1
|
Zhu B, Chen S, Bai Y, Chen H, Liao G, Mukherjee N, Vazquez G, McIlwain DR, Tzankov A, Lee IT, Matter MS, Goltsev Y, Ma Z, Nolan GP, Jiang S. Robust single-cell matching and multimodal analysis using shared and distinct features. Nat Methods 2023; 20:304-315. [PMID: 36624212 PMCID: PMC9911356 DOI: 10.1038/s41592-022-01709-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 10/31/2022] [Indexed: 01/10/2023]
Abstract
The ability to align individual cellular information from multiple experimental sources is fundamental for a systems-level understanding of biological processes. However, currently available tools are mainly designed for single-cell transcriptomics matching and integration, and generally rely on a large number of shared features across datasets for cell matching. This approach underperforms when applied to single-cell proteomic datasets due to the limited number of parameters simultaneously accessed and lack of shared markers across these experiments. Here, we introduce a cell-matching algorithm, matching with partial overlap (MARIO) that accounts for both shared and distinct features, while consisting of vital filtering steps to avoid suboptimal matching. MARIO accurately matches and integrates data from different single-cell proteomic and multimodal methods, including spatial techniques and has cross-species capabilities. MARIO robustly matched tissue macrophages identified from COVID-19 lung autopsies via codetection by indexing imaging to macrophages recovered from COVID-19 bronchoalveolar lavage fluid by cellular indexing of transcriptomes and epitopes by sequencing, revealing unique immune responses within the lung microenvironment of patients with COVID.
Collapse
Affiliation(s)
- Bokai Zhu
- grid.168010.e0000000419368956Department of Microbiology and Immunology, Stanford University, Stanford, CA USA ,grid.168010.e0000000419368956Department of Pathology, Stanford University, Stanford, CA USA
| | - Shuxiao Chen
- grid.25879.310000 0004 1936 8972Department of Statistics and Data Science, The Wharton School, University of Pennsylvania, PA, USA
| | - Yunhao Bai
- grid.168010.e0000000419368956Department of Pathology, Stanford University, Stanford, CA USA ,grid.168010.e0000000419368956Department of Chemistry, Stanford University, Stanford, CA USA
| | - Han Chen
- grid.168010.e0000000419368956Department of Pathology, Stanford University, Stanford, CA USA
| | - Guanrui Liao
- grid.239395.70000 0000 9011 8547Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA USA
| | - Nilanjan Mukherjee
- grid.168010.e0000000419368956Department of Pathology, Stanford University, Stanford, CA USA
| | - Gustavo Vazquez
- grid.168010.e0000000419368956Department of Pathology, Stanford University, Stanford, CA USA
| | - David R. McIlwain
- grid.168010.e0000000419368956Department of Pathology, Stanford University, Stanford, CA USA
| | - Alexandar Tzankov
- grid.6612.30000 0004 1937 0642Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Ivan T. Lee
- grid.168010.e0000000419368956Department of Pathology, Stanford University, Stanford, CA USA
| | - Matthias S. Matter
- grid.6612.30000 0004 1937 0642Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Yury Goltsev
- grid.168010.e0000000419368956Department of Pathology, Stanford University, Stanford, CA USA
| | - Zongming Ma
- Department of Statistics and Data Science, The Wharton School, University of Pennsylvania, PA, USA.
| | - Garry P. Nolan
- grid.168010.e0000000419368956Department of Pathology, Stanford University, Stanford, CA USA
| | - Sizun Jiang
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA. .,Department of Pathology, Dana Farber Cancer Institute, Boston, MA, USA. .,Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
2
|
Shinton SA, Brill-Dashoff J, Hayakawa K. Pla2g2a promotes innate Th2-type immunity lymphocytes to increase B1a cells. Sci Rep 2022; 12:14899. [PMID: 36050343 PMCID: PMC9437038 DOI: 10.1038/s41598-022-18876-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 08/22/2022] [Indexed: 11/09/2022] Open
Abstract
Newborns require early generation of effective innate immunity as a primary physiological mechanism for survival. The neonatal Lin28+Let7– developmental pathway allows increased generation of Th2-type cells and B1a (B-1 B) cells compared to adult cells and long-term maintenance of these initially generated innate cells. For initial B1a cell growth from the neonatal to adult stage, Th2-type IL-5 production from ILC2s and NKT2 cells is important to increase B1a cells. The Th17 increase is dependent on extracellular bacteria, and increased bacteria leads to lower Th2-type generation. Secreted group IIA-phospholipase A2 (sPLA2-IIA) from the Pla2g2a gene can bind to gram-positive bacteria and degrade bacterial membranes, controlling microbiota in the intestine. BALB/c mice are Pla2g2a+, and express high numbers of Th2-type cells and B1a cells. C57BL/6 mice are Pla2g2a-deficient and distinct from the SLAM family, and exhibit fewer NKT2 cells and fewer B1a cells from the neonatal to adult stage. We found that loss of Pla2g2a in the BALB/c background decreased IL-5 from Th2-type ILC2s and NKT2s but increased bacterial-reactive NKT17 cells and MAIT cells, and decreased the number of early-generated B1a cells and MZ B cells and the CD4/CD8 T cell ratio. Low IL-5 by decreased Th2-type cells in Pla2g2a loss led to low early-generated B1a cell growth from the neonatal to adult stage. In anti-thymocyte/Thy-1 autoreactive μκ transgenic (ATAμκ Tg) Pla2g2a+ BALB/c background C.B17 mice generated NKT2 cells that continuously control CD1d+ B1 B cells through old aging and lost CD1d in B1 B cells generating strong B1 ATA B cell leukemia/lymphoma. Pla2g2a-deficient ATAμκTg C57BL/6 mice suppressed the initial B1a cell increase, with low/negative spontaneous leukemia/lymphoma generation. These data confirmed that the presence of Pla2g2a to control bacteria is important to allow the neonatal to adult stage. Pla2g2a promotes innate Th2-type immunity lymphocytes to increase early generated B1a cells.
Collapse
Affiliation(s)
- Susan A Shinton
- Fox Chase Cancer Center, 333 Cottman Ave., Philadelphia, PA, 19111, USA
| | | | - Kyoko Hayakawa
- Fox Chase Cancer Center, 333 Cottman Ave., Philadelphia, PA, 19111, USA.
| |
Collapse
|
3
|
Regulation of the BCR signalosome by the class II peptide editor, H2-M, affects the development and repertoire of innate-like B cells. Cell Rep 2022; 38:110200. [DOI: 10.1016/j.celrep.2021.110200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 09/23/2021] [Accepted: 12/13/2021] [Indexed: 11/21/2022] Open
|
4
|
Govek KW, Troisi EC, Miao Z, Aubin RG, Woodhouse S, Camara PG. Single-cell transcriptomic analysis of mIHC images via antigen mapping. SCIENCE ADVANCES 2021; 7:eabc5464. [PMID: 33674303 PMCID: PMC7935366 DOI: 10.1126/sciadv.abc5464] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 01/21/2021] [Indexed: 06/12/2023]
Abstract
Highly multiplexed immunohistochemistry (mIHC) enables the staining and quantification of dozens of antigens in a tissue section with single-cell resolution. However, annotating cell populations that differ little in the profiled antigens or for which the antibody panel does not include specific markers is challenging. To overcome this obstacle, we have developed an approach for enriching mIHC images with single-cell RNA sequencing data, building upon recent experimental procedures for augmenting single-cell transcriptomes with concurrent antigen measurements. Spatially-resolved Transcriptomics via Epitope Anchoring (STvEA) performs transcriptome-guided annotation of highly multiplexed cytometry datasets. It increases the level of detail in histological analyses by enabling the systematic annotation of nuanced cell populations, spatial patterns of transcription, and interactions between cell types. We demonstrate the utility of STvEA by uncovering the architecture of poorly characterized cell types in the murine spleen using published cytometry and mIHC data of this organ.
Collapse
Affiliation(s)
- Kiya W Govek
- Department of Genetics and Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, 3700 Hamilton Walk, Philadelphia, PA 19104, USA
| | - Emma C Troisi
- Department of Genetics and Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, 3700 Hamilton Walk, Philadelphia, PA 19104, USA
| | - Zhen Miao
- Department of Genetics and Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, 3700 Hamilton Walk, Philadelphia, PA 19104, USA
| | - Rachael G Aubin
- Department of Genetics and Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, 3700 Hamilton Walk, Philadelphia, PA 19104, USA
| | - Steven Woodhouse
- Department of Genetics and Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, 3700 Hamilton Walk, Philadelphia, PA 19104, USA
| | - Pablo G Camara
- Department of Genetics and Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, 3700 Hamilton Walk, Philadelphia, PA 19104, USA.
| |
Collapse
|
5
|
Yang Y, Li X, Ma Z, Wang C, Yang Q, Byrne-Steele M, Hong R, Min Q, Zhou G, Cheng Y, Qin G, Youngyunpipatkul JV, Wing JB, Sakaguchi S, Toonstra C, Wang LX, Vilches-Moure JG, Wang D, Snyder MP, Wang JY, Han J, Herzenberg LA. CTLA-4 expression by B-1a B cells is essential for immune tolerance. Nat Commun 2021; 12:525. [PMID: 33483505 PMCID: PMC7822855 DOI: 10.1038/s41467-020-20874-x] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 12/17/2020] [Indexed: 01/11/2023] Open
Abstract
CTLA-4 is an important regulator of T-cell function. Here, we report that expression of this immune-regulator in mouse B-1a cells has a critical function in maintaining self-tolerance by regulating these early-developing B cells that express a repertoire enriched for auto-reactivity. Selective deletion of CTLA-4 from B cells results in mice that spontaneously develop autoantibodies, T follicular helper (Tfh) cells and germinal centers (GCs) in the spleen, and autoimmune pathology later in life. This impaired immune homeostasis results from B-1a cell dysfunction upon loss of CTLA-4. Therefore, CTLA-4-deficient B-1a cells up-regulate epigenetic and transcriptional activation programs and show increased self-replenishment. These activated cells further internalize surface IgM, differentiate into antigen-presenting cells and, when reconstituted in normal IgH-allotype congenic recipient mice, induce GCs and Tfh cells expressing a highly selected repertoire. These findings show that CTLA-4 regulation of B-1a cells is a crucial immune-regulatory mechanism.
