20
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      The Fundamental Role of Chromatin Loop Extrusion in Physiological V(D)J Recombination

      research-article

      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

          RAG endonuclease initiates IgH locus ( Igh) V(D)J assembly in progenitor (pro)-B cells by joining Ds to J Hs, before joining upstream V Hs to DJ H intermediates 1 . In mouse pro-B cells, the CTCF-binding element (CBE)-anchored chromatin loop domain 2 at the 3’end of Igh contains an internal sub-domain spanning the 5’CBE anchor (IGCR1) 3 , the D Hs, and a RAG-bound recombination center (RC) 4 . The RC comprises J H-proximal D (DQ52), 4 J Hs, and the intronic enhancer (“iEμ”) 5 . Robust RAG cleavage is restricted to paired V(D)J segments flanked by complementary recombination signal sequences (12RSSs and 23RSSs) 6 . Ds are flanked downstream and upstream by 12RSSs that, respectively, mediate deletional joining with convergently-oriented J H-23RSSs and V H-23RSSs 6 . Despite 12/23 compatibility, inversional D to J H joining via upstream D-12RSSs is rare 7, 8 . Plasmid-based assays attributed lack of inversional D to J H joining to sequence-based preference for downstream D-12RSSs 9 , as opposed to putative linear scanning mechanisms 10, 11 . Given recent findings that RAG linearly scans convergent CBE-anchored chromatin loops 4, 12- 14 , potentially formed by cohesin-mediated loop extrusion 15- 18 , we revisited a scanning role. Here, we report that J H-23RSS chromosomal orientation programs RC-bound RAG to linearly scan upstream chromatin in the 3’ Igh sub-domain for convergently-oriented D-12RSSs and, thereby, to mediate deletional joining of all Ds, except RC-based DQ52 that joins by a diffusion-related mechanism. In a DQ52-based RC, formed in the absence of J Hs, RAG bound by the downstream DQ52-RSS scans the downstream constant region exon-containing 3’ Igh sub-domain in which scanning can be impeded by targeted nuclease-dead Cas9 (dCas9) binding, by transcription through repetitive Igh switch sequences, and by the 3’ Igh CBE-based loop anchor. Notably, each scanning impediment focally increases RAG activity on potential substrate sequences within the impeded region. High resolution mapping of RC chromatin interactions reveals that such focal RAG targeting is associated with corresponding impediments to the loop extrusion process that drives chromatin past RC-bound RAG.

          Related collections

          Most cited references36

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

          Base-pair-resolution genome-wide mapping of active RNA polymerases using precision nuclear run-on (PRO-seq)

          We provide a protocol for precision nuclear run-on sequencing (PRO-seq) and its variant, PRO-cap, which map the location of active RNA polymerases (PRO-seq) or transcription start sites (TSSs) (PRO-cap) genome-wide at high resolution. The density of RNA polymerases at a particular genomic locus directly reflects the level of nascent transcription at that region. Nuclei are isolated from cells and, under nuclear run-on conditions, transcriptionally engaged RNA polymerases incorporate one or, at most, a few biotin-labeled nucleotide triphosphates (biotin-NTPs) into the 3' end of nascent RNA. The biotin-labeled nascent RNA is used to prepare sequencing libraries, which are sequenced from the 3' end to provide high-resolution positional information for the RNA polymerases. PRO-seq provides much higher sensitivity than ChIP-seq, and it generates a much larger fraction of usable sequence reads than ChIP-seq or NET-seq (native elongating transcript sequencing). Similarly to NET-seq, PRO-seq maps the RNA polymerase at up to base-pair resolution with strand specificity, but unlike NET-seq it does not require immunoprecipitation. With the protocol provided here, PRO-seq (or PRO-cap) libraries for high-throughput sequencing can be generated in 4-5 working days. The method has been applied to human, mouse, Drosophila melanogaster and Caenorhabditis elegans cells and, with slight modifications, to yeast.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Mechanisms of programmed DNA lesions and genomic instability in the immune system.

