1
|
Intrathymic dendritic cell-biased precursors promote human T cell lineage specification through IRF8-driven transmembrane TNF. Nat Immunol 2023; 24:474-486. [PMID: 36703005 DOI: 10.1038/s41590-022-01417-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 12/16/2022] [Indexed: 01/27/2023]
Abstract
The cross-talk between thymocytes and thymic stromal cells is fundamental for T cell development. In humans, intrathymic development of dendritic cells (DCs) is evident but its physiological significance is unknown. Here we showed that DC-biased precursors depended on the expression of the transcription factor IRF8 to express the membrane-bound precursor form of the cytokine TNF (tmTNF) to promote differentiation of thymus seeding hematopoietic progenitors into T-lineage specified precursors through activation of the TNF receptor (TNFR)-2 instead of TNFR1. In vitro recapitulation of TNFR2 signaling by providing low-density tmTNF or a selective TNFR2 agonist enhanced the generation of human T cell precursors. Our study shows that, in addition to mediating thymocyte selection and maturation, DCs function as hematopoietic stromal support for the early stages of human T cell development and provide proof of concept that selective targeting of TNFR2 can enhance the in vitro generation of T cell precursors for clinical application.
Collapse
|
2
|
Dutta B, Osato M. The RUNX Family, a Novel Multifaceted Guardian of the Genome. Cells 2023; 12:255. [PMID: 36672189 PMCID: PMC9856552 DOI: 10.3390/cells12020255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 12/24/2022] [Accepted: 12/30/2022] [Indexed: 01/11/2023] Open
Abstract
The DNA repair machinery exists to protect cells from daily genetic insults by orchestrating multiple intrinsic and extrinsic factors. One such factor recently identified is the Runt-related transcription factor (RUNX) family, a group of proteins that act as a master transcriptional regulator for multiple biological functions such as embryonic development, stem cell behaviors, and oncogenesis. A significant number of studies in the past decades have delineated the involvement of RUNX proteins in DNA repair. Alterations in RUNX genes cause organ failure and predisposition to cancers, as seen in patients carrying mutations in the other well-established DNA repair genes. Herein, we review the currently existing findings and provide new insights into transcriptional and non-transcriptional multifaceted regulation of DNA repair by RUNX family proteins.
Collapse
Affiliation(s)
- Bibek Dutta
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
| | - Motomi Osato
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan
| |
Collapse
|
3
|
LncRNA Profiling Reveals That the Deregulation of H19, WT1-AS, TCL6, and LEF1-AS1 Is Associated with Higher-Risk Myelodysplastic Syndrome. Cancers (Basel) 2020; 12:cancers12102726. [PMID: 32977510 PMCID: PMC7598221 DOI: 10.3390/cancers12102726] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/18/2020] [Accepted: 09/20/2020] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Although lncRNAs have been increasingly recognized as regulators of hematopoiesis, only several studies addressed their role in myelodysplastic syndrome (MDS). By genome-wide profiling, we identified lncRNAs deregulated in various groups of MDS patients. We computationally constructed lncRNA-protein coding gene networks to associate deregulated lncRNAs with cellular processes involved in MDS. We showed that expression of H19, WT1-AS, TCL6, and LEF1-AS1 lncRNAs associate with higher-risk MDS and proposed processes related with these transcripts. Abstract Background: myelodysplastic syndrome (MDS) is a hematopoietic stem cell disorder with an incompletely known pathogenesis. Long noncoding RNAs (lncRNAs) play multiple roles in hematopoiesis and represent a new class of biomarkers and therapeutic targets, but information on their roles in MDS is limited. Aims: here, we aimed to characterize lncRNAs deregulated in MDS that may function in disease pathogenesis. In particular, we focused on the identification of lncRNAs that could serve as novel potential biomarkers of adverse outcomes in MDS. Methods: we performed microarray expression profiling of lncRNAs and protein-coding genes (PCGs) in the CD34+ bone marrow cells of MDS patients. Expression profiles were analyzed in relation to different aspects of the disease (i.e., diagnosis, disease subtypes, cytogenetic and mutational aberrations, and risk of progression). LncRNA-PCG networks were constructed to link deregulated lncRNAs with regulatory mechanisms associated with MDS. Results: we found several lncRNAs strongly associated with disease pathogenesis (e.g., H19, WT1-AS, TCL6, LEF1-AS1, EPB41L4A-AS1, PVT1, GAS5, and ZFAS1). Of these, downregulation of LEF1-AS1 and TCL6 and upregulation of H19 and WT1-AS were associated with adverse outcomes in MDS patients. Multivariate analysis revealed that the predominant variables predictive of survival are blast count, H19 level, and TP53 mutation. Coexpression network data suggested that prognosis-related lncRNAs are predominantly related to cell adhesion and differentiation processes (H19 and WT1-AS) and mechanisms such as chromatin modification, cytokine response, and cell proliferation and death (LEF1-AS1 and TCL6). In addition, we observed that transcriptional regulation in the H19/IGF2 region is disrupted in higher-risk MDS, and discordant expression in this locus is associated with worse outcomes. Conclusions: we identified specific lncRNAs contributing to MDS pathogenesis and proposed cellular processes associated with these transcripts. Of the lncRNAs associated with patient prognosis, the level of H19 transcript might serve as a robust marker comparable to the clinical variables currently used for patient stratification.