Collapse
Affiliation(s)
- Yang Yang
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.
| | - Xiao Li
- The Center for RNA Science and Therapeutics, Case Western Reserve University, Cleveland, OH, USA
| | - Zhihai Ma
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | | | | | | | - Rongjian Hong
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Qing Min
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Gao Zhou
- The Center for RNA Science and Therapeutics, Case Western Reserve University, Cleveland, OH, USA
| | - Yong Cheng
- St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Guang Qin
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | | | - James B Wing
- Laboratory of Human Immunology (Single Cell Immunology), World Premier International Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Laboratory of Experimental Immunology, World Premier International Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Shimon Sakaguchi
- Laboratory of Experimental Immunology, World Premier International Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Christian Toonstra
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD, USA
| | - Lai-Xi Wang
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD, USA
| | - Jose G Vilches-Moure
- Department of Comparative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Denong Wang
- Tumor Glycomics Laboratory, SRI International Biosciences Division, Menlo Park, CA, USA
| | - Michael P Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Ji-Yang Wang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Department of Clinical Immunology, Children's Hospital of Fudan University, Shanghai, China
- Department of Microbiology and Immunology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Jian Han
- iRepertoire Inc, Huntsville, AL, USA
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
| | - Leonore A Herzenberg
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
6
|
Tsuji N, Rothstein TL, Holodick NE. Antigen Receptor Specificity and Cell Location Influence the Diversification and Selection of the B-1a Cell Pool with Age. THE JOURNAL OF IMMUNOLOGY 2020; 205:741-759. [PMID: 32561570 DOI: 10.4049/jimmunol.1901302] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 05/20/2020] [Indexed: 01/18/2023]
Abstract
B-1a cells provide immediate and essential protection from infection through production of natural Ig, which is germline-like due to minimal insertion of N region additions. We have previously demonstrated peritoneal B-1a cell-derived phosphorylcholine-specific and total IgM moves away from germline (as evidenced by an increase in N-additions) with age as a result of selection. In young mice, anti-phosphatidylcholine Abs, like anti-phosphorylcholine Abs, contain few N-additions, and have been shown to be essential in protection from bacterial sepsis. In this study, we demonstrate the germline-like status of phosphatidylcholine (PtC)-specific (PtC+) peritoneal B-1a cell IgM does not change with age. In direct contrast, the splenic PtC+ B-1a cell population does not preserve its IgM germline status in the aged mice. Furthermore, splenic PtC+ B-1a cells displayed more diverse variable gene segments of the H chain (VH) use in both the young and aged mice as compared with peritoneal PtC+ B-1a cells. Whereas the peritoneal PtC+ population increased VH12 use with age, we observed differential use of VH11, VH12, and VH2 between the peritoneal and splenic PtC+ populations with age. These results suggest disparate selection pressures occur with age upon B-1a cells expressing different specificities in distinct locations. Overall, these results illuminate the need to further elucidate how B-1a cells are influenced over time in terms of production and selection, both of which contribute to the actual and available natural IgM repertoire with increasing age. Such studies would aid in the development of more effective vaccination and therapeutic strategies in the aged population.
Collapse
Affiliation(s)
- Naomi Tsuji
- Center for Immunobiology, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI 49007; and Department of Investigative Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI 49007
| | - Thomas L Rothstein
- Center for Immunobiology, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI 49007; and Department of Investigative Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI 49007
| | - Nichol E Holodick
- Center for Immunobiology, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI 49007; and Department of Investigative Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI 49007
| |
Collapse
|
7
|
Honjo K, Won WJ, King RG, Ianov L, Crossman DK, Easlick JL, Shakhmatov MA, Khass M, Vale AM, Stephan RP, Li R, Davis RS. Fc Receptor-Like 6 (FCRL6) Discloses Progenitor B Cell Heterogeneity That Correlates With Pre-BCR Dependent and Independent Pathways of Natural Antibody Selection. Front Immunol 2020; 11:82. [PMID: 32117244 PMCID: PMC7033751 DOI: 10.3389/fimmu.2020.00082] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 01/13/2020] [Indexed: 11/24/2022] Open
Abstract
B-1a cells produce "natural" antibodies (Abs) to neutralize pathogens and clear neo self-antigens, but the fundamental selection mechanisms that shape their polyreactive repertoires are poorly understood. Here, we identified a B cell progenitor subset defined by Fc receptor-like 6 (FCRL6) expression, harboring innate-like defense, migration, and differentiation properties conducive for natural Ab generation. Compared to FCRL6- pro B cells, the repressed mitotic, DNA damage repair, and signaling activity of FCRL6+ progenitors, yielded VH repertoires with biased distal Ighv segment accessibility, constrained diversity, and hydrophobic and charged CDR-H3 sequences. Beyond nascent autoreactivity, VH11 productivity, which predominates phosphatidylcholine-specific B-1a B cell receptors (BCRs), was higher for FCRL6+ cells as was pre-BCR formation, which was required for Myc induction and VH11, but not VH12, B-1a development. Thus, FCRL6 revealed unexpected heterogeneity in the developmental origins, regulation, and selection of natural Abs at the pre-BCR checkpoint with implications for autoimmunity and lymphoproliferative disorders.
Collapse
MESH Headings
- Animals
- Antibodies/immunology
- Antibodies/metabolism
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Female
- Humans
- Immunoglobulin Heavy Chains/genetics
- Immunoglobulin Heavy Chains/immunology
- Immunoglobulin Variable Region/genetics
- Immunoglobulin Variable Region/immunology
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Phosphatidylcholines/immunology
- Phosphatidylcholines/metabolism
- Precursor Cells, B-Lymphoid/immunology
- Precursor Cells, B-Lymphoid/metabolism
- Receptors, Antigen, B-Cell/genetics
- Receptors, Antigen, B-Cell/immunology
- Receptors, Antigen, B-Cell/metabolism
- Receptors, Fc/genetics
- Receptors, Fc/immunology
- Receptors, Fc/metabolism
- Signal Transduction/genetics
- Signal Transduction/immunology
Collapse
Affiliation(s)
- Kazuhito Honjo
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Woong-Jai Won
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Rodney G. King
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Lara Ianov
- Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - David K. Crossman
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Juliet L. Easlick
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Mikhail A. Shakhmatov
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Mohamed Khass
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Genetic Engineering and Biotechnology Division, National Research Center, Cairo, Egypt
| | - Andre M. Vale
- Program in Immunobiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Robert P. Stephan
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Ran Li
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Randall S. Davis
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, United States
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
8
|
Hayakawa K, Li YS, Shinton SA, Bandi SR, Formica AM, Brill-Dashoff J, Hardy RR. Crucial Role of Increased Arid3a at the Pre-B and Immature B Cell Stages for B1a Cell Generation. Front Immunol 2019; 10:457. [PMID: 30930899 PMCID: PMC6428705 DOI: 10.3389/fimmu.2019.00457] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 02/20/2019] [Indexed: 02/03/2023] Open
Abstract
The Lin28b+Let7− axis in fetal/neonatal development plays a role in promoting CD5+ B1a cell generation as a B-1 B cell developmental outcome. Here we identify the Let7 target, Arid3a, as a crucial molecular effector of the B-1 cell developmental program. Arid3a expression is increased at pro-B cell stage and markedly increased at pre-B and immature B cell stages in the fetal/neonatal liver B-1 development relative to that in the Lin28b−Let7+ adult bone marrow (BM) B-2 cell development. Analysis of B-lineage restricted Lin28b transgenic (Tg) mice, Arid3a knockout and Arid3a Tg mice, confirmed that increased Arid3a allows B cell generation without requiring surrogate light chain (SLC) associated pre-BCR stage, and prevents MHC class II cell expression at the pre-B and newly generated immature B cell stages, distinct from pre-BCR dependent B development with MHC class II in adult BM. Moreover, Arid3a plays a crucial role in supporting B1a cell generation. The increased Arid3a leads higher Myc and Bhlhe41, and lower Siglec-G and CD72 at the pre-B and immature B cell stages than normal adult BM, to allow BCR signaling induced B1a cell generation. Arid3a-deficiency selectively blocks the development of B1a cells, while having no detectable effect on CD5− B1b, MZ B, and FO B cell generation resembling B-2 development outcome. Conversely, enforced expression of Arid3a by transgene is sufficient to promote the development of B1a cells from adult BM. Under the environment change between birth to adult, altered BCR repertoire in increased B1a cells occurred generated from adult BM. However, crossed with B1a-restricted VH/D/J IgH knock-in mice allowed to confirm that SLC-unassociated B1a cell increase and CLL/lymphoma generation can occur in aged from Arid3a increased adult BM. These results confirmed that in fetal/neonatal normal mice, increased Arid3a at the pre-B cell and immature B cell stages is crucial for generating B1a cells together with the environment for self-ligand reactive BCR selection, B1a cell maintenance, and potential for development of CLL/Lymphoma in aged mice.
Collapse
Affiliation(s)
- Kyoko Hayakawa
- Fox Chase Cancer Center, Philadelphia, PA, United States
| | - Yue-Sheng Li
- Fox Chase Cancer Center, Philadelphia, PA, United States
| | | | | | | | | | | |
Collapse
|
9
|
Hayakawa K, Formica AM, Nakao Y, Ichikawa D, Shinton SA, Brill-Dashoff J, Smith MR, Morse HC, Hardy RR. Early Generated B-1-Derived B Cells Have the Capacity To Progress To Become Mantle Cell Lymphoma-like Neoplasia in Aged Mice. THE JOURNAL OF IMMUNOLOGY 2018; 201:804-813. [PMID: 29898964 DOI: 10.4049/jimmunol.1800400] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 05/16/2018] [Indexed: 11/19/2022]
Abstract
In mice, fetal/neonatal B-1 cell development generates murine CD5+ B cells (B1a) with autoreactivity. We analyzed B1a cells at the neonatal stage in a VH11/D/JH knock-in mouse line (VH11t) that generates an autoreactive antiphosphatidylcholine BCR. Our study revealed that antiphosphatidylcholine B1a cells develop in liver, mature in spleen, and distribute in intestine/colon, mesenteric lymph node (mLN), and body cavity as the outcome of B-1 cell development before B-2 cell development. Throughout life, self-renewing B-1 B1a cells circulate through intestine, mesenteric vessel, and blood. The body cavity-deposited B1a cells also remigrate. In old age, some B1a cells proceed to monoclonal B cell lymphocytosis. When neonatal B-1 B1a cells express an antithymocyte/Thy-1 autoreactivity (ATA) BCR transgene in the C.B17 mouse background, ATA B cells increase in PBL and strongly develop lymphomas in aging mice that feature splenomegaly and mLN hyperplasia with heightened expression of CD11b, IL-10, and activated Stat3. At the adult stage, ATA B cells were normally present in the mantle zone area, including in intestine. Furthermore, frequent association with mLN hyperplasia suggests the influence by intestinal microenvironment on lymphoma development. When cyclin D1 was overexpressed by the Eμ-cyclin D1 transgene, ATA B cells progressed to further diffused lymphoma in aged mice, including in various lymph nodes with accumulation of IgMhiIgDloCD5+CD23-CD43+ cells, resembling aggressive human mantle cell lymphoma. Thus, our findings reveal that early generated B cells, as an outcome of B-1 cell development, can progress to become lymphocytosis, lymphoma, and mantle cell lymphoma-like neoplasia in aged mice.
