1
|
Brennecke AM, Düber S, Roy B, Thomsen I, Garbe AI, Klawonn F, Pabst O, Kretschmer K, Weiss S. Induced B Cell Development in Adult Mice. Front Immunol 2018; 9:2483. [PMID: 30429851 PMCID: PMC6220648 DOI: 10.3389/fimmu.2018.02483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 10/08/2018] [Indexed: 11/30/2022] Open
Abstract
We employed the B-Indu-Rag1 model in which the coding exon of recombination-activating gene 1 (Rag1) is inactivated by inversion. It is flanked by inverted loxP sites. Accordingly, B cell development is stopped at the pro/pre B-I cell precursor stage. A B cell-specific Cre recombinase fused to a mutated estrogen receptor allows the induction of RAG1 function and B cell development by application of Tamoxifen. Since Rag1 function is recovered in a non-self-renewing precursor cell, only single waves of development can be induced. Using this system, we could determine that B cells minimally require 5 days to undergo development from pro/preB-I cells to the large and 6 days to the small preB-II cell stage. First immature transitional (T) 1 and T2 B cells could be detected in the bone marrow at day 6 and day 7, respectively, while their appearance in the spleen took one additional day. We also tested a contribution of adult bone marrow to the pool of B-1 cells. Sublethally irradiated syngeneic WT mice were adoptively transferred with bone marrow of B-Indu-Rag1 mice and B cell development was induced after 6 weeks. A significant portion of donor derived B-1 cells could be detected in such adult mice. Finally, early VH gene usage was tested after induction of B cell development. During the earliest time points the VH genes proximal to D/J were found to be predominantly rearranged. At later time points, the large family of the most distal VH prevailed.
Collapse
Affiliation(s)
| | - Sandra Düber
- Molecular Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Bishnudeo Roy
- Molecular Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Irene Thomsen
- Medical School Hannover, Institute of Immunology, Hannover, Germany
| | - Annette I Garbe
- Molecular Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Osteoimmunology, DFG-Center for Regenerative Therapies Dresden, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Frank Klawonn
- Biostatistics Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Oliver Pabst
- Medical School Hannover, Institute of Immunology, Hannover, Germany.,Institute of Molecular Medicine, RWTH Aachen University, Aachen, Germany
| | - Karsten Kretschmer
- Molecular Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Molecular and Cellular Immunology/Immune Regulation, DFG-Center for Regenerative Therapies Dresden, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Siegfried Weiss
- Molecular Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Medical School Hannover, Institute of Immunology, Hannover, Germany
| |
Collapse
|
2
|
Bossen C, Mansson R, Murre C. Chromatin topology and the regulation of antigen receptor assembly. Annu Rev Immunol 2012; 30:337-56. [PMID: 22224771 DOI: 10.1146/annurev-immunol-020711-075003] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
During an organism's ontogeny and in the adult, each B and T lymphocyte generates a unique antigen receptor, thereby creating the organism's ability to respond to a vast number of different antigens. The antigen receptor loci are organized into distinct regions that contain multiple variable (V), diversity (D), and/or joining (J) and constant (C) coding elements that are scattered across large genomic regions. In this review, we discuss the epigenetic modifications that take place in the different antigen receptor loci, the chromatin structure adopted by the antigen receptor loci to allow recombination of elements separated by large genomic distances, and the relationship between epigenetics and chromatin structure and how they relate to the generation of antigen receptor diversity.
Collapse
Affiliation(s)
- Claudia Bossen
- Division of Biological Sciences, Department of Molecular Biology, University of California at San Diego, La Jolla, California 92093-0377, USA
| | | | | |
Collapse
|
3
|
Mechanistic basis for RAG discrimination between recombination sites and the off-target sites of human lymphomas. Mol Cell Biol 2011; 32:365-75. [PMID: 22064481 DOI: 10.1128/mcb.06187-11] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
During V(D)J recombination, RAG targeting to correct sites versus off-target sites relies on both DNA sequence features and on chromatin marks. Kinetic analysis using the first highly active full-length purified RAG1/RAG2 complexes has now allowed us to define the important catalytic features of this complex. We found that the overall rate of nicking, but not hairpinning, is critical for the discrimination between correct (optimal) versus off-target (suboptimal) sites used in human T-cell lymphomas, and we show that the C-terminal portion of RAG2 is required for this. This type of kinetic analysis permits us to analyze only the catalytically active RAG complex, in contrast to all other methods, which are unavoidably confounded by mixture with inactive RAG complexes. Moreover, we can distinguish the two major features of any enzymatic catalysis: the binding constant (K(D)) and the catalytic turnover rate, k(cat). Beyond a minimal essential threshold of heptamer quality, further suboptimal heptamer deviations primarily reduce the catalytic rate constant k(cat) for nicking. Suboptimal nonamers reduce not only the binding of the RAG complex to the recombination site (K(D)) but also the catalytic rate constant, consistent with a tight interaction between the RAG complex and substrate during catalysis. These features explain many aspects of RAG physiology and pathophysiology.
Collapse
|
4
|
Mercer EM, Lin YC, Murre C. Factors and networks that underpin early hematopoiesis. Semin Immunol 2011; 23:317-25. [PMID: 21930392 DOI: 10.1016/j.smim.2011.08.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2010] [Accepted: 08/19/2011] [Indexed: 01/08/2023]
Abstract
Multiple trajectories have recently been described through which hematopoietic progenitor cells travel prior to becoming lineage-committed effectors. A wide spectrum of transcription factors has recently been identified that modulate developmental progression along such trajectories. Here we describe how distinct families of transcription factors act and are linked together to orchestrate early hematopoiesis.