Collapse
|
4
|
RUNX1 mutations in blast-phase chronic myeloid leukemia associate with distinct phenotypes, transcriptional profiles, and drug responses. Leukemia 2020; 35:1087-1099. [PMID: 32782381 PMCID: PMC8024199 DOI: 10.1038/s41375-020-01011-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 07/26/2020] [Accepted: 07/29/2020] [Indexed: 02/07/2023]
Abstract
Blast-phase chronic myeloid leukemia (BP-CML) is associated with additional chromosomal aberrations, RUNX1 mutations being one of the most common. Tyrosine kinase inhibitor therapy has only limited efficacy in BP-CML, and characterization of more defined molecular subtypes is warranted in order to design better treatment modalities for this poor prognosis patient group. Using whole-exome and RNA sequencing we demonstrate that PHF6 and BCORL1 mutations, IKZF1 deletions, and AID/RAG-mediated rearrangements are enriched in RUNX1mut BP-CML leading to typical mutational signature. On transcriptional level interferon and TNF signaling were deregulated in primary RUNX1mut CML cells and stem cell and B-lymphoid factors upregulated giving a rise to distinct phenotype. This was accompanied with the sensitivity of RUNX1mut blasts to CD19-CAR T cells in ex vivo assays. High-throughput drug sensitivity and resistance testing revealed leukemia cells from RUNX1mut patients to be highly responsive for mTOR-, BCL2-, and VEGFR inhibitors and glucocorticoids. These findings were further investigated and confirmed in CRISPR/Cas9-edited homozygous RUNX1−/− and heterozygous RUNX1−/mut BCR-ABL positive cell lines. Overall, our study provides insights into the pathogenic role of RUNX1 mutations and highlights personalized targeted therapy and CAR T-cell immunotherapy as potentially promising strategies for treating RUNX1mut BP-CML patients.
Collapse
|
5
|
León TE, Rapoz-D'Silva T, Bertoli C, Rahman S, Magnussen M, Philip B, Farah N, Richardson SE, Ahrabi S, Guerra-Assunção JA, Gupta R, Nacheva EP, Henderson S, Herrero J, Linch DC, de Bruin RAM, Mansour MR. EZH2-Deficient T-cell Acute Lymphoblastic Leukemia Is Sensitized to CHK1 Inhibition through Enhanced Replication Stress. Cancer Discov 2020; 10:998-1017. [PMID: 32349972 PMCID: PMC7611258 DOI: 10.1158/2159-8290.cd-19-0789] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 03/13/2020] [Accepted: 04/24/2020] [Indexed: 11/16/2022]
Abstract
Loss-of-function mutations of EZH2, the enzymatic component of PRC2, have been associated with poor outcome and chemotherapy resistance in T-cell acute lymphoblastic leukemia (T-ALL). Using isogenic T-ALL cells, with and without CRISPR/Cas9-induced EZH2-inactivating mutations, we performed a cell-based synthetic lethal drug screen. EZH2-deficient cells exhibited increased sensitivity to structurally diverse inhibitors of CHK1, an interaction that could be validated genetically. Furthermore, small-molecule inhibition of CHK1 had efficacy in delaying tumor progression in isogenic EZH2-deficient, but not EZH2 wild-type, T-ALL cells in vivo, as well as in a primary cell model of PRC2-mutant ALL. Mechanistically, EZH2 deficiency resulted in a gene-expression signature of immature T-ALL cells, marked transcriptional upregulation of MYCN, increased replication stress, and enhanced dependency on CHK1 for cell survival. Finally, we demonstrate this phenotype is mediated through derepression of a distal PRC2-regulated MYCN enhancer. In conclusion, we highlight a novel and clinically exploitable pathway in high-risk EZH2-mutated T-ALL. SIGNIFICANCE: Loss-of-function mutations of PRC2 genes are associated with chemotherapy resistance in T-ALL, yet no specific therapy for this aggressive subtype is currently clinically available. Our work demonstrates that loss of EZH2 activity leads to MYCN-driven replication stress, resulting in increased sensitivity to CHK1 inhibition, a finding with immediate clinical relevance.This article is highlighted in the In This Issue feature, p. 890.
Collapse
Affiliation(s)
- Theresa E León
- Department of Haematology, UCL Cancer Institute, University College London, London, United Kingdom
| | - Tanya Rapoz-D'Silva
- Department of Haematology, UCL Cancer Institute, University College London, London, United Kingdom
| | - Cosetta Bertoli
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Sunniyat Rahman
- Department of Haematology, UCL Cancer Institute, University College London, London, United Kingdom
| | - Michael Magnussen
- Department of Haematology, UCL Cancer Institute, University College London, London, United Kingdom
| | - Brian Philip
- Department of Haematology, UCL Cancer Institute, University College London, London, United Kingdom
| | - Nadine Farah
- Department of Haematology, UCL Cancer Institute, University College London, London, United Kingdom
| | - Simon E Richardson
- Department of Haematology, UCL Cancer Institute, University College London, London, United Kingdom
| | - Sara Ahrabi
- Department of Haematology, UCL Cancer Institute, University College London, London, United Kingdom
| | | | - Rajeev Gupta
- Stem Cell Laboratory, UCL Cancer Institute, University College London, London, United Kingdom
| | - Elisabeth P Nacheva
- Health Service Laboratories LLP, UCL Cancer Institute, London, United Kingdom
| | - Stephen Henderson
- Bill Lyons Informatics Centre, UCL Cancer Institute, University College London, London, United Kingdom
| | - Javier Herrero
- Bill Lyons Informatics Centre, UCL Cancer Institute, University College London, London, United Kingdom
| | - David C Linch
- Department of Haematology, UCL Cancer Institute, University College London, London, United Kingdom
| | - Robertus A M de Bruin
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Marc R Mansour
- Department of Haematology, UCL Cancer Institute, University College London, London, United Kingdom.
