2
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      An Artificial CTCF Peptide Triggers Efficient Therapeutic Efficacy in Ocular Melanoma

      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

          Although CCCTC binding factor (CTCF) has been demonstrated to play a variety of often contradictory roles in tumorigenesis, little is known about its function in the tumorigenesis of ocular melanoma. Here, we generated two artificial CTCF peptides (Decoy-CTCFs) combining the zinc finger domain of wild-type CTCF and artificial marker region. This Decoy-CTCF retained the DNA binding region but lost the functional regions of wild-type CT CF. Transferring artificial CTCF into ocular melanoma cells suppressed proliferation and migration in the tumor cells, while no effect was observed in normal cells. Intriguingly, we first showed that decoy-CTCF inhibited tumorigenesis by preventing the histone acetyltransferase EP300 from binding to the promoter of SELL. Thus SELL was a novel oncogene in the tumorigenesis of ocular melanoma. These studies provide efficient decoy CTCF-based therapeutic concept in malignant ocular melanoma and reveal the potential mechanism underlying decoy-based tumor therapy.

          Graphical Abstract

          Abstract

          An artificial CTCF peptide (Decoy-CTCFs) prevents the histone acetyltransferase EP300 to interact the promoter of SELL for inactivating SELL expression and triggering efficient therapeutic efficacy in ocular tumors. These studies delineate a novel decoy CTCF-guided mechanism in tumor therapy and provide a promising therapeutic concept for ocular melanoma.

          Related collections

          Most cited references26

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

          Endogenous miRNA sponge lincRNA-RoR regulates Oct4, Nanog, and Sox2 in human embryonic stem cell self-renewal.

          The embryonic stem cell (ESC) transcriptional and epigenetic networks are controlled by a multilayer regulatory circuitry, including core transcription factors (TFs), posttranscriptional modifier microRNAs (miRNAs), and some other regulators. However, the role of large intergenic noncoding RNAs (lincRNAs) in this regulatory circuitry and their underlying mechanism remains undefined. Here, we demonstrate that a lincRNA, linc-RoR, may function as a key competing endogenous RNA to link the network of miRNAs and core TFs, e.g., Oct4, Sox2, and Nanog. We show that linc-RoR shares miRNA-response elements with these core TFs and that linc-RoR prevents these core TFs from miRNA-mediated suppression in self-renewing human ESC. We suggest that linc-RoR forms a feedback loop with core TFs and miRNAs to regulate ESC maintenance and differentiation. These results may provide insights into the functional interactions of the components of genetic networks during development and may lead to new therapies for many diseases. Copyright © 2013 Elsevier Inc. All rights reserved.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            The protein CTCF is required for the enhancer blocking activity of vertebrate insulators.

            An insulator is a DNA sequence that can act as a barrier to the influences of neighboring cis-acting elements, preventing gene activation, for example, when located between an enhancer and a promoter. We have identified a 42 bp fragment of the chicken beta-globin insulator that is both necessary and sufficient for enhancer blocking activity in human cells. We show that this sequence is the binding site for CTCF, a previously identified eleven-zinc finger DNA-binding protein that is highly conserved in vertebrates. CTCF sites are present in all of the vertebrate enhancer-blocking elements we have examined. We suggest that directional enhancer blocking by CTCF is a conserved component of gene regulation in vertebrates.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              CTCF mediates long-range chromatin looping and local histone modification in the beta-globin locus.

              CTCF (CCCTC-binding factor) binds sites around the mouse beta-globin locus that spatially cluster in the erythroid cell nucleus. We show that both conditional deletion of CTCF and targeted disruption of a DNA-binding site destabilize these long-range interactions and cause local loss of histone acetylation and gain of histone methylation, apparently without affecting transcription at the locus. Our data demonstrate that CTCF is directly involved in chromatin architecture and regulates local balance between active and repressive chromatin marks. We postulate that throughout the genome, relative position and stability of CTCF-mediated loops determine their effect on enhancer-promoter interactions, with gene insulation as one possible outcome.
                Bookmark

                Author and article information

                Contributors
                Journal
                Mol Ther Oncolytics
                Mol Ther Oncolytics
                Molecular Therapy Oncolytics
                American Society of Gene & Cell Therapy
                2372-7705
                09 July 2020
                25 September 2020
                09 July 2020
                : 18
                : 317-325
                Affiliations
                [1 ]Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200025, P.R. China
                [2 ]Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Science and Technology, Tongji University, Shanghai 200092, P.R. China
                [3 ]Frontier Science Research Center for Stem Cells, Tongji University, Shanghai 200092, P.R. China
                Author notes
                []Corresponding author: He Zhang, PhD, Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Science and Technology, Tongji University, Shanghai 200092, P.R. China. zhanghe@ 123456tongji.edu.cn
                [∗∗ ]Corresponding author: Xianqun Fan, MD, PhD, Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200025, P.R. China. fanxq@ 123456sjtu.edu.cn
                [4]

                These authors contributed equally to this work.

                Article
                S2372-7705(20)30108-X
                10.1016/j.omto.2020.07.004
                7394857
                e5b13cd1-0b03-43b9-aa61-3a57eb01c93b
                © 2020 The Authors

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

                History
                : 28 March 2020
                : 4 July 2020
                Categories
                Article

                decoy ctcf,ocular melanoma,therapeutic efficacy
                decoy ctcf, ocular melanoma, therapeutic efficacy

                Comments

                Comment on this article