Collapse
Affiliation(s)
| | | | - Yuka Nakao
- Fox Chase Cancer Center, Philadelphia, PA 19111
| | - Daiju Ichikawa
- Fox Chase Cancer Center, Philadelphia, PA 19111.,Division of Clinical Physiology and Therapeutics, Keio University Faculty of Pharmacy, Tokyo 105-8512, Japan
| | | | | | - Mitchell R Smith
- Fox Chase Cancer Center, Philadelphia, PA 19111.,George Washington University Cancer Center, Washington, DC 20052; and
| | - Herbert C Morse
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852
| | | |
Collapse
|
10
|
Chaparro M, Verreth A, Lobaton T, Gravito-Soares E, Julsgaard M, Savarino E, Magro F, Biron AI, Lopez-Serrano P, Casanova MJ, Gompertz M, Vitor S, Arroyo M, Pugliese D, Zabana Y, Vicente R, Aguas M, Shitrit BGA, Gutierrez A, Doherty GA, Fernandez-Salazar L, Cadilla MJ, Huguet JM, OʼToole A, Stasi E, Marcos MN, Villoria A, Karmiris K, Rahier JF, Rodriguez C, Palomares DLM, Fiorino G, Benitez JM, Principi M, Naftali T, Taxonera C, Mantzaris G, Sebkova L, Iade B, Lissner D, Bradley FI, Roman LSA, Marin-Jimenez I, Merino O, Sierra M, Van Domselaar M, Caprioli F, Guerra I, Peixe P, Piqueras M, Rodriguez-Lago I, Ber Y, van Hoeve K, Torres P, Gravito-Soares M, Rudbeck-Resdal D, Bartolo O, Peixoto A, Martin G, Armuzzi A, Garre A, Donday MG, de Carpi MFJ, Gisbert JP. Long-Term Safety of In Utero Exposure to Anti-TNFα Drugs for the Treatment of Inflammatory Bowel Disease: Results from the Multicenter European TEDDY Study. Am J Gastroenterol 2018; 113:396-403. [PMID: 29460920 DOI: 10.1038/ajg.2017.501] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Accepted: 11/26/2017] [Indexed: 12/11/2022]
Abstract
OBJECTIVES The long-term safety of exposure to anti-tumor necrosis factor (anti-TNFα) drugs during pregnancy has received little attention. We aimed to compare the relative risk of severe infections in children of mothers with inflammatory bowel disease (IBD) who were exposed to anti-TNFα drugs in utero with that of children who were not exposed to the drugs. METHODS Retrospective multicenter cohort study. Exposed cohort: children from mothers with IBD receiving anti-TNFα medication (with or without thiopurines) at any time during pregnancy or during the 3 months before conception. Non-exposed cohort: children from mothers with IBD not treated with anti-TNFα agents or thiopurines at any time during pregnancy or the 3 months before conception. The cumulative incidence of severe infections after birth was estimated using Kaplan-Meier curves, which were compared using the log-rank test. Cox-regression analysis was performed to identify potential predictive factors for severe infections in the offspring. RESULTS The study population comprised 841 children, of whom 388 (46%) had been exposed to anti-TNFα agents. Median follow-up after delivery was 47 months in the exposed group and 68 months in the non-exposed group. Both univariate and multivariate analysis showed the incidence rate of severe infections to be similar in non-exposed and exposed children (1.6% vs. 2.8% per person-year, hazard ratio 1.2 (95% confidence interval 0.8-1.8)). In the multivariate analysis, preterm delivery was the only variable associated with a higher risk of severe infection (2.5% (1.5-4.3)). CONCLUSIONS In utero exposure to anti-TNFα drugs does not seem to be associated with increased short-term or long-term risk of severe infections in children.
Collapse
Affiliation(s)
- M Chaparro
- Gastroenterology Units Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP) and Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - A Verreth
- Department of Gastroenterology and Department of Pediatric Gastroenterology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - T Lobaton
- Hospital Universitari Germans Trias i Pujol and CIBEREHD, Badalona, Spain
| | | | - M Julsgaard
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus, Denmark
| | | | - F Magro
- Centro Hospitalar São João, Porto, Portugal
| | - Avni I Biron
- Gastroenterology Devision, Rabin Medical Center, Petach Tikva, Israel
| | | | - M J Casanova
- Gastroenterology Units Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP) and Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - M Gompertz
- Hospital Clinic and CIBEREHD, Barcelona, Spain
| | - S Vitor
- Hospital de Santa Maria - Centro Hospitalar Lisboa Norte, Lisboa, Portugal
| | - M Arroyo
- Hospital Clinico Universitario Lozano Blesa, IIS Aragon, CIBEREHD, Zaragoza, Spain
| | - D Pugliese
- IBD Unit, Presidio Columbus, Fondazione Policlinico Gemelli Università Cattolica, Roma, Italy
| | - Y Zabana
- Hospital Universitari Mutua de Terrassa and CIBEREHD, Terrassa, Spain
| | - R Vicente
- Hospital Universitario Miguel Servet, Zaragoza, Spain
| | - M Aguas
- Hospital Universitario La Fe and CIBEREHD, Valencia, Spain
| | | | - A Gutierrez
- Hospital General Universitario de Alicante and Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Alicante, Spain
| | - G A Doherty
- St. Vincents University Hospital, Dublin, Ireland
| | | | | | - J M Huguet
- Hospital General Universitario de Valencia, Valencia, Spain
| | | | - E Stasi
- IRCCS Saverio de Bellis, Castellana Grotte, Italy
| | | | - A Villoria
- Hospital Universitari Parc Taulí.Institut d'Investigació i Innovació Parc Taulí. Departament de Medicina, Universitat Autònoma de Barcelona.CIBERehd, Instituto de Salud Carlos III, Sabadell, Spain
| | - K Karmiris
- Venizeleio General Hospital, Heraklion, Greece
| | | | - C Rodriguez
- Complejo Universitario de Navarra, Pamplona, Spain
| | | | - G Fiorino
- IBD Center, Humanitas Clinical and Research Institute, Rozzano, Milan, Italy and Department of Biomedical Sciences, Humanitas University, Rozzano, Milan, Italy
| | - J M Benitez
- Hospital Universitario Reina Sofia and IMIBIC, Córdoba, Spain
| | - M Principi
- Azienda Policlinico Ospedaliero-Universitaria di Bari, Bari, Italy
| | - T Naftali
- Meir Hospital Kfar saba Tel Aviv University, Tel Aviv, Israel
| | - C Taxonera
- Hospital Clínico San Carlos and IdISSC, Madrid, Spain
| | - G Mantzaris
- Evangelismos, Ophthalmiatreion Athinon and Polyclinic Hospitals, Athens, Greece
| | - L Sebkova
- Azienda Ospedaliera "Pugliese-Ciaccio", Catanzaro, Italy
| | - B Iade
- Hospital de Clinicas, Montevideo, Uruguay
| | - D Lissner
- Universitatsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | | | | | - I Marin-Jimenez
- Hospital General Universitario Gregorio Marañón and IiSGM, Madrid, Spain
| | - O Merino
- Hospital Universitario de Cruces, Baracaldo, Spain
| | - M Sierra
- Complejo Universitario de León, León, Spain
| | | | - F Caprioli
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Cà Granda, Ospedale Policlinico di Milano AND Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - I Guerra
- Hospital Universitario de Fuenlabrada, Fuenlabrada, Spain
| | - P Peixe
- Centro Hospitalar Lisboa Ocidental, Lisboa, Portugal
| | - M Piqueras
- Consorci Sanitari de Terrasa, Terrasa, Spain
| | | | - Y Ber
- Hospital San Jorge, Huesca, Spain
| | - K van Hoeve
- Department of Paediatrics, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - P Torres
- Hospital Universitari Germans Trias i Pujol and CIBEREHD, Badalona, Spain
| | | | - D Rudbeck-Resdal
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus, Denmark
| | | | - A Peixoto
- Centro Hospitalar São João, Porto, Portugal
| | - G Martin
- Gastroenterology Devision, Rabin Medical Center, Petach Tikva, Israel
| | - A Armuzzi
- IBD Unit, Presidio Columbus, Fondazione Policlinico Gemelli Università Cattolica, Roma, Italy
| | - A Garre
- Gastroenterology Units Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP) and Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - M G Donday
- Gastroenterology Units Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP) and Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | | | - J P Gisbert
- Gastroenterology Units Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP) and Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| |
Collapse
|
11
|
Zhiming W, Luman W, Tingting Q, Yiwei C. Chemokines and receptors in intestinal B lymphocytes. J Leukoc Biol 2018; 103:807-819. [PMID: 29443417 DOI: 10.1002/jlb.1ru0717-299rr] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Revised: 01/11/2018] [Accepted: 01/11/2018] [Indexed: 02/06/2023] Open
Abstract
Recent studies indicate that chemoattractant cytokines (chemokines) and their receptors modulate intestinal B lymphocytes in different ways, including regulating their maturity and differentiation in the bone marrow and homing to intestinal target tissues. Here, we review several important chemokine/chemokine receptor axes that guide intestinal B cells, focusing on the homing and migration of IgA antibody-secreting cells (IgA-ASCs) to intestinal-associated lymphoid tissues. We describe the selective regulation of these chemokine axes in coordinating the IgA-ASC trafficking in intestinal diseases. Finally, we discuss the role of B cells as chemokine producers serving dual roles in regulating the mucosal immune microenvironment.
Collapse
Affiliation(s)
- Wang Zhiming
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Wang Luman
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China.,Biotherapy Research Center, Fudan University, Shanghai, China
| | - Qian Tingting
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Chu Yiwei
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China.,Biotherapy Research Center, Fudan University, Shanghai, China
| |
Collapse
|
12
|
Mariette X, Förger F, Abraham B, Flynn AD, Moltó A, Flipo RM, van Tubergen A, Shaughnessy L, Simpson J, Teil M, Helmer E, Wang M, Chakravarty EF. Lack of placental transfer of certolizumab pegol during pregnancy: results from CRIB, a prospective, postmarketing, pharmacokinetic study. Ann Rheum Dis 2018; 77:228-233. [PMID: 29030361 PMCID: PMC5867410 DOI: 10.1136/annrheumdis-2017-212196] [Citation(s) in RCA: 246] [Impact Index Per Article: 35.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 09/22/2017] [Accepted: 10/01/2017] [Indexed: 12/31/2022]
Abstract
OBJECTIVES There is a need for effective and safe treatment during pregnancy in women with chronic inflammatory diseases. This study evaluated placental transfer of certolizumab pegol (CZP), an Fc-free anti-tumour necrosis factor drug, from CZP-treated pregnant women to their infants. METHODS CRIB was a pharmacokinetic (PK) study of women ≥30 weeks pregnant receiving commercial CZP for a locally approved indication (last dose ≤35 days prior to delivery). Blood samples were collected from mothers, umbilical cords and infants at delivery, and infants again at weeks 4 and 8 post-delivery. CZP plasma concentrations were measured with a highly sensitive and CZP-specific electrochemiluminescence immunoassay (lower limit of quantification 0.032 μg/mL). RESULTS Sixteen women entered and completed the study. Maternal CZP plasma levels at delivery were within the expected therapeutic range (median [range] 24.4 [5.0-49.4] μg/mL). Of the 16 infants, 2 were excluded from the per-protocol set: 1 due to missing data at birth and 1 due to implausible PK data. Of the remaining 14 infants, 13 had no quantifiable CZP levels at birth (<0.032 μg/mL), and 1 had a minimal CZP level of 0.042 μg/mL (infant/mother plasma ratio 0.0009); no infants had quantifiable CZP levels at weeks 4 and 8. Of 16 umbilical cord samples, 1 was excluded due to missing data; 3/15 had quantifiable CZP levels (maximum 0.048 μg/mL). CONCLUSIONS There was no to minimal placental transfer of CZP from mothers to infants, suggesting lack of in utero foetal exposure during the third trimester. These results support continuation of CZP treatment during pregnancy, when considered necessary. TRIAL REGISTRATION NUMBER NCT02019602; Results.