| |
Collapse
|
6
|
Seitz V, Kleo K, Dröge A, Schaper S, Elezkurtaj S, Bedjaoui N, Dimitrova L, Sommerfeld A, Berg E, von der Wall E, Müller U, Joosten M, Lenze D, Heimesaat MM, Baldus C, Zinser C, Cieslak A, Macintyre E, Stocking C, Hennig S, Hummel M. Evidence for a role of RUNX1 as recombinase cofactor for TCRβ rearrangements and pathological deletions in ETV6-RUNX1 ALL. Sci Rep 2020; 10:10024. [PMID: 32572036 PMCID: PMC7308335 DOI: 10.1038/s41598-020-65744-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 05/06/2020] [Indexed: 11/08/2022] Open
Abstract
T-cell receptor gene beta (TCRβ) gene rearrangement represents a complex, tightly regulated molecular mechanism involving excision, deletion and recombination of DNA during T-cell development. RUNX1, a well-known transcription factor for T-cell differentiation, has recently been described to act in addition as a recombinase cofactor for TCRδ gene rearrangements. In this work we employed a RUNX1 knock-out mouse model and demonstrate by deep TCRβ sequencing, immunostaining and chromatin immunoprecipitation that RUNX1 binds to the initiation site of TCRβ rearrangement and its homozygous inactivation induces severe structural changes of the rearranged TCRβ gene, whereas heterozygous inactivation has almost no impact. To compare the mouse model results to the situation in Acute Lymphoblastic Leukemia (ALL) we analyzed TCRβ gene rearrangements in T-ALL samples harboring heterozygous Runx1 mutations. Comparable to the Runx1+/- mouse model, heterozygous Runx1 mutations in T-ALL patients displayed no detectable impact on TCRβ rearrangements. Furthermore, we reanalyzed published sequence data from recurrent deletion borders of ALL patients carrying an ETV6-RUNX1 translocation. RUNX1 motifs were significantly overrepresented at the deletion ends arguing for a role of RUNX1 in the deletion mechanism. Collectively, our data imply a role of RUNX1 as recombinase cofactor for both physiological and aberrant deletions.
Collapse
Affiliation(s)
- V Seitz
- Charité University Medicine Berlin, Institute of Pathology, Berlin, Germany
- HS Diagnomics GmbH, Berlin, Germany
| | - K Kleo
- Charité University Medicine Berlin, Institute of Pathology, Berlin, Germany
| | - A Dröge
- HS Diagnomics GmbH, Berlin, Germany
| | | | - S Elezkurtaj
- Charité University Medicine Berlin, Institute of Pathology, Berlin, Germany
| | - N Bedjaoui
- University of Paris, Institute Necker-Enfants Malades (INEM), INSERM U1151, Laboratoire d'Onco-Hematology, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Necker Enfants-Malades, Paris, France
| | - L Dimitrova
- Charité University Medicine Berlin, Institute of Pathology, Berlin, Germany
| | - A Sommerfeld
- Charité University Medicine Berlin, Institute of Pathology, Berlin, Germany
| | - E Berg
- Charité University Medicine Berlin, Institute of Pathology, Berlin, Germany
| | - E von der Wall
- Charité University Medicine Berlin, Institute of Pathology, Berlin, Germany
| | - U Müller
- Heinrich-Pette-Institute, Leibniz-Institute for Experimental Virology, Hamburg, Germany
| | - M Joosten
- Charité University Medicine Berlin, Institute of Pathology, Berlin, Germany
| | - D Lenze
- Charité University Medicine Berlin, Institute of Pathology, Berlin, Germany
| | - M M Heimesaat
- Charité University Medicine Berlin, Institute of Microbiology, Infectious Diseases and Immunology, Berlin, Germany
| | - C Baldus
- University Medical Center Schleswig-Holstein, Department of Internal Medicine II, Kiel, Germany
| | - C Zinser
- Precigen Bioinformatics Germany GmbH, Munich, Germany
| | - A Cieslak
- University of Paris, Institute Necker-Enfants Malades (INEM), INSERM U1151, Laboratoire d'Onco-Hematology, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Necker Enfants-Malades, Paris, France
| | - E Macintyre
- University of Paris, Institute Necker-Enfants Malades (INEM), INSERM U1151, Laboratoire d'Onco-Hematology, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Necker Enfants-Malades, Paris, France
| | - C Stocking
- University Medical Center Eppendorf, Department of Stem Cell Transplantation, Hamburg, Germany
| | - S Hennig
- HS Diagnomics GmbH, Berlin, Germany
| | - M Hummel
- Charité University Medicine Berlin, Institute of Pathology, Berlin, Germany.
