1
|
Glynn RA, Hayer KE, Bassing CH. ATM-dependent Phosphorylation of Nemo SQ Motifs Is Dispensable for Nemo-mediated Gene Expression Changes in Response to DNA Double-Strand Breaks. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:628-640. [PMID: 39007641 PMCID: PMC11348802 DOI: 10.4049/jimmunol.2300139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 06/25/2024] [Indexed: 07/16/2024]
Abstract
In response to DNA double-strand breaks (DSBs), the ATM kinase activates NF-κB factors to stimulate gene expression changes that promote survival and allow time for cells to repair damage. In cell lines, ATM can activate NF-κB transcription factors via two independent, convergent mechanisms. One is ATM-mediated phosphorylation of nuclear NF-κB essential modulator (Nemo) protein, which leads to monoubiquitylation and export of Nemo to the cytoplasm where it engages the IκB kinase (IKK) complex to activate NF-κB. Another is DSB-triggered migration of ATM into the cytoplasm, where it promotes monoubiquitylation of Nemo and the resulting IKK-mediated activation of NF-κB. ATM has many other functions in the DSB response beyond activation of NF-κB, and Nemo activates NF-κB downstream of diverse stimuli, including developmental or proinflammatory stimuli such as LPSs. To elucidate the in vivo role of DSB-induced, ATM-dependent changes in expression of NF-κB-responsive genes, we generated mice expressing phosphomutant Nemo protein lacking consensus SQ sites for phosphorylation by ATM or related kinases. We demonstrate that these mice are viable/healthy and fertile and exhibit overall normal B and T lymphocyte development. Moreover, treatment of their B lineage cells with LPS induces normal NF-κB-regulated gene expression changes. Furthermore, in marked contrast to results from a pre-B cell line, primary B lineage cells expressing phosphomutant Nemo treated with the genotoxic drug etoposide induce normal ATM- and Nemo-dependent changes in expression of NF-κB-regulated genes. Our data demonstrate that ATM-dependent phosphorylation of Nemo SQ motifs in vivo is dispensable for DSB-signaled changes in expression of NF-κB-regulated genes.
Collapse
Affiliation(s)
- Rebecca A. Glynn
- Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Katharina E. Hayer
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
- Biomedical Engineering Doctoral Degree Program, School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104
- Department of Biomedical and Health Informatics, Children’s Hospital of Philadelphia, Perelman School of Medicine, Philadelphia, PA, 19104
| | - Craig H. Bassing
- Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
2
|
Huang X, Li Y, Zhang J, Yan L, Zhao H, Ding L, Bhatara S, Yang X, Yoshimura S, Yang W, Karol SE, Inaba H, Mullighan C, Litzow M, Zhu X, Zhang Y, Stock W, Jain N, Jabbour E, Kornblau SM, Konopleva M, Pui CH, Paietta E, Evans W, Yu J, Yang JJ. Single-cell systems pharmacology identifies development-driven drug response and combination therapy in B cell acute lymphoblastic leukemia. Cancer Cell 2024; 42:552-567.e6. [PMID: 38593781 PMCID: PMC11008188 DOI: 10.1016/j.ccell.2024.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 02/19/2024] [Accepted: 03/11/2024] [Indexed: 04/11/2024]
Abstract
Leukemia can arise at various stages of the hematopoietic differentiation hierarchy, but the impact of developmental arrest on drug sensitivity is unclear. Applying network-based analyses to single-cell transcriptomes of human B cells, we define genome-wide signaling circuitry for each B cell differentiation stage. Using this reference, we comprehensively map the developmental states of B cell acute lymphoblastic leukemia (B-ALL), revealing its strong correlation with sensitivity to asparaginase, a commonly used chemotherapeutic agent. Single-cell multi-omics analyses of primary B-ALL blasts reveal marked intra-leukemia heterogeneity in asparaginase response: resistance is linked to pre-pro-B-like cells, with sensitivity associated with the pro-B-like population. By targeting BCL2, a driver within the pre-pro-B-like cell signaling network, we find that venetoclax significantly potentiates asparaginase efficacy in vitro and in vivo. These findings demonstrate a single-cell systems pharmacology framework to predict effective combination therapies based on intra-leukemia heterogeneity in developmental state, with potentially broad applications beyond B-ALL.
Collapse
Affiliation(s)
- Xin Huang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, Anhui 230601, China
| | - Yizhen Li
- Division of Pharmaceutical Sciences, Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Hematology, Children's Hospital of Soochow University, Suzhou, Jiangsu 215003, China
| | - Jingliao Zhang
- Department of Pediatrics Blood Diseases Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Lei Yan
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Huanbin Zhao
- Division of Pharmaceutical Sciences, Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Liang Ding
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Sheetal Bhatara
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Xu Yang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Satoshi Yoshimura
- Division of Pharmaceutical Sciences, Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Wenjian Yang
- Division of Pharmaceutical Sciences, Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Seth E Karol
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Hiroto Inaba
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Charles Mullighan
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Hematological Malignancies Program, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Mark Litzow
- Division of Hematology, Mayo Clinic, Rochester, MN 55905, USA
| | - Xiaofan Zhu
- Department of Pediatrics Blood Diseases Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Yingchi Zhang
- Department of Pediatrics Blood Diseases Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Wendy Stock
- Department of Medicine Section of Hematology-Oncology, University of Chicago, Chicago, IL 60637, USA
| | - Nitin Jain
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Elias Jabbour
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Steven M Kornblau
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Marina Konopleva
- Department of Oncology and Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Ching-Hon Pui
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Hematological Malignancies Program, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Elisabeth Paietta
- Cancer Center, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - William Evans
- Division of Pharmaceutical Sciences, Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jiyang Yu
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| | - Jun J Yang
- Division of Pharmaceutical Sciences, Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Hematological Malignancies Program, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
3
|
Bristol JA, Nelson SE, Ohashi M, Casco A, Hayes M, Ranheim EA, Pawelski AS, Singh DR, Hodson DJ, Johannsen EC, Kenney SC. Latent Epstein-Barr virus infection collaborates with Myc over-expression in normal human B cells to induce Burkitt-like Lymphomas in mice. PLoS Pathog 2024; 20:e1012132. [PMID: 38620028 PMCID: PMC11045125 DOI: 10.1371/journal.ppat.1012132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 04/25/2024] [Accepted: 03/18/2024] [Indexed: 04/17/2024] Open
Abstract
Epstein-Barr virus (EBV) is an important cause of human lymphomas, including Burkitt lymphoma (BL). EBV+ BLs are driven by Myc translocation and have stringent forms of viral latency that do not express either of the two major EBV oncoproteins, EBNA2 (which mimics Notch signaling) and LMP1 (which activates NF-κB signaling). Suppression of Myc-induced apoptosis, often through mutation of the TP53 (p53) gene or inhibition of pro-apoptotic BCL2L11 (BIM) gene expression, is required for development of Myc-driven BLs. EBV+ BLs contain fewer cellular mutations in apoptotic pathways compared to EBV-negative BLs, suggesting that latent EBV infection inhibits Myc-induced apoptosis. Here we use an EBNA2-deleted EBV virus (ΔEBNA2 EBV) to create the first in vivo model for EBV+ BL-like lymphomas derived from primary human B cells. We show that cord blood B cells infected with both ΔEBNA2 EBV and a Myc-expressing vector proliferate indefinitely on a CD40L/IL21 expressing feeder layer in vitro and cause rapid onset EBV+ BL-like tumors in NSG mice. These LMP1/EBNA2-negative Myc-driven lymphomas have wild type p53 and very low BIM, and express numerous germinal center B cell proteins (including TCF3, BACH2, Myb, CD10, CCDN3, and GCSAM) in the absence of BCL6 expression. Myc-induced activation of Myb mediates expression of many of these BL-associated proteins. We demonstrate that Myc blocks LMP1 expression both by inhibiting expression of cellular factors (STAT3 and Src) that activate LMP1 transcription and by increasing expression of proteins (DNMT3B and UHRF1) known to enhance DNA methylation of the LMP1 promoters in human BLs. These results show that latent EBV infection collaborates with Myc over-expression to induce BL-like human B-cell lymphomas in mice. As NF-κB signaling retards the growth of EBV-negative BLs, Myc-mediated repression of LMP1 may be essential for latent EBV infection and Myc translocation to collaboratively induce human BLs.
Collapse
Affiliation(s)
- Jillian A. Bristol
- Department of Oncology, McArdle Laboratory for Cancer Research, School of Medicine and Public Health, University of Wisconsin Madison, Madison, Wisconsin, United States of America
| | - Scott E. Nelson
- Department of Oncology, McArdle Laboratory for Cancer Research, School of Medicine and Public Health, University of Wisconsin Madison, Madison, Wisconsin, United States of America
| | - Makoto Ohashi
- Department of Oncology, McArdle Laboratory for Cancer Research, School of Medicine and Public Health, University of Wisconsin Madison, Madison, Wisconsin, United States of America
| | - Alejandro Casco
- Department of Oncology, McArdle Laboratory for Cancer Research, School of Medicine and Public Health, University of Wisconsin Madison, Madison, Wisconsin, United States of America
| | - Mitchell Hayes
- Department of Oncology, McArdle Laboratory for Cancer Research, School of Medicine and Public Health, University of Wisconsin Madison, Madison, Wisconsin, United States of America
| | - Erik A. Ranheim
- Department of Pathology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Abigail S. Pawelski
- Department of Oncology, McArdle Laboratory for Cancer Research, School of Medicine and Public Health, University of Wisconsin Madison, Madison, Wisconsin, United States of America
| | - Deo R. Singh
- Department of Oncology, McArdle Laboratory for Cancer Research, School of Medicine and Public Health, University of Wisconsin Madison, Madison, Wisconsin, United States of America
| | - Daniel J. Hodson
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Eric C. Johannsen
- Department of Oncology, McArdle Laboratory for Cancer Research, School of Medicine and Public Health, University of Wisconsin Madison, Madison, Wisconsin, United States of America
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Shannon C. Kenney
- Department of Oncology, McArdle Laboratory for Cancer Research, School of Medicine and Public Health, University of Wisconsin Madison, Madison, Wisconsin, United States of America
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| |
Collapse
|
4
|
McCaleb MR, Miranda AM, Ratliff KC, Torres RM, Pelanda R. CD19 Is Internalized Together with IgM in Proportion to B Cell Receptor Stimulation and Is Modulated by Phosphatidylinositol 3-Kinase in Bone Marrow Immature B Cells. Immunohorizons 2023; 7:49-63. [PMID: 36637517 PMCID: PMC10074640 DOI: 10.4049/immunohorizons.2200092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 12/16/2022] [Indexed: 01/14/2023] Open
Abstract
Newly generated immature B cells that bind self-antigen with high avidity arrest in differentiation and undergo central tolerance via receptor editing and clonal deletion. These autoreactive immature B cells also express low surface levels of the coreceptor CD19, a key activator of the PI3K pathway. Signals emanating from both CD19 and PI3K are known to be critical for attenuating receptor editing and selecting immature B cells into the periphery. However, the mechanisms that modulate CD19 expression at this stage of B cell development have not yet been resolved. Using in vivo and in vitro models, we demonstrate that Cd19 de novo gene transcription and translation do not significantly contribute to the differences in CD19 surface expression in mouse autoreactive and nonautoreactive immature B cells. Instead, CD19 downregulation is induced by BCR stimulation in proportion to BCR engagement, and the remaining surface IgM and CD19 molecules promote intracellular PI3K-AKT activity in proportion to their level of expression. The internalized CD19 is degraded with IgM by the lysosome, but inhibiting lysosome-mediated protein degradation only slightly improves surface CD19. In fact, CD19 is restored only upon Ag removal. Our data also reveal that the PI3K-AKT pathway positively modulates CD19 surface expression in immature B cells via a mechanism that is independent of inhibition of FOXO1 and its role on Cd19 gene transcription while is dependent on mTORC1.