Collapse
Affiliation(s)
- Elinore M Mercer
- Department of Molecular Biology, University of California, San Diego, La Jolla, CA 92093, United States.
| | | | | |
Collapse
|
5
|
Lazorchak AS, Su B. Perspectives on the role of mTORC2 in B lymphocyte development, immunity and tumorigenesis. Protein Cell 2011; 2:523-30. [PMID: 21822797 DOI: 10.1007/s13238-011-1077-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2011] [Accepted: 06/30/2011] [Indexed: 12/20/2022] Open
Abstract
Mammalian target of rapamycin complex 2 (mTORC2) is a key downstream mediator of phosphoinositol-3-kinase (PI3K) dependent growth factor signaling. In lymphocytes, mTORC2 has emerged as an important regulator of cell development, homeostasis and immune responses. However, our current understanding of mTORC2 functions and the molecular mechanisms regulating mTORC2 signaling in B and T cells are still largely incomplete. Recent studies have begun to shed light on this important pathway. We have previously reported that mTORC2 mediates growth factor dependent phosphorylation of Akt and facilitates Akt dependent phosphorylation and inactivation of transcription factors FoxO1 and FoxO3a. We have recently explored the functions of mTORC2 in B cells and show that mTORC2 plays a key role in regulating survival and immunoglobulin (Ig) gene recombination of bone marrow B cells through an Akt2-FoxO1 dependent mechanism. Ig recombination is suppressed in proliferating B cells to ensure that DNA double strand breaks are not generated in actively dividing cells. Our results raise the possibility that genetic or pharmacologic inhibition of mTORC2 may promote B cell tumor development as a result of inefficient suppression of Ig recombination in dividing B cells. We also propose a novel strategy to treat cancers based on our recent discovery that mTORC2 regulates Akt protein stability.
Collapse
Affiliation(s)
- Adam S Lazorchak
- Department of Immunobiology and the Vascular Biology and Therapeutic Program, Yale School of Medicine, New Haven, CT 06519, USA
| | | |
Collapse
|
6
|
IL-7R expression and IL-7 signaling confer a distinct phenotype on developing human B-lineage cells. Blood 2011; 118:2116-27. [PMID: 21680796 DOI: 10.1182/blood-2010-08-302513] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
IL-7 is an important cytokine for lymphocyte differentiation. Similar to what occurs in vivo, human CD19⁺ cells developing in human/murine xenogeneic cultures show differential expression of the IL-7 receptor α (IL-7Rα) chain (CD127). We now describe the relationship between CD127 expression/signaling and Ig gene rearrangement. In the present study, < 10% of CD19⁺CD127⁺ and CD19⁺CD127⁻ populations had complete VDJ(H) rearrangements. IGH locus conformation measurements by 3D FISH revealed that CD127⁺ and CD127⁻ cells were less contracted than pediatric BM pro-B cells that actively rearrange the IGH locus. Complete IGH rearrangements in CD127⁺ and CD127⁻ cells had smaller CDR3 lengths and fewer N-nucleotide insertions than pediatric BM B-lineage cells. Despite the paucity of VDJ(H) rearrangements, microarray analysis indicated that CD127⁺ cells resembled large pre-B cells, which is consistent with their low level of Ig light-chain rearrangements. Unexpectedly, CD127⁻ cells showed extensive Ig light-chain rearrangements in the absence of IGH rearrangements and resembled small pre-B cells. Neutralization of IL-7 in xenogeneic cultures led to an increase in Ig light-chain rearrangements in CD127⁺ cells, but no change in complete IGH rearrangements. We conclude that IL-7-mediated suppression of premature Ig light-chain rearrangement is the most definitive function yet described for IL-7 in human B-cell development.
Collapse
|
7
|
Schatz DG, Ji Y. Recombination centres and the orchestration of V(D)J recombination. Nat Rev Immunol 2011; 11:251-63. [PMID: 21394103 DOI: 10.1038/nri2941] [Citation(s) in RCA: 420] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The initiation of V(D)J recombination by the recombination activating gene 1 (RAG1) and RAG2 proteins is carefully orchestrated to ensure that antigen receptor gene assembly occurs in the appropriate cell lineage and in the proper developmental order. Here we review recent advances in our understanding of how DNA binding and cleavage by the RAG proteins are regulated by the chromatin structure and architecture of antigen receptor genes. These advances suggest novel mechanisms for both the targeting and the mistargeting of V(D)J recombination, and have implications for how these events contribute to genome instability and lymphoid malignancy.
Collapse
Affiliation(s)
- David G Schatz
- Department of Immunobiology and Howard Hughes Medical Institute, Yale University School of Medicine, 300 Cedar Street, Box 208011, New Haven, Connecticut 06520-8011, USA.
| | | |
Collapse
|
8
|
Subrahmanyam R, Sen R. Epigenetic features that regulate IgH locus recombination and expression. Curr Top Microbiol Immunol 2011; 356:39-63. [PMID: 21779986 DOI: 10.1007/82_2011_153] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Precisely regulated rearrangements that yield imprecise recombination junctions are hallmarks of antigen receptor gene assembly. At the immunoglobulin heavy chain (IgH) gene locus this is initiated by rearrangement of a D (H) gene segment to a J (H) gene segment to generate DJ(H) junctions, followed by rearrangement of a V (H) gene segment to the DJ(H) junction to generate fully recombined VDJ alleles. In this review we discuss the regulatory features of each step of IgH gene assembly and the role of epigenetic mechanisms in achieving regulatory precision.
Collapse
Affiliation(s)
- Ramesh Subrahmanyam
- National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | | |
Collapse
|
9
|
Subrahmanyam R, Sen R. RAGs' eye view of the immunoglobulin heavy chain gene locus. Semin Immunol 2010; 22:337-45. [PMID: 20864355 DOI: 10.1016/j.smim.2010.08.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2010] [Accepted: 08/12/2010] [Indexed: 10/19/2022]
Abstract
The immunoglobulin heavy chain (IgH) gene locus is activated at a precise stage of B lymphocyte development to undergo gene rearrangements that assemble the functional gene. In this review we summarize our current understanding of the chromatin state of the IgH as it appears just prior to the initiation of V(D)J recombination, and the implications of this structure for regulation of recombination. We also examine the role of the intron enhancer, Eμ, in establishing the pre-rearrangement chromatin structure. The emerging picture shows that the IgH locus consists of independently regulated domains, each of which requires multiple levels of epigenetic changes to reach the fully activated state.