Collapse
Affiliation(s)
- Xavier Mariette
- Université Paris-Sud, Hôpitaux Universitaires Paris-Sud, INSERM, Le Kremlin-Bicêtre, France
| | - Frauke Förger
- Inselspital, University Hospital and University of Bern, Bern, Switzerland
| | | | - Ann D Flynn
- University of Utah Health, Salt Lake City, Utah, USA
| | - Anna Moltó
- Department of Rheumatology, Hôpital Cochin, Assistance Publique – Hôpitaux de Paris, INSERM, Paris, France
| | - René-Marc Flipo
- Centre Hospitalier Regional Universitaire de Lille, Lille, Nord-Pas-de-Calais, France
| | - Astrid van Tubergen
- Department of Medicine, Division of Rheumatology and CAPHRI - Care and Public Health Research Institute Maastricht, Maastricht University Medical Center, Maastricht, Netherlands
| | | | | | | | | | | | | |
Collapse
|
13
|
Shukla V, Shukla A, Joshi SS, Lu R. Interferon regulatory factor 4 attenuates Notch signaling to suppress the development of chronic lymphocytic leukemia. Oncotarget 2018; 7:41081-41094. [PMID: 27232759 PMCID: PMC5173044 DOI: 10.18632/oncotarget.9596] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 05/14/2016] [Indexed: 11/25/2022] Open
Abstract
Molecular pathogenesis of Chronic Lymphocytic Leukemia (CLL) is not fully elucidated. Genome wide association studies have linked Interferon Regulatory Factor 4 (IRF4) to the development of CLL. We recently established a causal relationship between low levels of IRF4 and development of CLL. However, the molecular mechanism through which IRF4 suppresses CLL development remains unclear. Deregulation of Notch signaling pathway has been identified as one of the most recurrent molecular anomalies in the pathogenesis of CLL. Yet, the role of Notch signaling as well as its regulation during CLL development remains poorly understood. Previously, we demonstrated that IRF4 deficient mice expressing immunoglobulin heavy chain Vh11 (IRF4−/−Vh11) developed spontaneous CLL with complete penetrance. In this study, we show that elevated Notch2 expression and the resulting hyperactivation of Notch signaling are common features of IRF4−/−Vh11 CLL cells. Our studies further reveal that Notch signaling is indispensable for CLL development in the IRF4−/−Vh11 mice. Moreover, we identify E3 ubiquitin ligase Nedd4, which targets Notch for degradation, as a direct target of IRF4 in CLL cells and their precursors. Collectively, our studies provide the first in vivo evidence for an essential role of Notch signaling in the development of CLL and establish IRF4 as a critical regulator of Notch signaling during CLL development.
Collapse
Affiliation(s)
- Vipul Shukla
- Department of Genetics Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Ashima Shukla
- Department of Genetics Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Shantaram S Joshi
- Department of Genetics Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Runqing Lu
- Department of Genetics Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
14
|
Hayakawa K, Formica AM, Zhou Y, Ichikawa D, Asano M, Li YS, Shinton SA, Brill-Dashoff J, Núñez G, Hardy RR. NLR Nod1 signaling promotes survival of BCR-engaged mature B cells through up-regulated Nod1 as a positive outcome. J Exp Med 2017; 214:3067-3083. [PMID: 28878001 PMCID: PMC5626402 DOI: 10.1084/jem.20170497] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 06/11/2017] [Accepted: 07/24/2017] [Indexed: 11/04/2022] Open
Abstract
The microenvironment, including microbial products, plays a role in mature B cell survival. Hayakawa et al. show that B cell antigen receptor ligand–mediated Nod1 up-regulation in vivo in B cell development leads to preferential mature B cell survival as a competitive survival, increasing the Nod1+ B cell pool with age. Although B cell development requires expression of the B cell antigen receptor (BCR), it remains unclear whether engagement of self-antigen provides a positive impact for most B cells. Here, we show that BCR engagement by self-ligand during development in vivo results in up-regulation of the Nod-like receptor member Nod1, which recognizes the products of intestinal commensal bacteria. In anti-thymocyte/Thy-1 autoreactive BCR knock-in mice lacking self–Thy-1 ligand, immunoglobulin light chain editing occurred, generating B cells with up-regulated Nod1, including follicular and marginal zone B cells with natural autoreactivity. This BCR editing with increased Nod1 resulted in preferential survival. In normal adult mice, most mature B cells are enriched for Nod1 up-regulated cells, and signaling through Nod1 promotes competitive survival of mature B cells. These findings demonstrate a role for microbial products in promoting survival of mature B cells through up-regulated Nod1, providing a positive effect of BCR engagement on development of most B cells.
Collapse
Affiliation(s)
| | | | - Yan Zhou
- Fox Chase Cancer Center, Philadelphia, PA
| | | | | | | | | | | | - Gabriel Núñez
- Department of Pathology and Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI
| | | |
Collapse
|
15
|
Hayakawa K, Formica AM, Brill-Dashoff J, Shinton SA, Ichikawa D, Zhou Y, Morse HC, Hardy RR. Early generated B1 B cells with restricted BCRs become chronic lymphocytic leukemia with continued c-Myc and low Bmf expression. J Exp Med 2016; 213:3007-3024. [PMID: 27899442 PMCID: PMC5154941 DOI: 10.1084/jem.20160712] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 09/01/2016] [Accepted: 10/21/2016] [Indexed: 01/22/2023] Open
Abstract
Hayakawa et al. show that distinctive B-lineage progression from B-1 development allows for generation of B1a cells with restricted BCRs and self-renewal capacity, both contributing to potential for CLL progression. In mice, generation of autoreactive CD5+ B cells occurs as a consequence of BCR signaling induced by (self)-ligand exposure from fetal/neonatal B-1 B cell development. A fraction of these cells self-renew and persist as a minor B1 B cell subset throughout life. Here, we show that transfer of early generated B1 B cells from Eμ-TCL1 transgenic mice resulted in chronic lymphocytic leukemia (CLL) with a biased repertoire, including stereotyped BCRs. Thus, B1 B cells bearing restricted BCRs can become CLL during aging. Increased anti-thymocyte/Thy-1 autoreactive (ATA) BCR cells in the B1 B cell subset by transgenic expression yielded spontaneous ATA B-CLL/lymphoma incidence, enhanced by TCL1 transgenesis. In contrast, ATA B-CLL did not develop from other B cell subsets, even when the identical ATA BCR was expressed on a Thy-1 low/null background. Thus, both a specific BCR and B1 B cell context were important for CLL progression. Neonatal B1 B cells and their CLL progeny in aged mice continued to express moderately up-regulated c-Myc and down-regulated proapoptotic Bmf, unlike most mature B cells in the adult. Thus, there is a genetic predisposition inherent in B-1 development generating restricted BCRs and self-renewal capacity, with both features contributing to potential for progression to CLL.
Collapse
Affiliation(s)
| | | | | | | | | | - Yan Zhou
- Fox Chase Cancer Center, Philadelphia, PA 19111
| | - Herbert C Morse
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852
| | | |
Collapse
|
16
|
Hayakawa K, Formica AM, Colombo MJ, Shinton SA, Brill-Dashoff J, Morse HC, Li YS, Hardy RR. Loss of a chromosomal region with synteny to human 13q14 occurs in mouse chronic lymphocytic leukemia that originates from early-generated B-1 B cells. Leukemia 2016; 30:1510-9. [PMID: 27055869 PMCID: PMC4979312 DOI: 10.1038/leu.2016.61] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Revised: 02/25/2016] [Accepted: 02/29/2016] [Indexed: 01/01/2023]
Abstract
A common feature of B-cell chronic lymphocytic leukemia (CLL) is chromosomal loss of 13q14, containing the miR15a/16-1 locus controlling B-cell proliferation. However, CLL etiology remains unclear. CLL is an adult leukemia with an incidence that increases with advancing age. A unique feature of CLL is biased B-cell antigen receptor (BCR) usage, autoreactivity with polyreactivity and CD5 expression, all suggest a role for the BCR in driving CLL pathogenesis. Among human CLLs, BCRs autoreactive with non-muscle myosin IIA (AMyIIA) are recurrent. Here we identify an unmutated AMyIIA BCR in mouse, with distinctive CDR3 segments capable of promoting leukemogenesis. B cells with this AMyIIA BCR are generated by BCR-dependent signaling during B-1 fetal/neonatal development with CD5 induction, but not in adults. These early-generated AMyIIA B-1 B cells self-renew, increase during aging and can progress to become monoclonal B-cell lymphocytosis, followed by aggressive CLL in aged mice, often with the loss of a chromosomal region containing the miR15a/16-1 locus of varying length, as in human CLL. Thus, the ability to generate this defined autoreactive BCR by B-1 B cells is a key predisposing step in mice, promoting progression to chronic leukemia.
Collapse
MESH Headings
- Animals
- B-Lymphocytes/pathology
- Cell Self Renewal
- Chromosome Deletion
- Chromosome Disorders
- Chromosomes, Human, Pair 13
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/etiology
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Mice
- Nonmuscle Myosin Type IIA/metabolism
- Receptors, Antigen, B-Cell/metabolism
- Synteny
Collapse
Affiliation(s)
- Kyoko Hayakawa
- Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111, USA
| | | | | | | | | | - Herbert C. Morse
- Laboratory of Immunogenetics, National Institute of Allergy and
Infectious Diseases, National Institutes of Health, Rockville, Maryland 20852,
USA
| | - Yue-Sheng Li
- Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111, USA
| | | |
Collapse
|
17
|
Sheibani S, Cohen R, Kane S, Dubinsky M, Church JA, Mahadevan U. The Effect of Maternal Peripartum Anti-TNFα Use on Infant Immune Response. Dig Dis Sci 2016; 61:1622-7. [PMID: 26725061 DOI: 10.1007/s10620-015-3992-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 12/08/2015] [Indexed: 01/11/2023]
Abstract
BACKGROUND Infliximab (IFX) and adalimumab (ADA) cross the placenta in the third trimester and can be detectable in infants for up to 12 months. AIM The aim of this study was to determine whether in utero IFX or ADA exposure results in an impaired immune response in infants, as measured by immunoglobulin levels and antibody responses to routine primary immunizations. METHODS Infants who were exposed to in utero anti-TNFα agents were prospectively evaluated. Immunoglobulin levels (IgG, IgM, IgA) and antibodies to standard vaccinations, including tetanus toxoid (tetanus) and Haemophilus influenza type b (Hib), were measured in infants of at least 6 months of age. RESULTS Twelve infants were prospectively studied: 10 exposed to in utero IFX and 2 exposed to ADA with at least one dose administered in the third trimester. Immunoglobulin levels were available on 10/12 patients, with all showing adequate immunoglobulin levels, except for low IgM levels in 5 (50 %) infants. Adequate responses to both the tetanus and Hib vaccines were seen in 11 of 12 (92 %) infants. CONCLUSIONS Infants exposed to anti-TNFα agents in utero demonstrate appropriate response to two commonly administered neonatal vaccines and show adequate immunoglobulin levels, except for IgM. Newborns with a history of exposure to anti-TNFα agents should follow a standard vaccination schedule for inactive vaccines.
