6
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      IKKα Kinase Regulates the DNA Damage Response and Drives Chemo-resistance in Cancer

      research-article

      Read this article at

      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.

          Summary

          Phosphorylated IKKα(p45) is a nuclear active form of the IKKα kinase that is induced by the MAP kinases BRAF and TAK1 and promotes tumor growth independent of canonical NF-κB signaling. Insights into the sources of IKKα(p45) activation and its downstream substrates in the nucleus remain to be defined. Here, we discover that IKKα(p45) is rapidly activated by DNA damage independent of ATM-ATR, but dependent on BRAF-TAK1-p38-MAPK, and is required for robust ATM activation and efficient DNA repair. Abolishing BRAF or IKKα activity attenuates ATM, Chk1, MDC1, Kap1, and 53BP1 phosphorylation, compromises 53BP1 and RIF1 co-recruitment to sites of DNA lesions, and inhibits 53BP1-dependent fusion of dysfunctional telomeres. Furthermore, IKKα or BRAF inhibition synergistically enhances the therapeutic potential of 5-FU and irinotecan to eradicate chemotherapy-resistant metastatic human tumors in vivo. Our results implicate BRAF and IKKα kinases in the DDR and reveal a combination strategy for cancer treatment.

          Graphical Abstract

          Highlights

          • IKKα kinase is activated by BRAF-TAK1-p38-MAPK in response to DNA damage

          • Loss of IKKα or BRAF attenuates ATM signaling and compromises DNA repair

          • Loss of IKKα or BRAF in combination with DNA damage potentiates tumor eradication

          • Combination treatment of patient-derived tumors prolongs survival in mice

          Abstract

          Colomer et al. discover that IKKα kinase contributes to the chemo- and radio-resistance of cancer cells by facilitating ATM activation and DNA repair. BRAF inhibitors prevent damage-induced IKKα activation, leading to the attenuation of ATM signaling and DNA repair. IKKα depletion or BRAF inhibitors combined with 5-FU and irinotecan synergistically enhance the killing of patient-derived xenograft tumors.

          Related collections

          Most cited references22

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

          Wild-type KRAS is required for panitumumab efficacy in patients with metastatic colorectal cancer.

          Panitumumab, a fully human antibody against the epidermal growth factor receptor (EGFR), has activity in a subset of patients with metastatic colorectal cancer (mCRC). Although activating mutations in KRAS, a small G-protein downstream of EGFR, correlate with poor response to anti-EGFR antibodies in mCRC, their role as a selection marker has not been established in randomized trials. KRAS mutations were detected using polymerase chain reaction on DNA from tumor sections collected in a phase III mCRC trial comparing panitumumab monotherapy to best supportive care (BSC). We tested whether the effect of panitumumab on progression-free survival (PFS) differed by KRAS status. KRAS status was ascertained in 427 (92%) of 463 patients (208 panitumumab, 219 BSC). KRAS mutations were found in 43% of patients. The treatment effect on PFS in the wild-type (WT) KRAS group (hazard ratio [HR], 0.45; 95% CI: 0.34 to 0.59) was significantly greater (P < .0001) than in the mutant group (HR, 0.99; 95% CI, 0.73 to 1.36). Median PFS in the WT KRAS group was 12.3 weeks for panitumumab and 7.3 weeks for BSC. Response rates to panitumumab were 17% and 0%, for the WT and mutant groups, respectively. WT KRAS patients had longer overall survival (HR, 0.67; 95% CI, 0.55 to 0.82; treatment arms combined). Consistent with longer exposure, more grade III treatment-related toxicities occurred in the WT KRAS group. No significant differences in toxicity were observed between the WT KRAS group and the overall population. Panitumumab monotherapy efficacy in mCRC is confined to patients with WT KRAS tumors. KRAS status should be considered in selecting patients with mCRC as candidates for panitumumab monotherapy.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            BRCA1-associated exclusion of 53BP1 from DNA damage sites underlies temporal control of DNA repair.