Collapse
Affiliation(s)
- Megan R. McCaleb
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO; and
| | - Anjelica M. Miranda
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO; and
| | - Kaysie C. Ratliff
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO; and
| | - Raul M. Torres
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO; and
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, CO
| | - Roberta Pelanda
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO; and
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, CO
| |
Collapse
|
5
|
Single-cell analysis identifies dynamic gene expression networks that govern B cell development and transformation. Nat Commun 2021; 12:6843. [PMID: 34824268 PMCID: PMC8617197 DOI: 10.1038/s41467-021-27232-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 11/02/2021] [Indexed: 12/22/2022] Open
Abstract
Integration of external signals and B-lymphoid transcription factor activities organise B cell lineage commitment through alternating cycles of proliferation and differentiation, producing a diverse repertoire of mature B cells. We use single-cell transcriptomics/proteomics to identify differentially expressed gene networks across B cell development and correlate these networks with subtypes of B cell leukemia. Here we show unique transcriptional signatures that refine the pre-B cell expansion stages into pre-BCR-dependent and pre-BCR-independent proliferative phases. These changes correlate with reciprocal changes in expression of the transcription factor EBF1 and the RNA binding protein YBX3, that are defining features of the pre-BCR-dependent stage. Using pseudotime analysis, we further characterize the expression kinetics of different biological modalities across B cell development, including transcription factors, cytokines, chemokines, and their associated receptors. Our findings demonstrate the underlying heterogeneity of developing B cells and characterise developmental nodes linked to B cell transformation.
Collapse
|
6
|
Dinh TN, Onea AS, Jazirehi AR. Combination of celecoxib (Celebrex ®) and CD19 CAR-redirected CTL immunotherapy for the treatment of B-cell non-Hodgkin's lymphomas. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL IMMUNOLOGY 2017; 6:27-42. [PMID: 28804691 PMCID: PMC5545683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 01/31/2017] [Indexed: 06/07/2023]
Abstract
The nonsteroidal anti-inflammatory drug (NSAID) Celecoxib (Celebrex®) received Food and Drug Administration (FDA) approval in 1998 for treatment of osteoarthritis and rheumatoid arthritis, and in recent years, its use has been extended to various types of malignancies, such as breast, colon, and urinary cancers. To maintain the survival of malignant B cells, non-Hodgkin's Lymphoma (NHL) is highly dependent on inflammatory microenvironment, and is inhibited by celecoxib. Celecoxib hinders tumor growth interacting with various apoptotic genes, such as cyclooxygenase-2 (Cox-2), B-cell lymphoma 2 (Bcl-2) family, phosphor-inositide-3 kinase/serine-threonine-specific protein kinase (PI3K/Akt), and inhibitors of apoptosis proteins (IAP) family. CD19-redirected chimeric antigen-receptor (CD19 CAR) T cell therapy has shown promise in the treatment of B cell malignancies. Considering its regulatory effect on apoptotic gene products in various tumor types, Celecoxib is a promising drug to be used in combination with CD19 CAR T cell therapy to optimize immunotherapy of NHL.
Collapse
Affiliation(s)
- Tam Nm Dinh
- Department of Surgery, Division of Surgical Oncology, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles (UCLA)CA 90095, Los Angeles, USA
| | - Alexandra S Onea
- Department of Surgery, Division of Surgical Oncology, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles (UCLA)CA 90095, Los Angeles, USA
| | - Ali R Jazirehi
- Department of Surgery, Division of Surgical Oncology, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles (UCLA)CA 90095, Los Angeles, USA
| |
Collapse
|
7
|
Fisher MR, Rivera-Reyes A, Bloch NB, Schatz DG, Bassing CH. Immature Lymphocytes Inhibit Rag1 and Rag2 Transcription and V(D)J Recombination in Response to DNA Double-Strand Breaks. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2017; 198:2943-2956. [PMID: 28213501 PMCID: PMC5360515 DOI: 10.4049/jimmunol.1601639] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 01/16/2017] [Indexed: 12/26/2022]
Abstract
Mammalian cells have evolved a common DNA damage response (DDR) that sustains cellular function, maintains genomic integrity, and suppresses malignant transformation. In pre-B cells, DNA double-strand breaks (DSBs) induced at Igκ loci by the Rag1/Rag2 (RAG) endonuclease engage this DDR to modulate transcription of genes that regulate lymphocyte-specific processes. We previously reported that RAG DSBs induced at one Igκ allele signal through the ataxia telangiectasia mutated (ATM) kinase to feedback-inhibit RAG expression and RAG cleavage of the other Igκ allele. In this article, we show that DSBs induced by ionizing radiation, etoposide, or bleomycin suppress Rag1 and Rag2 mRNA levels in primary pre-B cells, pro-B cells, and pro-T cells, indicating that inhibition of Rag1 and Rag2 expression is a prevalent DSB response among immature lymphocytes. DSBs induced in pre-B cells signal rapid transcriptional repression of Rag1 and Rag2, causing downregulation of both Rag1 and Rag2 mRNA, but only Rag1 protein. This transcriptional inhibition requires the ATM kinase and the NF-κB essential modulator protein, implicating a role for ATM-mediated activation of canonical NF-κB transcription factors. Finally, we demonstrate that DSBs induced in pre-B cells by etoposide or bleomycin inhibit recombination of Igκ loci and a chromosomally integrated substrate. Our data indicate that immature lymphocytes exploit a common DDR signaling pathway to limit DSBs at multiple genomic locations within developmental stages wherein monoallelic Ag receptor locus recombination is enforced. We discuss the implications of our findings for mechanisms that orchestrate the differentiation of monospecific lymphocytes while suppressing oncogenic Ag receptor locus translocations.
Collapse
Affiliation(s)
- Megan R Fisher
- Division of Cancer Pathobiology, Department of Pathology and Laboratory Medicine, Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA 19104
- Immunology Graduate Group, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104
- Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104
| | - Adrian Rivera-Reyes
- Division of Cancer Pathobiology, Department of Pathology and Laboratory Medicine, Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA 19104
- Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104
- Cancer Biology Program of the Cell and Molecular Biology Graduate Group, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104; and
| | - Noah B Bloch
- Division of Cancer Pathobiology, Department of Pathology and Laboratory Medicine, Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA 19104
| | - David G Schatz
- Department of Immunobiology, Yale University School of Medicine, Howard Hughes Medical Institute, New Haven, CT 06520
| | - Craig H Bassing
- Division of Cancer Pathobiology, Department of Pathology and Laboratory Medicine, Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA 19104;
- Immunology Graduate Group, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104
- Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104
- Cancer Biology Program of the Cell and Molecular Biology Graduate Group, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104; and
| |
Collapse
|
8
|
Kraus H, Kaiser S, Aumann K, Bönelt P, Salzer U, Vestweber D, Erlacher M, Kunze M, Burger M, Pieper K, Sic H, Rolink A, Eibel H, Rizzi M. A feeder-free differentiation system identifies autonomously proliferating B cell precursors in human bone marrow. THE JOURNAL OF IMMUNOLOGY 2013; 192:1044-54. [PMID: 24379121 DOI: 10.4049/jimmunol.1301815] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The peripheral B cell compartment is maintained by homeostatic proliferation and through replenishment by bone marrow precursors. Because hematopoietic stem cells cycle at a slow rate, replenishment must involve replication of precursor B cells. To study proliferation of early human B cell progenitors, we established a feeder cell-free in vitro system allowing the development of B cells from CD34(+) hematopoietic stem cells up to the stage of immature IgM(+) B cells. We found that pro-B and pre-B cells generated in vitro can proliferate autonomously and persist up to 7 wk in culture in the absence of signals induced by exogenously added cytokines. Nevertheless, addition of IL-7 enhanced pre-B cell expansion and inhibited maturation into IgM(+) B cells. The B cell precursor subsets replicating in vitro were highly similar to the bone marrow B cell precursors cycling in vivo. The autonomous proliferation of B cell precursor subsets in vitro and their long-term persistence implies that proliferation during pro-B and pre-B cell stages plays an important role in the homeostasis of the peripheral B cell compartment. Our in vitro culture can be used to study defects in B cell development or in reconstitution of the B cell pool after depletion and chemotherapy.