Collapse
Affiliation(s)
- Ramesh Subrahmanyam
- Gene Regulation Section, Laboratory of Cellular and Molecular Biology, National Institute on Aging, National Institutes of Health, 251 Bayview Blvd., Room 06C214, Baltimore, MD 21224, United States
| | | |
Collapse
|
10
|
Sikes ML, McMillan RE, Bradshaw JM. The center of accessibility: Dβ control of V(D)J recombination. Arch Immunol Ther Exp (Warsz) 2010; 58:427-33. [PMID: 20890731 DOI: 10.1007/s00005-010-0101-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2010] [Accepted: 07/23/2010] [Indexed: 12/26/2022]
Abstract
Developmental patterning of antigen receptor gene assembly in lymphocyte precursors correlates with decondensation of the chromatin surrounding individual gene segments. Ongoing V(D)J recombination is associated with hyperacetylation of histones H3 and H4 and the expression of sterile germline transcripts across the region of recombinational accessibility. Likewise, histone acetyltransferase and SWI/SNF chromatin remodeling complexes each appear to be required for recombination, and the PHD-finger of RAG-2 preferentially associates with recombination signal sequence (RSS) chromatin that contains H3 trimethylated on lysine 4. However, the regulatory mechanisms that direct chromatin alteration and rearrangement have proven elusive, due in large part to the interdependency of individual stages in gene activation, our limited understanding of functional significance of changes to the histone code, and the difficulty of modeling recombinational accessibility in existing experimental systems. Examining Tcrb assembly in developing thymocytes, we review the central roles of RSS elements and germline promoters as foci for epigenetic reorganization of recombinationally accessible gene segments in light of recent findings and persistent questions.
Collapse
Affiliation(s)
- Michael L Sikes
- Department of Microbiology, North Carolina State University, 100 Derieux Place, Campus Box 7615, Raleigh, NC 27695, USA.
| | | | | |
Collapse
|
11
|
Mendicino M, Ramsoondar J, Phelps C, Vaught T, Ball S, LeRoith T, Monahan J, Chen S, Dandro A, Boone J, Jobst P, Vance A, Wertz N, Bergman Z, Sun XZ, Polejaeva I, Butler J, Dai Y, Ayares D, Wells K. Generation of antibody- and B cell-deficient pigs by targeted disruption of the J-region gene segment of the heavy chain locus. Transgenic Res 2010; 20:625-41. [PMID: 20872248 PMCID: PMC7089184 DOI: 10.1007/s11248-010-9444-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2010] [Accepted: 09/13/2010] [Indexed: 01/22/2023]
Abstract
A poly(A)-trap gene targeting strategy was used to disrupt the single functional heavy chain (HC) joining region (JH) of swine in primary fibroblasts. Genetically modified piglets were then generated via somatic cell nuclear transfer (SCNT) and bred to yield litters comprising JH wild-type littermate (+/+), JH heterozygous knockout (±) and JH homozygous knockout (−/−) piglets in the expected Mendelian ratio of 1:2:1. There are only two other targeted loci previously published in swine, and this is the first successful poly(A)-trap strategy ever published in a livestock species. In either blood or secondary lymphoid tissues, flow cytometry, RT-PCR and ELISA detected no circulating IgM+ B cells, and no transcription or secretion of immunoglobulin (Ig) isotypes, respectively in JH −/− pigs. Histochemical and immunohistochemical (IHC) studies failed to detect lymph node (LN) follicles or CD79α+ B cells, respectively in JH −/− pigs. T cell receptor (TCR)β transcription and T cells were detected in JH −/− pigs. When reared conventionally, JH −/− pigs succumbed to bacterial infections after weaning. These antibody (Ab)- and B cell-deficient pigs have significant value as models for both veterinary and human research to discriminate cellular and humoral protective immunity to infectious agents. Thus, these pigs may aid in vaccine development for infectious agents such as the pandemic porcine reproductive and respiratory syndrome virus (PRRSV) and H1N1 swine flu. These pigs are also a first significant step towards generating a pig that expresses fully human, antigen-specific polyclonal Ab to target numerous incurable infectious diseases with high unmet clinical need.
Collapse
Affiliation(s)
- M Mendicino
- Revivicor, Inc., 1700 Kraft Drive, Blacksburg, VA 24060, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Ostrovnaya I, Nanjangud G, Olshen AB. A classification model for distinguishing copy number variants from cancer-related alterations. BMC Bioinformatics 2010; 11:297. [PMID: 20525196 PMCID: PMC2897829 DOI: 10.1186/1471-2105-11-297] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2009] [Accepted: 06/02/2010] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Both somatic copy number alterations (CNAs) and germline copy number variants (CNVs) that are prevalent in healthy individuals can appear as recurrent changes in comparative genomic hybridization (CGH) analyses of tumors. In order to identify important cancer genes CNAs and CNVs must be distinguished. Although the Database of Genomic Variants (DGV) contains a list of all known CNVs, there is no standard methodology to use the database effectively. RESULTS We develop a prediction model that distinguishes CNVs from CNAs based on the information contained in the DGV and several other variables, including segment's length, height, closeness to a telomere or centromere and occurrence in other patients. The models are fitted on data from glioblastoma and their corresponding normal samples that were collected as part of The Cancer Genome Atlas project and hybridized to Agilent 244 K arrays. CONCLUSIONS Using the DGV alone CNVs in the test set can be correctly identified with about 85% accuracy if the outliers are removed before segmentation and with 72% accuracy if the outliers are included, and additional variables improve the prediction by about 2-3% and 12%, respectively. Final models applied to data from ovarian tumors have about 90% accuracy with all the variables and 86% accuracy with the DGV alone.
Collapse
Affiliation(s)
- Irina Ostrovnaya
- Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, NY, USA.
| | | | | |
Collapse
|
13
|
Harding FA, Stickler MM, Razo J, DuBridge RB. The immunogenicity of humanized and fully human antibodies: residual immunogenicity resides in the CDR regions. MAbs 2010; 2:256-65. [PMID: 20400861 DOI: 10.4161/mabs.2.3.11641] [Citation(s) in RCA: 517] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Monoclonal antibodies represent an attractive therapeutic tool as they are highly specific for their targets, convey effector functions and enjoy robust manufacturing procedures. Humanization of murine monoclonal antibodies has vastly improved their in vivo tolerability. Humanization, the replacement of mouse constant regions and V framework regions for human sequences, results in a significantly less immunogenic product. However, some humanized and even fully human sequence-derived antibody molecules still carry immunological risk. To more fully understand the immunologic potential of humanized and human antibodies, we analyzed CD4(+) helper T cell epitopes in a set of eight humanized antibodies. The antibodies studied represented a number of different VH and VL family members carrying unique CDR regions. In spite of these differences, CD4(+) T cell epitopes were found only in CDR-sequence containing regions. We were able to incorporate up to two amino acid modifications in a single epitope that reduced the immunogenic potential while retaining full biologic function. We propose that immunogenicity will always be present in some antibody molecules due to the nature of the antigen-specific combining sites. A consequence of this result is modifications to reduce immunogenicity will be centered on the affinity-determining regions. Modifications to CDR regions can be designed that reduce the immunogenic potential while maintaining the bioactivity of the antibody molecule.