Collapse
Affiliation(s)
- Sarah Sheibani
- Division of Gastroenterology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA.
- Division of Gastroenterology, University of Southern California, Keck School of Medicine, 1520 San Pablo Street, Suite 1000, Los Angeles, CA, 90033-5312, USA.
| | - Russell Cohen
- Division of Gastroenterology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Sunanda Kane
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Marla Dubinsky
- Division of Gastroenterology, Department of Medicine, Mt. Sinai Medical Center, New York, NY, USA
| | - Joseph A Church
- Division of Clinical Immunology and Allergy, Department of Pediatrics, Children's Hospital Los Angeles and Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Uma Mahadevan
- Division of Gastroenterology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
18
|
Wu YY, Georg I, Díaz-Barreiro A, Varela N, Lauwerys B, Kumar R, Bagavant H, Castillo-Martín M, El Salem F, Marañón C, Alarcón-Riquelme ME. Concordance of increased B1 cell subset and lupus phenotypes in mice and humans is dependent on BLK expression levels. THE JOURNAL OF IMMUNOLOGY 2015; 194:5692-702. [PMID: 25972485 DOI: 10.4049/jimmunol.1402736] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 04/13/2015] [Indexed: 01/20/2023]
Abstract
Polymorphisms in the B lymphoid tyrosine kinase (BLK) gene have been associated with autoimmune diseases, including systemic lupus erythematosus, with risk correlating with reduced expression of BLK. How reduced expression of BLK causes autoimmunity is unknown. Using Blk(+/+) , Blk(+/-) , and Blk(-/-) mice, we show that aged female Blk(+/-) and Blk(-/-) mice produced higher anti-dsDNA IgG Abs and developed immune complex-mediated glomerulonephritis, compared with Blk(+/+) mice. Starting at young age, Blk(+/-) and Blk(-/-) mice accumulated increased numbers of splenic B1a cells, which differentiated into class-switched CD138(+) IgG-secreting B1a cells. Increased infiltration of B1a-like cells into the kidneys was also observed in aged Blk(+/-) and Blk(-/-) mice. In humans, we found that healthy individuals had BLK genotype-dependent levels of anti-dsDNA IgG Abs as well as increased numbers of a B1-like cell population, CD19(+)CD3(-)CD20(+)CD43(+)CD27(+), in peripheral blood. Furthermore, we describe the presence of B1-like cells in the tubulointerstitial space of human lupus kidney biopsies. Taken together, our study reveals a previously unappreciated role of reduced BLK expression on extraperitoneal accumulation of B1a cells in mice, as well as the presence of IgG autoantibodies and B1-like cells in humans.
Collapse
Affiliation(s)
- Ying-Yu Wu
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104;
| | - Ina Georg
- Centre for Genomics and Oncological Research (GENYO), Pfizer-University of Granada-Andalusian Regional Government, Health Sciences Technology Park, Granada 18016, Spain
| | - Alejandro Díaz-Barreiro
- Centre for Genomics and Oncological Research (GENYO), Pfizer-University of Granada-Andalusian Regional Government, Health Sciences Technology Park, Granada 18016, Spain
| | - Nieves Varela
- Centre for Genomics and Oncological Research (GENYO), Pfizer-University of Granada-Andalusian Regional Government, Health Sciences Technology Park, Granada 18016, Spain
| | - Bernard Lauwerys
- Pôle de Pathologies Rhumatismales, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, 1200 Brussels, Belgium; and
| | - Ramesh Kumar
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104
| | - Harini Bagavant
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104
| | | | - Fadi El Salem
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Concepción Marañón
- Centre for Genomics and Oncological Research (GENYO), Pfizer-University of Granada-Andalusian Regional Government, Health Sciences Technology Park, Granada 18016, Spain
| | - Marta E Alarcón-Riquelme
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104; Centre for Genomics and Oncological Research (GENYO), Pfizer-University of Granada-Andalusian Regional Government, Health Sciences Technology Park, Granada 18016, Spain;
| |
Collapse
|
19
|
Hayakawa K, Formica AM, Colombo MJ, Ichikawa D, Shinton SA, Brill-Dashoff J, Hardy RR. B cells generated by B-1 development can progress to chronic lymphocytic leukemia. Ann N Y Acad Sci 2015; 1362:250-5. [PMID: 25907284 DOI: 10.1111/nyas.12768] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
B cells generated early during fetal/neonatal B-1 development in mice include autoreactive cells with detectable CD5 upregulation induced by B cell receptor (BCR) signaling (B1a cells). A fraction of B1a cells are maintained by self-renewal for life, with the potential risk of dysregulated growth and progression to chronic lymphocytic leukemia (CLL)/lymphoma during aging. In studies using the Eμ-hTCL1 transgenic mouse system, it became clear that this B1a subset has a higher potential than other B cell subsets for progression to CLL. We have generated several autoreactive germline BCR gene models to compare B cells generated under conditions of natural exposure to autoantigen. Analysis of the mice has been key in understanding the importance of the BCR and BCR signaling for generating different B cell subsets and for investigating the cellular origin of B-CLL.
Collapse
Affiliation(s)
| | | | | | - Daiju Ichikawa
- Fox Chase Cancer Center, Philadelphia, Pennsylvania.,Keio University Faculty of Pharmacy, Juntendo University School of Medicine, Tokyo, Japan
| | | | | | | |
Collapse
|
20
|
Neely HR, Flajnik MF. CXCL13 responsiveness but not CXCR5 expression by late transitional B cells initiates splenic white pulp formation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2015; 194:2616-23. [PMID: 25662995 PMCID: PMC4355030 DOI: 10.4049/jimmunol.1401905] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Secondary lymphoid organs (SLO) provide the structural framework for coconcentration of Ag and Ag-specific lymphocytes required for an efficient adaptive immune system. The spleen is the primordial SLO, and evolved concurrently with Ig/TCR:pMHC-based adaptive immunity. The earliest cellular/histological event in the ontogeny of the spleen's lymphoid architecture, the white pulp (WP), is the accumulation of B cells around splenic vasculature, an evolutionarily conserved feature since the spleen's emergence in early jawed vertebrates such as sharks. In mammals, B cells are indispensable for both formation and maintenance of SLO microarchitecture; their expression of lymphotoxin α1β2 (LTα1β2) is required for the LTα1β2:CXCL13 positive feedback loop without which SLO cannot properly form. Despite the spleen's central role in the evolution of adaptive immunity, neither the initiating event nor the B cell subset necessary for WP formation has been identified. We therefore sought to identify both in mouse. We detected CXCL13 protein in late embryonic splenic vasculature, and its expression was TNF-α and RAG-2 independent. A substantial influx of CXCR5(+) transitional B cells into the spleen occurred 18 h before birth. However, these late embryonic B cells were unresponsive to CXCL13 (although responsive to CXCL12) and phenotypically indistinguishable from blood-derived B cells. Only after birth did B cells acquire CXCL13 responsiveness, accumulate around splenic vasculature, and establish the uniquely splenic B cell compartment, enriched for CXCL13-responsive late transitional cells. Thus, CXCL13 is the initiating component of the CXCL13:LTα1β2 positive feedback loop required for WP ontogeny, and CXCL13-responsive late transitional B cells are the initiating subset.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
- Chemokine CXCL13/genetics
- Chemokine CXCL13/immunology
- Chemokine CXCL13/metabolism
- DNA-Binding Proteins/deficiency
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/immunology
- Feedback, Physiological
- Female
- Gene Expression Regulation, Developmental
- Immunohistochemistry
- In Situ Hybridization
- Lymphotoxin alpha1, beta2 Heterotrimer/immunology
- Lymphotoxin alpha1, beta2 Heterotrimer/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Pregnancy
- Receptors, CXCR5/genetics
- Receptors, CXCR5/immunology
- Receptors, CXCR5/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Spleen/blood supply
- Spleen/embryology
- Spleen/immunology
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/immunology
- Tumor Necrosis Factor-alpha/metabolism
Collapse
Affiliation(s)
- Harold R Neely
- Department of Microbiology and Immunology, University of Maryland, Baltimore, MD 21201
| | - Martin F Flajnik
- Department of Microbiology and Immunology, University of Maryland, Baltimore, MD 21201
| |
Collapse
|
21
|
Ichikawa D, Asano M, Shinton SA, Brill-Dashoff J, Formica AM, Velcich A, Hardy RR, Hayakawa K. Natural anti-intestinal goblet cell autoantibody production from marginal zone B cells. THE JOURNAL OF IMMUNOLOGY 2014; 194:606-14. [PMID: 25480561 DOI: 10.4049/jimmunol.1402383] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Expression of a germline VH3609/D/JH2 IgH in mice results in the generation of B1 B cells with anti-thymocyte/Thy-1 glycoprotein autoreactivity by coexpression of Vk21-5/Jk2 L chain leading to production of serum IgM natural autoantibody. In these same mice, the marginal zone (MZ) B cell subset in spleen shows biased usage of a set of Ig L chains different from B1 B cells, with 30% having an identical Vk19-17/Jk1 L chain rearrangement. This VH3609/Vk19-17 IgM is reactive with intestinal goblet cell granules, binding to the intact large polymatrix form of mucin 2 glycoprotein secreted by goblet cells. Analysis of a μκ B cell AgR (BCR) transgenic (Tg) mouse with this anti-goblet cell/mucin2 autoreactive (AGcA) specificity demonstrates that immature B cells expressing the Tg BCR become MZ B cells in spleen by T cell-independent BCR signaling. These Tg B cells produce AGcA as the predominant serum IgM, but without enteropathy. Without the transgene, AGcA autoreactivity is low but detectable in the serum of BALB/c and C.B17 mice, and this autoantibody is specifically produced by the MZ B cell subset. Thus, our findings reveal that AGcA is a natural autoantibody associated with MZ B cells.
Collapse
Affiliation(s)
- Daiju Ichikawa
- Fox Chase Cancer Center, Philadelphia, PA 19111; Keio University Faculty of Pharmacy, Tokyo 105-8512, Japan
| | - Masanao Asano
- Fox Chase Cancer Center, Philadelphia, PA 19111; Juntendo University School of Medicine, Tokyo 113, Japan; and
| | | | | | | | - Anna Velcich
- Albert Einstein Cancer Center/Montefiore Medical Center, Bronx, NY 10467
| | | | | |
Collapse
|
22
|
Tarunina M, Hernandez D, Johnson CJ, Rybtsov S, Ramathas V, Jeyakumar M, Watson T, Hook L, Medvinsky A, Mason C, Choo Y. Directed differentiation of embryonic stem cells using a bead-based combinatorial screening method. PLoS One 2014; 9:e104301. [PMID: 25251366 PMCID: PMC4174505 DOI: 10.1371/journal.pone.0104301] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 07/07/2014] [Indexed: 01/25/2023] Open
Abstract
We have developed a rapid, bead-based combinatorial screening method to determine optimal combinations of variables that direct stem cell differentiation to produce known or novel cell types having pre-determined characteristics. Here we describe three experiments comprising stepwise exposure of mouse or human embryonic cells to 10,000 combinations of serum-free differentiation media, through which we discovered multiple novel, efficient and robust protocols to generate a number of specific hematopoietic and neural lineages. We further demonstrate that the technology can be used to optimize existing protocols in order to substitute costly growth factors with bioactive small molecules and/or increase cell yield, and to identify in vitro conditions for the production of rare developmental intermediates such as an embryonic lymphoid progenitor cell that has not previously been reported.