            Chromosomal translocations involving antigen receptor loci are common in lymphoid malignancies. Translocations require DNA double-strand breaks (DSBs) at two chromosomal sites, their physical juxtaposition, and their fusion by end-joining. Ability of lymphocytes to generate diverse repertoires of antigen receptors and effector antibodies derives from programmed genomic alterations that produce DSBs. We discuss these lymphocyte-specific processes, with a focus on mechanisms that provide requisite DSB target specificity and mechanisms that suppress DSB translocation. We also discuss recent work that provides new insights into DSB repair pathways and the influences of three-dimensional genome organization on physiological processes and cancer genomes. Copyright © 2013 Elsevier Inc. All rights reserved.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              The Energetics and Physiological Impact of Cohesin Extrusion

              Cohesin extrusion is thought to play a central role in establishing the architecture of mammalian genomes. However, extrusion has not been visualized in vivo, and thus, its functional impact and energetics are unknown. Using ultra-deep Hi-C, we show that loop domains form by a process that requires cohesin ATPases. Once formed, however, loops and compartments are maintained for hours without energy input. Strikingly, without ATP, we observe the emergence of hundreds of CTCF-independent loops that link regulatory DNA. We also identify architectural "stripes," where a loop anchor interacts with entire domains at high frequency. Stripes often tether super-enhancers to cognate promoters, and in B cells, they facilitate Igh transcription and recombination. Stripe anchors represent major hotspots for topoisomerase-mediated lesions, which promote chromosomal translocations and cancer. In plasmacytomas, stripes can deregulate Igh-translocated oncogenes. We propose that higher organisms have coopted cohesin extrusion to enhance transcription and recombination, with implications for tumor development.
                Bookmark

                Author and article information

                Journal
                0410462
                6011
                Nature
                Nature
                Nature
                0028-0836
                1476-4687
                6 November 2019
                11 September 2019
                September 2019
                11 March 2020
                : 573
                : 7775
                : 600-604
                Affiliations
                [1 ]Howard Hughes Medical Institute, Program in Cellular and Molecular Medicine, Boston Children’s Hospital, and Department of Genetics, Harvard Medical School, Boston, MA 02115, USA.
                [2 ]Immunobiology Center, Department of Biomedical Sciences, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI 49007, USA.
                [3 ]The Center for Genome Architecture, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA.
                [4 ]Department of Bioengineering, Rice University, Houston, TX 77030, USA.
                [5 ]Center for Theoretical Biological Physics and Department of Computer Science, Rice University, Houston, TX 77030, USA
                [6 ]Medical Scientist Training Program, Baylor College of Medicine, Houston, TX 77030, USA.
                [7 ]Department of Pediatrics, Texas Children's Hospital, Houston, TX 77030, USA.
                [8 ]Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA
                [9 ]Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
                Author notes
                Correspondence and requests for materials should be addressed to F.W.A ( alt@ 123456enders.tch.harvard.edu ).

                AUTHOR CONTRIBUTIONS

                Y.Z., X.Z., and F.W.A. designed the study; Y.Z., X.Z., Z.L., H.H., J.L, and E.D performed experiments, except for Hi-C experiments which were performed by A.P-A and analyzed by A.P-A, M.S.S. and E.L-A. Z.B. provided critical reagents and advice on 3C-HTGTS. N.K. and J.Z. designed some bioinformatics pipelines; Y.Z., X.Z., and F.W.A analyzed and interpreted all data other than Hi-C data. Y.Z., X.Z., and F.W.A. designed figures and wrote the manuscript. Z.B, Z.L, H.H, J.L, A.P-A, and E.L-A helped polish the manuscript.

                [10]

                These authors contributed equally: Yu Zhang, Xuefei Zhang.

                [*]

                Present address: Immunobiology Center, Department of Biomedical Sciences, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI 49007, USA.

                Article
                NIHMS1537247
                10.1038/s41586-019-1547-y
                6867615
                31511698
                ff2048ab-bb94-4e20-889e-49505f62e3ae

                Users may view, print, copy, and download text and data-mine the content in such documents, for the purposes of academic research, subject always to the full Conditions of use: http://www.nature.com/authors/editorial_policies/license.html#terms

                History
                Categories
                Article

                Uncategorized
                Uncategorized

                Comments

                Comment on this article