4000-520-616
欢迎来到免疫在线!(蚂蚁淘生物旗下平台)  请登录 |  免费注册 |  询价篮
主营:主营:分子类,蛋白类,抗体类,生化类试剂
咨询热线电话
4000-520-616
当前位置: 首页 > 新闻动态 >
新闻详情
...and genomic stability by RAD51AP1 via RAD51 recombinase...
来自 : 发布时间:2024-05-03
ActionsCite Favorites Display options Display options Format Promotion of homologous recombination and GEnomic stability by RAD51AP1 via RAD51 recombinase enhancement 1 Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA. Promotion of homologous recombination and genomic stability by RAD51AP1 via RAD51 recombinase enhancement 1 Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA. Homologous recombination (HR) repairs chromosome damage and is indispensable for tumor suppression in humans. RAD51 mediates the DNA strand-pairing step in HR. RAD51 associated protein 1 (RAD51AP1) is a RAD51-interacting protein whose function has remained elusive. Knockdown of RAD51AP1 in human cells by RNA interference engenders sensitivity to different types of genotoxic stress, and RAD51AP1 is epistatic to the HR protein XRCC3. Moreover, RAD51AP1-depleted cells are impaired for the recombinational repair of a DNA double-strand break and exhibit chromatid breaks both spontaneously and upon DNA-damaging treatment. Purified RAD51AP1 binds both dsDNA and a D loop structure and, only when able to interact with RAD51, greatly stimulates the RAD51-mediated D loop reaction. Biochemical and cytological results show that RAD51AP1 functions at a step subsequent to the assembly of the RAD51-ssDNA nucleoprotein filament. Our findings provide evidence that RAD51AP1 helps maintain genomic integrity via RAD51 recombinase enhancement. Figure 1. Induction of MMC, CPT and X-ray Sensitivity and Chromatid Breaks by RAD51AP1 Knockdown Figure 1. Induction of MMC, CPT and X-ray Sensitivity and Chromatid Breaks by RAD51AP1 Knockdown (A) Survival curves of RAD51AP1-depleted HeLa cells treated with MMC. Non-depleting negative controls: pRNAi, GFP shRNA and mutated shRNA #2. All points with error bars in panels A-E represent the average of at least three different experiments ± 1 SD. * Note: shRNA #1 was tested in triplicate but with 2 µM MMC only, and resulted in the same sensitization as shRNA #2 and #3. (B) Complementation of RAD51AP1-depletion in HeLa cells. Cells expressing both shRNA #2 and EGFP-RAD51AP1res are more resistant to MMC than cells expressing shRNA #2 only. (C) Epistasis between Rad51AP1 and XRCC3. HeLa cells depleted for both XRCC3 and RAD51AP1 show the same sensitivity to MMC as cells depleted for RAD51AP1 only. (D and E) Survival curves of RAD51AP1-depleted asynchronous HeLa cells treated with camptothecin or X-rays. Non-depleting negative controls are GFP shRNA and mutated shRNA #2 for D, or just mutated shRNA #2 for E. Note: shRNAs #1 and #3, which do not sensitize asynchronous HeLa cells to X-rays, do sensitize cells synchronized in S-phase (Fig. S2A). (F) Representative western blot analysis to show the extent of RAD51AP1-depletion observed in HeLa cells with three different shRNAs; probed with anti-GFP antibody. The EGFP-RAD51AP1 fusion protein is a surrogate marker for RAD51AP1 expression. pRNAi is a negative control and QM (a transcription factor) is a loading control. Additional western blots, also demonstrating depletion of endogenous RAD51AP1, are presented in Figs. 2, S1 and S2. (G and H) MMC (50nM)-induced and spontaneous