1
|
Fujisaki K, Okazaki S, Ogawa S, Takeda M, Sugihara E, Imai K, Mizuno S, Takahashi S, Goitsuka R. B Cells of Early-life Origin Defined by RAG2-based Lymphoid Cell Tracking under Native Hematopoietic Conditions. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:296-305. [PMID: 38874543 DOI: 10.4049/jimmunol.2400072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 05/22/2024] [Indexed: 06/15/2024]
Abstract
During the perinatal period, the immune system sets the threshold to select either response or tolerance to environmental Ags, which leads to the potential to provide a lifetime of protection and health. B-1a B cells have been demonstrated to develop during this perinatal time window, showing a unique and restricted BCR repertoire, and these cells play a major role in natural Ab secretion and immune regulation. In the current study, we developed a highly efficient temporally controllable RAG2-based lymphoid lineage cell labeling and tracking system and applied this system to understand the biological properties and contribution of B-1a cells generated at distinct developmental periods to the adult B-1a compartments. This approach revealed that B-1a cells with a history of RAG2 expression during the embryonic and neonatal periods dominate the adult B-1a compartment, including those in the bone marrow (BM), peritoneal cavity, and spleen. Moreover, the BCR repertoire of B-1a cells with a history of RAG2 expression during the embryonic period was restricted, becoming gradually more diverse during the neonatal period, and then heterogeneous at the adult stage. Furthermore, more than half of plasmablasts/plasma cells in the adult BM had embryonic and neonatal RAG2 expression histories. Moreover, BCR analysis revealed a high relatedness between BM plasmablasts/plasma cells and B-1a cells derived from embryonic and neonatal periods, suggesting that these cell types have a common origin. Taken together, these findings define, under native hematopoietic conditions, the importance in adulthood of B-1a cells generated during the perinatal period.
Collapse
Affiliation(s)
- Keiko Fujisaki
- Division of Cell Fate Regulation, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Shogo Okazaki
- Department of Microbiology and Immunology, Nihon University School of Dentistry, Tokyo, Japan
| | - Shuhei Ogawa
- Division of Integrated Research, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Miyama Takeda
- Division of Cell Fate Regulation, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Eiji Sugihara
- Open Facility Center and Cancer Center, Fujita Health University, Aichi, Japan
| | - Kenichi Imai
- Department of Microbiology and Immunology, Nihon University School of Dentistry, Tokyo, Japan
| | - Seiya Mizuno
- Laboratory Animal Resource Center, Transborder Medical Research Center, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Satoru Takahashi
- Laboratory Animal Resource Center, Transborder Medical Research Center, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
- Department of Anatomy and Embryology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Ryo Goitsuka
- Division of Cell Fate Regulation, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| |
Collapse
|
2
|
Yang Y, Liu L, Tucker HO. The malignant transformation potential of the oncogene STYK1/NOK at early lymphocyte development in transgenic mice. Biochem Biophys Rep 2024; 38:101709. [PMID: 38638675 PMCID: PMC11024497 DOI: 10.1016/j.bbrep.2024.101709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 04/01/2024] [Accepted: 04/05/2024] [Indexed: 04/20/2024] Open
Abstract
B-cell Chronic Lymphocytic Leukemia (B-CLL) is a malignancy caused by the clonal expansion of mature B lymphocytes bearing a CD5+CD19+ (B1) phenotype. However, the origin of B-CLL remains controversial. We showed previously that STYK1/NOK transgenic mice develop a CLL-like disease. Using this model system in this study, we attempt to define the stage of CLL initiation. Here, we show that the phenotype of STYK1/NOK-induced B-CLL is heterogeneous. The expanded B1 lymphocyte pool was detected within peripheral lymphoid organs and was frequently associated with the expansions of memory B cells. Despite this immunophenotypic heterogeneity, suppression of B cell development at an early stage consistently occurred within the bone marrow (BM) of STYK1/NOK-tg mice. Overall, we suggest that enforced expression of STYK1/NOK in transgenic mice might significantly predispose BM hematopoietic stem cells (HSCs) towards the development of B-CLL.
Collapse
Affiliation(s)
- Yin Yang
- Department of Pathogen Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
| | - Li Liu
- Department of Pathogen Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
| | - Haley O. Tucker
- Molecular Biosciences, Institute for Cellular and Molecular Biology, University of Texas at Austin, 1 University Station A5000, Austin, TX, 78712, USA
| |
Collapse
|
3
|
Luo S, Jing C, Ye AY, Kratochvil S, Cottrell CA, Koo JH, Chapdelaine Williams A, Francisco LV, Batra H, Lamperti E, Kalyuzhniy O, Zhang Y, Barbieri A, Manis JP, Haynes BF, Schief WR, Batista FD, Tian M, Alt FW. Humanized V(D)J-rearranging and TdT-expressing mouse vaccine models with physiological HIV-1 broadly neutralizing antibody precursors. Proc Natl Acad Sci U S A 2023; 120:e2217883120. [PMID: 36574685 PMCID: PMC9910454 DOI: 10.1073/pnas.2217883120] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 11/22/2022] [Indexed: 12/28/2022] Open
Abstract
Antibody heavy chain (HC) and light chain (LC) variable region exons are assembled by V(D)J recombination. V(D)J junctional regions encode complementarity-determining-region 3 (CDR3), an antigen-contact region immensely diversified through nontemplated nucleotide additions ("N-regions") by terminal deoxynucleotidyl transferase (TdT). HIV-1 vaccine strategies seek to elicit human HIV-1 broadly neutralizing antibodies (bnAbs), such as the potent CD4-binding site VRC01-class bnAbs. Mice with primary B cells that express receptors (BCRs) representing bnAb precursors are used as vaccination models. VRC01-class bnAbs uniformly use human HC VH1-2 and commonly use human LCs Vκ3-20 or Vκ1-33 associated with an exceptionally short 5-amino-acid (5-aa) CDR3. Prior VRC01-class models had nonphysiological precursor levels and/or limited precursor diversity. Here, we describe VRC01-class rearranging mice that generate more physiological primary VRC01-class BCR repertoires via rearrangement of VH1-2, as well as Vκ1-33 and/or Vκ3-20 in association with diverse CDR3s. Human-like TdT expression in mouse precursor B cells increased LC CDR3 length and diversity and also promoted the generation of shorter LC CDR3s via N-region suppression of dominant microhomology-mediated Vκ-to-Jκ joins. Priming immunization with eOD-GT8 60mer, which strongly engages VRC01 precursors, induced robust VRC01-class germinal center B cell responses. Vκ3-20-based responses were enhanced by N-region addition, which generates Vκ3-20-to-Jκ junctional sequence combinations that encode VRC01-class 5-aa CDR3s with a critical E residue. VRC01-class-rearranging models should facilitate further evaluation of VRC01-class prime and boost immunogens. These new VRC01-class mouse models establish a prototype for the generation of vaccine-testing mouse models for other HIV-1 bnAb lineages that employ different HC or LC Vs.
Collapse
Affiliation(s)
- Sai Luo
- HHMI, Boston Children's Hospital, Boston, MA 02115
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115
- Department of Genetics, Harvard Medical School, Boston, MA 02115
| | - Changbin Jing
- HHMI, Boston Children's Hospital, Boston, MA 02115
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115
- Department of Genetics, Harvard Medical School, Boston, MA 02115
| | - Adam Yongxin Ye
- HHMI, Boston Children's Hospital, Boston, MA 02115
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115
- Department of Genetics, Harvard Medical School, Boston, MA 02115
| | - Sven Kratochvil
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139
| | - Christopher A Cottrell
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, San Diego, CA 92037
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, San Diego, CA 92037
- Center for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, San Diego, CA 92037
| | - Ja-Hyun Koo
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139
| | - Aimee Chapdelaine Williams
- HHMI, Boston Children's Hospital, Boston, MA 02115
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115
- Department of Genetics, Harvard Medical School, Boston, MA 02115
| | - Lucas Vieira Francisco
- HHMI, Boston Children's Hospital, Boston, MA 02115
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115
- Department of Genetics, Harvard Medical School, Boston, MA 02115
| | - Himanshu Batra
- HHMI, Boston Children's Hospital, Boston, MA 02115
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115
- Department of Genetics, Harvard Medical School, Boston, MA 02115
| | - Edward Lamperti
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139
| | - Oleksandr Kalyuzhniy
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, San Diego, CA 92037
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, San Diego, CA 92037
- Center for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, San Diego, CA 92037
| | - Yuxiang Zhang
- HHMI, Boston Children's Hospital, Boston, MA 02115
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115
- Department of Genetics, Harvard Medical School, Boston, MA 02115
| | - Alessandro Barbieri
- Department of Laboratory Medicine, Boston Children's Hospital, Boston, MA 02115
| | - John P Manis
- Department of Laboratory Medicine, Boston Children's Hospital, Boston, MA 02115
| | - Barton F Haynes
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710
- Department of Immunology, Duke University School of Medicine, Durham, NC 27710
| | - William R Schief
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, San Diego, CA 92037
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, San Diego, CA 92037
- Center for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, San Diego, CA 92037
| | - Facundo D Batista
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139
- Department of Immunology, Harvard Medical School, Boston, MA 02115
- Department of Microbiology, Harvard Medical School, Boston, MA 02115
| | - Ming Tian
- HHMI, Boston Children's Hospital, Boston, MA 02115
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115
- Department of Genetics, Harvard Medical School, Boston, MA 02115
| | - Frederick W Alt
- HHMI, Boston Children's Hospital, Boston, MA 02115
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115
- Department of Genetics, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
4
|
A self-sustaining layer of early-life-origin B cells drives steady-state IgA responses in the adult gut. Immunity 2022; 55:1829-1842.e6. [PMID: 36115337 DOI: 10.1016/j.immuni.2022.08.018] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 05/20/2022] [Accepted: 08/24/2022] [Indexed: 12/12/2022]
Abstract
The adult immune system consists of cells that emerged at various times during ontogeny. We aimed to define the relationship between developmental origin and composition of the adult B cell pool during unperturbed hematopoiesis. Lineage tracing stratified murine adult B cells based on the timing of output, revealing that a substantial portion originated within a restricted neonatal window. In addition to B-1a cells, early-life time-stamped B cells included clonally interrelated IgA plasma cells in the gut and bone marrow. These were actively maintained by B cell memory within gut chronic germinal centers and contained commensal microbiota reactivity. Neonatal rotavirus infection recruited recurrent IgA clones that were distinct from those arising by infection with the same antigen in adults. Finally, gut IgA plasma cells arose from the same hematopoietic progenitors as B-1a cells during ontogeny. Thus, a complex layer of neonatally imprinted B cells confer unique antibody responses later in life.
Collapse
|
5
|
Nomura H, Wada N, Takahashi H, Kase Y, Yamagami J, Egami S, Iriki H, Mukai M, Kamata A, Ito H, Fujii H, Ishikura T, Koseki H, Watanabe T, Yamada T, Ohara O, Koyasu S, Amagai M. IgM to IgG Class Switching Is a Necessary Step for Pemphigus Phenotype Induction in Desmoglein 3-Specific B Cell Receptor Knock-in Mouse. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:582-593. [PMID: 34996836 DOI: 10.4049/jimmunol.2100781] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 11/22/2021] [Indexed: 01/03/2023]
Abstract
Pemphigus vulgaris is an autoimmune blistering disease caused by IgG targeting desmoglein 3 (Dsg3), an adhesion molecule of keratinocytes. Anti-Dsg3 IgG production is prevented in healthy individuals, but it is unclear how Dsg3-specific B cells are regulated. To clarify the immunological condition regulating Dsg3-specific B cells, a pathogenic anti-Dsg3 Ig (AK23) knock-in mouse was generated. AK23 knock-in B cells developed normally without undergoing deletion or acquiring an anergic phenotype in vivo. The knock-in B cells showed Ca2+ influx upon IgM cross-linking and differentiated into AK23-IgG+ B cells after LPS and IL-4 stimulation in vitro that induced a pemphigus phenotype after adoptive transfer into Rag2 -/- mice. However, the knock-in mouse itself produced AK23-IgM but little IgG without blisters in vivo. Dsg3 immunization and skin inflammation caused AK23-IgG production and a pemphigus phenotype in vivo. Furthermore, Fcgr2b deficiency or haploinsufficiency spontaneously induced AK23-IgG production and a pemphigus phenotype with poor survival rates in AK23 knock-in mice. To assess Fcgr2b involvement in Ig class-switch efficiency, postswitch transcripts of B cells were quantified and significantly higher in Fcgr2b -/- and Fcgr2b +/- mice than wild-type mice in a gene dose-dependent manner. Finally, RNA sequencing revealed reduced expression of FCGR2B and FcγRIIB-related genes in patient B cells. These results indicated that Dsg3-specific B cells do not spontaneously perform pathogenic class switching in vivo, and pemphigus phenotype induction was prevented under normal conditions. Attenuated FcγRIIB signaling is also one of the drivers for pathogenic class switching and is consistent with immunological features identified from clinical samples. This study unveiled a characteristic immune state silencing autoreactive B cells in mice.