Collapse
Affiliation(s)
- Helene Kraus
- Center for Chronic Immunodeficiency, University Medical Center Freiburg, 79108 Freiburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Steinel NC, Lee BS, Tubbs AT, Bednarski JJ, Schulte E, Yang-Iott KS, Schatz DG, Sleckman BP, Bassing CH. The ataxia telangiectasia mutated kinase controls Igκ allelic exclusion by inhibiting secondary Vκ-to-Jκ rearrangements. ACTA ACUST UNITED AC 2013; 210:233-9. [PMID: 23382544 PMCID: PMC3570110 DOI: 10.1084/jem.20121605] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
DNA double-strand breaks induced during Igκ recombination signal through ATM to suppress the initiation of additional Vκ-to-Jκ rearrangements. Allelic exclusion is enforced through the ability of antigen receptor chains expressed from one allele to signal feedback inhibition of V-to-(D)J recombination on the other allele. To achieve allelic exclusion by such means, only one allele can initiate V-to-(D)J recombination within the time required to signal feedback inhibition. DNA double-strand breaks (DSBs) induced by the RAG endonuclease during V(D)J recombination activate the Ataxia Telangiectasia mutated (ATM) and DNA-dependent protein kinase (DNA-PK) kinases. We demonstrate that ATM enforces Igκ allelic exclusion, and that RAG DSBs induced during Igκ recombination in primary pre–B cells signal through ATM, but not DNA-PK, to suppress initiation of additional Igκ rearrangements. ATM promotes high-density histone H2AX phosphorylation to create binding sites for MDC1, which functions with H2AX to amplify a subset of ATM-dependent signals. However, neither H2AX nor MDC1 is required for ATM to enforce Igκ allelic exclusion and suppress Igκ rearrangements. Upon activation in response to RAG Igκ cleavage, ATM signals down-regulation of Gadd45α with concomitant repression of the Gadd45α targets Rag1 and Rag2. Our data indicate that ATM kinases activated by RAG DSBs during Igκ recombination transduce transient H2AX/MDC1-independent signals that suppress initiation of further Igκ rearrangements to control Igκ allelic exclusion.
Collapse
Affiliation(s)
- Natalie C Steinel
- Immunology Graduate Group, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Abstract
A large antibody repertoire is generated in developing B cells in the bone marrow. Before these B cells achieve immunocompetence, those expressing autospecificities must be purged. To that end, B cells within the bone marrow and just following egress from the bone marrow are subject to tolerance induction. Once B cells achieve immunocompetence, the antibody repertoire can be further diversified by somatic hypermutation of immunoglobulin genes in B cells that have been activated by antigen and cognate T cell help and have undergone a germinal center (GC) response. This process also leads to the generation of autoreactive B cells which must be again purged to protect the host. Thus, B cells within the GC and just following egress from the GC are also subject to tolerance induction. Available data suggest that B cell intrinsic processes triggered by signaling through the B cell receptor activate tolerance mechanisms at both time points. Recent data suggest that GC and post-GC B cells are also subject to B cell extrinsic tolerance mechanisms mediated through soluble and membrane-bound factors derived from various T cell subsets.
Collapse
|
11
|
Bednarski JJ, Nickless A, Bhattacharya D, Amin RH, Schlissel MS, Sleckman BP. RAG-induced DNA double-strand breaks signal through Pim2 to promote pre-B cell survival and limit proliferation. ACTA ACUST UNITED AC 2011; 209:11-7. [PMID: 22201128 PMCID: PMC3260864 DOI: 10.1084/jem.20112078] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Interleukin 7 (IL-7) promotes pre-B cell survival and proliferation by activating the Pim1 and Akt kinases. These signals must be attenuated to induce G1 cell cycle arrest and expression of the RAG endonuclease, which are both required for IgL chain gene rearrangement. As lost IL-7 signals would limit pre-B cell survival, how cells survive during IgL chain gene rearrangement remains unclear. We show that RAG-induced DNA double-strand breaks (DSBs) generated during IgL chain gene assembly paradoxically promote pre-B cell survival. This occurs through the ATM-dependent induction of Pim2 kinase expression. Similar to Pim1, Pim2 phosphorylates BAD, which antagonizes the pro-apoptotic function of BAX. However, unlike IL-7 induction of Pim1, RAG DSB-mediated induction of Pim2 does not drive proliferation. Rather, Pim2 has antiproliferative functions that prevent the transit of pre-B cells harboring RAG DSBs from G1 into S phase, where these DNA breaks could be aberrantly repaired. Thus, signals from IL-7 and RAG DSBs activate distinct Pim kinase family members that have context-dependent activities in regulating pre-B cell proliferation and survival.
Collapse
Affiliation(s)
- Jeffrey J Bednarski
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | | | |
Collapse
|
12
|
Abstract
Expression of a BCR is critical for B-cell development and survival. We have identified 4 patients with agammaglobulinemia and markedly reduced but detectable B cells in the peripheral circulation. These B cells have an unusual phenotype characterized by increased expression of CD19 but no BCR. The cells are positive for CD20, CD22, and CD38, but not for Annexin 5 or activation markers, including CD69, CD83, or CD86. EBV lines derived from these B cells lack functionally rearranged immunoglobulin heavy-chain transcripts, as shown by PCR-rapid amplification of cDNA ends (PCR-RACE). Analysis of BM from 2 of the patients showed a severe reduction in the number of pro-B cells as well as pre-B cells. Functionally rearranged heavy-chain transcripts were identified, indicating that machinery to rearrange immunoglobulin genes was intact. Flow cytometry of B-lineage cells suggested accelerated acquisition of maturation markers in early B-cell precursors and increased phosphorylation of signal transduction molecules. Further, expression of TdT, a molecule that is normally down-regulated by a functional pre-BCR complex, was decreased. We hypothesize that the accelerated maturation, increased expression of CD19, and lack of a BCR were due to the constitutive activation of the BCR signal transduction pathway in these patients.
Collapse
|
13
|
Sekiguchi DR, Sutter JA, Rickels MR, Naji A, Liu C, Propert KJ, Rogers WT, Prak ETL. Analysis of B cell subsets following pancreatic islet cell transplantation in a patient with type 1 diabetes by cytometric fingerprinting. J Immunol Methods 2010; 363:233-44. [PMID: 20933515 DOI: 10.1016/j.jim.2010.09.038] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2010] [Revised: 08/13/2010] [Accepted: 09/10/2010] [Indexed: 01/28/2023]
Abstract
Manual gating of bivariate plots remains the most frequently used data analysis method in flow cytometry. However, gating is operator-dependent and cumbersome, particularly with the increasing complexity of modern multicolor immunophenotyping data. A method that can remove operator bias, enable systematic and thorough analysis of complex high-dimensional data, correlate temporal changes in different subsets and lead to biomarker discovery is needed. Here we apply such a method, called cytometric fingerprinting (CF), to data obtained on peripheral blood B cells from an adult patient with type-1 diabetes who underwent pancreatic islet transplantation. We establish that CF can be used to analyze longitudinal trends in immunophenotypic data, and show that results from CF are comparable to those obtained with traditional gating methods. Both methods reveal the appearance of transitional B cells and subsequent accumulation of more mature B cells following immunosuppression and transplantation. This pattern is consistent with a temporally ordered process of B cell auto-reconstitution. We also show the comparative efficiency of fingerprinting in recognizing relative changes in B cell subsets with respect to time, its ability to couple the data with statistical methods (agglomerative clustering) and its potential to define novel subsets.
Collapse
Affiliation(s)
- Debora R Sekiguchi
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Stromal cell independent B cell development in vitro: generation and recovery of autoreactive clones. J Immunol Methods 2010; 354:53-67. [PMID: 20109461 DOI: 10.1016/j.jim.2010.01.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2009] [Revised: 01/15/2010] [Accepted: 01/19/2010] [Indexed: 11/24/2022]
Abstract
We describe and characterize a stromal cell independent culture system that efficiently supports pro-B cell to IgM+ B cell development with near normal levels of IgH and Igkappa diversity. Pro-B cells present in non-adherent bone marrow cells proliferate in the presence of IL-7 and subsequent to the removal of IL-7 and addition of BAFF, differentiate normally into IgM+ B cells. B cell development in vitro closely follows the patterns of development in vivo with culture-derived (CD) B cells demonstrating characteristic patterns of surface antigen expression and gene activation. IgM+ CD B cells respond to TLR stimulation by proliferation and differentiation into antibody-secreting cells. Self-reactive IgM+ B cell development is blocked in 3H9 IgH knockin mice; however, cultures of 3H9 IgH knockin pro-B cells yields high frequencies of "forbidden", autoreactive IgM+ B cells. Furthermore, serum IgG autoantibody exceeded that present in autoimmune, C4(-/-) animals following the reconstitution of RAG1(-/-) mice with IgM+ CD cells derived from BL/6 mice.
Collapse
|
15
|
Hillion S, Rochas C, Youinou P, Jamin C. Signaling pathways regulating RAG expression in B lymphocytes. Autoimmun Rev 2009; 8:599-604. [PMID: 19393209 DOI: 10.1016/j.autrev.2009.02.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2009] [Accepted: 02/04/2009] [Indexed: 10/21/2022]
Abstract
Development of B-cell lymphopoiesis is dependent on the presence of recombination activating genes RAG1 and RAG2 enzymes. They control the rearrangements of immunoglobulin variable, diversity and joining region segments, and allow progression of the cellular maturation. RAG1 and RAG2 are successively up- and down-regulated at each B-cell stage to progressively generate a B-cell receptor for which unforeseeable antigenic specificity results from a stochastic process. Therefore, in autoreactive immature B cells, new round of RAG re-expression can be observed to eliminate self-reactivity. In some circumstances, RAG up-regulation can also be found in peripheral mature B lymphocytes, specifically in autoimmune diseases. It is therefore of utmost importance to unravel signaling pathways that trigger RAG induction in normal and pathological conditions. Therapeutic modulation of cytokines or intracellular contacts involved in RAG expression might restrict the development of inappropriate autoimmune repertoire.
Collapse
|
16
|
Wang YH, Diamond B. B cell receptor revision diminishes the autoreactive B cell response after antigen activation in mice. J Clin Invest 2008; 118:2896-907. [PMID: 18636122 DOI: 10.1172/jci35618] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2008] [Accepted: 06/11/2008] [Indexed: 12/16/2022] Open
Abstract
Autoreactive B cells are regulated in the BM during development through mechanisms, including editing of the B cell receptor (BCR), clonal deletion, and anergy. Peripheral B cell tolerance is also important for protection from autoimmune damage, although the mechanisms are less well defined. Here we demonstrated, using a mouse model of SLE-like serology, that during an autoimmune response, RAG was reinduced in antigen-activated early memory or preplasma B cells. Expression of RAG was specific to antigen-reactive B cells, required the function of the IL-7 receptor (IL-7R), and contributed to maintenance of humoral tolerance. We also showed that soluble antigen could diminish a non-autoreactive antibody response through induction of BCR revision. These data suggest that tolerance induction operates in B cells at a postactivation checkpoint and that BCR revision helps regulate autoreactivity generated during an ongoing immune response.