| |
Collapse
|
7
|
Patil P, Cieslak A, Bernhart SH, Toprak UH, Wagener R, López C, Wiehle L, Bens S, Altmüller J, Franitza M, Scholz I, Jayne S, Ahearne MJ, Scheffold A, Jebaraj BMC, Schneider C, Costa D, Braun T, Schrader A, Campo E, Dyer MJS, Nürnberg P, Dürig J, Johansson P, Böttcher S, Schlesner M, Herling M, Stilgenbauer S, Macintyre E, Siebert R. Reconstruction of rearranged T-cell receptor loci by whole genome and transcriptome sequencing gives insights into the initial steps of T-cell prolymphocytic leukemia. Genes Chromosomes Cancer 2019; 59:261-267. [PMID: 31677197 DOI: 10.1002/gcc.22821] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 10/29/2019] [Accepted: 10/29/2019] [Indexed: 12/20/2022] Open
Abstract
T-cell prolymphocytic leukemia (T-PLL) is an aggressive tumor with leukemic presentation of mature T-lymphocytes. Here, we aimed at characterizing the initial events in the molecular pathogenesis of T-PLL and particularly, at determining the point in T-cell differentiation when the hallmark oncogenic events, that is, inv(14)(q11q32)/t(14;14)(q11;q32) and t(X;14)(q28;q11) occur. To this end, we mined whole genome and transcriptome sequencing data of 17 and 11 T-PLL cases, respectively. Mapping of the 14q32.1 locus breakpoints identified only TCL1A, which was moreover significantly overexpressed in T-PLL as compared to benign CD4+ and CD8+ T-cells, as the only common oncogenic target of aberrations. In cases with t(14;14), the breakpoints mapped telomeric and in cases with inv(14) centromeric or in the 3'-untranslated region of TCL1A. Regarding the T-cell receptor alpha (TRA) locus-TCL1A breakpoint junctions, all 17 breakpoints involved recombination signal sequences and 15 junctions contained nontemplated (N-) nucleotides. All T-PLL cases studied carried in-frame TRA rearrangements on the intact allele, which skewed significantly toward usage of distal/central TRAV/TRAJ gene segments as compared to the illegitimate TRA rearrangements. Our findings suggest that the oncogenic TRA-TCL1A/MTCP1 rearrangements in T-PLL occur during opening of the TRA locus, that is, during the progression from CD4+ immature single positive to early double positive thymocyte stage, just before physiologic TCL1A expression is silenced. The cell carrying such an oncogenic event continues maturation and rearranges the second TRA allele to achieve a functional T-cell receptor. Thereafter, it switches off RAG and DNTT expression in line with the mature T-cell phenotype at presentation of T-PLL.
Collapse
Affiliation(s)
- Paurnima Patil
- Institute of Human Genetics, University of Ulm and University of Ulm Medical Center, Ulm, Germany
| | - Agata Cieslak
- Diagnostic Haematology, Necker-Enfants Malades Hospital, Assistance Publique Hôpitaux de Paris, Université Paris Descartes Sorbonne Cité, Institut Necker-Enfants Malades (INEM), Institut national de recherche médicale (INSERM), Paris, France
| | - Stephan H Bernhart
- Interdisciplinary Center for Bioinformatics, Transcriptome Bioinformatics, University of Leipzig, Leipzig, Germany
| | - Umut H Toprak
- Bioinformatics and Omics Data Analytics, German Cancer Research Center, Faculty of Biosciences, Heidelberg University, Heidelberg, Germany.,Division Neuroblastoma Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Hopp-Children's Cancer Center at the NCT Heidelberg (KiTZ), Heidelberg, Germany
| | - Rabea Wagener
- Institute of Human Genetics, University of Ulm and University of Ulm Medical Center, Ulm, Germany.,Institute for Human Genetics, Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Cristina López
- Institute of Human Genetics, University of Ulm and University of Ulm Medical Center, Ulm, Germany.,Institute for Human Genetics, Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Laura Wiehle
- Institute of Human Genetics, University of Ulm and University of Ulm Medical Center, Ulm, Germany
| | - Susanne Bens
- Institute of Human Genetics, University of Ulm and University of Ulm Medical Center, Ulm, Germany.,Institute for Human Genetics, Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Janine Altmüller
- Cologne Center for Genomics, Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Marek Franitza
- Cologne Center for Genomics, Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Ingrid Scholz
- Omics IT and Data Management Core Facility, German Cancer Research Center, Heidelberg, Germany
| | - Sandrine Jayne
- Ernest and Helen Scott Haematological Research Institute, Department of Cancer Studies, University of Leicester, Leicester, UK
| | - Matthew J Ahearne
- Ernest and Helen Scott Haematological Research Institute, Department of Cancer Studies, University of Leicester, Leicester, UK
| | - Annika Scheffold
- Department of Internal Medicine III, University of Ulm, Ulm, Germany
| | - Billy M C Jebaraj
- Department of Internal Medicine III, University of Ulm, Ulm, Germany
| | | | - Dolors Costa
- Haematopathology Section, Hospital Clínic, Institut d'Investigaciones Biomèdiques August Pi I Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Till Braun
- Department I of Internal Medicine, Center for Integrated Oncology Köln Bonn, Deutsche CLL Studiengruppe (DCLLSG), Excellence Cluster for Cellular Stress Response and Aging-Associated Diseases (CECAD), and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Alexandra Schrader
- Department I of Internal Medicine, Center for Integrated Oncology Köln Bonn, Deutsche CLL Studiengruppe (DCLLSG), Excellence Cluster for Cellular Stress Response and Aging-Associated Diseases (CECAD), and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Elias Campo
- Haematopathology Section, Hospital Clínic, Institut d'Investigaciones Biomèdiques August Pi I Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Martin J S Dyer
- Ernest and Helen Scott Haematological Research Institute, Department of Cancer Studies, University of Leicester, Leicester, UK
| | - Peter Nürnberg
- Cologne Center for Genomics, Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Jan Dürig
- Department of Hematology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Patricia Johansson
- Department of Hematology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Sebastian Böttcher
- Department III of Internal Medicine, University Hospital Rostock, Rostock, Germany
| | - Matthias Schlesner
- Bioinformatics and Omics Data Analytics, German Cancer Research Center, Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Marco Herling
- Department I of Internal Medicine, Center for Integrated Oncology Köln Bonn, Deutsche CLL Studiengruppe (DCLLSG), Excellence Cluster for Cellular Stress Response and Aging-Associated Diseases (CECAD), and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | | | - Elizabeth Macintyre
- Diagnostic Haematology, Necker-Enfants Malades Hospital, Assistance Publique Hôpitaux de Paris, Université Paris Descartes Sorbonne Cité, Institut Necker-Enfants Malades (INEM), Institut national de recherche médicale (INSERM), Paris, France
| | - Reiner Siebert
- Institute of Human Genetics, University of Ulm and University of Ulm Medical Center, Ulm, Germany.,Institute for Human Genetics, Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
8
|
Clave E, Araujo IL, Alanio C, Patin E, Bergstedt J, Urrutia A, Lopez-Lastra S, Li Y, Charbit B, MacPherson CR, Hasan M, Melo-Lima BL, Douay C, Saut N, Germain M, Trégouët DA, Morange PE, Fontes M, Duffy D, Di Santo JP, Quintana-Murci L, Albert ML, Toubert A. Human thymopoiesis is influenced by a common genetic variant within the TCRA-TCRD locus. Sci Transl Med 2018; 10:10/457/eaao2966. [DOI: 10.1126/scitranslmed.aao2966] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 04/11/2018] [Accepted: 07/25/2018] [Indexed: 12/12/2022]
Abstract
The thymus is the primary lymphoid organ where naïve T cells are generated; however, with the exception of age, the parameters that govern its function in healthy humans remain unknown. We characterized the variability of thymic function among 1000 age- and sex-stratified healthy adults of the Milieu Intérieur cohort, using quantification of T cell receptor excision circles (TRECs) in peripheral blood T cells as a surrogate marker of thymopoiesis. Age and sex were the only nonheritable factors identified that affect thymic function. TREC amounts decreased with age and were higher in women compared to men. In addition, a genome-wide association study revealed a common variant (rs2204985) within the T cell receptor TCRA-TCRD locus, between the DD2 and DD3 gene segments, which associated with TREC amounts. Strikingly, transplantation of human hematopoietic stem cells with the rs2204985 GG genotype into immunodeficient mice led to thymopoiesis with higher TRECs, increased thymocyte counts, and a higher TCR repertoire diversity. Our population immunology approach revealed a genetic locus that influences thymopoiesis in healthy adults, with potentially broad implications in precision medicine.