Collapse
Affiliation(s)
- Hisashi Nomura
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan
| | - Naoko Wada
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan.,Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Hayato Takahashi
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan
| | - Yuko Kase
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan
| | - Jun Yamagami
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan
| | - Shohei Egami
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan.,Laboratory for Skin Homeostasis, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Hisato Iriki
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan
| | - Miho Mukai
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan
| | - Aki Kamata
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan
| | - Hiromi Ito
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan
| | - Hideki Fujii
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan.,Department of Oral Microbiology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Tomoyuki Ishikura
- Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Haruhiko Koseki
- Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Takashi Watanabe
- Laboratory for Integrative Genomics, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Taketo Yamada
- Department of Pathology, Saitama Medical University, Saitama, Japan; and
| | - Osamu Ohara
- Laboratory for Integrative Genomics, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Shigeo Koyasu
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan; .,Laboratory for Immune Cell Systems, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Masayuki Amagai
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan; .,Laboratory for Skin Homeostasis, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| |
Collapse
|
6
|
Liu D, Lieber MR. The mechanisms of human lymphoid chromosomal translocations and their medical relevance. Crit Rev Biochem Mol Biol 2021; 57:227-243. [PMID: 34875186 DOI: 10.1080/10409238.2021.2004576] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The most common human lymphoid chromosomal translocations involve concurrent failures of the recombination activating gene (RAG) complex and Activation-Induced Deaminase (AID). These are two enzymes that are normally expressed for purposes of the two site-specific DNA recombination processes: V(D)J recombination and class switch recombination (CSR). First, though it is rare, a low level of expression of AID can introduce long-lived T:G mismatch lesions at 20-600 bp fragile zones. Second, the V(D)J recombination process can occasionally fail to rejoin coding ends, and this failure may permit an opportunity for Artemis:DNA-dependent kinase catalytic subunit (DNA-PKcs) to convert the T:G mismatch sites at the fragile zones into double-strand breaks. The 20-600 bp fragile zones must be, at least transiently, in a single-stranded DNA (ssDNA) state for the first step to occur, because AID only acts on ssDNA. Here we discuss the key DNA sequence features that lead to AID action at a fragile zone, which are (a) the proximity and density of strings of cytosine nucleotides (C-strings) that cause a B/A-intermediate DNA conformation; (b) overlapping AID hotspots that contain a methyl CpG (WRCG), which AID converts to a long-lived T:G mismatch; and (c) transcription, which, though not essential, favors increased ssDNA in the fragile zone. We also summarize chromosomal features of the focal fragile zones in lymphoid malignancies and discuss the clinical relevance of understanding the translocation mechanisms. Many of the key principles covered here are also relevant to chromosomal translocations in non-lymphoid somatic cells as well.
Collapse
Affiliation(s)
- Di Liu
- Department of Pathology & Laboratory Medicine, Department of Biochemistry & Molecular Biology, Department of Molecular Microbiology and Immunology, and Section of Computational Biology in the Department of Biological Sciences, USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Michael R Lieber
- Department of Pathology & Laboratory Medicine, Department of Biochemistry & Molecular Biology, Department of Molecular Microbiology and Immunology, and Section of Computational Biology in the Department of Biological Sciences, USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
7
|
An in vivo method for diversifying the functions of therapeutic antibodies. Proc Natl Acad Sci U S A 2021; 118:2025596118. [PMID: 33658386 DOI: 10.1073/pnas.2025596118] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
V(D)J recombination generates mature B cells that express huge repertoires of primary antibodies as diverse immunoglobulin (Ig) heavy chain (IgH) and light chain (IgL) of their B cell antigen receptors (BCRs). Cognate antigen binding to BCR variable region domains activates B cells into the germinal center (GC) reaction in which somatic hypermutation (SHM) modifies primary variable region-encoding sequences, with subsequent selection for mutations that improve antigen-binding affinity, ultimately leading to antibody affinity maturation. Based on these principles, we developed a humanized mouse model approach to diversify an anti-PD1 therapeutic antibody and allow isolation of variants with novel properties. In this approach, component Ig gene segments of the anti-PD1 antibody underwent de novo V(D)J recombination to diversify the anti-PD1 antibody in the primary antibody repertoire in the mouse models. Immunization of these mouse models further modified the anti-PD1 antibodies through SHM. Known anti-PD1 antibodies block interaction of PD1 with its ligands to alleviate PD1-mediated T cell suppression, thereby boosting antitumor T cell responses. By diversifying one such anti-PD1 antibody, we derived many anti-PD1 antibodies, including anti-PD1 antibodies with the opposite activity of enhancing PD1/ligand interaction. Such antibodies theoretically might suppress deleterious T cell activities in autoimmune diseases. The approach we describe should be generally applicable for diversifying other therapeutic antibodies.
Collapse
|
8
|
Baizan-Edge A, Stubbs BA, Stubbington MJT, Bolland DJ, Tabbada K, Andrews S, Corcoran AE. IL-7R signaling activates widespread V H and D H gene usage to drive antibody diversity in bone marrow B cells. Cell Rep 2021; 36:109349. [PMID: 34260907 PMCID: PMC8293627 DOI: 10.1016/j.celrep.2021.109349] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 04/05/2021] [Accepted: 06/15/2021] [Indexed: 01/09/2023] Open
Abstract
Generation of the primary antibody repertoire requires V(D)J recombination of hundreds of gene segments in the immunoglobulin heavy chain (Igh) locus. The role of interleukin-7 receptor (IL-7R) signaling in Igh recombination has been difficult to partition from its role in B cell survival and proliferation. With a detailed description of the Igh repertoire in murine IL-7Rα-/- bone marrow B cells, we demonstrate that IL-7R signaling profoundly influences VH gene selection during VH-to-DJH recombination. We find skewing toward 3' VH genes during de novo VH-to-DJH recombination more severe than the fetal liver (FL) repertoire and uncover a role for IL-7R signaling in DH-to-JH recombination. Transcriptome and accessibility analyses suggest reduced expression of B lineage transcription factors (TFs) and targets and loss of DH and VH antisense transcription in IL-7Rα-/- B cells. Thus, in addition to its roles in survival and proliferation, IL-7R signaling shapes the Igh repertoire by activating underpinning mechanisms.
Collapse
Affiliation(s)
- Amanda Baizan-Edge
- 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
| | - Michael J T Stubbington
- Nuclear Dynamics Programme, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Daniel J Bolland
- Nuclear Dynamics Programme, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK; Lymphocyte Signaling and Development Programme, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Kristina Tabbada
- Nuclear Dynamics Programme, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK; Lymphocyte Signaling and Development Programme, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Simon Andrews
- Bioinformatics Group, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Anne E Corcoran
- Nuclear Dynamics Programme, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK; Lymphocyte Signaling and Development Programme, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK.
| |
Collapse
|
9
|
Davila ML, Brentjens R, Wang X, Rivière I, Sadelain M. How do CARs work?: Early insights from recent clinical studies targeting CD19. Oncoimmunology 2021; 1:1577-1583. [PMID: 23264903 PMCID: PMC3525612 DOI: 10.4161/onci.22524] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Second-generation chimeric antigen receptors (CARs) are powerful tools to redirect antigen-specific T cells independently of HLA-restriction. Recent clinical studies evaluating CD19-targeted T cells in patients with B-cell malignancies demonstrate the potency of CAR-engineered T cells. With results from 28 subjects enrolled by five centers conducting studies in patients with chronic lymphocytic leukemia (CLL) or lymphoma, some insights into the parameters that determine T-cell function and clinical outcome of CAR-based approaches are emerging. These parameters involve CAR design, T-cell production methods, conditioning chemotherapy as well as patient selection. Here, we discuss the potential relevance of these findings and in particular the interplay between the adoptive transfer of T cells and pre-transfer patient conditioning.
Collapse
Affiliation(s)
- Marco L Davila
- Center for Cell Engineering; Department of Medicine; Molecular Pharmacology and Chemistry Program; Memorial Sloan-Kettering Cancer Center; New York, NY
| | | | | | | | | |
Collapse
|
10
|
Vergani S, Yuan J. Developmental changes in the rules for B cell selection. Immunol Rev 2021; 300:194-202. [PMID: 33501672 DOI: 10.1111/imr.12949] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/04/2021] [Accepted: 01/06/2021] [Indexed: 12/14/2022]
Abstract
The autoimmune checkpoint during B cell maturation eliminates self-antigen reactive specificities from the mature B cell repertoire. However, an exception to this rule is illustrated by B-1 cells, an innate-like self-reactive B cell subset that is positively selected into the mature B cell pool in a self-antigen-driven fashion. The mechanisms by which B-1 cells escape central tolerance have puzzled the field for decades. A key clue comes from their restricted developmental window during fetal and neonatal life. Here we use B-1 cells as a prototypic early life derived B cell subset to explore developmental changes in the constraints of B cell selection. We discuss recent advancements in the understanding of the molecular program, centered around the RNA binding protein Lin28b, that licenses self-reactive B-1 cell output during ontogeny. Finally, we speculate on the possible link between the unique rules of early life B cell tolerance and the establishment of B cell - microbial mutualism to propose an integrated model for how developmental and environmental cues come together to create a protective layer of B cell memory involved in neonatal immune imprinting.
Collapse
Affiliation(s)
- Stefano Vergani
- Developmental Immunology Unit, Division of Molecular Hematology, Department of Laboratory Medicine, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Joan Yuan
- Developmental Immunology Unit, Division of Molecular Hematology, Department of Laboratory Medicine, Lund Stem Cell Center, Lund University, Lund, Sweden
| |
Collapse
|
11
|
Lehrke MJ, Shapiro MJ, Rajcula MJ, Kennedy MM, McCue SA, Medina KL, Shapiro VS. The mitochondrial iron transporter ABCB7 is required for B cell development, proliferation, and class switch recombination in mice. eLife 2021; 10:69621. [PMID: 34762046 PMCID: PMC8585479 DOI: 10.7554/elife.69621] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 10/20/2021] [Indexed: 12/14/2022] Open
Abstract
Iron-sulfur (Fe-S) clusters are cofactors essential for the activity of numerous enzymes including DNA polymerases, helicases, and glycosylases. They are synthesized in the mitochondria as Fe-S intermediates and are exported to the cytoplasm for maturation by the mitochondrial transporter ABCB7. Here, we demonstrate that ABCB7 is required for bone marrow B cell development, proliferation, and class switch recombination, but is dispensable for peripheral B cell homeostasis in mice. Conditional deletion of ABCB7 using Mb1-cre resulted in a severe block in bone marrow B cell development at the pro-B cell stage. The loss of ABCB7 did not alter expression of transcription factors required for B cell specification or commitment. While increased intracellular iron was observed in ABCB7-deficient pro-B cells, this did not lead to increased cellular or mitochondrial reactive oxygen species, ferroptosis, or apoptosis. Interestingly, loss of ABCB7 led to replication-induced DNA damage in pro-B cells, independent of VDJ recombination, and these cells had evidence of slowed DNA replication. Stimulated ABCB7-deficient splenic B cells from CD23-cre mice also had a striking loss of proliferation and a defect in class switching. Thus, ABCB7 is essential for early B cell development, proliferation, and class switch recombination.
Collapse
Affiliation(s)
| | | | | | | | | | - Kay L Medina
- Department of Immunology, Mayo ClinicRochesterUnited States
| | | |
Collapse
|
12
|
Aslam MA, Alemdehy MF, Hao B, Krijger PHL, Pritchard CEJ, de Rink I, Muhaimin FI, Nurzijah I, van Baalen M, Kerkhoven RM, van den Berk PCM, Skok JA, Jacobs H. The Ig heavy chain protein but not its message controls early B cell development. Proc Natl Acad Sci U S A 2020; 117:31343-31352. [PMID: 33229554 PMCID: PMC7733823 DOI: 10.1073/pnas.2004810117] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Development of progenitor B cells (ProB cells) into precursor B cells (PreB cells) is dictated by immunoglobulin heavy chain checkpoint (IgHCC), where the IgHC encoded by a productively rearranged Igh allele assembles into a PreB cell receptor complex (PreBCR) to generate signals to initiate this transition and suppressing antigen receptor gene recombination, ensuring that only one productive Igh allele is expressed, a phenomenon known as Igh allelic exclusion. In contrast to a productively rearranged Igh allele, the Igh messenger RNA (mRNA) (IgHR) from a nonproductively rearranged Igh allele is degraded by nonsense-mediated decay (NMD). This fact prohibited firm conclusions regarding the contribution of stable IgHR to the molecular and developmental changes associated with the IgHCC. This point was addressed by generating the IghTer5H∆TM mouse model from IghTer5H mice having a premature termination codon at position +5 in leader exon of IghTer5H allele. This prohibited NMD, and the lack of a transmembrane region (∆TM) prevented the formation of any signaling-competent PreBCR complexes that may arise as a result of read-through translation across premature Ter5 stop codon. A highly sensitive sandwich Western blot revealed read-through translation of IghTer5H message, indicating that previous conclusions regarding a role of IgHR in establishing allelic exclusion requires further exploration. As determined by RNA sequencing (RNA-Seq), this low amount of IgHC sufficed to initiate PreB cell markers normally associated with PreBCR signaling. In contrast, the IghTer5H∆TM knock-in allele, which generated stable IgHR but no detectable IgHC, failed to induce PreB development. Our data indicate that the IgHCC is controlled at the level of IgHC and not IgHR expression.