Collapse
Affiliation(s)
- Marina Tarunina
- Plasticell Ltd, Stevenage Bioscience Catalyst, Stevenage, United Kingdom
| | - Diana Hernandez
- Plasticell Ltd, Stevenage Bioscience Catalyst, Stevenage, United Kingdom
- Advanced Centre for Biochemical Engineering, University College London, London, United Kingdom
| | | | - Stanislav Rybtsov
- MRC Centre for Regenerative Medicine/Institute of Stem cell Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Vidya Ramathas
- Plasticell Ltd, Stevenage Bioscience Catalyst, Stevenage, United Kingdom
| | | | - Thomas Watson
- Plasticell Ltd, Stevenage Bioscience Catalyst, Stevenage, United Kingdom
| | - Lilian Hook
- Plasticell Ltd, Stevenage Bioscience Catalyst, Stevenage, United Kingdom
| | - Alexander Medvinsky
- MRC Centre for Regenerative Medicine/Institute of Stem cell Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Chris Mason
- Advanced Centre for Biochemical Engineering, University College London, London, United Kingdom
| | - Yen Choo
- Plasticell Ltd, Stevenage Bioscience Catalyst, Stevenage, United Kingdom
- Progenitor Labs Ltd, Stevenage Bioscience Catalyst, Stevenage, United Kingdom
| |
Collapse
|
23
|
Heinig K, Gätjen M, Grau M, Stache V, Anagnostopoulos I, Gerlach K, Niesner RA, Cseresnyes Z, Hauser AE, Lenz P, Hehlgans T, Brink R, Westermann J, Dörken B, Lipp M, Lenz G, Rehm A, Höpken UE. Access to follicular dendritic cells is a pivotal step in murine chronic lymphocytic leukemia B-cell activation and proliferation. Cancer Discov 2014; 4:1448-65. [PMID: 25252690 DOI: 10.1158/2159-8290.cd-14-0096] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
UNLABELLED In human chronic lymphocytic leukemia (CLL) pathogenesis, B-cell antigen receptor signaling seems important for leukemia B-cell ontogeny, whereas the microenvironment influences B-cell activation, tumor cell lodging, and provision of antigenic stimuli. Using the murine Eμ-Tcl1 CLL model, we demonstrate that CXCR5-controlled access to follicular dendritic cells confers proliferative stimuli to leukemia B cells. Intravital imaging revealed a marginal zone B cell-like leukemia cell trafficking route. Murine and human CLL cells reciprocally stimulated resident mesenchymal stromal cells through lymphotoxin-β-receptor activation, resulting in CXCL13 secretion and stromal compartment remodeling. Inhibition of lymphotoxin/lymphotoxin-β-receptor signaling or of CXCR5 signaling retards leukemia progression. Thus, CXCR5 activity links tumor cell homing, shaping a survival niche, and access to localized proliferation stimuli. SIGNIFICANCE CLL and other indolent lymphoma are not curable and usually relapse after treatment, a process in which the tumor microenvironment plays a pivotal role. We dissect the consecutive steps of CXCR5-dependent tumor cell lodging and LTβR-dependent stroma-leukemia cell interaction; moreover, we provide therapeutic solutions to interfere with this reciprocal tumor-stroma cross-talk.
Collapse
Affiliation(s)
- Kristina Heinig
- Department of Tumor Genetics and Immunogenetics, Max-Delbrück-Center for Molecular Medicine, MDC, Berlin, Germany
| | - Marcel Gätjen
- Department of Hematology, Oncology and Tumorimmunology, Max-Delbrück-Center for Molecular Medicine, MDC, Berlin, Germany
| | - Michael Grau
- Department of Physics, Philipps-University Marburg, Marburg, Germany
| | - Vanessa Stache
- Department of Tumor Genetics and Immunogenetics, Max-Delbrück-Center for Molecular Medicine, MDC, Berlin, Germany
| | | | - Kerstin Gerlach
- Department of Hematology, Oncology and Tumorimmunology, Max-Delbrück-Center for Molecular Medicine, MDC, Berlin, Germany
| | | | - Zoltan Cseresnyes
- Deutsches Rheumaforschungszentrum, DRFZ, Berlin, Germany. Confocal and 2-Photon Microscopy Core Facility, Max-Delbrück-Center for Molecular Medicine, MDC, Berlin, Germany
| | - Anja E Hauser
- Deutsches Rheumaforschungszentrum, DRFZ, Berlin, Germany. Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Peter Lenz
- Department of Physics, Philipps-University Marburg, Marburg, Germany
| | - Thomas Hehlgans
- Institute for Immunology, University Regensburg, Regensburg, Germany
| | - Robert Brink
- Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Jörg Westermann
- Department of Hematology, Oncology and Tumorimmunology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
| | - Bernd Dörken
- Department of Hematology, Oncology and Tumorimmunology, Max-Delbrück-Center for Molecular Medicine, MDC, Berlin, Germany. Department of Hematology, Oncology and Tumorimmunology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
| | - Martin Lipp
- Department of Tumor Genetics and Immunogenetics, Max-Delbrück-Center for Molecular Medicine, MDC, Berlin, Germany
| | - Georg Lenz
- Department of Hematology, Oncology and Tumorimmunology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
| | - Armin Rehm
- Department of Hematology, Oncology and Tumorimmunology, Max-Delbrück-Center for Molecular Medicine, MDC, Berlin, Germany. Department of Hematology, Oncology and Tumorimmunology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany.
| | - Uta E Höpken
- Department of Tumor Genetics and Immunogenetics, Max-Delbrück-Center for Molecular Medicine, MDC, Berlin, Germany.
| |
Collapse
|
24
|
Abstract
Interferon Regulatory Factor 4 (IRF4) and IRF8 are critical regulators of immune system development and function. In B lymphocytes, IRF4 and IRF8 have been shown to control important events during their development and maturation including pre-B cell differentiation, induction of B cell tolerance pathways, marginal zone B cell development, germinal center reaction and plasma cell differentiation. Mechanistically, IRF4 and IRF8 are found to function redundantly to control certain stages of B cell development, but in other stages, they function nonredundantly to play distinct roles in B cell biology. In line with their essential roles in B cell development, deregulated expressions of IRF4 and IRF8 have been associated to the pathogenesis of several B cell malignancies and diseases. Recent studies have elucidated diverse transcriptional networks regulated by IRF4 and IRF8 at distinct B cell developmental stages and related malignancies. In this review we will discuss the recent advances for the roles of IRF4 and IRF8 during B cell development and associated diseases.
Collapse
|
25
|
Cruz-Leal Y, Machado Y, López-Requena A, Canet L, Laborde R, Álvares AM, Lucatelli Laurindo MF, Santo Tomas JF, Alonso ME, Alvarez C, Mortara RA, Popi AF, Mariano M, Pérez R, Lanio ME. Role of B-1 cells in the immune response against an antigen encapsulated into phosphatidylcholine-containing liposomes. Int Immunol 2014; 26:427-37. [PMID: 24618118 DOI: 10.1093/intimm/dxu042] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
B-1 lymphocytes comprise a unique subset of B cells that differ phenotypically, ontogenetically and functionally from conventional B-2 cells. A frequent specificity of the antibody repertoire of peritoneal B-1 cells is phosphatidylcholine. Liposomes containing phosphatidylcholine have been studied as adjuvants and their interaction with dendritic cells and macrophages has been demonstrated. However, the role of B-1 cells in the adjuvanticity of liposomes composed of phosphatidylcholine has not been explored. In the present work, we studied the contribution of B-1 cells to the humoral response against ovalbumin (OVA) encapsulated into dipalmitoylphosphatidylcholine (DPPC) and cholesterol-containing liposomes. BALB/X-linked immunodeficient (xid) mice, which are deficient in B-1 cells, showed quantitative and qualitative differences in the anti-OVA antibody response compared with wild-type animals after immunization with these liposomes. The OVA-specific immune response was significantly increased in the BALB/xid mice when reconstituted with B-1 cells from naive BALB/c mice. Our results indicate the internalization of DPPC-containing liposomes by these cells and their migration from the peritoneal cavity to the spleen. Phosphatidylcholine significantly contributed to the immunogenicity of liposomes, as DPPC-containing liposomes more effectively stimulated the anti-OVA response compared with vesicles composed of dipalmitoylphosphatidylglycerol. In conclusion, we present evidence for a cognate interaction between B-1 cells and phosphatidylcholine liposomes, modulating the immune response to encapsulated antigens. This provides a novel targeting approach to assess the role of B-1 cells in humoral immunity.
Collapse
Affiliation(s)
- Yoelys Cruz-Leal
- Center for Protein Studies, Faculty of Biology, University of Havana, Havana 10400, Cuba
| | - Yoan Machado
- Research and Development Division, Center of Molecular Immunology (CIM), Havana 11600, Cuba
| | | | - Liem Canet
- Center for Protein Studies, Faculty of Biology, University of Havana, Havana 10400, Cuba
| | - Rady Laborde
- Center for Protein Studies, Faculty of Biology, University of Havana, Havana 10400, Cuba
| | - Anuska Marcelino Álvares
- Discipline of Immunology, Department of Microbiology, Immunology and Parasitology, Universidade Federal de São Paulo, 4023-900, SP, Brazil
| | - María F Lucatelli Laurindo
- Discipline of Immunology, Department of Microbiology, Immunology and Parasitology, Universidade Federal de São Paulo, 4023-900, SP, Brazil
| | - Julio F Santo Tomas
- Research and Development Division, Center of Molecular Immunology (CIM), Havana 11600, Cuba
| | - María E Alonso
- Center for Protein Studies, Faculty of Biology, University of Havana, Havana 10400, Cuba
| | - Carlos Alvarez
- Center for Protein Studies, Faculty of Biology, University of Havana, Havana 10400, Cuba
| | - Renato A Mortara
- Department of Microbiology, Immunology and Parasitology, Universidade Federal de São Paulo, 4023-900, SP, Brazil and
| | - Ana F Popi
- Discipline of Immunology, Department of Microbiology, Immunology and Parasitology, Universidade Federal de São Paulo, 4023-900, SP, Brazil
| | - Mario Mariano
- Discipline of Immunology, Department of Microbiology, Immunology and Parasitology, Universidade Federal de São Paulo, 4023-900, SP, Brazil Universidade Paulista UNIP, São Paulo, Brazil
| | - Rolando Pérez
- Research and Development Division, Center of Molecular Immunology (CIM), Havana 11600, Cuba
| | - María E Lanio
- Center for Protein Studies, Faculty of Biology, University of Havana, Havana 10400, Cuba
| |
Collapse
|
26
|
Abstract
Interferon regulatory factor 4 (IRF4) is a critical transcriptional regulator of B-cell development and function. A recent genome-wide single-nucleotide polymorphism (SNP) association study identified IRF4 as a major susceptibility gene in chronic lymphocytic leukemia (CLL). Although the SNPs located in the IRF4 gene were linked to a downregulation of IRF4 in CLL patients, whether a low level of IRF4 is critical for CLL development remains unclear. In rodents, CLL cells are derived from B1 cells whose population is dramatically expanded in immunoglobulin heavy chain Vh11 knock-in mice. We bred a Vh11 knock-in allele into IRF4-deficient mice (IRF4(-/-)Vh11). Here, we report that IRF4(-/-)Vh11 mice develop spontaneous early-onset CLL with 100% penetrance. Further analysis shows that IRF4(-/-)Vh11 CLL cells proliferate predominantly in spleen and express high levels of Mcl-1. IRF4(-/-)Vh11 CLL cells are resistant to apoptosis but reconstitution of IRF4 expression in the IRF4(-/-)Vh11 CLL cells inhibits their survival. Thus, our study demonstrates for the first time a causal relationship between low levels of IRF4 and the development of CLL. Moreover, our findings establish IRF4(-/-)Vh11 mice as a novel mouse model of CLL that not only is valuable for dissecting molecular pathogenesis of CLL but could also be used for therapeutic purposes.
