Inviting an author to review:
Find an author and click ‘Invite to review selected article’ near their name.
Search for authorsSearch for similar articles
3
views
0
recommends
+1 Recommend
1 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      SETD2 mediates immunotherapy and radiotherapy efficacy via regulating DNA damage responses and genomic stability in lung adenocarcinoma

      brief-report

      Read this article at

      ScienceOpenPublisherPMC
      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          SETD2 is the main transferase for the trimethylation of histone H3 at lysine 36 (H3K36me3) in mammals. SETD2 plays important role in repairing DNA double strand breaks and maintaining chromatin integrity. 1 In renal carcinoma, SETD2 deficiency caused DNA replication fork instability and DNA damage. 2 The absence of SETD2 was also reported to have strong tumor-promoting effects in lung adenocarcinomas (LUAD). 3 However, the other roles of SETD2 remain poorly understood in LUAD. In this study, we utilized comprehensive omics-data analysis to determine that SETD2 was associated with DNA damage and immune-related signals in LUAD. SETD2 knockdown induced DNA damage and cGAS activation in LUAD cells, and reduced the number of cells at the G1 phase. Moreover, SETD2 deficiency was conducive to mutation burden, immune cell infiltration, and immunotherapy responses. High SETD2 expression was associated with high radiocurability. These findings suggest that SETD2 may be a promising biomarker of therapeutic responses for LUAD patients, and offer novel insights into immunotherapy and radiotherapy. In the combined analysis of RNA sequencing (RNA-seq) from non-small cell lung cancer in The Cancer Genome Atlas (TCGA NSCLC), assay for transposase-accessible chromatin (ATAC) sequencing and H3K36me3 chromatin immunoprecipitation sequencing from GSE110318 and Roadmap confirmed that SETD2 was associated with expression and promoter opening of DNA damage checkpoints (Fig. 1A), and that their promoters were covered by H3K36me3 (Fig. 1B). The cGAS/STING pathway senses double-stranded DNA (ds-DNA) and activates the type I interferon signaling and innate immunity. 4 We next analyzed the 3 RNA-seq datasets (GSE89214, GSE75234, and GSE150809) of shSETD2 or H3.3K36 mutation vs. controls. As expected, shSETD2 or H3.3K36 mutation promoted the type I interferon signaling and innate immunity (Fig. 1C–E). Figure 1 SETD2 deficiency induced DNA damage, promoted the cGAS/STING signaling, enhanced genomic mutations and immune cell infiltration, and was related to immunotherapy and radiotherapy. (A) Correlation between SETD2 expressions and ATAC signals of DNA damage checkpoints. (B) H3K36me3 signals of DNA damage checkpoints. (C) GSEA of shSETD2 vs. control in GSE89214. (D) GSEA of control vs. H3.3K36 mutations in GSE75234. (E) GSEA of shSETD2 vs. control in GSE150809. (F) Representative immunofluorescence (200 ×) indicated increased ds-DNA in siSETD2 H1975 cells. (G) The quantitative results of ds-DNA immunofluorescence. (H) Representative immunofluorescence (200 ×) indicated increased pH2AX in siSETD2 H1975 cells. (I) The quantitative results of pH2AX immunofluorescence. (J) qRT-PCR of cGAS/STING pathway components in H1975 cells with or without siSETD2. (K) The frequent genomic mutations in patients with or without SETD2 mutations in TCGA cohorts. (L) Proportion of lymphocyte infiltration of patients with or without SETD2 mutations in the TCGA LUAD cohort. (M–O) Kaplan–Meier analysis of SETD2 mutation vs. WT group in the ICI cohorts. (P) Correlation between SETD2 expression and survival fraction at 2 Gy in GSE20298. (Q) Kaplan–Meier analysis of SETD2 expression in the 3 cohorts with radiotherapy. (R) SETD2 expression and GMscore of patient clusters in the 3 cohorts with radiotherapy. Figure 1 H1975 cells had higher levels of SETD2 than the other NSCLC cells (Fig. S1). Immunofluorescence indicated that SETD2 knockdown significantly increased ds-DNA and pH2AX in H1975 cells, suggesting more DNA damage (Fig. 1F–I). As demonstrated in Figure 1J, siSETD2 had high efficiency, and SETD2 deficiency induced the cGAS/STING pathway. Moreover, SETD2 was associated with cell cycle checkpoints in TCGA NSCLC cohort (Fig. S2A, B). SETD2 had the lowest expression in the G2/M cluster from single cell RNA-seq data GSE131907 (Fig. S2C, D). Less siSETD2 cells were arrested at the G1 phase compared with the control group (Fig. S2E, F). The negative regulation of DNA damage by SETD2 may be potentially beneficial to genomic stability. We next explored the effects of SETD2 mutations on genomic mutation burden, which was a sign of genomic instability. In TCGA pan-cancer datasets, 513 SETD2 mutation sites were found, of which 188 sites with annotations were possibly oncogenic (Fig. S3), suggesting that SETD2 mutations might change its tumor suppressor function. Moreover, we found that driver mutations, but not variant