Collapse
Affiliation(s)
- Ying-Hua Wang
- Department of Microbiology, Columbia University Medical Center, New York, New York, USA
| | | |
Collapse
|
17
|
Milne CD, Corfe SA, Paige CJ. Heparan sulfate and heparin enhance ERK phosphorylation and mediate preBCR-dependent events during B lymphopoiesis. THE JOURNAL OF IMMUNOLOGY 2008; 180:2839-47. [PMID: 18292505 DOI: 10.4049/jimmunol.180.5.2839] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
As B lineage cells develop, they interact with cells, proteins, and extracellular matrix components of the surrounding microenvironment. In vitro, one critical checkpoint for developing cells occurs as they lose responsiveness to IL-7. These cells require contact with either stromal cells or other B lineage cells to mature. Our results demonstrate that heparan sulfate and heparin are able to promote this transition when added exogenously to the culture system or when heparan sulfate-bearing cell lines are cocultured with primary B cell progenitors. Addition of heparan sulfate or heparin to LPS-stimulated cultures of primary B cell progenitors resulted in more IgM secreted compared with untreated cultures. Heparan sulfate has been reported to be a ligand for the pre-B cell receptor (preBCR). Extending this observation, we found that treatment of preBCR+ cells with heparan sulfate before anti-micro stimulation leads to increased phosphorylation of ERK1/2. Consequently, preBCR+ cells proliferate more in the presence of IL-7 and heparan sulfate, whereas preBCR- cells are unaffected, suggesting that in these experiments, heparan sulfate is not directly affecting IL-7 activity. Heparin treatment of cultures induces many of the same biological effects as treatment with heparan sulfate, including elevated pERK levels in preBCR+ cells. However, heparin reduces the proliferation of cells expressing only the preBCR (opposed to both the preBCR and BCR) possibly due to internalization of the preBCR. Heparan sulfates are present on stromal cells and B lineage cells present in hemopoietic tissues and may provide stimulation to preB cells testing the signaling capacity of the preBCR.
Collapse
Affiliation(s)
- Craig D Milne
- Ontario Cancer Institute, University Health Network, Department of Immunology, University of Toronto, and Princess Margaret Hospital, Toronto, Ontario, Canada.
| | | | | |
Collapse
|
18
|
Amin RH, Schlissel MS. Foxo1 directly regulates the transcription of recombination-activating genes during B cell development. Nat Immunol 2008; 9:613-22. [PMID: 18469817 DOI: 10.1038/ni.1612] [Citation(s) in RCA: 219] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2007] [Accepted: 03/31/2008] [Indexed: 11/09/2022]
Abstract
Regulated expression of the recombinase RAG-1 and RAG-2 proteins is necessary for generating the vast repertoire of antigen receptors essential for adaptive immunity. Here, a retroviral cDNA library screen showed that the stress-regulated protein GADD45a activated transcription of the genes encoding RAG-1 and RAG-2 in transformed pro-B cells by a pathway requiring the transcription factor Foxo1. Foxo1 directly activated transcription of the Rag1-Rag2 locus throughout early B cell development, and a decrease in Foxo1 protein diminished the induction of Rag1 and Rag2 transcription in a model of receptor editing. We also found that transcription of Rag1 and Rag2 was repressed at the pro-B cell and immature B cell stages by the kinase Akt through its 'antagonism' of Foxo1 function. Thus, Foxo1 is a key regulator of Rag1 and Rag2 transcription in primary B cells.
Collapse
Affiliation(s)
- Rupesh H Amin
- Department of Molecular & Cell Biology, University of California at Berkeley, Berkeley, California 94720, USA
| | | |
Collapse
|
19
|
Hillion S, Dueymes M, Youinou P, Jamin C. IL-6 contributes to the expression of RAGs in human mature B cells. THE JOURNAL OF IMMUNOLOGY 2007; 179:6790-8. [PMID: 17982069 DOI: 10.4049/jimmunol.179.10.6790] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Mature B cells acquire the capacity to revise rearranged Ig V region genes in secondary lymphoid organs. In previous studies, we demonstrated that cross-linking the BCR and the CD40 induces the expression of the RAG1 and RAG2 enzymes and, thereby, secondary rearrangements. We examine herein the mechanism that underpins RAG1 and RAG2 expression in peripheral and tonsil B cells. Coordinated engagement of the BCR and CD40 promoted the synthesis of IL-6 and, thereby, up-regulation of its receptor on activated B lymphocytes. Furthermore, we provide evidence that IL-6 initiates the expression of RAGs in circulating B cells, and extends those in tonsil B cells. Thus, neutralization of IL-6 or blocking of its receptor inhibits RAG expression. Moreover, we demonstrate that IL-6 impedes BCR-mediated termination of RAG gene expression in both population of B cells. The recovered inhibition of RAG gene transcription by IL-6 receptor blockade supports the notion that once recombination is launched, its termination is also regulated by IL-6. Taken together, these studies provide new insight into the dual role of IL-6 in inducing and terminating expression of the recombinase machinery for secondary rearrangements in mature human B cells.
Collapse
Affiliation(s)
- Sophie Hillion
- Laboratory of Immunology, Brest University Medical School Hospital, Brest, France
| | | | | | | |
Collapse
|
20
|
Taguchi T, Takenouchi H, Shiozawa Y, Matsui J, Kitamura N, Miyagawa Y, Katagiri YU, Takahashi T, Okita H, Fujimoto J, Kiyokawa N. Interleukin-7 contributes to human pro–B-cell development in a mouse stromal cell–dependent culture system. Exp Hematol 2007; 35:1398-407. [PMID: 17656007 DOI: 10.1016/j.exphem.2007.05.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2007] [Revised: 05/10/2007] [Accepted: 05/31/2007] [Indexed: 10/23/2022]
Abstract
OBJECTIVE The role of interleukin (IL)-7 in human B lymphopoiesis is still controversial. We used an in vitro culture system to verify involvement of IL-7 in development of human pro-B cells from hematopoietic stem cells. MATERIALS AND METHODS Human CD34(+) bone marrow cells were cultured for 4 weeks on MS-5 mouse stromal cells to induce pro-B cells. Expression of IL-7 receptor alpha or other B-cell differentiation marker genes on cultured human CD34(+)bone marrow cells was investigated by reverse transcription polymerase chain reaction (RT-PCR). Colony assay of human CD34(+) bone marrow cells was also performed to determine the effect of IL-7 on colony-forming ability. Neutralizing antibody or reagent that eliminates the effect of IL-7 was added to the culture system, and the number of pro-B cells induced was estimated by flow cytometry. RESULTS RT-PCR analysis revealed mRNA expression of IL-7 receptor alpha as well as B-cell differentiation marker genes in not only CD19(+) pro-B cells but also CD19(-) CD33(-) cells induced from CD34(+) bone marrow cells after cultivation for 4 weeks on MS-5 cells. Addition of anti-mouse IL-7 antibody, anti-human IL-7 receptor alpha antibody, or JAK3 kinase inhibitor reduced the number of pro-B cells induced, demonstrating that elimination of IL-7 reduces pro-B-cell development. Addition of anti-mouse IL-7 antibody emphasized the colony-forming ability of burst-forming unit erythroid cells. CONCLUSIONS IL-7 produced by MS-5 cells is required for human pro-B-cell development from CD34(+)bone marrow cells in our culture system, and IL-7 appears to play a certain role in early human B lymphopoiesis.
Collapse
Affiliation(s)
- Tomoko Taguchi
- Department of Developmental Biology, National Research Institute for Child Health and Development, Setagaya-ku, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Hillion S, Youinou P, Jamin C. Peripheral expression of RAG in human B lymphocytes in normal and pathological conditions is dependent on interleukin-6. Autoimmun Rev 2007; 6:415-20. [PMID: 17537388 DOI: 10.1016/j.autrev.2007.01.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2007] [Accepted: 01/09/2007] [Indexed: 12/22/2022]
Abstract
Establishment of the B cell repertoire is regulated by recombination activating genes RAG1 and RAG2 proteins in the bone marrow. Tolerance of autoreactivity is mainly prevented by receptor editing, i.e. synthesis of a new B cell receptor following re-expression of RAG1 and RAG2. Numerous signals can lead to RAG up-regulation, all in association with soluble cytokines. In the periphery, autoreactive B cells or low-affinity B cell receptor synthesis may appear following antigenic immune response. Receptor revision, i.e. new immunoglobulin gene rearrangement can participate to the control of these lymphocytes following new RAG1 and RAG2 re-induction. Though signals leading to this peripheral RAG up-regulation are poorly described, IL-6 seems to have a preponderant role. Therefore, the elevated levels of IL-6 secreted by activated B cells in systemic lupus erythematosus might contribute to the maintenance of abnormal RAG expression, and in turn may participate to the emergence of autoreactive B cells in the periphery.
Collapse
Affiliation(s)
- Sophie Hillion
- Laboratory of Immunology Brest University Medical School, BP 824, F29609 Brest, France
| | | | | |
Collapse
|
22
|
Twohig J, Kulik L, Haluszczak C, Reuter J, Rossbach A, Bull M, Holers VM, Marchbank KJ. Defective B cell ontogeny and immune response in human complement receptor 2 (CR2, CD21) transgenic mice is partially recovered in the absence of C3. Mol Immunol 2007; 44:3434-44. [PMID: 17379312 PMCID: PMC1948027 DOI: 10.1016/j.molimm.2007.02.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2007] [Revised: 02/12/2007] [Accepted: 02/14/2007] [Indexed: 01/13/2023]
Abstract
Mice prematurely expressing human CR2 (hCR2) in the B cell lineage have a defective B cell ontogeny and immune response. Our recent analysis of this phenotype suggested that signaling through hCR2 and presumably mouse CD19 on the B cell surface, during bone marrow development, could result in the observed changes in B cell function in these mice. To test this hypothesis, we back crossed hCR2(high) transgenic mice onto the CD19(-/-) background. CD19(-/-)hCR2(high) mice were found to possess even fewer mature B cells than their CD19(+/+)hCR2(high) littermates, demonstrating that loss of CD19 exacerbated the effects elicited through hCR2. This data suggests that CD19 provides a survival signal during B cell development in this model. Next, we examined if the removal of the main ligand for CR2, namely C3d, through back-crossing onto the C3(-/-) background could restore normal B cell development. However, we found only minor recovery in peripheral B cell numbers and no obvious change in function. This was despite a three-fold increase in the level of hCR2 expression on B cells isolated from the spleen or bone marrow of C3(-/-)hCR2(high) mice when compared with C3 sufficient littermates. These data demonstrate that hCR2 is integrated in mouse B cell signaling and that the downstream effects of hCR2 expression during early B cell development are partially but not completely due to interaction with C3 fragments and signaling through CD19 in the bone marrow environment.