Collapse
|
27
|
Tian R, Fu M, Zhang Z, Ren J, An J, Liu Y, Li W. In situ IgM production and clonal expansion of B-1 cells in peritoneal cavity promote elimination of C. albicans infection in IgH transgenic mice with VH derived from a natural antibody. PLoS One 2013; 8:e60779. [PMID: 23565274 PMCID: PMC3614557 DOI: 10.1371/journal.pone.0060779] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Accepted: 03/02/2013] [Indexed: 01/15/2023] Open
Abstract
B-1 cells are innate-like cells that play important roles in host defense against infection. However, the function of B-1 cells in fungi infection remains unclear. Previously we produced IgH transgenic mice TgVH3B4 with VH derived from a natural antibody 3B4 that can identify C. albicans, and found that TgVH3B4 mice were resistant to intraperitoneal (i. p.) and intravenous C. albicans infection. Most of the peritoneal cavity (PEC) B-1 cells in TgVH3B4 mice express transgenic BCR that binds C. albicans. In the present study, we explored the response of B-1 cells to C. albicans infection by applying i. p. inoculation of fungi in TgVH3B4 mice. We found that C. albicans was cleared more efficiently in TgVH3B4 mice after i. p. inoculation than that of littermate control. The level of C. albicans-reactive IgM in PEC of TgVH3B4 mice was much higher than that of control, and the number of B-1a B cells was also elevated in TgVH3B4 mice, which was mainly due to enhanced proliferation of B-1 cells. Additionally, numbers of C. albicans-specific B cells increased greatly in TgVH3B4 mice after C. albicans inoculation. Our data suggested that in situ IgM production and clonal expansion of B-1 cells in PEC participate in host defense against C. albicans infection.
Collapse
Affiliation(s)
- Rong Tian
- Department of Dermatology, General Hospital of the Air Force, Beijing, P. R. China
- Department of Dermatology, Xijing Hospital, Xi’an, P. R. China
| | - Meng Fu
- Department of Dermatology, Xijing Hospital, Xi’an, P. R. China
| | - Zhuo Zhang
- Department of Neurology, Tangdu Hospital, Xi’an, P. R. China
| | - Jing Ren
- Department of Dermatology, Xijing Hospital, Xi’an, P. R. China
| | - Jingang An
- Department of Dermatology, Xijing Hospital, Xi’an, P. R. China
| | - Yufeng Liu
- Department of Dermatology, Xijing Hospital, Xi’an, P. R. China
| | - Wei Li
- Department of Dermatology, General Hospital of the Air Force, Beijing, P. R. China
- Department of Dermatology, Xijing Hospital, Xi’an, P. R. China
- * E-mail:
| |
Collapse
|
28
|
Expression of plasma cell alloantigen 1 defines layered development of B-1a B-cell subsets with distinct innate-like functions. Proc Natl Acad Sci U S A 2012; 109:20077-82. [PMID: 23169635 DOI: 10.1073/pnas.1212428109] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Innate-like B-1a cells contribute significantly to circulating natural antibodies and mucosal immunity as well as to immunoregulation. Here we show that these classic functions of B-1a cells segregate between two unique subsets defined by expression of plasma cell alloantigen 1 (PC1), also known as ectonucleotide pyrophosphatase phosphodiesterase 1 (ENPP1). These subsets, designated B-1a.PC1(lo) and B-1a.PC1(hi), differ significantly in IgH chain utilization. Adoptively transferred PC1(lo) cells secreted significantly more circulating natural IgM and intestinal IgA than PC1(hi) cells. In contrast, PC1(hi) cells produced more IL-10 than PC1(lo) cells when stimulated with LPS and phorbol 12-myristate 13-acetate (PMA). PC1(hi) cells were also more efficient than PC1(lo) cells in regulating Th1 cell differentiation, even though both B-1a subsets were comparably active in stimulating T-cell proliferation. Furthermore, PC1(lo) cells generated antigen-specific IgM responses to pneumococcal polysaccharide antigens, whereas PC1(hi) cells do not. We found that PC1(lo) cells develop from an early wave of B-1a progenitors in fetal life, whereas PC1(hi) cells are generated from a later wave after birth. We conclude that identification of B-1a.PC1(lo) and B-1a.PC1(hi) cells extends the concept of a layered immune system with important implications for developing effective vaccines and promoting the generation of immunoregulatory B cells.
Collapse
|
29
|
El Shikh MEM, Pitzalis C. Follicular dendritic cells in health and disease. Front Immunol 2012; 3:292. [PMID: 23049531 PMCID: PMC3448061 DOI: 10.3389/fimmu.2012.00292] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Accepted: 08/31/2012] [Indexed: 12/17/2022] Open
Abstract
Follicular dendritic cells (FDCs) are unique immune cells that contribute to the regulation of humoral immune responses. These cells are located in the B-cell follicles of secondary lymphoid tissues where they trap and retain antigens (Ags) in the form of highly immunogenic immune complexes (ICs) consisting of Ag plus specific antibody (Ab) and/or complement proteins. FDCs multimerize Ags and present them polyvalently to B-cells in periodically arranged arrays that extensively crosslink the B-cell receptors for Ag (BCRs). FDC-FcγRIIB mediates IC periodicity, and FDC-Ag presentation combined with other soluble and membrane bound signals contributed by FDCs, like FDC-BAFF, -IL-6, and -C4bBP, are essential for the induction of the germinal center (GC) reaction, the maintenance of serological memory, and the remarkable ability of FDC-Ags to induce specific Ab responses in the absence of cognate T-cell help. On the other hand, FDCs play a negative role in several disease conditions including chronic inflammatory diseases, autoimmune diseases, HIV/AIDS, prion diseases, and follicular lymphomas. Compared to other accessory immune cells, FDCs have received little attention, and their functions have not been fully elucidated. This review gives an overview of FDC structure, and recapitulates our current knowledge on the immunoregulatory functions of FDCs in health and disease. A better understanding of FDCs should permit better regulation of Ab responses to suit the therapeutic manipulation of regulated and dysregulated immune responses.
Collapse
Affiliation(s)
- Mohey Eldin M El Shikh
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London London, UK
| | | |
Collapse
|
30
|
TNF-α and the development of the neonatal immune system: implications for inhibitor use in pregnancy. Am J Gastroenterol 2011; 106:559-62. [PMID: 21468063 DOI: 10.1038/ajg.2011.5] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Inflammatory bowel diseases, Crohn's disease, and ulcerative colitis have an unpredictable course during and after pregnancy (1). There is a great deal of interest in treating moderate to severe active inflammatory bowel disease with anti-tumor necrosis factor (anti-TNF) biologics in pregnant women (2). We lack definitive information about the effects of these agents on the development of the immune system of the human fetus and the newborn baby. Anti-TNF agents fall within US Food and Drug Administration's (FDA) category B regarding fetal risk, indicating that no adequate and well-controlled studies have been conducted in pregnant or nursing women. Here, we review animal studies (of both mice and nonhuman primates) that examine the role of TNF and its inhibitors in the normal development of the immune system.
Collapse
|
31
|
Balogh P, Fisi V, Szakal AK. Fibroblastic reticular cells of the peripheral lymphoid organs: Unique features of a ubiquitous cell type. Mol Immunol 2008; 46:1-7. [DOI: 10.1016/j.molimm.2008.07.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2008] [Accepted: 07/06/2008] [Indexed: 10/21/2022]
|
32
|
Murakami T, Chen X, Hase K, Sakamoto A, Nishigaki C, Ohno H. Splenic CD19-CD35+B220+ cells function as an inducer of follicular dendritic cell network formation. Blood 2007; 110:1215-24. [PMID: 17519390 PMCID: PMC1939903 DOI: 10.1182/blood-2007-01-068387] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Follicular dendritic cells (FDCs) form a reticular FDC network in the lymphoid follicle that is essential for the retention and presentation of native antigens in the form of antigen-antibody immune complexes (ICs) to B cells during secondary immune response. Although the presence of migrating precursors of FDCs has been hypothesized, their entity has not been elucidated. Here we report the identification of murine splenic CD19(-)CD11c(-)CD35(+)B220(+) cells as an inducer of FDC network formation. We demonstrated that CD19(-)-CD11c(-)CD35(+)B220(+) cells, together with stromal cells, had the remarkable ability to form lymphoid-follicle-like structures that contained B220(+)FDC-M1(+) reticular cells originally derived from CD19(-)-CD11c(-)CD35(+)B220(+) cells in the CD35(+) reticulum. Our results indicate that CD19(-)CD11c(-)CD35(+)B220(+) cells function as an inducer of FDC network formation and that the interaction between CD19(-)CD11c(-)CD35(+)B220(+) cells and stromal cells is required to initiate lymphoid follicle formation.