Collapse
|
9
|
Bhat J, Helmuth J, Chitadze G, Kouakanou L, Peters C, Vingron M, Ammerpohl O, Kabelitz D. Stochastics of Cellular Differentiation Explained by Epigenetics: The Case of T-Cell Differentiation and Functional Plasticity. Scand J Immunol 2017; 86:184-195. [PMID: 28799233 DOI: 10.1111/sji.12589] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 08/06/2017] [Indexed: 12/19/2022]
Abstract
Epigenetic marks including histone modifications and DNA methylation are associated with the regulation of gene expression and activity. In addition, an increasing number of non-coding RNAs with regulatory activity on gene expression have been identified. Alongside, technological advancements allow for the analysis of these mechanisms with high resolution up to the single-cell level. For instance, the assay for transposase-accessible chromatin using sequencing (ATAC-seq) simultaneously probes for chromatin accessibility and nucleosome positioning. Thus, it provides information on two levels of epigenetic regulation. Development and differentiation of T cells into functional subset cells including memory T cells are dynamic processes driven by environmental signals. Here, we briefly review the current knowledge of how epigenetic regulation contributes to subset specification, differentiation and memory development in T cells. Specifically, we focus on epigenetic mechanisms differentially active in the two distinct T cell populations expressing αβ or γδ T cell receptors. We also discuss examples of epigenetic alterations of T cells in autoimmune diseases. DNA methylation and histone acetylation are subject to modification by several classes of 'epigenetic modifiers', some of which are in clinical use or in preclinical development. Therefore, we address the impact of some epigenetic modifiers on T-cell activation and differentiation, and discuss possible synergies with T cell-based immunotherapeutic strategies.
Collapse
Affiliation(s)
- J Bhat
- Institute of Immunology, University of Kiel and University Hospital Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - J Helmuth
- Otto-Warburg-Laboratories: Epigenomics, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - G Chitadze
- Institute of Immunology, University of Kiel and University Hospital Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - L Kouakanou
- Institute of Immunology, University of Kiel and University Hospital Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - C Peters
- Institute of Immunology, University of Kiel and University Hospital Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - M Vingron
- Department of Computational Molecular Biology, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - O Ammerpohl
- Institute of Human Genetics, University of Kiel and University Hospital Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - D Kabelitz
- Institute of Immunology, University of Kiel and University Hospital Schleswig-Holstein Campus Kiel, Kiel, Germany
| |
Collapse
|
10
|
Villarese P, Lours C, Trinquand A, Le Noir S, Belhocine M, Lhermitte L, Cieslak A, Tesio M, Petit A, LeLorch M, Spicuglia S, Ifrah N, Dombret H, Langerak AW, Boissel N, Macintyre E, Asnafi V. TCRα rearrangements identify a subgroup of NKL-deregulated adult T-ALLs associated with favorable outcome. Leukemia 2017; 32:61-71. [PMID: 28592888 DOI: 10.1038/leu.2017.176] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 05/16/2017] [Accepted: 05/25/2017] [Indexed: 12/18/2022]
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) results from leukemic transformation of T-cell precursors arrested at specific differentiation stages, including an 'early-cortical' thymic maturation arrest characterized by expression of cytoplasmic TCRβ but no surface T-cell receptor (TCR) and frequent ectopic expression of the TLX1/3 NK-like homeotic proteins (NKL). We designed a TCRα VJC PCR to identify clonal TCRα rearrangements in 32% of 127 T-ALLs, including 0/52 immature/TCRγδ lineage cases and 41/75 (55%) TCRαβ lineage cases. Amongst the latter, TCRα rearrangements were not identified in 30/54 (56%) of IMβ/pre-αβ early-cortical T-ALLs, of which the majority (21/30) expressed TLX1/3. We reasoned that the remaining T-ALLs might express other NKL proteins, so compared transcript levels of 46 NKL in T-ALL and normal thymic subpopulations. Ectopic overexpression of 10 NKL genes, of which six are unreported in T-ALL (NKX2-3, BARHL1, BARX2, EMX2, LBX2 and MSX2), was detectable in 17/104 (16%) T-ALLs. Virtually all NKL overexpressing T-ALLs were TCRα unrearranged and ectopic NKL transcript expression strongly repressed Eα activity, suggesting that ectopic NKL expression is the major determinant in early-cortical thymic T-ALL maturation arrest. This immunogenetic T-ALL subtype, defined by TCRβ VDJ but no TCRα VJ rearrangement, is associated with a favorable outcome in GRAALL-treated adult T-ALLs.