Collapse
Affiliation(s)
- Jason Twohig
- Department of Medical Biochemistry & Immunology, Cardiff University, Heath Park, Cardiff CF14 4XX, UK
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Clark MR, Cooper AB, Wang LD, Aifantis I. The pre-B cell receptor in B cell development: recent advances, persistent questions and conserved mechanisms. Curr Top Microbiol Immunol 2005; 290:87-103. [PMID: 16480040 DOI: 10.1007/3-540-26363-2_5] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
B cell development is a process tightly regulated by the orchestrated signaling of cytokine receptors, the pre-B cell receptor (BCR) and the B cell receptor (BCR). It commences with common lymphoid progenitors (CLP) up-regulating the expression of B cell-related genes and committing to the B cell lineage. Cytokine signaling (IL-7, stem cell factor, FLT3-L) is essential at this stage of development as it suppresses cell death, sustains proliferation and facilitates heavy chain rearrangements. As a result of heavy chain recombination, the pre-BCR is expressed, which then becomes the primary determiner of survival, cell cycle entry and allelic exclusion. In this review, we discuss the mechanisms of B cell lineage commitment and describe the signaling pathways that are initiated by the pre-BCR. Finally, we compare pre-BCR and pre-TCR structure, signal transduction and function, drawing parallels between early pre-B and pre-T cell development.
Collapse
Affiliation(s)
- M R Clark
- Section of Rheumatology, University of Chicago, IL 60637, USA.
| | | | | | | |
Collapse
|
24
|
Dong M, Fu Y, Yu C, Su J, Huang S, Wu X, Wei J, Yuan S, Shen Y, Xu A. Identification and characterisation of a homolog of an activation gene for the recombination activating gene 1 (RAG 1) in amphioxus. FISH & SHELLFISH IMMUNOLOGY 2005; 19:165-174. [PMID: 15752655 DOI: 10.1016/j.fsi.2004.11.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2004] [Revised: 10/27/2004] [Accepted: 11/15/2004] [Indexed: 05/24/2023]
Abstract
Expression of recombination activating genes (RAG) involved in the V (D) J recombination is regulated by the RAG1 gene activator (RGA) in mammals. The sequence of a cDNA clone from an amphioxus cDNA library was found to be homologous to that of RGA from mouse stromal cells with 45% identity. The full-length cDNA sequence comprises 1119 bp and encodes a putative protein of 210 amino acid residues. Characterisation of the amino acid sequence revealed that two MtN3 domains and seven transmembrane spans are present in this protein, indicating a potential role as a plasma membrane protein. This gene is expressed in many tissues and at differential developmental stages. A high expression level of RGA is detected in gonad tissues, and gastrula embryo and adult stages. The presence of the RGA gene in amphioxus suggests that the signal pathway required for the expression of RAG could exist in this primitive protochordate. It also implies that in the related molecules, primitive adaptive immunity may have existed in cephalochordate although the complete machinery of VDJ rearrangement may not be formed.
Collapse
Affiliation(s)
- Meiling Dong
- Department of Biochemistry, State Open Laboratory for Marine Function Genomics, Guangzhou Center for Bioinformatics, College of Life Sciences, Sun Yat-sen (Zhongshan) University, Guangzhou 510275, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Oliver PM, Wang M, Zhu Y, White J, Kappler J, Marrack P. Loss of Bim allows precursor B cell survival but not precursor B cell differentiation in the absence of interleukin 7. ACTA ACUST UNITED AC 2005; 200:1179-87. [PMID: 15520248 PMCID: PMC2211863 DOI: 10.1084/jem.20041129] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Interleukin (IL)-7 is a stromal cell-derived cytokine required for the survival, proliferation, and differentiation of B cell precursors. Members of the Bcl-2 family of proteins are known to have profound effects on lymphocyte survival, but not lymphocyte differentiation. To distinguish the relative dependence on IL-7 of B cell precursor survival versus B cell differentiation, the combined effects of lack of IL-7 and lack of the proapoptotic Bcl-2 relative, Bim, were studied. Bim is expressed to varying degrees in all B cell precursors and B cells. Lack of Bim compensated for lack of IL-7 in the survival of pro-, pre-, and immature B cells; however, lack of Bim did not substitute for the requirement for IL-7 in B cell precursor differentiation or B cell precursor proliferation. Precursor B cell survival is more dependent on sufficient levels of IL-7 than precursor B cell differentiation because the number of B cells and their precursors were reduced by half in mice heterozygous for IL-7 expression, but were restored to normal numbers in mice also lacking Bim. Hence, Bim and IL-7 work together to control the survival of B cell precursors and the number of B cells that exist in animals.
Collapse
Affiliation(s)
- Paula M Oliver
- Howard Hughes Medical Institute, Dept. of Immunology, 1400 Jackson St., Denver, CO 80206, USA
| | | | | | | | | | | |
Collapse
|
26
|
Lanvin O, Gouilleux F, Mullié C, Mazière C, Fuentes V, Bissac E, Dantin F, Mazière JC, Régnier A, Lassoued K, Gouilleux-Gruart V. Interleukin-7 induces apoptosis of 697 pre-B cells expressing dominant-negative forms of STAT5: evidence for caspase-dependent and -independent mechanisms. Oncogene 2004; 23:3040-7. [PMID: 15048088 DOI: 10.1038/sj.onc.1207450] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The transcription factors STAT5A and STAT5B (STAT: signal transducer and activator of transcription) play a major role in the signaling events elicited by a number of growth factor and cytokine receptors. In this work, we aimed to investigate the role of STAT5 in human precursor B cell survival by introducing dominant-negative (DN) forms of STAT5A or STAT5B in the 697 pre-B cell line. All clones expressing DN forms of either transcription factor exhibited a higher spontaneous apoptotic rate that was massively enhanced upon interleukin-7 (IL-7) stimulation. This was associated with caspase 8 cleavage, mitochondrial transmembrane potential disruption and caspase 3 activation. However, the DN forms of STAT5 did not alter the expression of Bcl-2, Bax, Bcl-x, Bim, A1 and Mcl1 proteins in IL-7-stimulated cells. The pancaspase inhibitor Z-Val-Ala-Asp-fluoromylmethyl ketone partially suppressed IL-7-mediated mitochondrial transmembrane potential disruption and cell death, suggesting that IL-7 induced the death of DN STAT5 expressing 697 cells through caspase-dependent and -independent mechanisms that both require mitochondrial activation.
Collapse
Affiliation(s)
- Olivia Lanvin
- Laboratoire d'Immunologie, INSERM, EMI 0351, 3 rue des Louvels, 80036 Amiens, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Galler GR, Mundt C, Parker M, Pelanda R, Mårtensson IL, Winkler TH. Surface mu heavy chain signals down-regulation of the V(D)J-recombinase machinery in the absence of surrogate light chain components. ACTA ACUST UNITED AC 2004; 199:1523-32. [PMID: 15173209 PMCID: PMC2211789 DOI: 10.1084/jem.20031523] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Early B cell development is characterized by stepwise, ordered rearrangement of the immunoglobulin (Ig) heavy (HC) and light (LC) chain genes. Only one of the two alleles of these genes is used to produce a receptor, a phenomenon referred to as allelic exclusion. It has been suggested that pre–B cell receptor (pre-BCR) signals are responsible for down-regulation of the VDJH-recombinase machinery (Rag1, Rag2, and terminal deoxynucleotidyl transferase [TdT]), thereby preventing further rearrangement on the second HC allele. Using a mouse model, we show that expression of an inducible μHC transgene in Rag2−/− pro–B cells induces down-regulation of the following: (a) TdT protein, (b) a transgenic green fluorescent protein reporter reflecting endogenous Rag2 expression, and (c) Rag1 primary transcripts. Similar effects were also observed in the absence of surrogate LC (SLC) components, but not in the absence of the signaling subunit Ig-α. Furthermore, in wild-type mice and in mice lacking either λ5, VpreB1/2, or the entire SLC, the TdT protein is down-regulated in μHC+LC− pre–B cells. Surprisingly, μHC without LC is expressed on the surface of pro–/pre–B cells from λ5−/−, VpreB1−/−VpreB2−/−, and SLC−/− mice. Thus, SLC or LC is not required for μHC cell surface expression and signaling in these cells. Therefore, these findings offer an explanation for the occurrence of HC allelic exclusion in mice lacking SLC components.
Collapse
Affiliation(s)
- Gunther R Galler
- Hematopoiesis Unit, Nikolaus-Fiebiger-Center, Friedrich-Alexander University, Glueckstrasse 6, 91054 Erlangen, Germany
| | | | | | | | | | | |
Collapse
|
28
|
Keren Z, Diamant E, Ostrovsky O, Bengal E, Melamed D. Modification of ligand-independent B cell receptor tonic signals activates receptor editing in immature B lymphocytes. J Biol Chem 2003; 279:13418-24. [PMID: 14668327 DOI: 10.1074/jbc.m311970200] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Maturation of B lymphocytes strictly depends on the signaling competence of the B cell antigen receptor (BCR). Autoreactive receptors undergo negative selection and can be replaced by receptor editing. In addition, the process of maturation of non-self B cells and migration to the spleen, referred to as positive selection, is limited by the signaling competence of the BCR. Using 3-83Tg mice deficient of CD19 we have shown that signaling incompetence not only blocks positive selection but also activates receptor editing. Here we study the role of ligand-independent BCR tonic tyrosine phosphorylation signals in activation of receptor editing. We find that editing, immature 3-83Tg B cells deficient of CD19 have elevated BCR tonic signals and that lowering these tonic signals effectively suppresses receptor editing. Furthermore, we show that elevation of BCR tonic signals in non-editing, immature 3-83Tg B cells stimulates significant receptor editing. We also show that positive selection and developmental progression from the bone marrow to the spleen are limited to cells capable of establishing appropriate tonic signals, as in contrast to immature cells, splenic 3-83Tg B cells deficient of CD19 have BCR tonic signals similar to those of the control 3-83Tg cells. This developmental progression is accompanied by activation of molecules signaling for growth and survival. Hence, we suggest that ligand-independent BCR tonic signals are required for promoting positive selection and suppressing the receptor-editing mechanism in immature B cells.