Collapse
Affiliation(s)
- Muhammad Assad Aslam
- Division of Tumor Biology and Immunology, Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
- Institute of Molecular Biology and Biotechnology, Bahauddin Zakariya University, 60800 Multan, Pakistan
| | - Mir Farshid Alemdehy
- Division of Tumor Biology and Immunology, Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Bingtao Hao
- Department of Pathology, New York University School of Medicine, New York, NY 10016
| | - Peter H L Krijger
- Hubrecht Institute-Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, 3584 CT Utrecht, The Netherlands
| | - Colin E J Pritchard
- Mouse Clinic for Cancer and Aging Transgenic Facility, Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Iris de Rink
- Genome Core Facility, Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | | | - Ika Nurzijah
- Division of Tumor Biology and Immunology, Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Martijn van Baalen
- Flow Cytometry Facility, Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Ron M Kerkhoven
- Genome Core Facility, Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Paul C M van den Berk
- Division of Tumor Biology and Immunology, Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Jane A Skok
- Department of Pathology, New York University School of Medicine, New York, NY 10016
| | - Heinz Jacobs
- Division of Tumor Biology and Immunology, Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands;
| |
Collapse
|
13
|
Zahid A, Siegler EL, Kenderian SS. CART Cell Toxicities: New Insight into Mechanisms and Management. Clin Hematol Int 2020; 2:149-155. [PMID: 33409484 PMCID: PMC7785104 DOI: 10.2991/chi.k.201108.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
T cells genetically engineered with chimeric antigen receptors (CART) have become a potent class of cancer immunotherapeutics. Numerous clinical trials of CART cells have revealed remarkable remission rates in patients with relapsed or refractory hematologic malignancies. Despite recent clinical success, CART cell therapy has also led to significant morbidity and occasional mortality from associated toxicities. Cytokine release syndrome (CRS) and Immune effector cell-associated neurotoxicity syndrome (ICANS) present barriers to the extensive use of CART cell therapy in the clinic. CRS can lead to fever, hypoxia, hypotension, coagulopathies, and multiorgan failure, and ICANS can result in cognitive dysfunction, seizures, and cerebral edema. The mechanisms of CRS and ICANS are becoming clearer, but many aspects remain unknown. Disease type and burden, peak serum CART cell levels, CART cell dose, CAR structure, elevated pro-inflammatory cytokines, and activated myeloid and endothelial cells all contribute to CART cell toxicity. Current guidelines for the management of toxicities associated with CART cell therapy vary between clinics, but are typically comprised of supportive care and treatment with corticosteroids or tocilizumab, depending on the severity of the symptoms. Acquiring a deeper understanding of CART cell toxicities and developing new management and prevention strategies are ongoing. In this review, we present findings in the mechanisms and management of CART cell toxicities.
Collapse
Affiliation(s)
- Anas Zahid
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, U.A.E
| | - Elizabeth L Siegler
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA.,Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Saad S Kenderian
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA.,Division of Hematology, Mayo Clinic, Rochester, MN, USA.,Department of Immunology, Mayo Clinic, Rochester, MN, USA.,Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
14
|
Jacobsen JA, Bartom ET, Sigvardsson M, Kee BL. Ezh2 Represses Transcription of Innate Lymphoid Genes in B Lymphocyte Progenitors and Maintains the B-2 Cell Fate. THE JOURNAL OF IMMUNOLOGY 2020; 204:1760-1769. [PMID: 32094206 DOI: 10.4049/jimmunol.1901188] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 01/28/2020] [Indexed: 02/05/2023]
Abstract
Lymphocyte lineage specification and commitment requires the activation of lineage-specific genes and repression of alternative lineage genes, respectively. The mechanisms governing alternative lineage gene repression and commitment in lymphocytes are largely unknown. In this study, we demonstrate that Ezh2, which represses gene expression through methylation of histone 3 lysine 27, was essential for repression of numerous genes, including genes encoding innate lymphocyte transcription factors, specifically in murine B lymphocyte progenitors, but these cells maintained their B lymphocyte identity. However, adult Ezh2-deficient B lymphocytes expressed Lin28b, which encodes an RNA-binding protein associated with fetal hematopoietic gene expression programs, and these cells acquired a fetal B-1 lymphocyte phenotype in vitro and in vivo. Therefore, Ezh2 coordinates the repression of multiple gene programs in B lymphocytes and maintains the adult B-2 cell fate.
Collapse
Affiliation(s)
| | - Elizabeth T Bartom
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Mikael Sigvardsson
- Division of Molecular Hematology, Lund University, Lund SE-221 00, Sweden; and
| | - Barbara L Kee
- Committee on Immunology, The University of Chicago, Chicago, IL 60657; .,Department of Pathology, The University of Chicago, Chicago, IL 60657
| |
Collapse
|
15
|
Wong JB, Hewitt SL, Heltemes-Harris LM, Mandal M, Johnson K, Rajewsky K, Koralov SB, Clark MR, Farrar MA, Skok JA. B-1a cells acquire their unique characteristics by bypassing the pre-BCR selection stage. Nat Commun 2019; 10:4768. [PMID: 31628339 PMCID: PMC6802180 DOI: 10.1038/s41467-019-12824-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Accepted: 09/24/2019] [Indexed: 12/23/2022] Open
Abstract
B-1a cells are long-lived, self-renewing innate-like B cells that predominantly inhabit the peritoneal and pleural cavities. In contrast to conventional B-2 cells, B-1a cells have a receptor repertoire that is biased towards bacterial and self-antigens, promoting a rapid response to infection and clearing of apoptotic cells. Although B-1a cells are known to primarily originate from fetal tissues, the mechanisms by which they arise has been a topic of debate for many years. Here we show that in the fetal liver versus bone marrow environment, reduced IL-7R/STAT5 levels promote immunoglobulin kappa gene recombination at the early pro-B cell stage. As a result, differentiating B cells can directly generate a mature B cell receptor (BCR) and bypass the requirement for a pre-BCR and pairing with surrogate light chain. This 'alternate pathway' of development enables the production of B cells with self-reactive, skewed specificity receptors that are peculiar to the B-1a compartment. Together our findings connect seemingly opposing lineage and selection models of B-1a cell development and explain how these cells acquire their unique properties.
Collapse
MESH Headings
- Animals
- B-Lymphocyte Subsets/immunology
- B-Lymphocyte Subsets/metabolism
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
- Bone Marrow/immunology
- Bone Marrow/metabolism
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Immunoglobulin Light Chains, Surrogate/genetics
- Immunoglobulin Light Chains, Surrogate/immunology
- Immunoglobulin Light Chains, Surrogate/metabolism
- Liver/embryology
- Liver/immunology
- Liver/metabolism
- Lymphocyte Activation/genetics
- Lymphocyte Activation/immunology
- Mice, Inbred C57BL
- Mice, Knockout
- Pre-B Cell Receptors/genetics
- Pre-B Cell Receptors/immunology
- Pre-B Cell Receptors/metabolism
- Receptors, Antigen, B-Cell/genetics
- Receptors, Antigen, B-Cell/immunology
- Receptors, Antigen, B-Cell/metabolism
- Receptors, Interleukin-7/genetics
- Receptors, Interleukin-7/immunology
- Receptors, Interleukin-7/metabolism
- STAT5 Transcription Factor/genetics
- STAT5 Transcription Factor/immunology
- STAT5 Transcription Factor/metabolism
Collapse
Affiliation(s)
- Jason B Wong
- Department of Pathology, New York University School of Medicine, New York University, New York, NY, USA
| | - Susannah L Hewitt
- Department of Pathology, New York University School of Medicine, New York University, New York, NY, USA
| | - Lynn M Heltemes-Harris
- Department of Laboratory Medicine and Pathology, Center for Immunology, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Malay Mandal
- Department of Medicine, Section of Rheumatology and Gwen Knapp Center for Lupus and Immunology Research, University of Chicago, Chicago, IL, USA
| | - Kristen Johnson
- Department of Pathology, New York University School of Medicine, New York University, New York, NY, USA
| | - Klaus Rajewsky
- Max Delbrück Center for Molecular Medicine, 13092, Berlin, Germany
| | - Sergei B Koralov
- Department of Pathology, New York University School of Medicine, New York University, New York, NY, USA
| | - Marcus R Clark
- Department of Medicine, Section of Rheumatology and Gwen Knapp Center for Lupus and Immunology Research, University of Chicago, Chicago, IL, USA
| | - Michael A Farrar
- Department of Laboratory Medicine and Pathology, Center for Immunology, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Jane A Skok
- Department of Pathology, New York University School of Medicine, New York University, New York, NY, USA.
| |
Collapse
|
16
|
Nouri Rouzbahani F, Shirkhoda M, Memari F, Dana H, Mahmoodi Chalbatani G, Mahmoodzadeh H, Samarghandi N, Gharagozlou E, Mohammadi Hadloo MH, Maleki AR, Sadeghian E, Nia E, Nia N, Hadjilooei F, Rezaeian O, Meghdadi S, Miri S, Jafari F, Rayzan E, Marmari V. Immunotherapy a New Hope for Cancer Treatment: A Review. Pak J Biol Sci 2019; 21:135-150. [PMID: 30187723 DOI: 10.3923/pjbs.2018.135.150] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Cancer is a major burden of disease worldwide with considerable impact on society. The tide of immunotherapy has finally changed after decades of disappointing results and has become a clinically validated treatment for many cancers. Immunotherapy takes many forms in cancer treatment, including the adoptive transfer of ex vivo activated T cells, oncolytic viruses, natural killer cells, cancer vaccines and administration of antibodies or recombinant proteins that either costimulate cells or block the so-called immune checkpoint pathways. Recently, cancer immunotherapy has received a high degree of attention, which mainly contains the treatments for programmed death ligand 1 (PD-L1), programmed death 1 (PD-1), chimeric antigen receptors (CARs) and cytotoxic T lymphocyte-associated antigen 4 (CTLA-4). Here, this paper reviewed the current understandings of the main strategies in cancer immunotherapy (adoptive cellular immunotherapy, immune checkpoint blockade, oncolytic viruses and cancer vaccines) and discuss the progress in the synergistic design of immune-targeting combination therapies.
Collapse
|
17
|
McBride A, Houtmann S, Wilde L, Vigil C, Eischen CM, Kasner M, Palmisiano N. The Role of Inhibition of Apoptosis in Acute Leukemias and Myelodysplastic Syndrome. Front Oncol 2019; 9:192. [PMID: 30972300 PMCID: PMC6445951 DOI: 10.3389/fonc.2019.00192] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 03/06/2019] [Indexed: 12/24/2022] Open
Abstract
Avoidance of apoptosis is a key mechanism that malignancies, including acute leukemias and MDS, utilize in order to proliferate and resist chemotherapy. Recently, venetoclax, an inhibitor of the anti-apoptotic protein BCL-2, has been approved for the treatment of upfront AML in an unfit, elderly population. This paper reviews the pre-clinical and clinical data for apoptosis inhibitors currently in development for the treatment of AML, ALL, and MDS.
Collapse
Affiliation(s)
- Amanda McBride
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| | - Sarah Houtmann
- Department of Medicine, Thomas Jefferson University, Philadelphia, PA, United States
| | - Lindsay Wilde
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| | - Carlos Vigil
- Division of Hematology, Oncology and Blood and Marrow Transplantation, Department of Internal Medicine, University of Iowa, Iowa City, IA, United States
| | - Christine M Eischen
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| | - Margaret Kasner
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| | - Neil Palmisiano
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
18
|
Hayakawa K, Li YS, Shinton SA, Bandi SR, Formica AM, Brill-Dashoff J, Hardy RR. Crucial Role of Increased Arid3a at the Pre-B and Immature B Cell Stages for B1a Cell Generation. Front Immunol 2019; 10:457. [PMID: 30930899 PMCID: PMC6428705 DOI: 10.3389/fimmu.2019.00457] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 02/20/2019] [Indexed: 02/03/2023] Open
Abstract
The Lin28b+Let7− axis in fetal/neonatal development plays a role in promoting CD5+ B1a cell generation as a B-1 B cell developmental outcome. Here we identify the Let7 target, Arid3a, as a crucial molecular effector of the B-1 cell developmental program. Arid3a expression is increased at pro-B cell stage and markedly increased at pre-B and immature B cell stages in the fetal/neonatal liver B-1 development relative to that in the Lin28b−Let7+ adult bone marrow (BM) B-2 cell development. Analysis of B-lineage restricted Lin28b transgenic (Tg) mice, Arid3a knockout and Arid3a Tg mice, confirmed that increased Arid3a allows B cell generation without requiring surrogate light chain (SLC) associated pre-BCR stage, and prevents MHC class II cell expression at the pre-B and newly generated immature B cell stages, distinct from pre-BCR dependent B development with MHC class II in adult BM. Moreover, Arid3a plays a crucial role in supporting B1a cell generation. The increased Arid3a leads higher Myc and Bhlhe41, and lower Siglec-G and CD72 at the pre-B and immature B cell stages than normal adult BM, to allow BCR signaling induced B1a cell generation. Arid3a-deficiency selectively blocks the development of B1a cells, while having no detectable effect on CD5− B1b, MZ B, and FO B cell generation resembling B-2 development outcome. Conversely, enforced expression of Arid3a by transgene is sufficient to promote the development of B1a cells from adult BM. Under the environment change between birth to adult, altered BCR repertoire in increased B1a cells occurred generated from adult BM. However, crossed with B1a-restricted VH/D/J IgH knock-in mice allowed to confirm that SLC-unassociated B1a cell increase and CLL/lymphoma generation can occur in aged from Arid3a increased adult BM. These results confirmed that in fetal/neonatal normal mice, increased Arid3a at the pre-B cell and immature B cell stages is crucial for generating B1a cells together with the environment for self-ligand reactive BCR selection, B1a cell maintenance, and potential for development of CLL/Lymphoma in aged mice.