Collapse
|
14
|
Abstract
Antigen receptors on the surface of B lymphocytes trigger adaptive immune responses after encountering their cognate antigens but also control a series of antigen-independent checkpoints during B cell development. These physiological processes are regulated by the expression and function of cell surface receptors, intracellular signaling molecules, and transcription factors. The function of these proteins can be altered by a dynamic array of post-translational modifications, using two interconnected mechanisms. These modifications can directly induce an altered conformational state in the protein target of the modification itself. In addition, they can create new binding sites for other protein partners, thereby contributing to where and when such multiple protein assemblies are activated within cells. As a new type of post-transcriptional regulator, microRNAs have emerged to influence the development and function of B cells by affecting the expression of target mRNAs.
Collapse
|
15
|
Yunk L, Meng W, Cohen PL, Eisenberg RA, Luning Prak ET. Antibodies in a heavy chain knock-in mouse exhibit characteristics of early heavy chain rearrangement. THE JOURNAL OF IMMUNOLOGY 2009; 183:452-61. [PMID: 19542457 DOI: 10.4049/jimmunol.0804060] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Studies in autoantibody transgenic mice have demonstrated receptor editing rearrangements at Ab H and L chain loci. However, the physiologic role of H chain editing (V(H) replacement and rearrangement on the second allele) has been called into question. It is unclear if additional rounds of H chain rearrangement are driven by BCR specificity. In this study, we analyze the manner in which B cells undergo additional H chain rearrangements in an anti-DNA H chain knock-in mouse, B6.56R. We find that rearrangements in 56R(+) B cells tend to involve the D gene locus on both alleles and the most J(H)-proximal V(H) gene segments on the endogenous allele. As a result, some B cells exhibit V(D)J rearrangements on both H chain alleles, yet allelic exclusion is tightly maintained in mature 56R B cells. As B cells mature, a higher proportion expresses the nontransgenic H chain allele. Rearrangements on both H chain alleles exhibit junctional diversity consistent with TdT-mediated N-addition, and TdT RNA is expressed exclusively at the pro-B cell stage in B6.56R. Collectively, these findings favor a single, early window of H chain rearrangement in B6.56R that precedes the expression of a functional BCR. B cells that happen to successfully rearrange another H chain may be favored in the periphery.
Collapse
Affiliation(s)
- Lenka Yunk
- Department of Pathology, University of Pennsylvania School of Medicine, Philadelphia, 19104, USA
| | | | | | | | | |
Collapse
|
16
|
Jhunjhunwala S, van Zelm MC, Peak MM, Murre C. Chromatin architecture and the generation of antigen receptor diversity. Cell 2009; 138:435-48. [PMID: 19665968 PMCID: PMC2726833 DOI: 10.1016/j.cell.2009.07.016] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The adaptive immune system generates a specific response to a vast spectrum of antigens. This remarkable property is achieved by lymphocytes that each express single and unique antigen receptors. During lymphocyte development, antigen receptor coding elements are assembled from widely dispersed gene segments. The assembly of antigen receptors is controlled at multiple levels, including epigenetic marking, nuclear location, and chromatin topology. Here, we review recently uncovered mechanisms that underpin long-range genomic interactions and the generation of antigen receptor diversity.
Collapse
Affiliation(s)
- Suchit Jhunjhunwala
- Division of Biological Sciences, Department of Molecular Biology, University of California, San Diego, La Jolla, CA 92093, USA
| | | | | | | |
Collapse
|
17
|
Wright SM, Woo YH, Alley TL, Shirley BJ, Akeson EC, Snow KJ, Maas SA, Elwell RL, Foreman O, Mills KD. Complex oncogenic translocations with gene amplification are initiated by specific DNA breaks in lymphocytes. Cancer Res 2009; 69:4454-60. [PMID: 19435904 DOI: 10.1158/0008-5472.can-08-4515] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Chromosomal instability is a hallmark of many tumor types. Complex chromosomal rearrangements with associated gene amplification, known as complicons, characterize many hematologic and solid cancers. Whereas chromosomal aberrations, including complicons, are useful diagnostic and prognostic cancer markers, their molecular origins are not known. Although accumulating evidence has implicated DNA double-strand break repair in suppression of oncogenic genome instability, the genomic elements required for chromosome rearrangements, especially complex lesions, have not been elucidated. Using a mouse model of B-lineage lymphoma, characterized by complicon formation involving the immunoglobulin heavy chain (Igh) locus and the c-myc oncogene, we have now investigated the requirement for specific genomic segments as donors for complex rearrangements. We now show that specific DNA double-strand breaks, occurring within a narrow segment of Igh, are necessary to initiate complicon formation. By contrast, neither specific DNA breaks nor the powerful intronic enhancer Emu are required for complicon-independent oncogenesis. This study is the first to delineate mechanisms of complex versus simple instability and the first to identify specific chromosomal elements required for complex chromosomal aberrations. These findings will illuminate genomic cancer susceptibility and risk factors.
Collapse
|
18
|
Abstract
The adaptive immune system of jawed vertebrates is based on a vast, anticipatory repertoire of specific antigen receptors, immunoglobulins (Ig) in B-lymphocytes and T-cell receptors (TCR) in T-lymphocytes. The Ig and TCRdiversity is generated by a process called V(D)J recombination, which is initiated by the RAG recombinase. Although RAG activity is very well conserved, the regulated accessibility of the antigen receptor genes to RAG has evolved with the species' organizational structure, which differs most significantly between fishes and tetrapods. V(D)J recombination was primarily characterized in developing lymphocytes of mice and human beings and is often described as an ordered, two-stage program. Studies in rabbit, chicken and shark show that this process does not have to be ordered, nor does it need to take place in two stages to generate a diverse repertoire and enable the expression of a single species of antigen receptor per cell, a restriction called allelic exclusion.