Collapse
Affiliation(s)
- P Villarese
- Université Paris Descartes Sorbonne Cité, Institut Necker Enfants-Malades (INEM), Institut National de Recherche Médicale (INSERM) U1151, Paris, France.,Laboratory of Onco-Hematology, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Necker Enfants-Malades, Paris, France
| | - C Lours
- Université Paris Descartes Sorbonne Cité, Institut Necker Enfants-Malades (INEM), Institut National de Recherche Médicale (INSERM) U1151, Paris, France.,Laboratory of Onco-Hematology, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Necker Enfants-Malades, Paris, France
| | - A Trinquand
- Université Paris Descartes Sorbonne Cité, Institut Necker Enfants-Malades (INEM), Institut National de Recherche Médicale (INSERM) U1151, Paris, France.,Laboratory of Onco-Hematology, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Necker Enfants-Malades, Paris, France
| | - S Le Noir
- Université Paris Descartes Sorbonne Cité, Institut Necker Enfants-Malades (INEM), Institut National de Recherche Médicale (INSERM) U1151, Paris, France
| | - M Belhocine
- Université Paris Descartes Sorbonne Cité, Institut Necker Enfants-Malades (INEM), Institut National de Recherche Médicale (INSERM) U1151, Paris, France.,Laboratory of Onco-Hematology, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Necker Enfants-Malades, Paris, France.,Aix Marseille Univ, INSERM, TAGC UMR1090, Marseille, France
| | - L Lhermitte
- Université Paris Descartes Sorbonne Cité, Institut Necker Enfants-Malades (INEM), Institut National de Recherche Médicale (INSERM) U1151, Paris, France.,Laboratory of Onco-Hematology, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Necker Enfants-Malades, Paris, France
| | - A Cieslak
- Université Paris Descartes Sorbonne Cité, Institut Necker Enfants-Malades (INEM), Institut National de Recherche Médicale (INSERM) U1151, Paris, France
| | - M Tesio
- Université Paris Descartes Sorbonne Cité, Institut Necker Enfants-Malades (INEM), Institut National de Recherche Médicale (INSERM) U1151, Paris, France
| | - A Petit
- Department of Hematology and Oncologie Pédiatrique, Hôpital Trousseau Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - M LeLorch
- Laboratory of Cytogenetics, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Necker Enfants-Malades, Paris, France
| | - S Spicuglia
- Aix Marseille Univ, INSERM, TAGC UMR1090, Marseille, France
| | - N Ifrah
- Department of Hematology, Centre Hospitalier, Angers, France
| | - H Dombret
- University Paris 7, Hôpital Saint-Louis, AP-HP, Department of Hematology and Institut Universitaire d'Hématologie, Paris, France
| | - A W Langerak
- Department of Immunology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - N Boissel
- University Paris 7, Hôpital Saint-Louis, AP-HP, Department of Hematology and Institut Universitaire d'Hématologie, Paris, France
| | - E Macintyre
- Université Paris Descartes Sorbonne Cité, Institut Necker Enfants-Malades (INEM), Institut National de Recherche Médicale (INSERM) U1151, Paris, France
| | - V Asnafi
- Université Paris Descartes Sorbonne Cité, Institut Necker Enfants-Malades (INEM), Institut National de Recherche Médicale (INSERM) U1151, Paris, France.,Laboratory of Onco-Hematology, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Necker Enfants-Malades, Paris, France
| |
Collapse
|
11
|
Malcolm TIM, Hodson DJ, Macintyre EA, Turner SD. Challenging perspectives on the cellular origins of lymphoma. Open Biol 2016; 6:rsob.160232. [PMID: 27683157 PMCID: PMC5043587 DOI: 10.1098/rsob.160232] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 09/02/2016] [Indexed: 12/18/2022] Open
Abstract
Both B and T lymphocytes have signature traits that set them apart from other cell types. They actively and repeatedly rearrange their DNA in order to produce a unique and functional antigen receptor, they have potential for massive clonal expansion upon encountering antigen via this receptor or its precursor, and they have the capacity to be extremely long lived as ‘memory’ cells. All three of these traits are fundamental to their ability to function as the adaptive immune response to infectious agents, but concurrently render these cells vulnerable to transformation. Thus, it is classically considered that lymphomas arise at a relatively late stage in a lymphocyte's development during the process of modifying diversity within antigen receptors, and when the cell is capable of responding to stimulus via its receptor. Attempts to understand the aetiology of lymphoma have reinforced this notion, as the most notable advances to date have shown chronic stimulation of the antigen receptor by infectious agents or self-antigens to be key drivers of these diseases. Despite this, there is still uncertainty about the cell of origin in some lymphomas, and increasing evidence that a subset arises in a more immature cell. Specifically, a recent study indicates that T-cell lymphoma, in particular nucleophosmin-anaplastic lymphoma kinase-driven anaplastic large cell lymphoma, may originate in T-cell progenitors in the thymus.