Collapse
Affiliation(s)
- Kyoko Hayakawa
- Fox Chase Cancer Center, Philadelphia, PA, United States
| | - Yue-Sheng Li
- Fox Chase Cancer Center, Philadelphia, PA, United States
| | | | | | | | | | | |
Collapse
|
19
|
Winkler TH, Mårtensson IL. The Role of the Pre-B Cell Receptor in B Cell Development, Repertoire Selection, and Tolerance. Front Immunol 2018; 9:2423. [PMID: 30498490 PMCID: PMC6249383 DOI: 10.3389/fimmu.2018.02423] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 10/01/2018] [Indexed: 11/18/2022] Open
Abstract
Around four decades ago, it had been observed that there were cell lines as well as cells in the fetal liver that expressed antibody μ heavy (μH) chains in the apparent absence of bona fide light chains. It was thus possible that these cells expressed another molecule(s), that assembled with μH chains. The ensuing studies led to the discovery of the pre-B cell receptor (pre-BCR), which is assembled from Ig μH and surrogate light (SL) chains, together with the signaling molecules Igα and β. It is expressed on a fraction of pro-B (pre-BI) cells and most large pre-B(II) cells, and has been implicated in IgH chain allelic exclusion and down-regulation of the recombination machinery, assessment of the expressed μH chains and shaping the IgH repertoire, transition from the pro-B to pre-B stage, pre-B cell expansion, and cessation.
Collapse
Affiliation(s)
- Thomas H Winkler
- Chair of Genetics, Department of Biology, Nikolaus-Fiebiger-Center for Molecular Medicine, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Inga-Lill Mårtensson
- Department of Rheumatology and Inflammation Research, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
20
|
Sohur US, Dixit MN, Chen CL, Byrom MW, Kerr LD. Rel/NF-kappaB represses bcl-2 transcription in pro-B lymphocytes. Gene Expr 2018; 8:219-29. [PMID: 10794524 PMCID: PMC6157363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
The mechanisms controlling programmed cell death (PCD) during early B cell development are not well understood. Members of both the Bcl-2 family of apoptosis-related proteins and the nuclear factor-kappa B/Rel (NF-kappaB/Rel) family of transcription factors are expressed differentially during B cell development. To date, however, no direct interactions between these two families have been demonstrated. The FL5.12 cell line represents a model for progenitor B cell development. Such cells reproducibly undergo PCD upon IL-3 withdrawal. The signal to enter the apoptotic pathway is mediated by a shift in the ratio of Bcl-2:Bax. While bax levels remain constant, bcl-2 transcription rate, steady-state mRNA, and protein levels decrease. Analysis of the bcl-2 promoter reveals 3 kappaB sites functionally able to bind kappaB factors from FL5.12 nuclear extracts. Cotransfection studies demonstrate that NF-kappaB factors can repress bcl-2 transcription and that site-directed mutagenesis of the kappaB motifs abolishes this repression. These studies suggest that NF-kappaB mediates PCD in pro-B cells through transcriptional repression of the survival gene bcl-2, thus shifting the bcl-2:bax ratio in favor of death-promoting complexes.
Collapse
Affiliation(s)
- U. Shivraj Sohur
- *Department of Microbiology & Immunology, Vanderbilt University School of Medicine, 1161 21st Ave. South, Nashville, TN 37232-2363
| | - Mrinalini N. Dixit
- †Department of Cell Biology, Vanderbilt University School of Medicine, 1161 21st Ave. South, Nashville, TN 37232-2363
| | - Chih-Li Chen
- *Department of Microbiology & Immunology, Vanderbilt University School of Medicine, 1161 21st Ave. South, Nashville, TN 37232-2363
| | - Mike W. Byrom
- *Department of Microbiology & Immunology, Vanderbilt University School of Medicine, 1161 21st Ave. South, Nashville, TN 37232-2363
| | - Lawrence D. Kerr
- *Department of Microbiology & Immunology, Vanderbilt University School of Medicine, 1161 21st Ave. South, Nashville, TN 37232-2363
- †Department of Cell Biology, Vanderbilt University School of Medicine, 1161 21st Ave. South, Nashville, TN 37232-2363
- Address correspondence to Lawrence D. Kerr, Vanderbilt University, School of Medicine, MCN A-4314, 1161 21st Ave. South, Nashville, TN 37232-2363. Tel: (615) 343-2568; Fax: (615) 343-2569; E-mail:
| |
Collapse
|
21
|
Kim SK, Knight DA, Jones LR, Vervoort S, Ng AP, Seymour JF, Bradner JE, Waibel M, Kats L, Johnstone RW. JAK2 is dispensable for maintenance of JAK2 mutant B-cell acute lymphoblastic leukemias. Genes Dev 2018; 32:849-864. [PMID: 29907650 PMCID: PMC6049517 DOI: 10.1101/gad.307504.117] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 05/07/2018] [Indexed: 11/24/2022]
Abstract
Kim et al. show that while expression of mutant Jak2 is necessary for B-cell acute lymphoblastic leukemia induction, neither its continued expression nor enzymatic activity is required to maintain leukemia survival and rapid proliferation. Activating JAK2 point mutations are implicated in the pathogenesis of myeloid and lymphoid malignancies, including high-risk B-cell acute lymphoblastic leukemia (B-ALL). In preclinical studies, treatment of JAK2 mutant leukemias with type I JAK2 inhibitors (e.g., Food and Drug Administration [FDA]-approved ruxolitinib) provided limited single-agent responses, possibly due to paradoxical JAK2Y1007/1008 hyperphosphorylation induced by these agents. To determine the importance of mutant JAK2 in B-ALL initiation and maintenance, we developed unique genetically engineered mouse models of B-ALL driven by overexpressed Crlf2 and mutant Jak2, recapitulating the genetic aberrations found in human B-ALL. While expression of mutant Jak2 was necessary for leukemia induction, neither its continued expression nor enzymatic activity was required to maintain leukemia survival and rapid proliferation. CRLF2/JAK2 mutant B-ALLs with sustained depletion or pharmacological inhibition of JAK2 exhibited enhanced expression of c-Myc and prominent up-regulation of c-Myc target genes. Combined indirect targeting of c-Myc using the BET bromodomain inhibitor JQ1 and direct targeting of JAK2 with ruxolitinib potently killed JAK2 mutant B-ALLs.
Collapse
Affiliation(s)
- Sang-Kyu Kim
- The Peter MacCallum Cancer Centre, Melbourne, 3000 Victoria, Australia.,The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, 3052 Victoria, Australia
| | - Deborah A Knight
- The Peter MacCallum Cancer Centre, Melbourne, 3000 Victoria, Australia
| | - Lisa R Jones
- The Peter MacCallum Cancer Centre, Melbourne, 3000 Victoria, Australia.,The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, 3052 Victoria, Australia
| | - Stephin Vervoort
- The Peter MacCallum Cancer Centre, Melbourne, 3000 Victoria, Australia.,The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, 3052 Victoria, Australia
| | - Ashley P Ng
- Division of Cancer and Haematology, The Walter and Eliza Hall Institute of Medical Research, Parkville, 3052 Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, 3010 Victoria, Australia
| | - John F Seymour
- The Peter MacCallum Cancer Centre, Melbourne, 3000 Victoria, Australia.,The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, 3052 Victoria, Australia
| | - James E Bradner
- Novartis Institutes for BioMedical (NIBR) Research, Cambridge, Massachusetts 02139, USA
| | - Michaela Waibel
- The Peter MacCallum Cancer Centre, Melbourne, 3000 Victoria, Australia.,The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, 3052 Victoria, Australia
| | - Lev Kats
- The Peter MacCallum Cancer Centre, Melbourne, 3000 Victoria, Australia.,The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, 3052 Victoria, Australia
| | - Ricky W Johnstone
- The Peter MacCallum Cancer Centre, Melbourne, 3000 Victoria, Australia.,The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, 3052 Victoria, Australia
| |
Collapse
|
22
|
Jensen CT, Åhsberg J, Sommarin MNE, Strid T, Somasundaram R, Okuyama K, Ungerbäck J, Kupari J, Airaksinen MS, Lang S, Bryder D, Soneji S, Karlsson G, Sigvardsson M. Dissection of progenitor compartments resolves developmental trajectories in B-lymphopoiesis. J Exp Med 2018; 215:1947-1963. [PMID: 29899037 PMCID: PMC6028518 DOI: 10.1084/jem.20171384] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 03/12/2018] [Accepted: 05/18/2018] [Indexed: 01/22/2023] Open
Abstract
Jensen et al. report the identification and characterization of novel lymphoid progenitor populations in the mouse bone marrow. The work resolves the complexity of the BLP/pre-pro–B/Fraction A compartments and provides a developmental trajectory for early B cell development. To understand the developmental trajectories in early lymphocyte differentiation, we identified differentially expressed surface markers on lineage-negative lymphoid progenitors (LPs). Single-cell polymerase chain reaction experiments allowed us to link surface marker expression to that of lineage-associated transcription factors (TFs) and identify GFRA2 and BST1 as markers of early B cells. Functional analyses in vitro and in vivo as well as single-cell gene expression analyses supported that surface expression of these proteins defined distinct subpopulations that include cells from both the classical common LPs (CLPs) and Fraction A compartments. The formation of the GFRA2-expressing stages of development depended on the TF EBF1, critical both for the activation of stage-specific target genes and modulation of the epigenetic landscape. Our data show that consecutive expression of Ly6D, GFRA2, and BST1 defines a developmental trajectory linking the CLP to the CD19+ progenitor compartment.
Collapse
Affiliation(s)
| | | | | | - Tobias Strid
- Division of Molecular Hematology, Lund University, Lund, Sweden.,Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Rajesh Somasundaram
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Kazuki Okuyama
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Jonas Ungerbäck
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Jussi Kupari
- Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | | | - Stefan Lang
- Division of Molecular Hematology, Lund University, Lund, Sweden
| | - David Bryder
- Division of Molecular Hematology, Lund University, Lund, Sweden
| | - Shamit Soneji
- Division of Molecular Hematology, Lund University, Lund, Sweden
| | - Göran Karlsson
- Division of Molecular Hematology, Lund University, Lund, Sweden
| | - Mikael Sigvardsson
- Division of Molecular Hematology, Lund University, Lund, Sweden .,Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| |
Collapse
|
23
|
Loc'h J, Delarue M. Terminal deoxynucleotidyltransferase: the story of an untemplated DNA polymerase capable of DNA bridging and templated synthesis across strands. Curr Opin Struct Biol 2018; 53:22-31. [PMID: 29656238 DOI: 10.1016/j.sbi.2018.03.019] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 03/27/2018] [Accepted: 03/30/2018] [Indexed: 01/08/2023]
Abstract
Terminal deoxynucleotidyltransferase (TdT) is a member of the polX family which is involved in DNA repair. It has been known for years as an untemplated DNA polymerase used during V(D)J recombination to generate diversity at the CDR3 region of immunoglobulins and T-cell receptors. Recently, however, TdT was crystallized in the presence of a complete DNA synapsis made of two double-stranded DNA (dsDNA), each with a 3' protruding end, and overlapping with only one micro-homology base-pair, thus giving structural insight for the first time into DNA synthesis across strands. It was subsequently shown that TdT indeed has an in trans template-dependent activity in the presence of an excess of the downstream DNA duplex. A possible biological role of this dual activity is discussed.
Collapse
Affiliation(s)
- Jérôme Loc'h
- Unit of Structural Dynamics of Biological Macromolecules and UMR 3528 du CNRS, Institut Pasteur, 75015 Paris, France
| | - Marc Delarue
- Unit of Structural Dynamics of Biological Macromolecules and UMR 3528 du CNRS, Institut Pasteur, 75015 Paris, France.
| |
Collapse
|
24
|
Kreslavsky T, Wong JB, Fischer M, Skok JA, Busslinger M. Control of B-1a cell development by instructive BCR signaling. Curr Opin Immunol 2018; 51:24-31. [PMID: 29414528 PMCID: PMC5943138 DOI: 10.1016/j.coi.2018.01.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 12/27/2017] [Accepted: 01/02/2018] [Indexed: 12/13/2022]
Abstract
B-1a cells remain one of the most enigmatic lymphocyte subsets. In this review, we discuss recent advances in our understanding of the development of these cells and their regulation by the transcription factors Bhlhe41 and Arid3a as well as by the RNA-binding protein Lin28b. A large body of literature supports an instructive role of BCR signaling in B-1a cell development and lineage commitment, which is initiated only after signaling from an autoreactive BCR. While both fetal and adult hematopoiesis can generate B-1a cells, the contribution of adult hematopoiesis to the B-1a cell compartment is low under physiological conditions. We discuss several models that can reconcile the instructive role of BCR signaling with this fetal bias in B-1a cell development.
Collapse
Affiliation(s)
- Taras Kreslavsky
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Campus-Vienna-Biocenter 1, A-1030 Vienna, Austria.
| | - Jason B Wong
- Department of Pathology, New York Medical Center, New York University, New York, USA
| | - Maria Fischer
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Campus-Vienna-Biocenter 1, A-1030 Vienna, Austria
| | - Jane A Skok
- Department of Pathology, New York Medical Center, New York University, New York, USA
| | - Meinrad Busslinger
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Campus-Vienna-Biocenter 1, A-1030 Vienna, Austria.
| |
Collapse
|
25
|
Abstract
Chimeric antigen receptor (CAR) T-cells are redirected T-cells that can recognize cancer antigens in a major histocompatibility complex (MHC)-independent fashion. A typical CAR is comprised of two main functional domains: an extracellular antigen recognition domain, called a single-chain variable fragment (scFv), and an intracellular signaling domain. Based on the number of intracellular signaling molecules, CARs are categorized into four generations. CAR T-cell therapy has become a promising treatment for hematologic malignancies. However, results of its clinical trials on solid tumors have not been encouraging. Here, we described the structure of CARs and summarized the clinical trials of CD19-targeted CAR T-cells. The side effects, safety management, challenges, and future prospects of CAR T-cells for the treatment of cancer, particularly for solid tumors, were also discussed.