Collapse
|
19
|
Achour I, Cavelier P, Tichit M, Bouchier C, Lafaye P, Rougeon F. Tetrameric and Homodimeric Camelid IgGs Originate from the Same IgH Locus. THE JOURNAL OF IMMUNOLOGY 2008; 181:2001-9. [DOI: 10.4049/jimmunol.181.3.2001] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
20
|
Liu H, Schmidt-Supprian M, Shi Y, Hobeika E, Barteneva N, Jumaa H, Pelanda R, Reth M, Skok J, Rajewsky K, Shi Y. Yin Yang 1 is a critical regulator of B-cell development. Genes Dev 2008; 21:1179-89. [PMID: 17504937 PMCID: PMC1865490 DOI: 10.1101/gad.1529307] [Citation(s) in RCA: 200] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The role of the transcription factor Yin Yang 1 (YY1) in development is largely unknown. Here we show that specific ablation of YY1 in mouse B cells caused a defect in somatic rearrangement in the immunoglobulin heavy-chain (IgH) locus and a block in the progenitor-B-to-precursor-B-cell transition, which was partially rescued by a prerearranged IgH transgene. Three-dimensional DNA fluorescence in situ hybridization analysis revealed an important function for YY1 in IgH locus contraction, a process indispensable for distal V(H) to D(H)J(H) recombination. We provide evidence that YY1 binds the intronic Ei mu enhancer within the IgH locus, consistent with a direct role for YY1 in V(H)D(H)J(H) recombination. These findings identified YY1 as a critical regulator of early B-cell development.
Collapse
Affiliation(s)
- Huifei Liu
- Department of Pathology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Marc Schmidt-Supprian
- Department of Pathology, Harvard Medical School, Boston, Massachusetts 02115, USA
- CBR Institute for Biomedical Research, Inc., Boston, Massachusetts 02115, USA
| | - Yujiang Shi
- Department of Pathology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Elias Hobeika
- Institute for Biology III, Albert-Ludwigs University of Freiburg and Max Planck Institute for Immunobiology, 79108 Freiburg, Germany
| | - Natasha Barteneva
- Department of Pathology, Harvard Medical School, Boston, Massachusetts 02115, USA
- Flow cytometry core facility, CBR Institute for Biomedical Research, Inc., Boston, Massachusetts 02115, USA
| | - Hassan Jumaa
- Institute for Biology III, Albert-Ludwigs University of Freiburg and Max Planck Institute for Immunobiology, 79108 Freiburg, Germany
| | - Roberta Pelanda
- Institute for Biology III, Albert-Ludwigs University of Freiburg and Max Planck Institute for Immunobiology, 79108 Freiburg, Germany
| | - Michael Reth
- Institute for Biology III, Albert-Ludwigs University of Freiburg and Max Planck Institute for Immunobiology, 79108 Freiburg, Germany
| | - Jane Skok
- Department of Immunology and Molecular Pathology, Division of Infection and Immunity, University College London, London W1T 4JF, United Kingdom
| | - Klaus Rajewsky
- Department of Pathology, Harvard Medical School, Boston, Massachusetts 02115, USA
- CBR Institute for Biomedical Research, Inc., Boston, Massachusetts 02115, USA
| | - Yang Shi
- Department of Pathology, Harvard Medical School, Boston, Massachusetts 02115, USA
- Corresponding author.E-MAIL ; FAX (617) 432-6687
| |
Collapse
|
21
|
Chakraborty T, Chowdhury D, Keyes A, Jani A, Subrahmanyam R, Ivanova I, Sen R. Repeat organization and epigenetic regulation of the DH-Cmu domain of the immunoglobulin heavy-chain gene locus. Mol Cell 2007; 27:842-50. [PMID: 17803947 DOI: 10.1016/j.molcel.2007.07.010] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2007] [Revised: 05/30/2007] [Accepted: 07/09/2007] [Indexed: 11/22/2022]
Abstract
The first steps of murine immunoglobulin heavy-chain (IgH) gene recombination take place within a chromosomal domain that contains diversity (D(H)) and joining (J(H)) gene segments, but not variable (V(H)) gene segments. Here we show that the chromatin state of this domain is markedly heterogeneous. Specifically, only 5'- and 3'-most D(H) gene segments carry active chromatin modifications, whereas intervening D(H)s are associated with heterochromatic marks that are maintained by ongoing histone deacetylation. The intervening D(H)s form part of a tandemly repeated sequence that expresses tissue-specific, antisense oriented transcripts. We propose that the intervening D(H) genes are actively suppressed by repeat-induced epigenetic silencing, which is reflected in their infrequent representation in DJ(H) junctions compared to the flanking D(H) genes.
Collapse
Affiliation(s)
- Tirtha Chakraborty
- Laboratory of Cellular and Molecular Biology, National Institute on Aging, Baltimore, MD 21224, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Abstract
The mechanism of recombination-activating gene (RAG)-mediated rearrangement exists in all jawed vertebrates, but the organization and structure of immunoglobulin (Ig) genes, as they differ in fish and among fish species, reveal their capability for rapid evolution. In systems where there can exist 100 Ig loci, exon restructuring and sequence changes of the constant regions led to divergence of effector functions. Recombination among these loci created hybrid genes, the strangest of which encode variable (V) regions that function as part of secreted molecules and, as the result of an ancient translocation, are also grafted onto the T-cell receptor. Genomic changes in V-gene structure, created by RAG recombinase acting on germline recombination signal sequences, led variously to the generation of fixed receptor specificities, pseudogene templates for gene conversion, and ultimately to Ig sequences that evolved away from Ig function. The presence of so many Ig loci in fishes raises interesting questions not only as to how their regulation is achieved but also how successive whole-locus duplications are accommodated by a system whose function in other vertebrates is based on clonal antigen receptor expression.