of uncertain significance, were significantly associated with lower expression levels of SETD2 in TCGA cohorts (Fig. S4). The whole gene mutation rates were increased in patients with SETD2 mutations in the TCGA LUAD, LUSC, and pan-cancer cohorts (Fig. 1K). In MSK-IMPACT cohort (pan-cancer, n = 10,945; LUAD, n = 1357; LUSC, n = 170), patients with SETD2 non-synonymous mutations had high genomic mutations (Fig. S5A). Similar results were found in the other 3 independent data sets (Broad, LUAD, n = 183; MSS Mixed Solid Tumors, pan-cancer, n = 249; Cancer Cell Line Encyclopedia, pan-cancer, n = 1739). In colon cancer of the CPTAC-2 cohort, SETD2-mutated patients had higher percentages of microsatellite instability (Fig. S5B). Next, we fairly checked the relations between each cut-off point of SETD2 expressions and genome altered fractions. In Cancer Cell Line Encyclopedia pan-cancer data (n = 1739) and NSCLC data (n = 135, Fig. S6), low expression of SETD2 was associated with increased genome altered fraction in the majority of cut-off points and all significant points. Similar results were also found in TCGA NSCLC (Fig. S7). The cGAS/STING pathway activation and unstable genome may favor immune infiltration. A total of 100 pathological slides were obtained from 10 randomly selected patients with or without SETD2 mutations in TCGA LUAD cohort. We selected 10 images of 910 × 910 pixels in the region of tumor cells for each patient. The representative images were shown in Figure S8. Patients with SETD2 mutations had higher proportions of lymphocyte infiltration (Fig. 1L) and fewer tumor cells, but they were not related to macrophages and stromal cells (Fig. S9). Clinically, Kaplan–Meier analysis indicated that the upregulated SETD2 was associated with longer survival in TCGA LUAD, GSE50081 (NSCLC, n = 172), and Raponi (LUSC, n = 130) cohorts (Fig. S10). Using multivariate Cox regression to correct covariates (pathological stage, age, and gender) in TCGA LUAD, SETD2 expression (P = 0.16), age (P = 0.13) and pathological stage were related to the prognosis (Table S1). High immune infiltration and mutation burden may be beneficial to immune checkpoint inhibitor (ICI) responses. As expected, the SETD2 mutation group showed the favorable prognosis in the ICI cohorts (MSK-IMPACT with anti-PD-1/PD-L1/CTLA-4, pan-cancer, n = 1661; CheckMate-012 with anti-PD1 plus anti-CTLA-4, NSCLC, n = 75; DFCI GSE4573 with anti-CTLA-4, melanoma, n = 110, Fig. 1M−O). Patients with low SETD2 expression had the favorable prognosis in the DFCI cohort with immunotherapy (Fig. S11). High expression of SETD2 attenuated the immunotherapy responses in all the significant cut-off points of SETD2 in the GSE35640 cohort (melanoma with MAGE-A3, n = 65, Fig. S12). Since DNA damage is related to radiation, we speculated that SETD2 was associated with radiotherapy responses. SETD2 expression showed a strong negative correlation with the survival fraction at 2 Gy in colorectal cancer cell data GSE20298 (P = 0.0004, Fig. 1P). The high levels of SETD2 were correlated with superior prognosis in the 3 cohorts with radiotherapy (Fig. 1Q). In terms of multivariate Cox regression, SETD2 expression and pathological stages were the predictors of radiocurability (Table S2). Previous studies suggested that an unstable genome contributed to radiotherapy resistance. 5 To expand the use scenario of genomic mutation, we developed a transcriptome-based genomic mutation score (GMscore). The design process was outlined in Figure S13A. Using only TCGA LUAD for training, our GMscore significantly discriminated between the high- and low-frequent mutation groups in TCGA LUAD, LUSC, BRCA, and SKCM cohorts (Fig. S13B). In the 3 cohorts with radiotherapy, patients were divided into 3 groups (Fig. S13C). The high survival group was associated with the highest SETD2 expression and lowest GMscore in comparison with the middle and low survival groups (Fig. 1R; Fig. S13D). Generally, our work demonstrated that defective SETD2 induced DNA damage, activated the cGAS/STING pathway, and reduced G1 phase cells in LUAD. SETD2 downregulation favored immune infiltration and immunotherapy responses. Moreover, deficient SETD2 was positively correlated with genomic mutations, which was associated with unfavorable radiotherapy prognosis. Conflict of interests The authors declare that they have no competing interests. Funding This work was supported by National Natural Science Foundation, China (No. 81972852, 81800429); Key Research & Development Project of Hubei Province, China (No. 2020BCA069); Nature Science Foundation of Hubei Province, China (No. 2020CFB612); Young and Middle-Aged Medical Backbone Talents of Wuhan, China (No. WHQG201902); Application Foundation Frontier Project of Wuhan, China (No. 2020020601012221); Translational Medicine and Interdisciplinary Research Joint Fund of Zhongnan Hospital of Wuhan University, China (No. ZNJC201922, ZNJC202007).