chromatid breaks in RAD51AP1-depleted cells. Mutated shRNA #2 is a non-depleting negative control. In panel H, spontaneous ctbs were scored 72 h and 96 h after RAD51AP1-depletion. Error bars: ± 1 SE for two (G) or three (H) independent experiments. (I) Giemsa-stained metaphase spread of MMC-treated RAD51AP1-depleted HeLa cells. Black arrows: chromatid breaks; red arrow: chromatid-type exchange aberration.Figure 2. RAD51AP1-depletion Impairs Homologous Recombination but not RAD51 DNA Repair Foci Figure 2. RAD51AP1-depletion Impairs Homologous Recombination but not RAD51 DNA Repair Foci (A) TK6-DRGFP cells depleted for RAD51AP1 (shRNA #2 or shRNA #3) or XRCC3 show ∼2-to 2.5-fold lower levels of GFP+ cells (i.e. homologous recombinants) than control cells transfected with either pRNAi or mutated shRNA #2. In four experiments, both negative controls were used and both gave very similar numbers (for example, see Fig. S3C). Therefore, their values were averaged and this average was set to 1.0. In the remaining experiments only mut. shRNA was used as a negative control and this number was set at 1.0. Data for the depleted cells are the mean of the relative fraction of GFP+ cells from 5-7 independent experiments ± 1 SE (see also Fig. S3C). (B and C) Western blot analyses to show the extent of EGFP-RAD51AP1 (here: AP1)-depletion (B) or XRCC3 (here: X3)-depletion (C) in TK6-DRGFP cells. Two hairpins were tested for each gene (lanes 2 and 3 corresponding to shRNA #2 and shRNA #3, respectively for RAD51AP1) and compared to control cells (lanes 1: transfected with mutated shRNA #2). QM: loading control. (D) RAD51 foci form normally in RAD51AP1-depleted HeLa cells after 8 Gy X-rays. Control transfected HeLa cells (mutated shRNA #2; panels A and B) and RAD51AP1-depleted cells (panels C-F) display bright RAD51 foci in ∼ 80 % of the cells analyzed. In contrast, HeLa cells depleted for either RAD51C (panels G and H) or XRCC3 (panels I and J) show impaired RAD51 foci formation (i.e. only some cells form overall smaller and less intense RAD51 foci). The single cell with large foci in panel J may not have been transfected with the XRCC3 shRNA. Cells were fixed and stained at 8 h after 8 Gy X-rays. Two panels for each sample from different areas of the chamber slide are shown. (E) Western blot analysis demonstrating the depletion of RAD51AP1, XRCC3 or RAD51C for the experiment shown in panel D. RAD51 acts as both a non-depleted negative control and as a loading standard. XRCC3 and Rad51C exist as a complex (Wiese et al., 2002a), and depletion of XRCC3 reduces the level of RAD51C and vice versa, as has previously been reported (Lio et al., 2004).(A) Purified GST-RAD51AP1, RAD51AP1, MBP-RAD51AP1, MBP-RAD51AP1 L319Q, MBP-RAD51AP1 H329A,… (A) Purified GST-RAD51AP1, RAD51AP1, MBP-RAD51AP1, MBP-RAD51AP1 L319Q, MBP-RAD51AP1 H329A, and MBP-RAD51AP1 CΔ25 were analyzed by SDS-PAGE and Coomassie Blue staining. (B) GST-tagged RAD51AP1 or GST was incubated with RAD51 or yRad51 and glutathione Sepharose beads were used to capture any protein complex that had formed. The beads were washed and treated with SDS to elute the bound proteins. The supernatant (S), wash (W) and SDS eluate (E) were analyzed by SDS-PAGE with Coomassie Blue staining. (C) MBP-tagged wild type, L319Q, H329A, or CΔ25 RAD51AP1 protein was incubated with RAD51 and amylose