Collapse
Affiliation(s)
- Zohar Keren
- Technion, Faculty of Medicine, Departments of Immunology and Biochemistry, Rappaport Family Institute for Research in the Medical Sciences, Haifa 31096, Israel
| | | | | | | | | |
Collapse
|
29
|
Wang LD, Clark MR. B-cell antigen-receptor signalling in lymphocyte development. Immunology 2003; 110:411-20. [PMID: 14632637 PMCID: PMC1783068 DOI: 10.1111/j.1365-2567.2003.01756.x] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2003] [Revised: 09/15/2003] [Accepted: 09/15/2003] [Indexed: 12/11/2022] Open
Abstract
Signalling through the B-cell antigen receptor (BCR) is required throughout B-cell development and peripheral maturation. Targeted disruption of BCR components or downstream effectors indicates that specific signalling mechanisms are preferentially required for central B-cell development, peripheral maturation and repertoire selection. Additionally, the avidity and the context in which antigen is encountered determine both cell fate and differentiation in the periphery. Although the signalling and receptor components required at each stage have been largely elucidated, the molecular mechanisms through which specific signalling are evoked at each stage are still obscure. In particular, it is not known how the pre-BCR initiates the signals required for normal development or how immature B cells regulate the signalling pathways that determine cell fate. In this review, we will summarize the recent studies that have defined the molecules required for B-cell development and maturation as well as the theories on how signals may be regulated at each stage.
Collapse
Affiliation(s)
- Leo D Wang
- Section of Rheumatology and Committee on Immunology, Biological Sciences Division and Pritzker School of Medicine, University of Chicago, Chicago, IL 60637, USA.
| | | |
Collapse
|
30
|
Bertrand FE, Spengeman JD, Shah N, LeBien TW. B-cell development in the presence of the MLL/AF4 oncoprotein proceeds in the absence of HOX A7 and HOX A9 expression. Leukemia 2003; 17:2454-9. [PMID: 14562113 DOI: 10.1038/sj.leu.2403178] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Infant acute lymphoblastic leukemia (ALL) is frequently characterized by the t(4;11)(q21;q23) cytogenetic abnormality encoding the MLL/AF4 oncogene, increased HOX gene expression and a pro-B/monocytoid phenotype. We have previously established a novel MLL/AF4-positive cell line, B-lineage 3 (BLIN-3), which retains several features of normal B-lineage development (functional Ig gene rearrangement and apoptotic sensitivity to stromal cell withdrawal) not generally observed in infant ALL. We now use microarray analysis to identify patterns of gene expression in BLIN-3 that may modulate MLL/AF4 oncogenesis and contribute to the retention of normal B-lineage developmental characteristics. Comparison of 6815 expressed genes in BLIN-3 with published microarray data on leukemic blasts from t(4;11) patients indicated that BLIN-3 was unique in lacking the expression of certain HOX-A cluster genes. These results were validated by RT-PCR showing no expression of HOX A7 or HOX A9 in BLIN-3. A HOX C8 promoter reporter was active in BLIN-3, indicating that lack of HOX gene expression in BLIN-3 was not due to a nonfunctional MLL/AF4. Our results suggest that B-lineage development can proceed in t(4;11) leukemic blasts in the absence of HOX-A gene expression.
Collapse
Affiliation(s)
- F E Bertrand
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC 27858, USA
| | | | | | | |
Collapse
|
31
|
Abstract
Hematopoiesis is regulated by a variety of signals that either originate within a developing cell or are supplied by the surrounding environment in secreted- or contact-dependent forms. This review discusses the effects of one secreted factor, interleukin-7, on the development of B lymphocytes. We describe a molecular mechanism for a crucial checkpoint during B lineage maturation, based on the integration of signals mediated by the pre-B cell receptor, the interleukin-7 receptor, and the environment in which these signals are received.
Collapse
Affiliation(s)
- Heather E Fleming
- Ontario Cancer Institute, Princess Margaret Hospital, University Health Network, Department of Immunology, University of Toronto, 610 University Avenue, 7-504 Toronto, Ont., Canada M5G 2M9.
| | | |
Collapse
|
32
|
Marchbank KJ, Kulik L, Gipson MG, Morgan BP, Holers VM. Expression of human complement receptor type 2 (CD21) in mice during early B cell development results in a reduction in mature B cells and hypogammaglobulinemia. THE JOURNAL OF IMMUNOLOGY 2002; 169:3526-35. [PMID: 12244142 DOI: 10.4049/jimmunol.169.7.3526] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Complement receptor (CR) type 2 (CR2/CD21) is normally expressed only during the immature and mature stages of B cell development. In association with CD19, CR2 plays an important role in enhancing mature B cell responses to foreign Ag. We used a murine Vlambda2 promoter/Vlambda2-4 enhancer minigene to develop transgenic mice that initiate expression of human CR2 (hCR2) during the CD43(+)CD25(-) late pro-B cell stage of development. We found peripheral blood B cell numbers reduced by 60% in mice expressing high levels of hCR2 and by 15% in mice with intermediate receptor expression. Splenic B cell populations were altered with an expansion of marginal zone cells, and basal serum IgG levels as well as T-dependent immune responses were also significantly decreased in transgenic mice. Mice expressing the highest levels of hCR2 demonstrated in the bone marrow a slight increase in B220(int)CD43(+)CD25(-) B cells in association with a substantial decrease in immature and mature B cells, indicative of a developmental block in the pro-B cell stage. These data demonstrate that stage-specific expression of CR2 is necessary for normal B cell development, as premature receptor expression substantially alters this process. Alterations in B cell development are most likely due to engagement of pre-B cell receptor-mediated or other regulatory pathways by hCR2 in a CD19- and possibly C3 ligand-dependent manner.
Collapse
Affiliation(s)
- Kevin J Marchbank
- Complement Biology Group, Department of Medical Biochemistry, University of Wales College of Medicine, Heath Park, Cardiff, United Kingdom
| | | | | | | | | |
Collapse
|
33
|
Affiliation(s)
- Jane Seagal
- Department of Immunology, Bruce Rappaport Faculty of Medicine and Rappaport Family Institute for Research in the Medical Sciences, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | | |
Collapse
|
34
|
Shivtiel S, Leider N, Sadeh O, Kraiem Z, Melamed D. Impaired light chain allelic exclusion and lack of positive selection in immature B cells expressing incompetent receptor deficient of CD19. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:5596-604. [PMID: 12023356 DOI: 10.4049/jimmunol.168.11.5596] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Positive signaling is now thought to be important for B cell maturation, although the nature of such signals has not yet been defined. We are studying the regulatory role of B cell Ag receptor (BCR) signaling in mediating positive selection of immature B cells. To do so, we use Ig transgenic mice (3-83Tg) that are deficient in CD19, thus generating a monoclonal immature B cell population expressing signaling-incompetent BCR. Immature 3-83Tg CD19(-/-) B cells undergo developmental arrest in the bone marrow, allowing maturation only to cells that effectively compensate for the compromised receptor by elevated levels of BCR. We find that developmentally arrested 3-83Tg CD19(-/-) B cells fail to impose L chain allelic exclusion and undergo intensive V(D)J recombination to edit their BCR. Furthermore, immature 3-83Tg CD19(-/-) B cells, which were grown in vitro, failed to undergo positive selection and to survive when adoptively transferred into normal recipients. However, elevation of BCR expression levels, obtained by transgene homozygosity, effectively compensated for the compromised BCR and completely restored BCR-mediated Ca(2+) influx, allelic exclusion, and positive selection. Our results suggest that the BCR signaling threshold mediates positive selection of developing B cells, and that a receptor-editing mechanism has an important role in rescuing cells that fail positive selection because of incompetent receptors.
Collapse
Affiliation(s)
- Shoham Shivtiel
- Department of Immunology, Faculty of Medicine, Technion, and Endocrine Research Unit, Carmel Medical Center, Haifa, Israel
| | | | | | | | | |
Collapse
|
35
|
Bertrand FE, Vogtenhuber C, Shah N, LeBien TW. Pro-B-cell to pre-B-cell development in B-lineage acute lymphoblastic leukemia expressing the MLL/AF4 fusion protein. Blood 2001; 98:3398-405. [PMID: 11719380 DOI: 10.1182/blood.v98.12.3398] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The most common chromosomal abnormality of infant acute lymphoblastic leukemia (ALL) is the t(4;11)(q21;q23) that gives rise to the MLL/AF4 fusion gene. Leukemic blasts expressing MLL/AF4 are arrested at an early progenitor stage with lymphoid or monocytoid characteristics. A novel B-lineage ALL cell line termed B-lineage-3 (BLIN-3) requiring human bone marrow (BM) stromal cell contact and interleukin-7 (IL-7) for optimal proliferation has been established. BLIN-3 cells have a CD19(+)/CD10(-) phenotype typical of infant ALL, and they harbor the t(4;11)(q21;q23) chromosomal translocation. Reverse transcription-polymerase chain reaction and Western blot analysis confirmed the presence of the MLL/AF4 fusion mRNA and protein in BLIN-3. Initial BLIN-3 cultures had a pro-B cell phenotype and did not express cytoplasmic or surface mu heavy chain. After approximately 5 months in culture on BM stromal cells plus IL-7, BLIN-3 sublines emerged expressing mu heavy chain and VpreB on the cell surfaces (ie, pre-B-cell receptor [BCR](+)). BLIN-3 cells expressing pre-BCR had the t(4;11)(q21;q23) translocation and expressed the MLL/AF4 fusion protein. Cross-linking the BLIN-3 pre-BCR led to enhanced cell proliferation, demonstrating that BLIN-3 expressed a functional pre-BCR. Increased acquisition of surface pre-BCR in BLIN-3 sublines was associated with loss of DJ rearrangements and the appearance of VDJ rearrangements. These results indicate that expression of the MLL/AF4 fusion protein is compatible with BM stromal cell and cytokine dependency, functional immunoglobulin gene segment rearrangement, and subsequent expression of a potentially diverse antigen receptor repertoire. Thus, the expression of MLL/AF4 is compatible with the normal developmental program of human B-lineage cells.
Collapse
Affiliation(s)
- F E Bertrand
- University of Minnesota Cancer Center and the Department of Laboratory Medicine and Pathology, Minneapolis 55455, USA.
| | | | | | | |
Collapse
|
36
|
de Fougerolles AR, Batista F, Johnsson E, Fearon DT. IgM and stromal cell-associated heparan sulfate/heparin as complement-independent ligands for CD19. Eur J Immunol 2001; 31:2189-99. [PMID: 11449373 DOI: 10.1002/1521-4141(200107)31:7<2189::aid-immu2189>3.0.co;2-v] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The more severe phenotype of mice lacking CD19 as compared to CD21 suggests that a complement-independent ligand for the CD19/CD21 complex exists. We sought ligands for CD19 by examining binding reactions with fusion proteins comprised of the extracellular region of CD19 and the Fc region of IgG1. A fusion protein containing the third extracellular domain (D3-Fc) bound to WEHI-231 cells, and this was competed by soluble IgM. This function of IgM was confirmed by the binding of D3-Fc to beads coated with IgM. A second ligand for D3-Fc was found on stromal cells, and was shown to be heparin/heparan sulfate. These two ligands would be considered to reside on follicular dendritic cells, and may account for the observed ability of D3-Fc to bind to sites in germinal centers containing these cells.