Collapse
Affiliation(s)
- Niaz Muhammad
- a Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences , Shaanxi Normal University , Xi'an , P.R. China
| | - Qinwen Mao
- b Department of Pathology , Northwestern University Feinberg School of Medicine , Chicago , IL , USA
| | - Haibin Xia
- a Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences , Shaanxi Normal University , Xi'an , P.R. China
| |
Collapse
|
26
|
Böiers C, Richardson SE, Laycock E, Zriwil A, Turati VA, Brown J, Wray JP, Wang D, James C, Herrero J, Sitnicka E, Karlsson S, Smith AJH, Jacobsen SEW, Enver T. A Human IPS Model Implicates Embryonic B-Myeloid Fate Restriction as Developmental Susceptibility to B Acute Lymphoblastic Leukemia-Associated ETV6-RUNX1. Dev Cell 2017; 44:362-377.e7. [PMID: 29290585 PMCID: PMC5807056 DOI: 10.1016/j.devcel.2017.12.005] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 08/04/2017] [Accepted: 12/01/2017] [Indexed: 12/15/2022]
Abstract
ETV6-RUNX1 is associated with childhood acute B-lymphoblastic leukemia (cALL) functioning as a first-hit mutation that initiates a clinically silent pre-leukemia in utero. Because lineage commitment hierarchies differ between embryo and adult, and the impact of oncogenes is cell-context dependent, we hypothesized that the childhood affiliation of ETV6-RUNX1 cALL reflects its origins in a progenitor unique to embryonic life. We characterize the first emerging B cells in first-trimester human embryos, identifying a developmentally restricted CD19-IL-7R+ progenitor compartment, which transitions from a myeloid to lymphoid program during ontogeny. This developmental series is recapitulated in differentiating human pluripotent stem cells (hPSCs), thereby providing a model for the initiation of cALL. Genome-engineered hPSCs expressing ETV6-RUNX1 from the endogenous ETV6 locus show expansion of the CD19-IL-7R+ compartment, show a partial block in B lineage commitment, and produce proB cells with aberrant myeloid gene expression signatures and potential: features (collectively) consistent with a pre-leukemic state.
Collapse
Affiliation(s)
- Charlotta Böiers
- Department of Cancer Biology, UCL Cancer Institute, UCL, London, UK; Lund Stem Cell Center, Lund University, Lund, Sweden
| | | | - Emma Laycock
- Department of Cancer Biology, UCL Cancer Institute, UCL, London, UK
| | - Alya Zriwil
- Lund Stem Cell Center, Lund University, Lund, Sweden
| | | | - John Brown
- Department of Cancer Biology, UCL Cancer Institute, UCL, London, UK
| | - Jason P Wray
- Department of Cancer Biology, UCL Cancer Institute, UCL, London, UK
| | - Dapeng Wang
- Department of Cancer Biology, UCL Cancer Institute, UCL, London, UK
| | - Chela James
- Department of Cancer Biology, UCL Cancer Institute, UCL, London, UK
| | - Javier Herrero
- Department of Cancer Biology, UCL Cancer Institute, UCL, London, UK
| | - Ewa Sitnicka
- Lund Stem Cell Center, Lund University, Lund, Sweden
| | | | - Andrew J H Smith
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK; MRC Molecular Haematology Unit, University of Oxford, Oxford, UK
| | - Sten Erik W Jacobsen
- MRC Molecular Haematology Unit, University of Oxford, Oxford, UK; Departments of Cell and Molecular Biology and Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden; Haematopoietic Stem Cell Laboratory, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK; Karolinska University Hospital, Stockholm, Sweden
| | - Tariq Enver
- Department of Cancer Biology, UCL Cancer Institute, UCL, London, UK; Lund Stem Cell Center, Lund University, Lund, Sweden.
| |
Collapse
|
27
|
On being the right size: antibody repertoire formation in the mouse and human. Immunogenetics 2017; 70:143-158. [DOI: 10.1007/s00251-017-1049-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 12/04/2017] [Indexed: 01/01/2023]
|
28
|
Cao Q, Zhao X, Bai J, Gery S, Sun H, Lin DC, Chen Q, Chen Z, Mack L, Yang H, Deng R, Shi X, Chong LW, Cho H, Xie J, Li QZ, Müschen M, Atkins AR, Liddle C, Yu RT, Alkan S, Said JW, Zheng Y, Downes M, Evans RM, Koeffler HP. Circadian clock cryptochrome proteins regulate autoimmunity. Proc Natl Acad Sci U S A 2017; 114:12548-12553. [PMID: 29109286 PMCID: PMC5703267 DOI: 10.1073/pnas.1619119114] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The circadian system regulates numerous physiological processes including immune responses. Here, we show that mice deficient of the circadian clock genes Cry1 and Cry2 [Cry double knockout (DKO)] develop an autoimmune phenotype including high serum IgG concentrations, serum antinuclear antibodies, and precipitation of IgG, IgM, and complement 3 in glomeruli and massive infiltration of leukocytes into the lungs and kidneys. Flow cytometry of lymphoid organs revealed decreased pre-B cell numbers and a higher percentage of mature recirculating B cells in the bone marrow, as well as increased numbers of B2 B cells in the peritoneal cavity of Cry DKO mice. The B cell receptor (BCR) proximal signaling pathway plays a critical role in autoimmunity regulation. Activation of Cry DKO splenic B cells elicited markedly enhanced tyrosine phosphorylation of cellular proteins compared with cells from control mice, suggesting that overactivation of the BCR-signaling pathway may contribute to the autoimmunity phenotype in the Cry DKO mice. In addition, the expression of C1q, the deficiency of which contributes to the pathogenesis of systemic lupus erythematosus, was significantly down-regulated in Cry DKO B cells. Our results suggest that B cell development, the BCR-signaling pathway, and C1q expression are regulated by circadian clock CRY proteins and that their dysregulation through loss of CRY contributes to autoimmunity.
Collapse
Affiliation(s)
- Qi Cao
- Department of Hematology and Oncology, Cedars-Sinai Medical Center, Los Angeles, CA 90048;
- Department of Pathology and Laboratory Medicine, LAC+USC Medical Center, Los Angeles, CA 90033
| | - Xuan Zhao
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Jingwen Bai
- Department of Hematology and Oncology, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Oncology, Xiang An Hospital of Xiamen University, Xiamen 361102, China
| | - Sigal Gery
- Department of Hematology and Oncology, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Haibo Sun
- Department of Hematology and Oncology, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - De-Chen Lin
- Department of Hematology and Oncology, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Qi Chen
- Department of Endocrinology, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Zhengshan Chen
- Department of Pathology and Laboratory Medicine, LAC+USC Medical Center, Los Angeles, CA 90033
- Department of Systems Biology, Beckman Research Institute, City of Hope National Medical Center, Pasadena, CA 91016
| | - Lauren Mack
- Nomis Foundation Laboratories for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Henry Yang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599
| | - Ruishu Deng
- Sanford Burnham Preybs Medical Discovery Institute, La Jolla, CA 92037
| | - Xianping Shi
- Department of Hematology and Oncology, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Ling-Wa Chong
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Han Cho
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Jianjun Xie
- Department of Hematology and Oncology, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Quan-Zhen Li
- Department of Immunology, Microarray Core Facility, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Markus Müschen
- Department of Systems Biology, Beckman Research Institute, City of Hope National Medical Center, Pasadena, CA 91016
| | - Annette R Atkins
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Christopher Liddle
- Storr Liver Centre, Westmead Institute for Medical Research and Sydney Medical School, University of Sydney, Westmead Hospital, Westmead, NSW 2145, Australia
| | - Ruth T Yu
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Serhan Alkan
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Jonathan W Said
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles Medical Center, Los Angeles, CA 90095
| | - Ye Zheng
- Nomis Foundation Laboratories for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Michael Downes
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037;
| | - Ronald M Evans
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037;
- Howard Hughes Medical Institute, Salk Institute for Biological Studies, La Jolla, CA 92037
| | - H Phillip Koeffler
- Department of Hematology and Oncology, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599
| |
Collapse
|
29
|
Matheson LS, Bolland DJ, Chovanec P, Krueger F, Andrews S, Koohy H, Corcoran AE. Local Chromatin Features Including PU.1 and IKAROS Binding and H3K4 Methylation Shape the Repertoire of Immunoglobulin Kappa Genes Chosen for V(D)J Recombination. Front Immunol 2017; 8:1550. [PMID: 29204143 PMCID: PMC5698286 DOI: 10.3389/fimmu.2017.01550] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 10/31/2017] [Indexed: 11/25/2022] Open
Abstract
V(D)J recombination is essential for the generation of diverse antigen receptor (AgR) repertoires. In B cells, immunoglobulin kappa (Igκ) light chain recombination follows immunoglobulin heavy chain (Igh) recombination. We recently developed the DNA-based VDJ-seq assay for the unbiased quantitation of Igh VH and DH repertoires. Integration of VDJ-seq data with genome-wide datasets revealed that two chromatin states at the recombination signal sequence (RSS) of VH genes are highly predictive of recombination in mouse pro-B cells. It is unknown whether local chromatin states contribute to Vκ gene choice during Igκ recombination. Here we adapt VDJ-seq to profile the Igκ VκJκ repertoire and present a comprehensive readout in mouse pre-B cells, revealing highly variable Vκ gene usage. Integration with genome-wide datasets for histone modifications, DNase hypersensitivity, transcription factor binding and germline transcription identified PU.1 binding at the RSS, which was unimportant for Igh, as highly predictive of whether a Vκ gene will recombine or not, suggesting that it plays a binary, all-or-nothing role, priming genes for recombination. Thereafter, the frequency with which these genes recombine was shaped both by the presence and level of enrichment of several other chromatin features, including H3K4 methylation and IKAROS binding. Moreover, in contrast to the Igh locus, the chromatin landscape of the promoter, as well as of the RSS, contributes to Vκ gene recombination. Thus, multiple facets of local chromatin features explain much of the variation in Vκ gene usage. Together, these findings reveal shared and divergent roles for epigenetic features and transcription factors in AgR V(D)J recombination and provide avenues for further investigation of chromatin signatures that may underpin V(D)J-mediated chromosomal translocations.
Collapse
Affiliation(s)
- Louise S Matheson
- Nuclear Dynamics Programme, Babraham Institute, Cambridge, United Kingdom
| | - Daniel J Bolland
- Nuclear Dynamics Programme, Babraham Institute, Cambridge, United Kingdom
| | - Peter Chovanec
- Nuclear Dynamics Programme, Babraham Institute, Cambridge, United Kingdom
| | - Felix Krueger
- Bioinformatics Group, Babraham Institute, Cambridge, United Kingdom
| | - Simon Andrews
- Bioinformatics Group, Babraham Institute, Cambridge, United Kingdom
| | - Hashem Koohy
- Nuclear Dynamics Programme, Babraham Institute, Cambridge, United Kingdom
| | - Anne E Corcoran
- Nuclear Dynamics Programme, Babraham Institute, Cambridge, United Kingdom
| |
Collapse
|
30
|
Liu X, Li YS, Shinton SA, Rhodes J, Tang L, Feng H, Jette CA, Look AT, Hayakawa K, Hardy RR. Zebrafish B Cell Development without a Pre-B Cell Stage, Revealed by CD79 Fluorescence Reporter Transgenes. THE JOURNAL OF IMMUNOLOGY 2017; 199:1706-1715. [PMID: 28739882 DOI: 10.4049/jimmunol.1700552] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 06/27/2017] [Indexed: 11/19/2022]
Abstract
CD79a and CD79b proteins associate with Ig receptors as integral signaling components of the B cell Ag receptor complex. To study B cell development in zebrafish, we isolated orthologs of these genes and performed in situ hybridization, finding that their expression colocalized with IgH-μ in the kidney, which is the site of B cell development. CD79 transgenic lines were made by linking the promoter and upstream regulatory segments of CD79a and CD79b to enhanced GFP to identify B cells, as demonstrated by PCR analysis of IgH-μ expression in sorted cells. We crossed these CD79-GFP lines to a recombination activating gene (Rag)2:mCherry transgenic line to identify B cell development stages in kidney marrow. Initiation of CD79:GFP expression in Rag2:mCherry+ cells and the timing of Ig H and L chain expression revealed simultaneous expression of both IgH-μ- and IgL-κ-chains, without progressing through the stage of IgH-μ-chain alone. Rag2:mCherry+ cells without CD79:GFP showed the highest Rag1 and Rag2 mRNAs compared with CD79a and CD79b:GFP+ B cells, which showed strongly reduced Rag mRNAs. Thus, B cell development in zebrafish does not go through a Raghi CD79+IgH-μ+ pre-B cell stage, different from mammals. After the generation of CD79:GFP+ B cells, decreased CD79 expression occurred upon differentiation to Ig secretion, as detected by alteration from membrane to secreted IgH-μ exon usage, similar to in mammals. This confirmed a conserved role for CD79 in B cell development and differentiation, without the requirement of a pre-B cell stage in zebrafish.
