Sixth People's Hospital. The studies are strictly in compliance with the Declaration of Helsinki. Bone marrow (BM) samples from subjects were obtained via aspiration, and the lymphocyte subsets were analyzed by FACS. The mononuclear cells were collected by density gradient centrifugation, and subsequently, the genomic DNA was extracted and sequenced by Kingmed Diagnostics. The blood samples from subjects were used for routine analysis, and the serum samples were used to detect antibody levels by ELISA.
Statistical analysis
All the experiments were independently repeated at least twice, and no inconsistent results were observed. GraphPad Prism 8.0 software was used to perform statistical analyses. Data are presented as the mean ± S.D. The box borders in the boxplots and violin plots represent the upper and lower quartiles (25th and 75th percentiles), and the centre line denotes the median. *p < 0.05, **p < 0.01, ****p < 0.0001, and p > 0.05 is considered as non-significant. All the data were reproducible, and details of replicates are described in the figure legends.