Collapse
Affiliation(s)
- A R de Fougerolles
- Wellcome Trust Immunology Unit, Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, GB, UK
| | | | | | | |
Collapse
|
37
|
Fearon DT, Carroll MC. Regulation of B lymphocyte responses to foreign and self-antigens by the CD19/CD21 complex. Annu Rev Immunol 2000; 18:393-422. [PMID: 10837064 DOI: 10.1146/annurev.immunol.18.1.393] [Citation(s) in RCA: 359] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The membrane protein complex CD19/CD21 couples the innate immune recognition of microbial antigens by the complement system to the activation of B cells. CD21 binds the C3d fragment of activated C3 that becomes covalently attached to targets of complement activation, and CD19 co-stimulates signaling through the antigen receptor, membrane immunoglobulin. CD21 is also expressed by follicular dendritic cells and mediates the long-term retention of antigen that is required for the maintenance of memory B cells. Understanding of the biology of this receptor complex has been enriched by analyses of genetically modified mice; these analyses have uncovered roles not only in positive responses to foreign antigens, but also in the development of tolerance to self-antigens. Studies of signal transduction have begun to determine the basis for the coreceptor activities of CD19. The integration of innate and adaptive immune recognition at this molecular site on the B cell guides the appropriate selection of antigen by adaptive immunity and emphasizes the importance of this coreceptor complex.
Collapse
Affiliation(s)
- D T Fearon
- Wellcome Trust Immunology Unit, Department of Medicine, School of Clinical Medicine, University of Cambridge, United Kingdom.
| | | |
Collapse
|
38
|
Abstract
Abstract
Development of mammalian B-lineage cells is characterized by progression through a series of checkpoints defined primarily by rearrangement and expression of immunoglobulin genes. Progression through these checkpoints is also influenced by stromal cells in the microenvironment of the primary tissues wherein B-cell development occurs, ie, fetal liver and bone marrow and adult bone marrow. This review focuses on the developmental biology of human bone marrow B-lineage cells, including perturbations that contribute to the origin and evolution of B-lineage acute lymphoblastic leukemia and primary immunodeficiency diseases characterized by agammaglobulinemia. Recently described in vitro and in vivo models that support development and expansion of human B-lineage cells through multiple checkpoints provide new tools for identifying the bone marrow stromal cell–derived molecules necessary for survival and proliferation. Mutations in genes encoding subunits of the pre-B cell receptor and molecules involved in pre-B cell receptor signaling culminate in X-linked and non–X-linked agammaglobulinemia. A cardinal feature of these immunodeficiencies is an apparent apoptotic sensitivity of B-lineage cells at the pro-B to pre-B transition. On the other end of the spectrum is the apoptotic resistance that accompanies the development of B-lineage acute lymphoblastic leukemia, potentially a reflection of genetic abnormalities that subvert normal apoptotic programs. The triad of laboratory models that mimic the bone marrow microenvironment, immunodeficiency diseases with specific defects in B-cell development, and B-lineage acute lymphoblastic leukemia can now be integrated to deepen our understanding of human B-cell development.
Collapse
|
39
|
Abstract
Development of mammalian B-lineage cells is characterized by progression through a series of checkpoints defined primarily by rearrangement and expression of immunoglobulin genes. Progression through these checkpoints is also influenced by stromal cells in the microenvironment of the primary tissues wherein B-cell development occurs, ie, fetal liver and bone marrow and adult bone marrow. This review focuses on the developmental biology of human bone marrow B-lineage cells, including perturbations that contribute to the origin and evolution of B-lineage acute lymphoblastic leukemia and primary immunodeficiency diseases characterized by agammaglobulinemia. Recently described in vitro and in vivo models that support development and expansion of human B-lineage cells through multiple checkpoints provide new tools for identifying the bone marrow stromal cell–derived molecules necessary for survival and proliferation. Mutations in genes encoding subunits of the pre-B cell receptor and molecules involved in pre-B cell receptor signaling culminate in X-linked and non–X-linked agammaglobulinemia. A cardinal feature of these immunodeficiencies is an apparent apoptotic sensitivity of B-lineage cells at the pro-B to pre-B transition. On the other end of the spectrum is the apoptotic resistance that accompanies the development of B-lineage acute lymphoblastic leukemia, potentially a reflection of genetic abnormalities that subvert normal apoptotic programs. The triad of laboratory models that mimic the bone marrow microenvironment, immunodeficiency diseases with specific defects in B-cell development, and B-lineage acute lymphoblastic leukemia can now be integrated to deepen our understanding of human B-cell development.
Collapse
|
40
|
Affiliation(s)
- S Desiderio
- Department of Molecular Biology and Genetics, Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | |
Collapse
|
41
|
Genetic Modification of Human B-Cell Development: B-Cell Development Is Inhibited by the Dominant Negative Helix Loop Helix Factor Id3. Blood 1999. [DOI: 10.1182/blood.v94.8.2637.420k22_2637_2646] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Transgenic and gene targeted mice have contributed greatly to our understanding of the mechanisms underlying B-cell development. We describe here a model system that allows us to apply molecular genetic techniques to the analysis of human B-cell development. We constructed a retroviral vector with a multiple cloning site connected to a gene encoding green fluorescent protein by an internal ribosomal entry site. Human CD34+CD38− fetal liver cells, cultured overnight in a combination of stem cell factor and interleukin-7 (IL-7), could be transduced with 30% efficiency. We ligated the gene encoding the dominant negative helix loop helix (HLH) factor Id3 that inhibits many enhancing basic HLH transcription factors into this vector. CD34+CD38− FL cells were transduced with Id3-IRES-GFP and cultured with the murine stromal cell line S17. In addition, we cultured the transduced cells in a reaggregate culture system with an SV-transformed human fibroblast cell line (SV19). It was observed that overexpression of Id3 inhibited development of B cells in both culture systems. B-cell development was arrested at a stage before expression of the IL-7R. The development of CD34+CD38− cells into CD14+ myeloid cells in the S17 system was not inhibited by overexpression of Id3. Moreover, Id3+ cells, although inhibited in their B-cell development, were still able to develop into natural killer (NK) cells when cultured in a combination of Flt-3L, IL-7, and IL-15. These findings confirm the essential role of bHLH factors in B-cell development and demonstrate the feasibility of retrovirus-mediated gene transfer as a tool to genetically modify human B-cell development.
Collapse
|
42
|
Kurosaka D, LeBien TW, Pribyl JA. Comparative studies of different stromal cell microenvironments in support of human B-cell development. Exp Hematol 1999; 27:1271-81. [PMID: 10428504 DOI: 10.1016/s0301-472x(99)00067-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
This study compared human murine stromal cells for their capacity to support human hematopoietic stem cell (HSC) development into the B lineage. FACS sorted human fetal bone marrow (BM) HSC (CD34+CD19- or CD34+/CD10-/CD19-/CD45RA) were cultured on human fetal BM stromal cells, human skin fibroblasts, or murine S17 stromal cells and analyzed by flow cytometry or reverse transcriptase polymerase chain reaction. CD34+CD19- HSC on human BM stromal cells or fibroblasts differentiated into B-lineage cells with a continuum in density of surface CD19 expression, and some cells expressing micro/kappa or micro/lambda B-cell receptors. In contrast, CD19+ cells from S17 cultures had two- to fourfold higher levels of CD19, but no cells expressing B-cell receptors. The number and percentage of CD19+ cells was high, intermediate, or low in the human BM, human fibroblast, or murine S17 stromal cell cultures, respectively. Reverse transcriptase polymerase chain reaction analysis showed that TdT, CD19, and DHQ52-J(H) rearrangements were expressed at comparable levels when CD34+/CD19- HSC were plated on human or murine stromal cells. In contrast, CD34+/CD10-/CD19-/CD45RA HSC plated on human or murine stromal cells expressed CD19 in both cultures, but TdT was only expressed in human stromal cell cultures. We conclude that human BM stromal cell, human skin fibroblasts, and murine S17 stromal cell cultures can provide complementary and comparative tools for identification of stromal cell ligands with potentially unique functions in regulating human B-cell development.
Collapse
Affiliation(s)
- D Kurosaka
- Department of Internal Medicine, The Jikei University of Medicine, Tokyo, Japan
| | | | | |
Collapse
|
43
|
Shiokawa S, Mortari F, Lima JO, Nuñez C, Bertrand FE, Kirkham PM, Zhu S, Dasanayake AP, Schroeder HW. IgM Heavy Chain Complementarity-Determining Region 3 Diversity Is Constrained by Genetic and Somatic Mechanisms Until Two Months After Birth. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.10.6060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
Due to the greater range of lengths available to the third complementarity determining region of the heavy chain (HCDR3), the Ab repertoire of normal adults includes larger Ag binding site structures than those seen in first and second trimester fetal tissues. Transition to a steady state range of HCDR3 lengths is not complete until the infant reaches 2 mo of age. Fetal constraints on length begin with a genetic predilection for use of short DH (D7-27 or DQ52) gene segments and against use of long DH (e.g., D3 or DXP) and JH (JH6) gene segments in both fetal liver and fetal bone marrow. Further control of length is achieved through DH-specific limitations in N addition, with D7-27 DJ joins including extensive N addition and D3-containing DJ joins showing a paucity of N addition. DH-specific constraints on N addition are no longer apparent in adult bone marrow. Superimposed upon these genetic mechanisms to control length is a process of somatic selection that appears to ensure expression of a restricted range of HCDR3 lengths in both fetus and adult. B cells that express Abs of an “inappropriate” length appear to be eliminated when they first display IgM on their cell surface. Control of N addition appears aberrant in X-linked agammaglobulinemia, which may exacerbate the block in B cell development seen in this disease. Restriction of the fetal repertoire appears to be an active process, forcing limits on the diversity, and hence range of Ab specificities, available to the young.