Collapse
Affiliation(s)
- Ellen Hsu
- Department of Physiology and Pharmacology, State University of New York Health Science Center at Brooklyn, Brooklyn, NY 11203, USA.
| | | | | | | |
Collapse
|
23
|
Lin D, Ippolito GC, Zong RT, Bryant J, Koslovsky J, Tucker P. Bright/ARID3A contributes to chromatin accessibility of the immunoglobulin heavy chain enhancer. Mol Cancer 2007; 6:23. [PMID: 17386101 PMCID: PMC1852116 DOI: 10.1186/1476-4598-6-23] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2007] [Accepted: 03/26/2007] [Indexed: 01/27/2023] Open
Abstract
Bright/ARID3A is a nuclear matrix-associated transcription factor that stimulates immunoglobulin heavy chain (IgH) expression and Cyclin E1/E2F-dependent cell cycle progression. Bright positively activates IgH transcriptional initiation by binding to ATC-rich P sites within nuclear matrix attachment regions (MARs) flanking the IgH intronic enhancer (Eμ). Over-expression of Bright in cultured B cells was shown to correlate with DNase hypersensitivity of Eμ. We report here further efforts to analyze Bright-mediated Eμ enhancer activation within the physiological constraints of chromatin. A system was established in which VH promoter-driven in vitro transcription on chromatin- reconstituted templates was responsive to Eμ. Bright assisted in blocking the general repression caused by nucleosome assembly but was incapable of stimulating transcription from prebound nucleosome arrays. In vitro transcriptional derepression by Bright was enhanced on templates in which Eμ is flanked by MARs and was inhibited by competition with high affinity Bright binding (P2) sites. DNase hypersensitivity of chromatin-reconstituted Eμ was increased when prepackaged with B cell nuclear extract supplemented with Bright. These results identify Bright as a contributor to accessibility of the IgH enhancer.
Collapse
Affiliation(s)
- Danjuan Lin
- Section of Molecular Genetics and Microbiology and Institute of Cell and Molecular Biology, University of Texas at Austin, Austin, Texas, USA
| | - Gregory C Ippolito
- Section of Molecular Genetics and Microbiology and Institute of Cell and Molecular Biology, University of Texas at Austin, Austin, Texas, USA
| | - Rui-Ting Zong
- Section of Molecular Genetics and Microbiology and Institute of Cell and Molecular Biology, University of Texas at Austin, Austin, Texas, USA
| | - James Bryant
- Section of Molecular Genetics and Microbiology and Institute of Cell and Molecular Biology, University of Texas at Austin, Austin, Texas, USA
| | - Janet Koslovsky
- Section of Molecular Genetics and Microbiology and Institute of Cell and Molecular Biology, University of Texas at Austin, Austin, Texas, USA
| | - Philip Tucker
- Section of Molecular Genetics and Microbiology and Institute of Cell and Molecular Biology, University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
24
|
Raffel GD, Mercher T, Shigematsu H, Williams IR, Cullen DE, Akashi K, Bernard OA, Gilliland DG. Ott1(Rbm15) has pleiotropic roles in hematopoietic development. Proc Natl Acad Sci U S A 2007; 104:6001-6. [PMID: 17376872 PMCID: PMC1851606 DOI: 10.1073/pnas.0609041104] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
OTT1(RBM15) was originally described as a 5' translocation partner of the MAL(MKL1) gene in t(1,22)(p13;q13) infant acute mega karyocytic leukemia. OTT1 has no established physiological function, but it shares homology with the spen/Mint/SHARP family of proteins defined by three amino-terminal RNA recognition motifs and a carboxyl-terminal SPOC (Spen paralog and ortholog carboxyl-terminal) domain believed to act as a transcriptional repressor. To define the role of OTT1 in hematopoiesis and help elucidate the mechanism of t(1,22) acute megakaryocytic leukemia pathogenesis, a conditional allele of Ott1 was generated in mice. Deletion of Ott1 in adult mice caused a loss of peripheral B cells due to a block in pro/pre-B differentiation. There is myeloid and megakaryocytic expansion in spleen and bone marrow, an increase in the Lin(-)Sca-1(+)c-Kit(+) compartment that includes hematopoietic stem cells, and a shift in progenitor fate toward granulocyte differentiation. These data show a requirement for Ott1 in B lymphopoiesis, and inhibitory roles in the myeloid, megakaryocytic, and progenitor compartments. The ability of Ott1 to affect hematopoietic cell fate and expansion in multiple lineages is a novel attribute for a spen family member and delineates Ott1 from other known effectors of hematopoietic development. It is plausible that dysregulation of Ott1-dependent hematopoietic developmental pathways, in particular those affecting the megakaryocyte lineage, may contribute to OTT1-MAL-mediated leukemogenesis.
Collapse
Affiliation(s)
- Glen D. Raffel
- *Division of Hematology–Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
- Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
- To whom correspondence may be addressed at:
Division of Hematology–Oncology, SCC-9, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA 02215. E-mail:
| | - Thomas Mercher
- Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - Hirokazu Shigematsu
- Department of Cancer, Immunology, and AIDS, Dana–Farber Cancer Institute, Boston, MA 02115
| | - Ifor R. Williams
- Department of Pathology, Emory University School of Medicine, Atlanta, GA 30322
| | - Dana E. Cullen
- Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - Koichi Akashi
- Department of Cancer, Immunology, and AIDS, Dana–Farber Cancer Institute, Boston, MA 02115
| | - Olivier A. Bernard
- Institut National de la Santé et de la Recherche Médicale, E0210, 75743 Paris, France
- **Université René Descartes, 75270 Paris, France; and
| | - D. Gary Gilliland
- Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
- Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115
- To whom correspondence may be addressed at:
Division of Hematology, Brigham and Women's Hospital, 1 Blackfan Circle, Boston, MA 02115. E-mail:
| |
Collapse
|
25
|
Lutz J, Müller W, Jäck HM. VH replacement rescues progenitor B cells with two nonproductive VDJ alleles. THE JOURNAL OF IMMUNOLOGY 2007; 177:7007-14. [PMID: 17082616 DOI: 10.4049/jimmunol.177.10.7007] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Inaccurate VDJ rearrangements generate a large number of progenitor (pro)-B cells with two nonproductive IgH alleles. Such cells lack essential survival signals mediated by surface IgM heavy chain (muH chain) expression and are normally eliminated. However, secondary rearrangements of upstream VH gene segments into assembled VDJ exons have been described in mice transgenic for productive muH chains, a process known as VH replacement. If VH replacement was independent of muH chain signals, it could also modify nonproductive VDJ exons and thus rescue pro-B cells with unsuccessful rearrangements on both alleles. To test this hypothesis, we homologously replaced the JH cluster of a mouse with a nonproductive VDJ exon. Surprisingly, B cell development in IgHVDJ-/VDJ- mice was only slightly impaired and significant numbers of IgM-positive B cells were produced. DNA sequencing confirmed that all VDJ sequences from muH chain-positive B lymphoid cells were generated by VH replacement in a RAG-dependent manner. Another unique feature of our transgenic mice was the presence of IgH chains with unusually long CDR3-H regions. Such IgH chains were functional and only modestly counter-selected, arguing against a strict length constraint for CDR3-H regions. In conclusion, VH replacement can occur in the absence of a muH chain signal and provides a potential rescue mechanism for pro-B cells with two nonproductive IgH alleles.