Collapse
Affiliation(s)
- Xingjun Liu
- Fox Chase Cancer Center, Philadelphia, PA 19111
| | - Yue-Sheng Li
- Fox Chase Cancer Center, Philadelphia, PA 19111.,DNA Sequencing and Genomic Core, National Institutes of Health, Bethesda, MD 20892
| | | | | | | | - Hui Feng
- The Center for Cancer Research, Boston University School of Medicine, Boston, MA 02118
| | - Cicely A Jette
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84103; and
| | - A Thomas Look
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02215
| | | | | |
Collapse
|
31
|
Fernando TR, Contreras JR, Zampini M, Rodriguez-Malave NI, Alberti MO, Anguiano J, Tran TM, Palanichamy JK, Gajeton J, Ung NM, Aros CJ, Waters EV, Casero D, Basso G, Pigazzi M, Rao DS. The lncRNA CASC15 regulates SOX4 expression in RUNX1-rearranged acute leukemia. Mol Cancer 2017; 16:126. [PMID: 28724437 PMCID: PMC5517805 DOI: 10.1186/s12943-017-0692-x] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 07/03/2017] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs) play a variety of cellular roles, including regulation of transcription and translation, leading to alterations in gene expression. Some lncRNAs modulate the expression of chromosomally adjacent genes. Here, we assess the roles of the lncRNA CASC15 in regulation of a chromosomally nearby gene, SOX4, and its function in RUNX1/AML translocated leukemia. RESULTS CASC15 is a conserved lncRNA that was upregulated in pediatric B-acute lymphoblastic leukemia (B-ALL) with t (12; 21) as well as pediatric acute myeloid leukemia (AML) with t (8; 21), both of which are associated with relatively better prognosis. Enforced expression of CASC15 led to a myeloid bias in development, and overall, decreased engraftment and colony formation. At the cellular level, CASC15 regulated cellular survival, proliferation, and the expression of its chromosomally adjacent gene, SOX4. Differentially regulated genes following CASC15 knockdown were enriched for predicted transcriptional targets of the Yin and Yang-1 (YY1) transcription factor. Interestingly, we found that CASC15 enhances YY1-mediated regulation of the SOX4 promoter. CONCLUSIONS Our findings represent the first characterization of this CASC15 in RUNX1-translocated leukemia, and point towards a mechanistic basis for its action.
Collapse
Affiliation(s)
- Thilini R Fernando
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, USA.,Present Address: Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA
| | - Jorge R Contreras
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, USA.,Cellular and Molecular Pathology Ph.D. Program, UCLA, Los Angeles, USA
| | - Matteo Zampini
- Women and Child Health Department- Hematology-Oncology laboratory, University of Padova, Padova, Italy
| | - Norma I Rodriguez-Malave
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, USA.,Cellular and Molecular Pathology Ph.D. Program, UCLA, Los Angeles, USA.,Present Address: Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA
| | - Michael O Alberti
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, USA.,Present Address: Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jaime Anguiano
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, USA.,Present Address: University of San Francisco, 2130 Fulton St, San Francisco, CA, 94117, USA
| | - Tiffany M Tran
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, USA.,Molecular, Cellular and Integrative Physiology Ph.D. program, UCLA, Los Angeles, USA
| | - Jayanth K Palanichamy
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, USA.,Present Address: All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Jasmine Gajeton
- Microbiology, Immunology and Molecular Genetics Program, UCLA, Los Angeles, USA.,Present Address: Department of Molecular Cardiology Lerner Research Institute, 9500 Euclid Avenue. Cleveland, Cleveland, OH, 44195, USA
| | - Nolan M Ung
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, USA
| | - Cody J Aros
- Medical Scientist Training Program, David Geffen School of Medicine, UCLA, Los Angeles, USA
| | - Ella V Waters
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, USA.,Present Address: Department of Molecular and Cell Biology, UC Berkeley, Berkeley, USA
| | - David Casero
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, USA
| | - Giuseppe Basso
- Women and Child Health Department- Hematology-Oncology laboratory, University of Padova, Padova, Italy
| | - Martina Pigazzi
- Women and Child Health Department- Hematology-Oncology laboratory, University of Padova, Padova, Italy
| | - Dinesh S Rao
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, USA. .,Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, USA. .,Broad Stem Cell Research Center, UCLA, 650 Charles E. Young Drive, Factor 12-272, Los Angeles, CA, 90095, USA.
| |
Collapse
|
32
|
Krasnov A, Jørgensen SM, Afanasyev S. Ig-seq: Deep sequencing of the variable region of Atlantic salmon IgM heavy chain transcripts. Mol Immunol 2017. [PMID: 28623734 DOI: 10.1016/j.molimm.2017.06.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Immunoglobulin M plays a key role in systemic protection of Atlantic salmon against pathogens. Until recent, studies have focused on antigen-specific antibodies and little is known about the IgM repertoire: its size, developmental changes and responses to antigens. We report the development of deep sequencing protocol to characterize the repertoire of IgM heavy chain variable region. Its structure and changes were examined at the early stages of life and after infection with virus of cardiac myopathy. Clonotypes are identified by the V and J gene segments and amino acid sequences of CDR3, which determine the contribution of the heavy chain to the antigen binding properties. A major fraction of transcripts are functional while the rest are either sterile (transcribed from noncoding parts of Ig loci) or include stop codons. Despite marked difference in frequencies of combinations of V and J genes, the size of repertoire is large. The IgM diversity steadily increases after hatch followed with temporal reduction during smoltification and recovery after seawater transfer. Most clonotypes are present only in one fish. However multiple transcripts in uninfected fish are produced exclusively from a small fraction of shared clonotypes. While only 4.7% of clonotypes are detected in three and more fish, they comprise 35% of transcripts. Increased frequencies of most abundant clonotypes were detected in the head kidney and blood at ten weeks after viral infection and all were shared. Occurrence of the same clonotypes in multiple individuals can be explained with either their simple structure or exposure to common antigens. Complexity of CDR3 assessed by contents of non complementary nucleotides is slightly lower in shared clonotypes but difference is small. High nucleotide diversity of CDR3 with identical amino acid sequences suggests selection.
Collapse
Affiliation(s)
| | | | - Sergey Afanasyev
- Nofima AS, PO Box 210, NO-1431 Ås, Norway; Sechenov Institute of Evolutionary Physiology and Biochemistry, M. Toreza av. 44, Saint Petersburg 194223, Russia
| |
Collapse
|
33
|
Adoptive immunotherapy for hematological malignancies: Current status and new insights in chimeric antigen receptor T cells. Blood Cells Mol Dis 2016; 62:49-63. [DOI: 10.1016/j.bcmd.2016.11.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Revised: 11/05/2016] [Accepted: 11/06/2016] [Indexed: 12/20/2022]
|
34
|
Li S, Liu W, Li Y, Zhao S, Liu C, Hu M, Yue W, Liu Y, Wang Y, Yang R, Xiang R, Liu F. Contribution of secondary Igkappa rearrangement to primary immunoglobulin repertoire diversification. Mol Immunol 2016; 78:193-206. [PMID: 27665270 DOI: 10.1016/j.molimm.2016.09.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 09/01/2016] [Accepted: 09/06/2016] [Indexed: 10/21/2022]
Abstract
Abs reactive to DNA and DNA/histone complexes are a distinguished characteristic of primary immunoglobulin repertoires in autoimmune B6.MRL-Faslpr and MRL/MpJ-Faslpr mice. These mice are defective in Fas receptor, which is critical for the apoptosis of autoreactive B cells by an extrinsic pathway. In the present study, we explored the possibility that bone marrow small pre-B and immature B cells from adult B6.MRL-Faslpr mice and MRL/MpJ-Faslpr mice respectively, which contain autoreactive B-cell antigen receptors (BCR) and manifest autoimmune syndromes, exhibit enhanced receptor editing patterns. Indeed, FASlpr pre B and immature B cells were shown to possess more ongoing replacements of non-productive (nP) than productive (P) primary VκJκ rearrangements. Significantly, the P vs nP ratios of these replaced primary rearrangements were 1:2, thus indicating that κ light-chain production appears not to inhibit secondary rearrangements. In addition, we identified multiple atypical rearrangements, such as Vκ cRS (cryptic recombination signals) cleavages. These results suggest that the onset of light chain secondary rearrangements persists similarly as a non-selected mode and independent of BCR autoreactivity during certain developmental windows of bone marrow B cells in lupus-prone mice and control, and leads us to propose the function of secondary, de novo Igκ rearrangements to increase BCR diversity.
Collapse
Affiliation(s)
- Shufang Li
- Department of Immunology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Wei Liu
- Tianjin Entry-Exit Inspection and Quarantine Bureau, Tianjin 300308, China
| | - Yinghui Li
- Department of Immunology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Shaorong Zhao
- Department of Immunology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Can Liu
- Department of Immunology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Mengyun Hu
- Collage of Life Science, Nankai University, Tianjin, 300071, China
| | - Wei Yue
- Department of Neurology, Huanhu Hospital, Tianjin 300060, China
| | - Yanhua Liu
- Department of Immunology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Yue Wang
- Department of Immunology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Rongcun Yang
- Department of Immunology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Rong Xiang
- Department of Immunology, School of Medicine, Nankai University, Tianjin 300071, China.
| | - Feifei Liu
- Department of Immunology, School of Medicine, Nankai University, Tianjin 300071, China.
| |
Collapse
|
35
|
Abstract
Early in embryonic development of mice, from day 12.5 after conception, myeloid-lymphoid bipotent progenitors, expressing receptors both for IL7 and CSF-1, migrate from embryonic blood into developing fetal liver. These progenitors also express multiple chemokine receptors, i.e., CCR7, CXCR3, CXCR4, and CXCR5, all on one cell. Their migration through LYVE-1+ vascular endothelium is guided by CCR7, recognizing the chemokine CCL19, and by CXCR3, recognizing CXCL10/11, chemokines which are both produced by the endothelium. Once inside fetal liver, the progenitors are attracted by the chemokine CXCL12 to ALCAM+ liver mesenchyme, which produces not only this chemokine, but also the myeloid differentiation-inducing cytokine CSF-1 and the lymphoid differentiation-inducing cytokine IL7. In this mesenchymal environment B-lymphocyte lineage progenitors are then induced by IL7 to enter differentiation and Ig gene rearrangements. Within 3-4 days surface IgM+ immature B-cells develop, which are destined to enter the B1-cell compartments in the peripheral lymphoid organs.
Collapse
Affiliation(s)
- K Kajikhina
- Research Group on "Lymphocyte Development," Max Planck Institute for Infection Biology, Berlin, Germany
| | - M Tsuneto
- Research Group on "Lymphocyte Development," Max Planck Institute for Infection Biology, Berlin, Germany; Reproductive Centre, Mio Fertility Clinic, Yonago, Japan
| | - F Melchers
- Research Group on "Lymphocyte Development," Max Planck Institute for Infection Biology, Berlin, Germany.
| |
Collapse
|
36
|
Jensen CT, Lang S, Somasundaram R, Soneji S, Sigvardsson M. Identification of Stage-Specific Surface Markers in Early B Cell Development Provides Novel Tools for Identification of Progenitor Populations. THE JOURNAL OF IMMUNOLOGY 2016; 197:1937-44. [PMID: 27456481 DOI: 10.4049/jimmunol.1600297] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 06/22/2016] [Indexed: 11/19/2022]
Abstract
Whereas the characterization of B lymphoid progenitors has been facilitated by the identification of lineage- and stage-specific surface markers, the continued identification of differentially expressed proteins increases our capacity to explore normal and malignant B cell development. To identify novel surface markers with stage-specific expression patterns, we explored the reactivity of CD19(+) B cell progenitor cells to Abs targeted to 176 surface proteins. Markers with stage-specific expression were identified using a transgenic reporter gene system subdividing the B cell progenitors into four surface IgM(-) stages. This approach affirmed the utility of known stage-specific markers, as well as identifying additional proteins that selectively marked defined stages of B cell development. Among the stage-specific markers were the cell adhesion proteins CD49E, CD11A, and CD54 that are highly expressed selectively on the most immature progenitors. This work identifies a set of novel stage-specific surface markers that can be used as a complement to the classical staining protocols to explore B lymphocyte development.
Collapse
Affiliation(s)
- Christina T Jensen
- Department of Molecular Hematology, Lund University, 22184 Lund, Sweden; and
| | - Stefan Lang
- Department of Molecular Hematology, Lund University, 22184 Lund, Sweden; and
| | - Rajesh Somasundaram
- Department of Clinical and Experimental Medicine, Linköping University, 58185 Linköping, Sweden
| | - Shamit Soneji
- Department of Molecular Hematology, Lund University, 22184 Lund, Sweden; and
| | - Mikael Sigvardsson
- Department of Molecular Hematology, Lund University, 22184 Lund, Sweden; and Department of Clinical and Experimental Medicine, Linköping University, 58185 Linköping, Sweden
| |
Collapse
|
37
|
Ghosh D, Wikenheiser DJ, Kennedy B, McGovern KE, Stuart JD, Wilson EH, Stumhofer JS. An Atypical Splenic B Cell Progenitor Population Supports Antibody Production during Plasmodium Infection in Mice. THE JOURNAL OF IMMUNOLOGY 2016; 197:1788-800. [PMID: 27448588 DOI: 10.4049/jimmunol.1502199] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 06/17/2016] [Indexed: 12/26/2022]
Abstract
Hematopoietic stem and progenitor cells (HSPCs) function to replenish the immune cell repertoire under steady-state conditions and in response to inflammation due to infection or stress. Whereas the bone marrow serves as the primary niche for hematopoiesis, extramedullary mobilization and differentiation of HSPCs occur in the spleen during acute Plasmodium infection, a critical step in the host immune response. In this study, we identified an atypical HSPC population in the spleen of C57BL/6 mice, with a lineage(-)Sca-1(+)c-Kit(-) (LSK(-)) phenotype that proliferates in response to infection with nonlethal Plasmodium yoelii 17X. Infection-derived LSK(-) cells upon transfer into naive congenic mice were found to differentiate predominantly into mature follicular B cells. However, when transferred into infection-matched hosts, infection-derived LSK(-) cells gave rise to B cells capable of entering into a germinal center reaction, and they developed into memory B cells and Ab-secreting cells that were capable of producing parasite-specific Abs. Differentiation of LSK(-) cells into B cells in vitro was enhanced in the presence of parasitized RBC lysate, suggesting that LSK(-) cells expand and differentiate in direct response to the parasite. However, the ability of LSK(-) cells to differentiate into B cells was not dependent on MyD88, as myd88(-/-) LSK(-) cell expansion and differentiation remained unaffected after Plasmodium infection. Collectively, these data identify a population of atypical lymphoid progenitors that differentiate into B lymphocytes in the spleen and are capable of contributing to the ongoing humoral immune response against Plasmodium infection.