          Related collections

          Most cited references5

          • Record: found
          • Abstract: found
          • Article: not found

          Cyclic GMP-AMP synthase is a cytosolic DNA sensor that activates the type I interferon pathway.

          The presence of DNA in the cytoplasm of mammalian cells is a danger signal that triggers host immune responses such as the production of type I interferons. Cytosolic DNA induces interferons through the production of cyclic guanosine monophosphate-adenosine monophosphate (cyclic GMP-AMP, or cGAMP), which binds to and activates the adaptor protein STING. Through biochemical fractionation and quantitative mass spectrometry, we identified a cGAMP synthase (cGAS), which belongs to the nucleotidyltransferase family. Overexpression of cGAS activated the transcription factor IRF3 and induced interferon-β in a STING-dependent manner. Knockdown of cGAS inhibited IRF3 activation and interferon-β induction by DNA transfection or DNA virus infection. cGAS bound to DNA in the cytoplasm and catalyzed cGAMP synthesis. These results indicate that cGAS is a cytosolic DNA sensor that induces interferons by producing the second messenger cGAMP.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            SETD2 is required for DNA double-strand break repair and activation of the p53-mediated checkpoint

            Histone modifications establish the chromatin states that coordinate the DNA damage response. In this study, we show that SETD2, the enzyme that trimethylates histone H3 lysine 36 (H3K36me3), is required for ATM activation upon DNA double-strand breaks (DSBs). Moreover, we find that SETD2 is necessary for homologous recombination repair of DSBs by promoting the formation of RAD51 presynaptic filaments. In agreement, SETD2-mutant clear cell renal cell carcinoma (ccRCC) cells displayed impaired DNA damage signaling. However, despite the persistence of DNA lesions, SETD2-deficient cells failed to activate p53, a master guardian of the genome rarely mutated in ccRCC and showed decreased cell survival after DNA damage. We propose that this novel SETD2-dependent role provides a chromatin bookmarking instrument that facilitates signaling and repair of DSBs. In ccRCC, loss of SETD2 may afford an alternative mechanism for the inactivation of the p53-mediated checkpoint without the need for additional genetic mutations in TP53. DOI: http://dx.doi.org/10.7554/eLife.02482.001
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              SETD2 loss-of-function promotes renal cancer branched evolution through replication stress and impaired DNA repair

              Defining mechanisms that generate intratumour heterogeneity and branched evolution may inspire novel therapeutic approaches to limit tumour diversity and adaptation. SETD2 (Su(var), Enhancer of zeste, Trithorax-domain containing 2) trimethylates histone-3 lysine-36 (H3K36me3) at sites of active transcription and is mutated in diverse tumour types, including clear cell renal carcinomas (ccRCCs). Distinct SETD2 mutations have been identified in spatially separated regions in ccRCC, indicative of intratumour heterogeneity. In this study, we have addressed the consequences of SETD2 loss-of-function through an integrated bioinformatics and functional genomics approach. We find that bi-allelic SETD2 aberrations are not associated with microsatellite instability in ccRCC. SETD2 depletion in ccRCC cells revealed aberrant and reduced nucleosome compaction and chromatin association of the key replication proteins minichromosome maintenance complex component (MCM7) and DNA polymerase δ hindering replication fork progression, and failure to load lens epithelium-derived growth factor and the Rad51 homologous recombination repair factor at DNA breaks. Consistent with these data, we observe chromosomal breakpoint locations are biased away from H3K36me3 sites in SETD2 wild-type ccRCCs relative to tumours with bi-allelic SETD2 aberrations and that H3K36me3-negative ccRCCs display elevated DNA damage in vivo. These data suggest a role for SETD2 in maintaining genome integrity through nucleosome stabilization, suppression of replication stress and the coordination of DNA repair.
                Bookmark

                Author and article information

                Contributors
                Journal
                Genes Dis
                Genes Dis
                Genes & Diseases
                Chongqing Medical University
                2352-4820
                2352-3042
                28 March 2022
                March 2023
                28 March 2022
                : 10
                : 2
                : 336-339
                Affiliations
                [a ]Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
                [b ]Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
                [c ]Tumor Precision Diagnosis and Treatment Technology and Translational Medicine, Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
                [d ]Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
                [e ]Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
                Author notes
                []Corresponding author. 169 Donghu Road, Wuhan, Hubei 430071, China. Fax: +86 27 6781 2892. chxie_65@ 123456whu.edu.cn
                [∗∗ ]Corresponding author. 169 Donghu Road, Wuhan, Hubei 430071, China. Fax: +86 27 6781 1471. yan.gong@ 123456whu.edu.cn
                Article
                S2352-3042(22)00072-1
                10.1016/j.gendis.2022.02.016
                10201589
                10e5c4f0-4ed4-4540-b130-ca8aa7a37611
                © 2022 The Authors. Publishing services by Elsevier B.V. on behalf of KeAi Communications Co., Ltd.

                This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

                History
                : 19 December 2021
                : 21 February 2022
                : 22 February 2022
                Categories
                Rapid Communication

                Comments

                Comment on this article