Collapse
Affiliation(s)
- Satoshi Shiokawa
- *Division of Developmental and Clinical Immunology, Departments of
- †Medicine and
| | - Frank Mortari
- *Division of Developmental and Clinical Immunology, Departments of
- †Medicine and
| | - Jose O. Lima
- *Division of Developmental and Clinical Immunology, Departments of
- †Medicine and
| | - César Nuñez
- *Division of Developmental and Clinical Immunology, Departments of
- †Medicine and
| | - Fred E. Bertrand
- *Division of Developmental and Clinical Immunology, Departments of
- ‡Microbiology, and
| | - Perry M. Kirkham
- *Division of Developmental and Clinical Immunology, Departments of
- ‡Microbiology, and
| | - Shigui Zhu
- *Division of Developmental and Clinical Immunology, Departments of
- †Medicine and
| | - Ananda P. Dasanayake
- §Specialized Center for Caries Research, University of Alabama, Birmingham, AL 35294
| | - Harry W. Schroeder
- *Division of Developmental and Clinical Immunology, Departments of
- †Medicine and
- ‡Microbiology, and
| |
Collapse
|
44
|
Luce MJ, Burrows PD. The neuronal EGF-related genes NELL1 and NELL2 are expressed in hemopoietic cells and developmentally regulated in the B lineage. Gene 1999; 231:121-6. [PMID: 10231576 DOI: 10.1016/s0378-1119(99)00093-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
NELL1 and NELL2 (neural epidermal growth factor-like 1 and 2) are recently described members of the epidermal growth factor gene family that have previously been shown to be expressed almost exclusively in brain tissue. Here we demonstrate regulated expression of NELL1 and NELL2 in human hematopoietic cells. Mature NELL1 mRNA is not detected in any normal hemopoietic cell type, although the gene is transcribed during a narrow window of pre-B cell development, and cell lines at the same developmental stage express the NELL1 mRNA. The related NELL2 gene is expressed by all nucleated peripheral blood cells examined (B, T, monocyte, and natural killer cells), but not in any of the bone marrow B lineage cells at earlier stages of development. However, leukemic cell lines corresponding to the same early differentiation stages express abundant NELL2 mRNA. These results suggest normal and possible oncogenic roles for the NELL proteins in hemopoietic cells.
Collapse
Affiliation(s)
- M J Luce
- University of Alabama at Birmingham, Department of Microbiology, Division of Developmental and Clinical Immunology, Birmingham, AL, USA
| | | |
Collapse
|
45
|
Deguchi T, Komada Y, Sugiyama K, Zhang XL, Azuma E, Yamamoto H, Sakurai M. Expression of homing-associated cell adhesion molecule (H-CAM/CD44) on human CD34+ hematopoietic progenitor cells. Exp Hematol 1999; 27:542-52. [PMID: 10089918 DOI: 10.1016/s0301-472x(98)00047-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
We investigated the expression of CD44 molecule on CD34+ hematopoietic progenitor cells. Significantly lower expression of CD44 was observed on bone marrow (BM) CD34+ cells compared with circulating CD34+ cells in cord blood and peripheral blood. Using fluorescence-activated cell sorting, human CD34+ BM cells were fractionated into CD44+ and CD44- populations. Immunofluorescence analysis revealed that the majority of CD34+CD44- cells expressed B-lymphocyte-associated CD10 and CD19 antigens, whereas only a part of CD34+CD44+ cells were positive for CD19. Myeloid and erythroid progenitor cells were found predominantly in CD34+ CD44+ cell fractions. In short-term suspension cultures, cell proliferation and G1-->S transition in the cell cycle were enhanced in CD34+CD44+ cells. In contrast, a large part of CD34+CD44- cells underwent apoptotic cell death. Although co-culture with BM stromal cells could partially prevent CD34+CD44- cells from undergoing apoptosis, significant increase of apoptotic cells was consistently observed. Furthermore, CD34+CD44- cells plated on BM stromal cells could differentiate into CD34-CD44-CD10-CD19+ cells. These findings suggest that CD34+CD44- cells expressing CD19 would represent unique B-lymphocyte-committed precursors in BM, which might undergo apoptotic cell death in the early steps of B-cell differentiation.
Collapse
Affiliation(s)
- T Deguchi
- Department of Pediatrics, Mie University School of Medicine, Japan
| | | | | | | | | | | | | |
Collapse
|
46
|
Fearon DT. Non-structural determinants of immunogenicity and the B cell co-receptors, CD19, CD21, and CD22. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 1999; 452:181-4. [PMID: 9889972 DOI: 10.1007/978-1-4615-5355-7_20] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Affiliation(s)
- D T Fearon
- Wellcome Trust Immunology Unit, University of Cambridge School of Clinical Medicine, United Kingdom
| |
Collapse
|
47
|
Levin SD, Koelling RM, Friend SL, Isaksen DE, Ziegler SF, Perlmutter RM, Farr AG. Thymic Stromal Lymphopoietin: A Cytokine That Promotes the Development of IgM+ B Cells In Vitro and Signals Via a Novel Mechanism. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.2.677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
A novel cytokine from a thymic stromal cell line (thymic stromal lymphopoietin (TSLP)) promotes the development of B220+/IgM+ immature B cells when added to fetal liver cultures, long term bone marrow cultures, or bone marrow cells plated in semisolid medium. Because the activities of TSLP overlap with those of IL-7 in some in vitro assays, we compared the signaling mechanisms employed by TSLP and IL-7. Proliferation of a factor-dependent pre-B cell line (NAG8/7) in response to either TSLP or IL-7 was inhibited by anti-IL-7Rα mAbs, suggesting that the functional TSLP receptor complex uses IL-7Rα. In contrast, three different Abs to the common cytokine receptor γ-chain had no effect on the response of these cells to TSLP, indicating that the functional TSLP receptor complex does not use the common cytokine receptor γ-chain. Both cytokines induced activation of Stat5, but only IL-7 induced activation of the Janus family kinases Jak1 and Jak3. In fact, TSLP failed to activate any of the four known Janus family kinases, suggesting that Stat5 phosphorylation is mediated by a novel mechanism. Taken together, these data support the idea that TSLP can make unique contributions to B lymphopoiesis and indicate that it does so by mechanisms distinct from IL-7.
Collapse
Affiliation(s)
| | - Ray M. Koelling
- †Biological Structure, University of Washington, Seattle, WA 98195
| | | | | | | | | | - Andrew G. Farr
- *Immunology and
- †Biological Structure, University of Washington, Seattle, WA 98195
| |
Collapse
|
48
|
Abstract
Genes encoding the heavy chain portion of immunoglobulin molecules arise from the combinatorial association of V, D and J gene segments, which occurs during discrete stages of B lineage development in the bone marrow. Recently, V(H) replacement, a form of receptor editing, has been described, in which the variable region of an existing VDJ(H) rearrangement is replaced by another V(H) gene segment in a recombination event believed to involve an embedded heptamer within the coding region of the V(H). Studies of transgenic mice with "knocked-in" VDJ(H) genes encoding anti-DNA specificity have demonstrated that receptor editing of the heavy chain is one mechanism by which autoreactive B cell receptors can be modified. Another mouse, the "quasi-monoclonal", which encodes a "knocked-in" VDJ(H) for the hapten NP also contains B lineage cells that undergo V(H) replacement. This suggests that V(H) replacement may play a role in the normal diversification of the antibody repertoire. Using a ligation-mediated PCR assay, we have identified V(QM) double-stranded DNA breaks indicative of V(H) replacement intermediates from bone marrow and splenic B lineage cells of quasi-monoclonal mice in the absence of immunization. V(QM) to J558 recombination deletion products consistent with V(H) replacement were also detected in both the bone marrow and spleen of non-immunized quasi-monoclonal mice. Moreover, RAG-1 transcripts were detected in the spleen. These data suggest that V(H) replacement can be part of the mechanism(s) used by B lineage cells to generate diversity throughout B lineage development, including later stages occurring in secondary lymphoid tissues.
Collapse
Affiliation(s)
- F E Bertrand
- The Ontario Cancer Institute and Department of Immunology, University of Toronto, Canada.
| | | | | |
Collapse
|
49
|
Ghia P, ten Boekel E, Rolink AG, Melchers F. B-cell development: a comparison between mouse and man. IMMUNOLOGY TODAY 1998; 19:480-5. [PMID: 9785673 DOI: 10.1016/s0167-5699(98)01330-9] [Citation(s) in RCA: 94] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A common variable immunodeficiency (CVID) patient, who carries mutations on both alleles of the gene encoding the surrogate light chain component lambda 5/14.1, shows a similar phenotype of B-cell deficiency as the lambda 5-deficient mutant mouse. As discussed here by Paolo Ghia and colleagues, this points to a remarkably similar developmental pathway of B cells in humans and mice.
Collapse
Affiliation(s)
- P Ghia
- Dana Farber Cancer Institute Boston, MA, USA
| | | | | | | |
Collapse
|
50
|
Muraguchi A, Tagoh H, Kitagawa T, Nagata T, Kishi H. Stromal cells and cytokines in the induction of recombination activating gene (RAG) expression in a human lymphoid progenitor cell. Leuk Lymphoma 1998; 30:73-85. [PMID: 9669678 DOI: 10.3109/10428199809050931] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The activation of recombination activating genes (RAGs) plays critical roles in the V(D)J gene recombination machinery and lymphocyte repertoire formation. However, the regulation of RAG gene expression in humans as well as animals is poorly understood. We show that RAG gene expression is activated in a human lymphoid progenitor cell line (FL8.2.4.4) by coculturing them on a bone marrow-derived stromal cell line (PA6) in the presence of cytokines. The RAG transcripts become detectable in 12 hours after initiation of culture, and the increased level is sustained at 24 hours. Among the cytokines, IL-3, IL-6, and IL-7, but not IL-2, IL-4, SCF, GM-CSF induces RAG activation. IL-3, IL-6, and IL-7 exert their effect synergistically on RAG activation. A cognate interaction between FL8.2.4.4 cells and PA6 stromal cells seems to be prerequisite for RAG activation. RAG transcripts are inducible in FL8.2.4.4 cells when cocultured on paraformaldehyde fixed-PA6 stromal cells in the presence of cytokines. These data indicate that two separate signals are both required for induction of RAG activation in lymphoid progenitors; one from the cell surface molecule(s) on stromal cells, and the other from recombinant cytokine(s). The expression of RAG mRNA in FL8.2.4.4 cells is concomitant with induction of recombinase activity. Thus, this system may provide a useful means for further understanding of the mechanisms controlling RAG activation and lymphocyte development in human system.
Collapse
Affiliation(s)
- A Muraguchi
- Department of Immunology, Faculty of Medicine, Toyama Medical and Pharmaceutical University, Japan.
| | | | | | | | | |
Collapse
|