Collapse
Affiliation(s)
- Johannes Lutz
- Division of Molecular Immunology, Department of Internal Medicine III, Nikolaus-Fiebiger-Center of Molecular Medicine, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | | | | |
Collapse
|
26
|
Pawlitzky I, Angeles CV, Siegel AM, Stanton ML, Riblet R, Brodeur PH. Identification of a candidate regulatory element within the 5' flanking region of the mouse Igh locus defined by pro-B cell-specific hypersensitivity associated with binding of PU.1, Pax5, and E2A. THE JOURNAL OF IMMUNOLOGY 2006; 176:6839-51. [PMID: 16709844 DOI: 10.4049/jimmunol.176.11.6839] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The Igh locus is controlled by cis-acting elements, including Emu and the 3' IgH regulatory region which flank the C region genes within the well-studied 3' part of the locus. Although the presence of additional control elements has been postulated to regulate rearrangements of the VH gene array that extends to the 5' end of the locus, the 5' border of Igh and its flanking region have not been characterized. To facilitate the analysis of this unexplored region and to identify potential novel control elements, we physically mapped the most D-distal VH segments and scanned 46 kb of the immediate 5' flanking region for DNase I hypersensitive sites. Our studies revealed a cluster of hypersensitive sites 30 kb upstream of the most 5' VH gene. Detection of one site, HS1, is restricted to pro-B cell lines and HS1 is accessible to restriction enzyme digestion exclusively in normal pro-B cells, the stage defined by actively rearranging Igh-V loci. Sequence motifs within HS1 for PU.1, Pax5, and E2A bind these proteins in vitro and these factors are recruited to HS1 sequence only in pro-B cells. Transient transfection assays indicate that the Pax5 binding site is required for the repression of transcriptional activity of HS1-containing constructs. Thus, our characterization of the region 5' of the VH gene cluster demonstrated the presence of a single cluster of DNase I hypersensitive sites within the 5' flanking region, and identified a candidate Igh regulatory region defined by pro-B cell-specific hypersensitivity and interaction with factors implicated in regulating VDJ recombination.
Collapse
Affiliation(s)
- Inka Pawlitzky
- Immunology Program, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111, USA
| | | | | | | | | | | |
Collapse
|
27
|
Jung D, Giallourakis C, Mostoslavsky R, Alt FW. Mechanism and control of V(D)J recombination at the immunoglobulin heavy chain locus. Annu Rev Immunol 2006; 24:541-70. [PMID: 16551259 DOI: 10.1146/annurev.immunol.23.021704.115830] [Citation(s) in RCA: 408] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
V(D)J recombination assembles antigen receptor variable region genes from component germline variable (V), diversity (D), and joining (J) gene segments. For B cells, such rearrangements lead to the production of immunoglobulin (Ig) proteins composed of heavy and light chains. V(D)J is tightly controlled at the Ig heavy chain locus (IgH) at several different levels, including cell-type specificity, intra- and interlocus ordering, and allelic exclusion. Such controls are mediated at the level of gene segment accessibility to V(D)J recombinase activity. Although much has been learned, many long-standing questions regarding the regulation of IgH locus rearrangements remain to be elucidated. In this review, we summarize advances that have been made in understanding how V(D)J recombination at the IgH locus is controlled and discuss important areas for future investigation.
Collapse
Affiliation(s)
- David Jung
- Howard Hughes Medical Institute, Children's Hospital, CBR Institute for Biomedical Research, and Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA.
| | | | | | | |
Collapse
|
28
|
Maës J, Chappaz S, Cavelier P, O'Neill L, Turner B, Rougeon F, Goodhardt M. Activation of V(D)J recombination at the IgH chain JH locus occurs within a 6-kilobase chromatin domain and is associated with nucleosomal remodeling. THE JOURNAL OF IMMUNOLOGY 2006; 176:5409-17. [PMID: 16622008 DOI: 10.4049/jimmunol.176.9.5409] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
IgH genes are assembled during early B cell development by a series of regulated DNA recombination reactions in which DH and JH segments are first joined followed by V(H) to DJH rearrangement. Recent studies have highlighted the role of chromatin structure in the control of V(D)J recombination. In this study, we show that, in murine pro-B cell precursors, the JH segments are located within a 6-kb DNase I-sensitive chromatin domain containing acetylated histones H3 and H4, which is delimited 5' by the DQ52 promoter element and 3' by the intronic enhancer. Within this domain, the JH segments are covered by phased nucleosomes. High-resolution mapping of nucleosomes reveals that, in pro-B cells, unlike recombination refractory nonlymphoid cells, the recombination signal sequences flanking the four JH segments are located in regions of enhanced micrococcal nuclease and restriction enzyme accessibility, corresponding to either nucleosome-free regions or DNA rendered accessible within a nucleosome. These results support the idea that nucleosome remodeling provides an additional level of control in the regulation of Ig locus accessibility to recombination factors in B cell precursors.
Collapse
Affiliation(s)
- Jérôme Maës
- Unité de Génétique et Biochimie du Développement, Unité de Recherche Associée Centre National de la Recherche Scientifique 1960, Département d'Immunologie, Institut Pasteur, Paris, France
| | | | | | | | | | | | | |
Collapse
|
29
|
Sen R, Oltz E. Genetic and epigenetic regulation of IgH gene assembly. Curr Opin Immunol 2006; 18:237-42. [PMID: 16616470 DOI: 10.1016/j.coi.2006.03.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2006] [Accepted: 03/27/2006] [Indexed: 12/19/2022]
Abstract
Precursor B cells assemble a diverse repertoire of immunoglobulin (Ig) genes by the process of V(D)J recombination. Assembly of IgH genes is regulated in a tissue- and stage-specific manner via the activation and then the inactivation of distinct regions within the one megabase IgH locus. Recent studies have shown that regional control is achieved using a combination of genetic and epigenetic strategies, which modulate chromatin accessibility to V(D)J recombinase, relocate IgH loci within the nucleus, and promote changes in locus conformation that alter the spatial proximity of target gene segments. Orchestration of these regulatory processes is crucial for the generation of a functional B cell repertoire.