Collapse
Affiliation(s)
- Debopam Ghosh
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR 72205; and
| | - Daniel J Wikenheiser
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR 72205; and
| | - Brian Kennedy
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR 72205; and
| | - Kathryn E McGovern
- Division of Biomedical Sciences, University of California, Riverside, CA 92521
| | - Johnasha D Stuart
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR 72205; and
| | - Emma H Wilson
- Division of Biomedical Sciences, University of California, Riverside, CA 92521
| | - Jason S Stumhofer
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR 72205; and
| |
Collapse
|
38
|
Hayakawa K, Formica AM, Colombo MJ, Shinton SA, Brill-Dashoff J, Morse HC, Li YS, Hardy RR. Loss of a chromosomal region with synteny to human 13q14 occurs in mouse chronic lymphocytic leukemia that originates from early-generated B-1 B cells. Leukemia 2016; 30:1510-9. [PMID: 27055869 PMCID: PMC4979312 DOI: 10.1038/leu.2016.61] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Revised: 02/25/2016] [Accepted: 02/29/2016] [Indexed: 01/01/2023]
Abstract
A common feature of B-cell chronic lymphocytic leukemia (CLL) is chromosomal loss of 13q14, containing the miR15a/16-1 locus controlling B-cell proliferation. However, CLL etiology remains unclear. CLL is an adult leukemia with an incidence that increases with advancing age. A unique feature of CLL is biased B-cell antigen receptor (BCR) usage, autoreactivity with polyreactivity and CD5 expression, all suggest a role for the BCR in driving CLL pathogenesis. Among human CLLs, BCRs autoreactive with non-muscle myosin IIA (AMyIIA) are recurrent. Here we identify an unmutated AMyIIA BCR in mouse, with distinctive CDR3 segments capable of promoting leukemogenesis. B cells with this AMyIIA BCR are generated by BCR-dependent signaling during B-1 fetal/neonatal development with CD5 induction, but not in adults. These early-generated AMyIIA B-1 B cells self-renew, increase during aging and can progress to become monoclonal B-cell lymphocytosis, followed by aggressive CLL in aged mice, often with the loss of a chromosomal region containing the miR15a/16-1 locus of varying length, as in human CLL. Thus, the ability to generate this defined autoreactive BCR by B-1 B cells is a key predisposing step in mice, promoting progression to chronic leukemia.
Collapse
MESH Headings
- Animals
- B-Lymphocytes/pathology
- Cell Self Renewal
- Chromosome Deletion
- Chromosome Disorders
- Chromosomes, Human, Pair 13
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/etiology
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Mice
- Nonmuscle Myosin Type IIA/metabolism
- Receptors, Antigen, B-Cell/metabolism
- Synteny
Collapse
Affiliation(s)
- Kyoko Hayakawa
- Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111, USA
| | | | | | | | | | - Herbert C. Morse
- Laboratory of Immunogenetics, National Institute of Allergy and
Infectious Diseases, National Institutes of Health, Rockville, Maryland 20852,
USA
| | - Yue-Sheng Li
- Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111, USA
| | | |
Collapse
|
39
|
Abstract
Ying Yang 1 (YY1) is a ubiquitously expressed transcription factor shown to be essential for pro-B-cell development. However, the role of YY1 in other B-cell populations has never been investigated. Recent bioinformatics analysis data have implicated YY1 in the germinal center (GC) B-cell transcriptional program. In accord with this prediction, we demonstrated that deletion of YY1 by Cγ1-Cre completely prevented differentiation of GC B cells and plasma cells. To determine if YY1 was also required for the differentiation of other B-cell populations, we deleted YY1 with CD19-Cre and found that all peripheral B-cell subsets, including B1 B cells, require YY1 for their differentiation. Transitional 1 (T1) B cells were the most dependent upon YY1, being sensitive to even a half-dosage of YY1 and also to short-term YY1 deletion by tamoxifen-induced Cre. We show that YY1 exerts its effects, in part, by promoting B-cell survival and proliferation. ChIP-sequencing shows that YY1 predominantly binds to promoters, and pathway analysis of the genes that bind YY1 show enrichment in ribosomal functions, mitochondrial functions such as bioenergetics, and functions related to transcription such as mRNA splicing. By RNA-sequencing analysis of differentially expressed genes, we demonstrated that YY1 normally activates genes involved in mitochondrial bioenergetics, whereas it normally down-regulates genes involved in transcription, mRNA splicing, NF-κB signaling pathways, the AP-1 transcription factor network, chromatin remodeling, cytokine signaling pathways, cell adhesion, and cell proliferation. Our results show the crucial role that YY1 plays in regulating broad general processes throughout all stages of B-cell differentiation.
Collapse
|
40
|
Macrophage colony-stimulating factor receptor marks and regulates a fetal myeloid-primed B-cell progenitor in mice. Blood 2016; 128:217-26. [PMID: 27207794 DOI: 10.1182/blood-2016-01-693887] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 05/07/2016] [Indexed: 12/24/2022] Open
Abstract
Although it is well established that unique B-cell lineages develop through distinct regulatory mechanisms during embryonic development, much less is understood about the differences between embryonic and adult B-cell progenitor cells, likely to underpin the genetics and biology of infant and childhood PreB acute lymphoblastic leukemia (PreB-ALL), initiated by distinct leukemia-initiating translocations during embryonic development. Herein, we establish that a distinct subset of the earliest CD19(+) B-cell progenitors emerging in the E13.5 mouse fetal liver express the colony-stimulating factor-1 receptor (CSF1R), previously thought to be expressed, and play a lineage-restricted role in development of myeloid lineages, and macrophages in particular. These early embryonic CSF1R(+)CD19(+) ProB cells also express multiple other myeloid genes and, in line with this, possess residual myeloid as well as B-cell, but not T-cell lineage potential. Notably, these CSF1R(+) myeloid-primed ProB cells are uniquely present in a narrow window of embryonic fetal liver hematopoiesis and do not persist in adult bone marrow. Moreover, analysis of CSF1R-deficient mice establishes a distinct role of CSF1R in fetal B-lymphopoiesis. CSF1R(+) myeloid-primed embryonic ProB cells are relevant for infant and childhood PreB-ALLs, which frequently have a bi-phenotypic B-myeloid phenotype, and in which CSF1R-rearrangements have recently been reported.
Collapse
|
41
|
Hershberg U, Luning Prak ET. The analysis of clonal expansions in normal and autoimmune B cell repertoires. Philos Trans R Soc Lond B Biol Sci 2016; 370:rstb.2014.0239. [PMID: 26194753 PMCID: PMC4528416 DOI: 10.1098/rstb.2014.0239] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Clones are the fundamental building blocks of immune repertoires. The number of different clones relates to the diversity of the repertoire, whereas their size and sequence diversity are linked to selective pressures. Selective pressures act both between clones and within different sequence variants of a clone. Understanding how clonal selection shapes the immune repertoire is one of the most basic questions in all of immunology. But how are individual clones defined? Here we discuss different approaches for defining clones, starting with how antibodies are diversified during different stages of B cell development. Next, we discuss how clones are defined using different experimental methods. We focus on high-throughput sequencing datasets, and the computational challenges and opportunities that these data have for mining the antibody repertoire landscape. We discuss methods that visualize sequence variants within the same clone and allow us to consider collections of shared mutations to determine which sequences share a common ancestry. Finally, we comment on features of frequently encountered expanded B cell clones that may be of particular interest in the setting of autoimmunity and other chronic conditions.
Collapse
Affiliation(s)
- Uri Hershberg
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Bossone 7-711, 3141 Chestnut Street, Philadelphia, PA 19104, USA Department of Immunology and Microbiology, College of Medicine, Drexel University, Bossone 7-711, 3141 Chestnut Street, Philadelphia, PA 19104, USA
| | - Eline T Luning Prak
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, 405B Stellar Chance Labs, 422 Curie Boulevard, Philadelphia, PA 19104, USA
| |
Collapse
|
42
|
Bauer E, Schlederer M, Scheicher R, Horvath J, Aigner P, Schiefer AI, Kain R, Regele H, Hoermann G, Steiner G, Kenner L, Sexl V, Villunger A, Moriggl R, Stoiber D. Cooperation of ETV6/RUNX1 and BCL2 enhances immunoglobulin production and accelerates glomerulonephritis in transgenic mice. Oncotarget 2016; 7:12191-205. [PMID: 26919255 PMCID: PMC4914278 DOI: 10.18632/oncotarget.7687] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 01/31/2016] [Indexed: 01/15/2023] Open
Abstract
The t(12;21) translocation generating the ETV6/RUNX1 fusion gene represents the most frequent chromosomal rearrangement in childhood leukemia. Presence of ETV6/RUNX1 alone is usually not sufficient for leukemia onset, and additional genetic alterations have to occur in ETV6/RUNX1-positive cells to cause transformation. We have previously generated an ETV6/RUNX1 transgenic mouse model where the expression of the fusion gene is restricted to CD19-positive B cells. Since BCL2 family members have been proposed to play a role in leukemogenesis, we investigated combined effects of ETV6/RUNX1 with exogenous expression of the antiapoptotic protein BCL2 by crossing ETV6/RUNX1 transgenic animals with Vav-BCL2 transgenic mice. Strikingly, co-expression of ETV6/RUNX1 and BCL2 resulted in significantly shorter disease latency in mice, indicating oncogene cooperativity. This was associated with faster development of follicular B cell lymphoma and exacerbated immune complex glomerulonephritis. ETV6/RUNX1-BCL2 double transgenic animals displayed increased B cell numbers and immunoglobulin titers compared to Vav-BCL2 transgenic mice. This led to pronounced deposition of immune complexes in glomeruli followed by accelerated development of immune complex glomerulonephritis. Thus, our study reveals a previously unrecognized synergism between ETV6/RUNX1 and BCL2 impacting on malignant disease and autoimmunity.
Collapse
Affiliation(s)
- Eva Bauer
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria
| | - Michaela Schlederer
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | - Ruth Scheicher
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Jaqueline Horvath
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Petra Aigner
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria
| | - Ana-Iris Schiefer
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | - Renate Kain
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | - Heinz Regele
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | - Gregor Hoermann
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Günter Steiner
- Cluster Arthritis and Rehabilitation, Ludwig Boltzmann Society, Vienna, Austria
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Lukas Kenner
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
- Unit of Pathology of Laboratory Animals, University of Veterinary Medicine, Vienna, Austria
| | - Veronika Sexl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Andreas Villunger
- Division of Developmental Immunology, Biocenter, Medical University Innsbruck, Innsbruck, Austria
- Tyrolean Cancer Research Institute, Innsbruck, Austria
| | - Richard Moriggl
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, Vienna, Austria
| | - Dagmar Stoiber
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
43
|
De Grandis M, Lhoumeau AC, Mancini SJC, Aurrand-Lions M. Adhesion receptors involved in HSC and early-B cell interactions with bone marrow microenvironment. Cell Mol Life Sci 2016; 73:687-703. [PMID: 26495446 PMCID: PMC11108274 DOI: 10.1007/s00018-015-2064-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 09/16/2015] [Accepted: 10/08/2015] [Indexed: 02/06/2023]
Abstract
Hematopoiesis takes place in the bone marrow of adult mammals and is the process by which blood cells are replenished every day throughout life. Differentiation of hematopoietic cells occurs in a stepwise manner through intermediates of differentiation that could be phenotypically identified. This has allowed establishing hematopoietic cell classification with hematopoietic stem cells (HSCs) at the top of the hierarchy. HSCs are mostly quiescent and serve as a reservoir for maintenance of lifelong hematopoiesis. Over recent years, it has become increasingly clear that HSC quiescence is not only due to intrinsic properties, but is also mediated by cognate interactions between HSCs and surrounding cells within micro-anatomical sites called “niches”. This hematopoietic/stromal crosstalk model also applies to more mature progenitors such as B cell progenitors, which are thought to reside in distinct “niches”. This prompted many research teams to search for specific molecular mechanisms supporting leuko-stromal crosstalk in the bone marrow and acting at specific stage of differentiation to regulate hematopoietic homeostasis. Here, we review recent data on adhesion mechanisms involved in HSCs and B cell progenitors interactions with surrounding bone marrow stromal cells.
