7
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      IKZF1 selectively enhances homologous recombination repair by interacting with CtIP and USP7 in multiple myeloma

      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.

          Abstract

          Rationale: In multiple myeloma (MM), the activities of non-homologous end joining (NHEJ) and homologous recombination repair (HR) are increased compared with healthy controls. Whether and how IKZF1 as an enhancer of MM participates in the DNA repair pathway of tumor cells remains elusive.

          Methods: We used an endonuclease AsiSI-based system and quantitative chromatin immunoprecipitation assay (qChIP) analysis to test whether IKZF1 is involved in DNA repair. Immunopurification and mass spectrometric (MS) analysis were performed in MM1.S cells to elucidate the molecular mechanism that IKZF1 promotes DNA damage repair. The combination effect of lenalidomide or USP7 inhibitor with PARP inhibitor on cell proliferation was evaluated using MM cells in vitro and in vivo.

          Results: We demonstrate that IKZF1 specifically promotes homologous recombination DNA damage repair in MM cells, which is regulated by its interaction with CtIP and USP7. In this process, USP7 could regulate the stability of IKZF1 through its deubiquitinating activity. The N-terminal zinc finger domains of IKZF1 and the ubiquitin-like domain of USP7 are necessary for their interaction. Furthermore, targeted inhibition IKZF1 or USP7 could sensitize MM cells to PARP inhibitor treatment in vitro and in vivo.

          Conclusions: Our findings identify USP7 as a deubiquitinating enzyme for IKZF1 and uncover a new function of IKZF1 in DNA damage repair. In translational perspective, the combination inhibition of IKZF1 or USP7 with PARP inhibitor deserves further evaluation in clinical trials for the treatment of MM.

          Related collections

          Most cited references44

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

          PARP inhibitors: Synthetic lethality in the clinic.

          PARP inhibitors (PARPi), a cancer therapy targeting poly(ADP-ribose) polymerase, are the first clinically approved drugs designed to exploit synthetic lethality, a genetic concept proposed nearly a century ago. Tumors arising in patients who carry germline mutations in either BRCA1 or BRCA2 are sensitive to PARPi because they have a specific type of DNA repair defect. PARPi also show promising activity in more common cancers that share this repair defect. However, as with other targeted therapies, resistance to PARPi arises in advanced disease. In addition, determining the optimal use of PARPi within drug combination approaches has been challenging. Nevertheless, the preclinical discovery of PARPi synthetic lethality and the route to clinical approval provide interesting lessons for the development of other therapies. Here, we discuss current knowledge of PARP inhibitors and potential ways to maximize their clinical effectiveness.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            DNA double-strand break repair-pathway choice in somatic mammalian cells

            The major pathways of DNA double strand break (DSB) repair have key roles in suppressing genomic instability. However, if deployed in an inappropriate cellular context, these same repair functions can mediate chromosome rearrangements that underlie various human diseases, ranging from developmental disorders to cancer. Two major mechanisms of DSB repair predominate in mammalian cells, namely homologous recombination and non-homologous end joining. In this Review, we outline a ‘decision tree’ of DSB repair pathway choice in somatic mammalian cells, and consider how DSB repair dysfunction can lead to genomic instability. Stalled or broken replication forks present a distinctive challenge to the DSB repair system. Emerging evidence suggests that the ‘rules’ governing stalled fork repair pathway choice differ from those that operate at a conventional DSB.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Regulation of DNA double-strand break repair pathway choice.

              DNA double-strand breaks (DSBs) are critical lesions that can result in cell death or a wide variety of genetic alterations including large- or small-scale deletions, loss of heterozygosity, translocations, and chromosome loss. DSBs are repaired by non-homologous end-joining (NHEJ) and homologous recombination (HR), and defects in these pathways cause genome instability and promote tumorigenesis. DSBs arise from endogenous sources including reactive oxygen species generated during cellular metabolism, collapsed replication forks, and nucleases, and from exogenous sources including ionizing radiation and chemicals that directly or indirectly damage DNA and are commonly used in cancer therapy. The DSB repair pathways appear to compete for DSBs, but the balance between them differs widely among species, between different cell types of a single species, and during different cell cycle phases of a single cell type. Here we review the regulatory factors that regulate DSB repair by NHEJ and HR in yeast and higher eukaryotes. These factors include regulated expression and phosphorylation of repair proteins, chromatin modulation of repair factor accessibility, and the availability of homologous repair templates. While most DSB repair proteins appear to function exclusively in NHEJ or HR, a number of proteins influence both pathways, including the MRE11/RAD50/NBS1(XRS2) complex, BRCA1, histone H2AX, PARP-1, RAD18, DNA-dependent protein kinase catalytic subunit (DNA-PKcs), and ATM. DNA-PKcs plays a role in mammalian NHEJ, but it also influences HR through a complex regulatory network that may involve crosstalk with ATM, and the regulation of at least 12 proteins involved in HR that are phosphorylated by DNA-PKcs and/or ATM.
                Bookmark

                Author and article information

                Journal
                Int J Biol Sci
                Int J Biol Sci
                ijbs
                International Journal of Biological Sciences
                Ivyspring International Publisher (Sydney )
                1449-2288
                2022
                21 March 2022
                : 18
                : 6
                : 2515-2526
                Affiliations
                [1 ]Hongqiao International Institute of Medicine, Shanghai Tongren Hospital / Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
                [2 ]Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.
                [3 ]Department of Ultrasound, Second Affiliated Hospital of Zhejiang University, Hangzhou 310009, China.
                [4 ]Department of Hematology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province 213003, China.
                [5 ]Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.
                [6 ]Department of Oncology, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201999, China.
                [7 ]Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai 200433, China.
                Author notes
                ✉ Corresponding authors: Yingli Wu, M.D., Ph.D., Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China. Phone: 0086-21-63846590-776916; Fax: 0086-21-64154900, E-mail: wuyingli@ 123456shsmu.edu.cn ; Fenghou Gao, Ph.D., Department of Oncology, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201999, China. Phone: 0086-21-56691101-6964; Fax: 0086-21-56693614, E-mail: fenghougao@ 123456163.com ; Wei Liu, Ph.D., Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China. Phone: 0086-21-36126284; Fax: 0086-21-63243749, E-mail: bsjys@ 123456shsmu.edu.cn .

                #These authors contributed equally to this work.

                Competing Interests: The authors have declared that no competing interest exists.

                Article
                ijbsv18p2515
                10.7150/ijbs.70960
                8990476
                35414773
                7f15ec72-8d10-4293-89e0-ea7ebadfcc87
                © The author(s)

                This is an open access article distributed under the terms of the Creative Commons Attribution License ( https://creativecommons.org/licenses/by/4.0/). See http://ivyspring.com/terms for full terms and conditions.

                History
                : 12 January 2022
                : 7 March 2022
                Categories
                Research Paper

                Life sciences
                ikzf1,dna repair,hr,usp7,multiple myeloma
                Life sciences
                ikzf1, dna repair, hr, usp7, multiple myeloma

                Comments

                Comment on this article