Collapse
Affiliation(s)
- Ranjan Sen
- Laboratory of Cellular and Molecular Biology, National Institute on Aging/National Institutes of Health, 5600 Nathan Shock Drive, Baltimore, MD 21224, USA
| | | |
Collapse
|
30
|
Roessler S, Grosschedl R. Role of transcription factors in commitment and differentiation of early B lymphoid cells. Semin Immunol 2006; 18:12-9. [PMID: 16431127 DOI: 10.1016/j.smim.2005.12.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
B lymphopoiesis is a differentiation process in which hematopoietic stem cells are converted into antibody-producing plasma cells. B cell differentiation involves multiple steps, including cell specification, commitment to the B cell lineage, immunoglobulin rearrangements, maturation of B cells and terminal differentiation into plasma cells. Each of these steps is controlled by signaling pathways and transcription factors that act in synergy, feedback-loops or cross-antagonism to generate complex regulatory networks that allow for plasticity and stability of B cell differentiation.
Collapse
Affiliation(s)
- Stephanie Roessler
- Max-Planck Institute of Immunobiology, Department of Cellular and Molecular Immunology, Stubeweg 51, 79108 Freiburg, Germany
| | | |
Collapse
|
31
|
Abstract
RNA interference (RNAi) is an ancient and evolutionarily conserved process. In some eukaryotes, RNAi silences parasitic genetic elements. In plants, RNAi serves as an immune system against RNA viruses and transgenes and in worms, RNAi silences transposons. In mammals, RNAi has yet unknown functions. However, emerging roles for short RNAs and the factors that interact with them in other eukaryotes include chromatin modification, DNA deletion and DNA methylation, which may provide clues to the roles for short RNA function in mammals. For example, antigen receptor expression in lymphocytes is a highly regulated process and although much is known about chromatin modification and DNA deletion in the immune system, several molecular details of chromatin regulation remain elusive. This review compares emerging roles for short RNA function to processes required for antigen receptor expression in mammalian lymphocytes and predicts that short RNAs direct events required for successful lymphocyte-restricted gene expression.
Collapse
Affiliation(s)
- Dipanjan Chowdhury
- Center for Blood Research, Harvard Medical School, Boston, MA 02115, USA
| | | |
Collapse
|
32
|
Perlot T, Alt FW, Bassing CH, Suh H, Pinaud E. Elucidation of IgH intronic enhancer functions via germ-line deletion. Proc Natl Acad Sci U S A 2005; 102:14362-7. [PMID: 16186486 PMCID: PMC1242331 DOI: 10.1073/pnas.0507090102] [Citation(s) in RCA: 136] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Studies of chimeric mice demonstrated that the core Ig heavy chain (IgH) intronic enhancer (iEmu) functions in V(D)J and class switch recombination at the IgH locus. To more fully evaluate the role of this element in these and other processes, we generated mice homozygous for germ-line mutations in which the core sequences of iEmu (cEmu) were either deleted (cEmu(Delta/Delta) mice) or replaced with a pgk-Neo(R) cassette (cEmu(N/N) mice). The cEmu(Delta/Delta) mice had reduced B cell numbers, in association with impaired D to J(H) and V(H) to DJ(H) rearrangement, whereas cEmu(N/N) mice had a complete block in IgH V(D)J(H) recombination, confirming that additional cis elements cooperate with iEmu to enforce D to J(H) recombination. In addition, developing cEmu(Delta/Delta) and cEmu(N/N) B lineage cells had correspondingly decreased levels of germ-line transcripts from the J(H) region of the IgH locus (mu0 and Imu transcripts); although both had normal levels of germ-line V(H) transcripts, suggesting that cEmu may influence IgH locus V(D)J recombination by influencing accessibility of J(H) proximal regions of the locus. Consistent with chimera studies, peripheral cEmu(Delta/Delta) B cells had normal surface Ig and relatively normal class switch recombination. However, cEmu(Delta/Delta) B cells also had relatively normal somatic hypermutation of their IgH variable region genes, showing unexpectedly that the cEmu is not required for this process. The availability of mice with the iEmu mutation in their germ line will facilitate future studies to elucidate the roles of iEmu in V(H)(D)J(H) recombination in the context of IgH chromatin structure and germ-line transcription.
Collapse
Affiliation(s)
- Thomas Perlot
- The CBR Institute for Biomedical Research, Inc., and Department of Genetics, Harvard Medical School, 200 Longwood Avenue, Boston, MA 02115, USA
| | | | | | | | | |
Collapse
|
33
|
Abstract
Lymphocytes are characterised by monoclonal expression of antigen receptors. This is achieved by silencing of one of two homologous antigen receptor alleles, a process known as allelic exclusion. This process is regulated both before and after V(D)J recombination, by a variety of mechanisms. These include nuclear localisation, changes in chromatin structure and histone modifications, non-coding sense and antisense RNA transcription, epigenetic alterations at the DNA level, feedback signalling from expressed alleles, locus contraction and decontraction, recruitment to heterochromatin. This review will focus on recent advances in the immunoglobulin heavy and kappa light chain loci. The current picture is of a complex, temporally ordered sequence of events, in which these loci share many contributory mechanisms, but clear and intriguing differences are emerging.
Collapse
Affiliation(s)
- Anne E Corcoran
- Laboratory of Chromatin and Gene Expression, The Babraham Institute, Cambridge CB24AT, UK.
| |
Collapse
|
34
|
Abstract
When considering the daunting complexity of eukaryotic genomes, some comfort can be found in the fact that the human genome may contain only 30,000 to 40,000 genes. Moreover, growing evidence suggests that genomes may be organized in such a way as to take advantage of space. A gene's location in the linear DNA sequence and its position in the three-dimensional nucleus can both be important in its regulation. Contrary to prevailing notions in this postgenomic era, the bacteriophage lambda, a paragon of simplicity, may still have a few things to teach us with respect to these facets of nonrandom genomes.
Collapse
Affiliation(s)
- Steven T Kosak
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue North, Seattle, WA 98109, USA
| | | |
Collapse
|