Collapse
Affiliation(s)
- Maria De Grandis
- Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, Inserm U1068, CNRS UMR7258, Aix-Marseille Université UM105, Marseille, France
| | - Anne-Catherine Lhoumeau
- Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, Inserm U1068, CNRS UMR7258, Aix-Marseille Université UM105, Marseille, France
| | - Stéphane J. C. Mancini
- Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, Inserm U1068, CNRS UMR7258, Aix-Marseille Université UM105, Marseille, France
| | - Michel Aurrand-Lions
- Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, Inserm U1068, CNRS UMR7258, Aix-Marseille Université UM105, Marseille, France
| |
Collapse
|
44
|
Dai H, Wang Y, Lu X, Han W. Chimeric Antigen Receptors Modified T-Cells for Cancer Therapy. J Natl Cancer Inst 2016; 108:djv439. [PMID: 26819347 PMCID: PMC4948566 DOI: 10.1093/jnci/djv439] [Citation(s) in RCA: 187] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 12/21/2015] [Indexed: 02/06/2023] Open
Abstract
The genetic modification and characterization of T-cells with chimeric antigen receptors (CARs) allow functionally distinct T-cell subsets to recognize specific tumor cells. The incorporation of costimulatory molecules or cytokines can enable engineered T-cells to eliminate tumor cells. CARs are generated by fusing the antigen-binding region of a monoclonal antibody (mAb) or other ligand to membrane-spanning and intracellular-signaling domains. They have recently shown clinical benefit in patients treated with CD19-directed autologous T-cells. Recent successes suggest that the modification of T-cells with CARs could be a powerful approach for developing safe and effective cancer therapeutics. Here, we briefly review early studies, consider strategies to improve the therapeutic potential and safety, and discuss the challenges and future prospects for CAR T-cells in cancer therapy.
Collapse
Affiliation(s)
- Hanren Dai
- Affiliations of authors: Department of Immunology (HD, YW, WH) and Department of Molecular Biology (WH), Institute of Basic Medicine, School of Life Sciences, Department of Bio-therapeutic (HD, YW, WH), and Department of Hematology (XL), Chinese PLA General Hospital, Beijing, China
| | - Yao Wang
- Affiliations of authors: Department of Immunology (HD, YW, WH) and Department of Molecular Biology (WH), Institute of Basic Medicine, School of Life Sciences, Department of Bio-therapeutic (HD, YW, WH), and Department of Hematology (XL), Chinese PLA General Hospital, Beijing, China
| | - Xuechun Lu
- Affiliations of authors: Department of Immunology (HD, YW, WH) and Department of Molecular Biology (WH), Institute of Basic Medicine, School of Life Sciences, Department of Bio-therapeutic (HD, YW, WH), and Department of Hematology (XL), Chinese PLA General Hospital, Beijing, China
| | - Weidong Han
- Affiliations of authors: Department of Immunology (HD, YW, WH) and Department of Molecular Biology (WH), Institute of Basic Medicine, School of Life Sciences, Department of Bio-therapeutic (HD, YW, WH), and Department of Hematology (XL), Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
45
|
Xu K, Liu H, Shi Z, Song G, Zhu X, Jiang Y, Zhou Z, Liu X. Disruption of the RAG2 zinc finger motif impairs protein stability and causes immunodeficiency. Eur J Immunol 2015; 46:1011-9. [PMID: 26692406 DOI: 10.1002/eji.201545896] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2015] [Revised: 11/18/2015] [Accepted: 12/15/2015] [Indexed: 12/28/2022]
Abstract
Although the RAG2 core domain is the minimal region required for V(D)J recombination, the noncore region also plays important roles in the regulation of recombination, and mutations in this region are often related to severe combined immunodeficiency. A complete understanding of the functions of the RAG2 noncore region and the potential contributions of its individual residues has not yet been achieved. Here, we show that the zinc finger motif within the noncore region of RAG2 is indispensable for maintaining the stability of the RAG2 protein. The zinc finger motif in the noncore region of RAG2 is highly conserved from zebrafish to humans. Knock-in mice carrying a zinc finger mutation (C478Y) exhibit decreased V(D)J recombination efficiency and serious impairment in T/B-cell development due to RAG2 instability. Further studies also reveal the importance of the zinc finger motif for RAG2 stability. Moreover, mice harboring a RAG2 noncore region mutation (N474S), which is located near C478 but is not zinc-binding, exhibit no impairment in either RAG2 stability or T/B-cell development. Taken together, our findings contribute to defining critical functions of the RAG2 zinc finger motif and provide insights into the relationships between the mutations within this motif and immunodeficiency diseases.
Collapse
Affiliation(s)
- Ke Xu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Haifeng Liu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zhubing Shi
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Guangrong Song
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xiaoyan Zhu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yuzhang Jiang
- Department of Medical Laboratory, Huaian First People's Hospital, Nanjing Medical University, Huaian, Jiangsu, China
| | - Zhaocai Zhou
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xiaolong Liu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
46
|
Iacoangeli A, Lui A, Naik U, Ohta Y, Flajnik M, Hsu E. Biased Immunoglobulin Light Chain Gene Usage in the Shark. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2015; 195:3992-4000. [PMID: 26342033 PMCID: PMC4592821 DOI: 10.4049/jimmunol.1501426] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 08/04/2015] [Indexed: 12/13/2022]
Abstract
This study of a large family of κ L chain clusters in nurse shark completes the characterization of its classical Ig gene content (two H chain isotypes, μ and ω, and four L chain isotypes, κ, λ, σ, and σ-2). The shark κ clusters are minigenes consisting of a simple VL-JL-CL array, where V to J recombination occurs over an ~500-bp interval, and functional clusters are widely separated by at least 100 kb. Six out of ~39 κ clusters are prerearranged in the germline (germline joined). Unlike the complex gene organization and multistep assembly process of Ig in mammals, each shark Ig rearrangement, somatic or in the germline, appears to be an independent event localized to the minigene. This study examined the expression of functional, nonproductive, and sterile transcripts of the κ clusters compared with the other three L chain isotypes. κ cluster usage was investigated in young sharks, and a skewed pattern of split gene expression was observed, one similar in functional and nonproductive rearrangements. These results show that the individual activation of the spatially distant κ clusters is nonrandom. Although both split and germline-joined κ genes are expressed, the latter are prominent in young animals and wane with age. We speculate that, in the shark, the differential activation of the multiple isotypes can be advantageously used in receptor editing.
Collapse
Affiliation(s)
- Anna Iacoangeli
- Department of Physiology and Pharmacology, State University of New York Health Science Center at Brooklyn, Brooklyn, NY 11203; and
| | - Anita Lui
- Department of Physiology and Pharmacology, State University of New York Health Science Center at Brooklyn, Brooklyn, NY 11203; and
| | - Ushma Naik
- Department of Physiology and Pharmacology, State University of New York Health Science Center at Brooklyn, Brooklyn, NY 11203; and
| | - Yuko Ohta
- Department of Microbiology and Immunology, University of Maryland, Baltimore, MD 21201
| | - Martin Flajnik
- Department of Microbiology and Immunology, University of Maryland, Baltimore, MD 21201
| | - Ellen Hsu
- Department of Physiology and Pharmacology, State University of New York Health Science Center at Brooklyn, Brooklyn, NY 11203; and
| |
Collapse
|
47
|
Abstract
Second-generation chimeric antigen receptors (CARs) retarget and reprogramme T cells to augment their antitumour efficacy. The combined activating and co-stimulatory domains incorporated in these CARs critically determine the function, differentiation, metabolism and persistence of engineered T cells. CD19-targeted CARs that incorporate CD28 or 4-1BB signalling domains are the best known to date. Both have shown remarkable complete remission rates in patients with refractory B cell malignancies. Recent data indicate that CD28-based CARs direct a brisk proliferative response and boost effector functions, whereas 4-1BB-based CARs induce a more progressive T cell accumulation that may compensate for less immediate potency. These distinct kinetic features can be exploited to further develop CAR-based T cell therapies for a variety of cancers. A new field of immunopharmacology is emerging.
Collapse
|
48
|
Holodick NE, Rothstein TL. B cells in the aging immune system: time to consider B-1 cells. Ann N Y Acad Sci 2015; 1362:176-87. [PMID: 26194480 DOI: 10.1111/nyas.12825] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 05/15/2015] [Accepted: 05/27/2015] [Indexed: 02/05/2023]
Abstract
The investigation of immune senescence has uncovered many changes in B cell development, maintenance, and function with increasing age. However, most of these studies have focused on conventional B cell subsets in the spleen. The B-1 cell subset is an essential arm of the innate immune system, which in general has been understudied in terms of immune senescence. Here, we review what is currently known about B cells during aging and go on to describe why B-1 cell biology is an important component of the aging immune system in the context of diseases that most affect the aged population.
Collapse
Affiliation(s)
- Nichol E Holodick
- Center for Oncology and Cell Biology, The Feinstein Institute for Medical Research, Manhasset, New York
| | - Thomas L Rothstein
- Center for Oncology and Cell Biology, The Feinstein Institute for Medical Research, Manhasset, New York.,Departments of Medicine and Molecular Medicine, The Hofstra North Shore-LIJ School of Medicine, Manhasset, New York
| |
Collapse
|
49
|
Xing Y, Ji Q, Lin Y, Fu M, Gao J, Zhang P, Hu X, Feng L, Liu Y, Han H, Li W. Positive selection of natural poly-reactive B cells in the periphery occurs independent of heavy chain allelic inclusion. PLoS One 2015; 10:e0125747. [PMID: 25993514 PMCID: PMC4437983 DOI: 10.1371/journal.pone.0125747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2014] [Accepted: 03/25/2015] [Indexed: 11/28/2022] Open
Abstract
Natural autoreactive B cells are important mediators of autoimmune diseases. Receptor editing is known to play an important role in both central and peripheral B cell tolerance. However, the role of allelic inclusion in the development of natural autoreactive B cells is not clear. Previously, we generated μ chain (TgVH3B4I) and μ/κ chains (TgVH/L3B4) transgenic mice using transgene derived from the 3B4 hybridoma, which produce poly-reactive natural autoantibodies. In this study, we demonstrate that a considerable population of B cells edited their B cells receptors (BCRs) via light chain or heavy chain allelic inclusion during their development in TgVH3B4I mice. Additionally, allelic inclusion occurred more frequently in the periphery and promoted the differentiation of B cells into marginal zone or B-1a cells in TgVH3B4I mice. B cells from TgVH/L3B4 mice expressing the intact transgenic 3B4 BCR without receptor editing secreted poly-reactive 3B4 antibody. Interestingly, however, B cell that underwent allelic inclusion in TgVH3B4I mice also produced poly-reactive autoantibodies in vivo and in vitro. Our findings suggest that receptor editing plays a minor role in the positive selection of B cells expressing natural poly-reactive BCRs, which can be positively selected through heavy chain allelic inclusion to retain their poly-reactivity in the periphery.
Collapse
Affiliation(s)
- Ying Xing
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China; Department of Endocrinology and Metabolism Disease, Xijing Hospital, Fourth Military Medical University, Xi'an, China; State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, China
| | - Qiuhe Ji
- Department of Endocrinology and Metabolism Disease, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Ying Lin
- Department of Otolaryngology Head and Neck surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Meng Fu
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jixin Gao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Ping Zhang
- Department of Otolaryngology Head and Neck surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xingbin Hu
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, China
| | - Lei Feng
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, China
| | - Yufeng Liu
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Hua Han
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, China
| | - Wei Li
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
50
|
Zhou Y, Li YS, Bandi SR, Tang L, Shinton SA, Hayakawa K, Hardy RR. Lin28b promotes fetal B lymphopoiesis through the transcription factor Arid3a. ACTA ACUST UNITED AC 2015; 212:569-80. [PMID: 25753579 PMCID: PMC4387290 DOI: 10.1084/jem.20141510] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 02/11/2015] [Indexed: 01/24/2023]
Abstract
Zhou et al. demonstrate a requirement for the Let-7–Lin28b axis regulating a shift in development between fetal liver and bone marrow B lymphocyte progenitors in the generation of B1 versus B2 B cells. Specifically, the transcription factor Arid3a, induced by Lin28b and a target of Let-7 miRNA, is sufficient to recapitulate fetal B cell development from bone marrow progenitors. Mouse B cell precursors from fetal liver and adult bone marrow (BM) generate distinctive B cell progeny when transplanted into immunodeficient recipients, supporting a two-pathway model for B lymphopoiesis, fetal “B-1” and adult “B-2.” Recently, Lin28b was shown to be important for the switch between fetal and adult pathways; however, neither the mechanism of Lin28b action nor the importance of B cell antigen receptor (BCR) signaling in this process was addressed. Here, we report key advances in our understanding of the regulation of B-1/B-2 development. First, modulation of Let-7 in fetal pro-B cells is sufficient to alter fetal B-1 development to produce B cells resembling the progeny of adult B-2 development. Second, intact BCR signaling is required for the generation of B1a B cells from Lin28b-transduced BM progenitors, supporting a requirement for ligand-dependent selection, as is the case for normal B1a B cells. Third, the VH repertoire of Lin28b-induced BM B1a B cells differs from that of normal B1a, suggesting persisting differences from fetal progenitors. Finally, we identify the Arid3a transcription factor as a key target of Let-7, whose ectopic expression is sufficient to induce B-1 development in adult pro-B cells and whose silencing by knockdown blocks B-1 development in fetal pro-B cells.
Collapse
Affiliation(s)
- Yan Zhou
- Fox Chase Cancer Center, Philadelphia, PA 19111
| | | | | | | | | | | | | |
Collapse
|