Collapse
Affiliation(s)
- Tim I M Malcolm
- Division of Molecular Histopathology, Department of Pathology, University of Cambridge, Lab Block Level 3, Box 231, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Daniel J Hodson
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Elizabeth A Macintyre
- Hematology and INSERM1151, Institut Necker-Enfants Malades, Université Sorbonne Paris Cité at Descartes and Assistance Publique-Hôpitaux de Paris, Paris 75743 Cedex 15, France
| | - Suzanne D Turner
- Division of Molecular Histopathology, Department of Pathology, University of Cambridge, Lab Block Level 3, Box 231, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| |
Collapse
|
12
|
Affiliation(s)
- Agata Cieslak
- Université Paris Descartes Sorbonne Cité, Institut Necker-Enfants Malades (INEM), Institut National de Recherche Médicale (INSERM) U1151, and Laboratory Paris (AP-HP), Hôpital Necker Enfants-Malades, Paris, France
| | - Dominique Payet-Bornet
- Centre d'Immunologie de Marseille-Luminy (CIML), Institut National de la Santé et de la Recherche Médicale (Inserm U631), CNRS UMR6102, Université de la Méditerranée, Marseille, France
| | - Vahid Asnafi
- Université Paris Descartes Sorbonne Cité, Institut Necker-Enfants Malades (INEM), Institut National de Recherche Médicale (INSERM) U1151, and Laboratory Paris (AP-HP), Hôpital Necker Enfants-Malades, Paris, France
| |
Collapse
|
13
|
Zhong X, Wu Y, Liu Y, Zhu F, Li X, Li D, Li Z, Zeng L, Qiao J, Chen X, Xu K. Increased RUNX1 expression in patients with immune thrombocytopenia. Hum Immunol 2016; 77:687-691. [PMID: 27288310 DOI: 10.1016/j.humimm.2016.06.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 06/06/2016] [Accepted: 06/07/2016] [Indexed: 01/10/2023]
Abstract
Immune thrombocytopenia (ITP) is a heterogeneous autoimmune disease, characterized by dysregulation of cellular immunity. Th17 and associated IL-17 were involved in the pathogenesis of ITP. Runt-related transcription factor 1 (RUNX1), a member of the runt domain-containing family of transcription factors, is required for Th17 differentiation. Whether RUNX1 was involved in the pathogenesis of ITP remains poorly understood. In this study, 30 active ITP patients, 20 ITP in remission and 20 age and gender matched healthy controls were included. Peripheral blood mononuclear cells (PBMCs) were isolated to measure mRNA level of RUNX1 and retinoic acid receptor-related orphan receptor-γt (RORγt) by quantitative real-time PCR and Th17 cells by flow cytometry. Meanwhile, plasma was extracted for measurement of IL-17 level by ELISA. Our results showed a significantly higher expression of RUNX1, RORγt, Th17 cells and plasma level of IL-17 in active ITP patients than that in healthy controls. No differences of expression of RUNX1, RORγt and Th17 cells were observed between remission patients and controls. Furthermore, a significantly positive correlation of RUNX1 with RORγt was found in active ITP patients. In conclusion, RUNX1 was associated with the pathogenesis of ITP possibly through regulation of Th17 cell differentiation and therapeutically targeting it might be a novel approach in ITP treatment.
Collapse
Affiliation(s)
- Xiaomin Zhong
- Department of Medical Oncology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an 223300, China
| | - Yulu Wu
- Blood Diseases Institute, Xuzhou Medical College, Xuzhou 221002, China
| | - Yun Liu
- Blood Diseases Institute, Xuzhou Medical College, Xuzhou 221002, China
| | - Feng Zhu
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou 221002, China
| | - Xiaoqian Li
- Blood Diseases Institute, Xuzhou Medical College, Xuzhou 221002, China
| | - Depeng Li
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou 221002, China
| | - Zhenyu Li
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou 221002, China
| | - Lingyu Zeng
- Blood Diseases Institute, Xuzhou Medical College, Xuzhou 221002, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou 221002, China
| | - Jianlin Qiao
- Blood Diseases Institute, Xuzhou Medical College, Xuzhou 221002, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou 221002, China
| | - Xiaofei Chen
- Department of Medical Oncology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an 223300, China.
| | - Kailin Xu
- The First Clinical Medical College, Nanjing Medical University, Nanjing 210029, China; Blood Diseases Institute, Xuzhou Medical College, Xuzhou 221002, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou 221002, China.
| |
Collapse
|
14
|
Bolland DJ, Koohy H, Wood AL, Matheson LS, Krueger F, Stubbington MJT, Baizan-Edge A, Chovanec P, Stubbs BA, Tabbada K, Andrews SR, Spivakov M, Corcoran AE. Two Mutually Exclusive Local Chromatin States Drive Efficient V(D)J Recombination. Cell Rep 2016; 15:2475-87. [PMID: 27264181 PMCID: PMC4914699 DOI: 10.1016/j.celrep.2016.05.020] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 03/01/2016] [Accepted: 05/02/2016] [Indexed: 12/02/2022] Open
Abstract
Variable (V), diversity (D), and joining (J) (V(D)J) recombination is the first determinant of antigen receptor diversity. Understanding how recombination is regulated requires a comprehensive, unbiased readout of V gene usage. We have developed VDJ sequencing (VDJ-seq), a DNA-based next-generation-sequencing technique that quantitatively profiles recombination products. We reveal a 200-fold range of recombination efficiency among recombining V genes in the primary mouse Igh repertoire. We used machine learning to integrate these data with local chromatin profiles to identify combinatorial patterns of epigenetic features that associate with active VH gene recombination. These features localize downstream of VH genes and are excised by recombination, revealing a class of cis-regulatory element that governs recombination, distinct from expression. We detect two mutually exclusive chromatin signatures at these elements, characterized by CTCF/RAD21 and PAX5/IRF4, which segregate with the evolutionary history of associated VH genes. Thus, local chromatin signatures downstream of VH genes provide an essential layer of regulation that determines recombination efficiency. VDJ-seq enables precise quantification of antibody V(D)J recombination products Two distinct cis-regulatory designs characterize actively recombining V genes Putative recombination regulatory elements map downstream of mouse Igh V genes Recombination regulatory architecture reflects the V genes’ evolutionary history
Collapse
Affiliation(s)
- Daniel J Bolland
- Nuclear Dynamics Programme, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Hashem Koohy
- Nuclear Dynamics Programme, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Andrew L Wood
- Nuclear Dynamics Programme, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Louise S Matheson
- Nuclear Dynamics Programme, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Felix Krueger
- Bioinformatics Group, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Michael J T Stubbington
- Nuclear Dynamics Programme, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Amanda Baizan-Edge
- Nuclear Dynamics Programme, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Peter Chovanec
- Nuclear Dynamics Programme, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Bryony A Stubbs
- Nuclear Dynamics Programme, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Kristina Tabbada
- Nuclear Dynamics Programme, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Simon R Andrews
- Bioinformatics Group, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Mikhail Spivakov
- Nuclear Dynamics Programme, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK.