Collapse
MESH Headings
- Animals
- Antigens, CD19/metabolism
- Cell Differentiation
- Cell Lineage
- Dendritic Cells, Follicular/cytology
- Dendritic Cells, Follicular/metabolism
- Flow Cytometry
- Green Fluorescent Proteins/genetics
- Green Fluorescent Proteins/metabolism
- Immunophenotyping
- Killer Cells, Natural/classification
- Killer Cells, Natural/immunology
- Leukocyte Common Antigens/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Receptors, Complement 3b/metabolism
- Spleen/cytology
- Stromal Cells/cytology
- Stromal Cells/immunology
- Stromal Cells/metabolism
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/physiology
Collapse
Affiliation(s)
- Takaya Murakami
- Laboratory for Epithelial Immunobiology, Rikagaku Kenkyusho (RIKEN) Research Center for Allergy and Immunology, Yokohama, Japan
| | | | | | | | | | | |
Collapse
|
33
|
Ha SA, Tsuji M, Suzuki K, Meek B, Yasuda N, Kaisho T, Fagarasan S. Regulation of B1 cell migration by signals through Toll-like receptors. ACTA ACUST UNITED AC 2006; 203:2541-50. [PMID: 17060475 PMCID: PMC2118139 DOI: 10.1084/jem.20061041] [Citation(s) in RCA: 228] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Peritoneal B1 cells are known to generate large amounts of antibodies outside their residential site. These antibodies play an important role in the early defense against bacteria and viruses, before the establishment of adaptive immune responses. Although many stimuli, including antigen, lipopolysaccharide, or cytokines, have been shown to activate B1 cells and induce their differentiation into plasma cells, the molecular signals required for their egress from the peritoneal cavity are not understood. We demonstrate here that direct signals through Toll-like receptors (TLRs) induce specific, rapid, and transient down-regulation of integrins and CD9 on B1 cells, which is required for detachment from local matrix and a high velocity movement of cells in response to chemokines. Thus, we revealed an unexpected role for TLRs in governing the interplay between integrins, tetraspanins, and chemokine receptors required for B1 cell egress and, as such, in facilitating appropriate transition from innate to adaptive immune responses.
Collapse
Affiliation(s)
- Seon-ah Ha
- Laboratory for Mucosal Immunity, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | | | | | | | | | | | | |
Collapse
|
34
|
Shimizu I, Kawahara T, Haspot F, Bardwell PD, Carroll MC, Sykes M. B-cell extrinsic CR1/CR2 promotes natural antibody production and tolerance induction of anti-alphaGAL-producing B-1 cells. Blood 2006; 109:1773-81. [PMID: 17023586 PMCID: PMC1794072 DOI: 10.1182/blood-2006-02-002386] [Citation(s) in RCA: 18] [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
B-1b cells produce IgM natural antibodies against alpha1-3Galbeta1-4GlcNAc (alphaGal). These can be tolerized by nonmyeloablative induction of mixed chimerism using alphaGal-positive (alphaGal+) donor marrow. We assessed the role of CR1/2 in this model for induction of tolerance of B-1b cells. Mixed hematopoietic chimerism was induced in alpha1-3galactosyltransferase (GalT-/-) and GalT-/-Cr2-/- mice with alphaGal+ BALB/c marrow donors. Anti-alphaGal Ab and anti-alphaGal Ab-producing B cells became undetectable in GalT-/- chimeras, whereas they persisted in chimeric GalT-/-Cr2-/- mice. To determine whether CR1/2 expression on stromal cells and/or hematopoietic cells was critical for B-1-cell tolerance, we generated GalT-/- radiation chimeras in which CR1/CR2 was expressed on either stromal cells, hematopoietic cells, neither, or both. After induction of mixed chimerism from alphaGal+ allogeneic bone marrow (BM) donors, anti-alphaGal-producing B cells were rendered tolerant in reconstituted recipients expressing only stromal CR1/CR2. Our results suggest a possible role for follicular dendritic cells that pick up immune complexes via CR1/CR2 receptors in the tolerization of B-1b cells.
Collapse
Affiliation(s)
- Ichiro Shimizu
- Bone Marrow Transplantation Section, Transplantation Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | | | | | | | | | | |
Collapse
|
35
|
Tomita Y, Shimizu I, Iwai T, Zhang QW, Okano S, Kajiwara T, Onzuka T, Tominaga R. Application of chimerism-based drug-induced tolerance to rat into mouse xenotransplantation. Scand J Immunol 2006; 64:392-7. [PMID: 16970680 DOI: 10.1111/j.1365-3083.2006.01831.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The current critical shortage of human donor organs has stimulated the feasibility of the xenogenic transplantation, such as swine to primate. We have previously reported the induction of donor-specific tolerance in MHC-disparated recipient mice by using our cyclophosphamide (CP)-induced tolerance conditioning. In this study, we examined the efficacy of our CP-induced tolerance conditioning in xenogenic transplantation model. F344 rats and B10 mice were used as donors and recipients. Recipient mice were treated with donor spleen cells, CP, Busulfan and bone marrow cells, with or without prior NK-cell depletion. Donor mixed chimerism, and the presence of donor reactive T-cell population were analysed by flow cytometry. The survival of the donor skin grafts were observed after the conditioning. Donor mixed chimerism was temporary induced but terminated at 10 weeks after treatments. Donor-specific prolongation of the skin graft survival was observed after the treatments, however, grafts were rejected in the long term. NK-cell depletion, prior to the treatments, did not affect the levels of the mixed chimerism or graft prolongation. The donor-reactive recipient T-cell population was remained the same level as the untreated mice, suggesting the failure of the induction of the central T-cell tolerance. Thus, partial efficacy of our CP-induced tolerance treatments in the rat to mice xenotransplantation was observed. Our results suggested that the additional treatments were required to establish the stable xenogenic tolerance.
Collapse
Affiliation(s)
- Y Tomita
- Department of Cardiovascular Surgery, Faculty of Medicine, Kyushu University, Fukuoka, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Hardy RR. B-1 B cells: development, selection, natural autoantibody and leukemia. Curr Opin Immunol 2006; 18:547-55. [PMID: 16879952 DOI: 10.1016/j.coi.2006.07.010] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2006] [Accepted: 07/20/2006] [Indexed: 01/22/2023]
Abstract
B-1 (CD5+) B cells constitute a phenotypic and functionally distinct population of B cells in mouse that show enriched expression of autoreactive B-cell antigen receptors and that produce several types of natural autoantibodies. Recently, there has been much progress in this field of research. Evidence has appeared for the existence of distinctive B-cell precursors that preferentially generate B-1 B cells, and the crucial requirement for strong B-cell antigen receptor signaling in the maturation of B-1 B cells has been established. Other work focuses on a phenotypically similar population that lacks CD5, termed 'B-1b', which shows similarities and differences from most CD5+ B cells in both development and function. The relationship of normal B-1 cells with B-cell lymphomas and leukemias continues to be a subject of interest and debate.
Collapse
Affiliation(s)
- Richard R Hardy
- Division of Basic Science, Fox Chase Cancer Center, Philadelphia, PA 19111, USA.
| |
Collapse
|
37
|
Moritoki Y, Lian ZX, Ohsugi Y, Ueno Y, Gershwin ME. B cells and autoimmune liver diseases. Autoimmun Rev 2006; 5:449-57. [PMID: 16920571 DOI: 10.1016/j.autrev.2006.02.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2006] [Accepted: 02/16/2006] [Indexed: 02/07/2023]
Abstract
Autoimmune hepatitis (AIH), primary biliary cirrhosis (PBC) and primary sclerosing cholangitis (PSC) are the three major autoimmune diseases affecting the liver. They are all characterized by the presence of a variety of autoantibodies, some of which are found in all three diseases, whereas others are restricted to one or two of them or are even specific for the particular disease. In this review we will first provide details of the serological features of these three autoimmune diseases that target the liver. In addition, we will highlight the possible pathogenic roles of autoreactive B cells, focusing on their immunological functions as autoantibody producing cells and as antigen-presenting cells for T cell priming. As well, we will describe the contribution of toll-like receptor (TLR) signaling to the activation of autoimmune B cells and the putative role of defects in regulatory T cell function in the development of autoimmune liver diseases.
Collapse
Affiliation(s)
- Yuki Moritoki
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis School of Medicine, 451 E. Health Sciences Drive, Suite 6510, Davis, CA 95616, USA
| | | | | | | | | |
Collapse
|
38
|
Shimizu I, Tomita Y, Iwai T, Kajiwara T, Okano S, Nomoto K, Tominaga R. Sequential analysis of anti-alpha Gal natural antibody-producing B cells in GalT knockout mice in cyclophosphamide-induced tolerance. Scand J Immunol 2006; 63:435-43. [PMID: 16764697 DOI: 10.1111/j.1365-3083.2006.001763.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Previously, we have shown that cyclophosphamide (CP)-induced tolerance, marked by permanent acceptance of donor skin graft and establishment of donor mixed chimerism, was readily induced with treatment with donor spleen cells (SC), CP, busulfan (BU) and donor bone marrow cells (BMC). Here, we investigated the mechanism of anti-donor natural antibody (nAb) producing B-cell tolerance in our CP-induced tolerance systems in alpha1,3-galactosyltransferase-deficient knockout mice (GalT KO; GalT-/-, H-2(b/d)). After induction of tolerance using donor AKR SC and BMC, survival of donor heart and skin grafts and production of anti-Galalpha1-3Galbeta1-4GlcNAc (anti-alphaGal) Ab in recipient GalT KO mice were analyzed. In addition, the production of anti-alphaGal Ab and the presence of Gal-BSA binding B cells in GalT KO mice were analyzed by flow cytometry (FCM) after treatments with rabbit red blood cells (RRBC) and CP. Permanent acceptance of donor skin and heart grafts and abrogation of anti-alphaGal Ab were achieved in GalT KO mice treated with donor SC + CP/BU + BMC. However, in the GalT KO mice treated with donor SC and CP, donor skin grafts were acutely rejected, even though anti-alphaGal Ab was undetectable. Similarly, anti-alphaGal Ab was undetectable in GalT KO mice treated with RRBC and CP. Our data strongly indicated the following mechanisms: the clonal destruction in the early stage and the clonal anergy or ignorance in the late stage after conventional conditioning with RRBC and CP. In conclusion, our drug-induced tolerance protocols are effective to induce tolerance in recipients that produce anti-donor nAb.
Collapse
Affiliation(s)
- Ichiro Shimizu
- Department of Cardiovascular Surgery, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | |
Collapse
|
39
|
Wen L, Brill-Dashoff J, Shinton SA, Asano M, Hardy RR, Hayakawa K. Evidence of marginal-zone B cell-positive selection in spleen. Immunity 2005; 23:297-308. [PMID: 16169502 DOI: 10.1016/j.immuni.2005.08.007] [Citation(s) in RCA: 116] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2005] [Revised: 08/12/2005] [Accepted: 08/17/2005] [Indexed: 10/25/2022]
Abstract
Antigen receptor-mediated signaling is critical for the development and survival of B cells. However, it has not been established whether B cell development requires a signal from self-ligand engagement at the immature stage, a process known as "positive selection." Here, using a monoclonal B cell receptor (BCR) mouse line, specific for the self-Thy-1/CD90 glycoprotein, we demonstrate that BCR crosslinking by low-dose self-antigen promotes survival of immature B cells in culture. In spleen, an increase in BCR signaling strength, induced by low-dose self-antigen, directed naive immature B cells to mature, not into the default follicular B cell fate, but instead into the marginal-zone B cell subset. These data indicate that positive selection can occur in developing B cells and that BCR signal strength is a key factor in deciding between two functionally distinct mature B cell compartments in the microenvironment of the spleen.
Collapse
Affiliation(s)
- Lijun Wen
- Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA
| | | | | | | | | | | |
Collapse
|