            Following irradiation, numerous DNA-damage-responsive proteins rapidly redistribute into microscopically visible subnuclear aggregates, termed ionising-radiation-induced foci (IRIF). How the enrichment of proteins on damaged chromatin actually relates to DNA repair remains unclear. Here, we use super-resolution microscopy to examine the spatial distribution of BRCA1 and 53BP1 proteins within single IRIF at subdiffraction-limit resolution, yielding an unprecedented increase in detail that was not previously apparent by conventional microscopy. Consistent with a role for 53BP1 in promoting DNA double-strand break repair by non-homologous end joining, 53BP1 enrichment in IRIF is most prominent in the G0/G1 cell cycle phases, where it is enriched in dense globular structures. By contrast, as cells transition through S phase, the recruitment of BRCA1 into the core of IRIF is associated with an exclusion of 53BP1 to the focal periphery, leading to an overall reduction of 53BP1 occupancy at DNA damage sites. Our data suggest that the BRCA1-associated IRIF core corresponds to chromatin regions associated with repair by homologous recombination, and the enrichment of BRCA1 in IRIF represents a temporal switch in the DNA repair program. We propose that BRCA1 antagonises 53BP1-dependent DNA repair in S phase by inhibiting its interaction with chromatin proximal to damage sites. Furthermore, the genomic instability exhibited by BRCA1-deficient cells might result from a failure to efficiently exclude 53BP1 from such regions during S phase.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Nuclear cytokine-activated IKKalpha controls prostate cancer metastasis by repressing Maspin.

              Inflammation enhances tumour promotion through NF-kappaB-dependent mechanisms. NF-kappaB was also proposed to promote metastatogenesis through epithelial-mesenchymal transition. Yet a mechanistic link between inflammation and metastasis is missing. We identified a role for IkappaB kinase alpha (IKKalpha), activated by receptor activator of NF-kappaB (RANK/TNFRSF11A), in mammary epithelial proliferation during pregnancy. Owing to similarities between mammary and prostate epithelia, we examined IKKalpha involvement in prostate cancer and its progression. Here we show that a mutation that prevents IKKalpha activation slows down CaP growth and inhibits metastatogenesis in TRAMP mice, which express SV40 T antigen in the prostate epithelium. Decreased metastasis correlated with elevated expression of the metastasis suppressor Maspin, the ablation of which restored metastatic activity. IKKalpha activation by RANK ligand (RANKL/TNFSF11) inhibits Maspin expression in prostate epithelial cells, whereas repression of Maspin transcription requires nuclear translocation of active IKKalpha. The amount of active nuclear IKKalpha in mouse and human prostate cancer correlates with metastatic progression, reduced Maspin expression and infiltration of prostate tumours with RANKL-expressing inflammatory cells. We propose that tumour-infiltrating RANKL-expressing cells lead to nuclear IKKalpha activation and inhibition of Maspin transcription, thereby promoting the metastatic phenotype.
                Bookmark

                Author and article information

                Contributors
                Journal
                Mol Cell
                Mol. Cell
                Molecular Cell
                Cell Press
                1097-2765
                1097-4164
                22 August 2019
                22 August 2019
                : 75
                : 4
                : 669-682.e5
                Affiliations
                [1 ]Cancer Research Program, Institut Mar d’Investigacions Mèdiques, CIBERONC, Hospital del Mar, Doctor Aiguader 88, Barcelona 08003, Spain
                [2 ]DSB Repair Metabolism Laboratory, The Francis Crick Institute, London NW1 1AT, UK
                [3 ]Translational Research Laboratory, Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), Institut Català d’Oncologia, Hospitalet, Barcelona 08907, Spain
                [4 ]Department of Pathology, Institut Mar d’Investigacions Mèdiques, CIBERONC, Universitat Autònoma de Barcelona, Barcelona 08003, Spain
                [5 ]Department of Oncology, Institut Mar d’Investigacions Mèdiques, Universitat Pompeu Fabra, CIBERONC, Barcelona 08003, Spain
                [6 ]Faculty of Science and Technology, Bioinformatics and Medical Statistics Group, University of Vic-Central University of Catalonia, Vic 08500, Spain
                [7 ]Proteomics Unit, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona 08003, Spain
                [8 ]Proteomics Unit, Universitat Pompeu Fabra, Barcelona 08003, Spain
                Author notes
                []Corresponding author abigas@ 123456imim.es
                [∗∗ ]Corresponding author simon.boulton@ 123456crick.ac.uk
                [∗∗∗ ]Corresponding author lespinosa@ 123456imim.es
                [9]

                Lead Contact

                Article
                S1097-2765(19)30433-2
                10.1016/j.molcel.2019.05.036
                6715775
                31302002
                9ee3f66e-3792-42d4-aeef-0f2955aa5341
                © 2019 The Author(s)

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

                History
                : 25 April 2018
                : 13 March 2019
                : 30 May 2019
                Categories
                Article

                Molecular biology
                ikk,braf,combination therapy,cancer treatment,atm,phosphorylation,dna-damage repair,therapy resistance,patient-derived organoids,ortho-xenografts

                Comments

                Comment on this article