| | - Anne E Corcoran
- Nuclear Dynamics Programme, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK.
| |
Collapse
|
15
|
Voon DCC, Hor YT, Ito Y. The RUNX complex: reaching beyond haematopoiesis into immunity. Immunology 2015; 146:523-36. [PMID: 26399680 DOI: 10.1111/imm.12535] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 07/13/2015] [Accepted: 07/15/2015] [Indexed: 12/24/2022] Open
Abstract
Among their diverse roles as transcriptional regulators during development and cell fate specification, the RUNX transcription factors are best known for the parts they play in haematopoiesis. RUNX proteins are expressed throughout all haematopoietic lineages, being necessary for the emergence of the first haematopoietic stem cells to their terminal differentiation. Although much progress has been made since their discoveries almost two decades ago, current appreciation of RUNX in haematopoiesis is largely grounded in their lineage-specifying roles. In contrast, the importance of RUNX to immunity has been mostly obscured for historic, technical and conceptual reasons. However, this paradigm is likely to shift over time, as a primary purpose of haematopoiesis is to resource the immune system. Furthermore, recent evidence suggests a role for RUNX in the innate immunity of non-haematopoietic cells. This review takes a haematopoiesis-centric approach to collate what is known of RUNX's contribution to the overall mammalian immune system and discuss their growing prominence in areas such as autoimmunity, inflammatory diseases and mucosal immunity.
Collapse
Affiliation(s)
- Dominic Chih-Cheng Voon
- Institute for Frontier Science Initiative, Kanazawa University, Kanazawa, Ishikawa, Japan.,Division of Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa, Japan
| | | | - Yoshiaki Ito
- Cancer Biology Programme, Cancer Science Institute of Singapore, Singapore
| |
Collapse
|
16
|
Recruitment of RAG1 and RAG2 to Chromatinized DNA during V(D)J Recombination. Mol Cell Biol 2015; 35:3701-13. [PMID: 26303526 DOI: 10.1128/mcb.00219-15] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 06/09/2015] [Indexed: 11/20/2022] Open
Abstract
V(D)J recombination is initiated by the binding of the RAG1 and RAG2 proteins to recombination signal sequences (RSSs) that consist of conserved heptamer and nonamer sequences separated by a spacer of either 12 or 23 bp. Here, we used RAG-inducible pro-B v-Abl cell lines in conjunction with chromatin immunoprecipitation to better understand the protein and RSS requirements for RAG recruitment to chromatin. Using a catalytic mutant form of RAG1 to prevent recombination, we did not observe cooperation between RAG1 and RAG2 in their recruitment to endogenous Jκ gene segments over a 48-h time course. Using retroviral recombination substrates, we found that RAG1 was recruited inefficiently to substrates lacking an RSS or containing a single RSS, better to substrates with two 12-bp RSSs (12RSSs) or two 23-bp RSSs (23RSSs), and more efficiently to a substrate with a 12/23RSS pair. RSS mutagenesis demonstrated a major role for the nonamer element in RAG1 binding, and correspondingly, a cryptic RSS consisting of a repeat of CA dinucleotides, which poorly re-creates the nonamer, was ineffective in recruiting RAG1. Our findings suggest that 12RSS-23RSS cooperation (the "12/23 rule") is important not only for regulating RAG-mediated DNA cleavage but also for the efficiency of RAG recruitment to chromatin.
Collapse
|
17
|
Abstract
The modular, noncontiguous architecture of the antigen receptor genes necessitates their assembly through V(D)J recombination. This program of DNA breakage and rejoining occurs during early lymphocyte development, and depends on the RAG1 and RAG2 proteins, whose collaborative endonuclease activity targets specific DNA motifs enriched in the antigen receptor loci. This essential gene shuffling reaction requires lymphocytes to traverse several developmental stages wherein DNA breakage is tolerated, while minimizing the expense to overall genome integrity. Thus, RAG activity is subject to stringent temporal and spatial regulation. The RAG proteins themselves also contribute autoregulatory properties that coordinate their DNA cleavage activity with target chromatin structure, cell cycle status, and DNA repair pathways. Even so, lapses in regulatory restriction of RAG activity are apparent in the aberrant V(D)J recombination events that underlie many lymphomas. In this review, we discuss the current understanding of the RAG endonuclease, its widespread binding in the lymphocyte genome, its noncleavage activities that restrain its enzymatic potential, and the growing evidence of its evolution from an ancient transposase.
Collapse
|
18
|
Carico Z, Krangel MS. Chromatin Dynamics and the Development of the TCRα and TCRδ Repertoires. Adv Immunol 2015; 128:307-61. [DOI: 10.1016/bs.ai.2015.07.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|