agarose beads were used to capture any protein complex that had formed. The analysis was as in (B). (D) RAD51AP1 (0.03 to 1.5 µM) was incubated with ssDNA and dsDNA (panel I) or with dsDNA and the D-loop substrate (panel III). The mobility shift of the DNA substrates was analyzed in a 10% (panel I) or 5% (panel III) polyacrylamide gel. The asterisk denotes the position of the 5′ 32P label. The results from panels I and III were plotted in panels II and IV, respectively.Figure 4. Specific Enhancement of the RAD51-mediated D-loop Reaction by RAD51AP1 (A) Schematic of the D-loop assay. (B) D-loop reactions mediated by combinations of RAD51 (0.8 µM) and RAD51AP1 (0.05 to 1 µM). ATP was omitted from the reaction in lane 10. The results were plotted. (C) D-loop reactions mediated by combinations of RAD51 K133R (0.8 µM) and RAD51AP1 (0.05 to 1 µM). ATP was omitted from the reaction in lane 10. The results were plotted. (D) D-loop reactions mediated by combinations of RAD51 or RAD51 K133R (0.8 µM each) and the MBP tagged form of wild type or mutant RAD51AP1 (0.2 µM each). The results were plotted. Modesti M, Budzowska M, Baldeyron C, Demmers JA, Ghirlando R, Kanaar R. Modesti M, et al. Mol Cell. 2007 Nov 9;28(3):468-81. doi: 10.1016/j.molcel.2007.08.025. Mol Cell. 2007. PMID: 17996710 Liang F, Longerich S, Miller AS, Tang C, Buzovetsky O, Xiong Y, Maranon DG, Wiese C, Kupfer GM, Sung P. Liang F, et al. Cell Rep. 2016 Jun 7;15(10):2118-2126. doi: 10.1016/j.celrep.2016.05.007. Epub 2016 May 26. Cell Rep. 2016. PMID: 27239033 Free PMC article. Liang F, Miller AS, Tang C, Maranon D, Williamson EA, Hromas R, Wiese C, Zhao W, Sung P, Kupfer GM. Liang F, et al. J Biol Chem. 2020 Jun 12;295(24):8186-8194. doi: 10.1074/jbc.RA120.013714. Epub 2020 Apr 29. J Biol Chem. 2020. PMID: 32350107 Free PMC article. Pires E, et al. DNA Repair (Amst). 2017 Nov;59:76-81. doi: 10.1016/j.dnarep.2017.09.008. Epub 2017 Sep 22. DNA Repair (Amst). 2017. PMID: 28963981 Free PMC article. Review. Suwaki N, et al. Semin Cell Dev Biol. 2011 Oct;22(8):898-905. doi: 10.1016/j.semcdb.2011.07.019. Epub 2011 Jul 28. Semin Cell Dev Biol. 2011. PMID: 21821141 Pires E, Sharma N, Selemenakis P, Wu B, Huang Y, Alimbetov DS, Zhao W, Wiese C. Pires E, et al. J Biol Chem. 2021 Jul;297(1):100844. doi: 10.1016/j.jbc.2021.100844. Epub 2021 May 28. J Biol Chem. 2021. PMID: 34058198 Free PMC article. Ouyang J, Yadav T, Zhang JM, Yang H, Rheinbay E, Guo H, Haber DA, Lan L, Zou L. Ouyang J, et al. Nature. 2021 Jun;594(7862):283-288. doi: 10.1038/s41586-021-03538-8. Epub 2021 May 12. Nature. 2021. PMID: 33981036 Dou J, et al. Mol Cell Endocrinol. 2021 Mar 1;523:111137. doi: 10.1016/j.mce.2020.111137. Epub 2020 Dec 25. Mol Cell Endocrinol. 2021. PMID: 33359827 Liu CC, Veeraraghavan J, Tan Y, Kim JA, Wang X, Loo SK, Lee S, Hu Y, Wang XS. Liu CC, et al. Clin Cancer Res. 2021 Feb 1;27(3):785-798. doi: 10.1158/1078-0432.CCR-20-2769. Epub 2020 Nov 10. Clin Cancer Res. 2021. PMID: 33172895 Free PMC article. Bisteau X, Lee J, Srinivas V, Lee JHS, Niska-Blakie J, Tan G, Yap SYX, Hom KW, Wong CK, Chae J, Wang LC, Kim J, Rancati G, Sobota RM, Tan CSH, Kaldis P. Bisteau X, et al. Oncogene. 2020 Oct;39(44):6816-6840. doi: 10.1038/s41388-020-01470-1. Epub 2020 Sep 25. Oncogene. 2020. PMID: 32978522 Free PMC article.

本文链接: http://ge.immuno-online.com/view-7033.html

发布于 : 2024-05-03 阅读()