1
|
Elsner RA, Shlomchik MJ. Coordinated Regulation of Extrafollicular B Cell Responses by IL-12 and IFNγ. Immunol Rev 2025; 331:e70027. [PMID: 40211749 PMCID: PMC11986407 DOI: 10.1111/imr.70027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Accepted: 03/28/2025] [Indexed: 04/14/2025]
Abstract
Upon activation, B cells undergo either the germinal center (GC) or extrafollicular (EF) response. While GC are known to generate high-affinity memory B cells and long-lived plasma cells, the role of the EF response is less well understood. Initially, it was thought to be limited to that of a source of fast but lower-quality antibodies until the GC can form. However, recent evidence strongly supports the EF response as an important component of the humoral response to infection. EF responses are now also recognized as a source of pathogenic B cells in autoimmune diseases. The EF response itself is dynamic and regulated by pathways that are only recently being uncovered. We have identified that the cytokine IL-12 acts as a molecular switch, enhancing the EF response and suppressing GC through multiple mechanisms. These include direct effects on both B cells themselves and the coordinated differentiation of helper CD4 T cells. Here, we explore this pathway in relation to other recent advancements in our understanding of the EF response's role and highlight areas for future research. A better understanding of how the EF response forms and is regulated is essential for advancing treatments for many disease states.
Collapse
Affiliation(s)
- Rebecca A. Elsner
- Department of ImmunologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Mark J. Shlomchik
- Department of ImmunologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| |
Collapse
|
2
|
da Graça CG, Sheikh AA, Newman DM, Wen L, Li S, Shen J, Zhang Y, Gabriel SS, Chisanga D, Seow J, Poch A, Rausch L, Nguyen MHT, Singh J, Su CH, Cluse LA, Tsui C, Burn TN, Park SL, Von Scheidt B, Mackay LK, Vasanthakumar A, Bending D, Shi W, Cui W, Schröder J, Johnstone RW, Kallies A, Utzschneider DT. Stem-like memory and precursors of exhausted T cells share a common progenitor defined by ID3 expression. Sci Immunol 2025; 10:eadn1945. [PMID: 39888981 PMCID: PMC7617396 DOI: 10.1126/sciimmunol.adn1945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 12/23/2024] [Indexed: 02/02/2025]
Abstract
Stem-like T cells are attractive immunotherapeutic targets in patients with cancer given their ability to proliferate and differentiate into effector progeny. Thus, identifying T cells with enhanced stemness and understanding their developmental requirements are of broad clinical and therapeutic interest. Here, we demonstrate that during acute infection, the transcriptional regulator inhibitor of DNA binding 3 (ID3) identifies stem-like T cells that are uniquely adapted to generate precursors of exhausted T (Tpex) cells in response to chronic infection or cancer. Expression of ID3 itself enables Tpex cells to sustain T cell responses in chronic infection or cancer, whereas loss of ID3 results in impaired maintenance of CD8 T cell immunity. Furthermore, we demonstrate that interleukin-1 (IL-1) family members, including IL-36β and IL-18, promote the generation of ID3+ T cells that mediate superior tumor control. Overall, we identify ID3 as a common denominator of stem-like T cells in both acute and chronic infections that is specifically required to sustain T cell responses to chronic stimulation.
Collapse
Affiliation(s)
- Catarina Gago da Graça
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Amania A. Sheikh
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Dane M. Newman
- Cancer Biology and Therapeutics, Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Lifen Wen
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Sining Li
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Jian Shen
- Department of Pathology, Northwestern University, Chicago, IL
| | - Yuqi Zhang
- Department of Pathology, Northwestern University, Chicago, IL
| | - Sarah S. Gabriel
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - David Chisanga
- Olivia Newton-John Cancer Research Institute, Heidelberg, Australia
| | - Justine Seow
- Computational Sciences Initiative (CSI), The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Annika Poch
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Lisa Rausch
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Minh-Hanh T. Nguyen
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Jayendra Singh
- Olivia Newton-John Cancer Research Institute, Heidelberg, Australia
| | - Chun-Hsi Su
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Leonie A. Cluse
- Cancer Biology and Therapeutics, Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Carlson Tsui
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Thomas N. Burn
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Simone L. Park
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Bianca Von Scheidt
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Laura K. Mackay
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | | | - David Bending
- Department of Immunology and Immunotherapy, College of Medicine and Health, University of Birmingham, BirminghamB15 2TT, UK
| | - Wei Shi
- Olivia Newton-John Cancer Research Institute, Heidelberg, Australia
- School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - Weiguo Cui
- Department of Pathology, Northwestern University, Chicago, IL
| | - Jan Schröder
- Computational Sciences Initiative (CSI), The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Ricky W. Johnstone
- Cancer Biology and Therapeutics, Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Axel Kallies
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Daniel T. Utzschneider
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| |
Collapse
|
3
|
Kabrani E, Rahjouei A, Berruezo-Llacuna M, Ebeling S, Saha T, Altwasser R, Delgado-Benito V, Pavri R, Di Virgilio M. RIF1 integrates DNA repair and transcriptional requirements during the establishment of humoral immune responses. Nat Commun 2025; 16:777. [PMID: 39824820 PMCID: PMC11742068 DOI: 10.1038/s41467-025-56166-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 01/09/2025] [Indexed: 01/20/2025] Open
Abstract
The establishment of protective immune responses relies on the ability of terminally differentiated B cells to secrete a broad variety of antigen-specific antibodies with different effector functions. RIF1 is a multifunctional protein that promotes antibody isotype diversification via its DNA end protection activity during class switch recombination. In this study, we showed that RIF1 ablation resulted in increased plasmablast formation ex vivo and enhanced terminal differentiation into plasma cells upon immunization. Mechanistically, this phenotype is independent from RIF1's role in DNA repair and class switch recombination, and reflects its ability to modulate the transcriptional status of a subset of BLIMP1 target genes. Therefore, here we show that, in addition to promoting antibody diversification, RIF1 fine-tunes the kinetics of late B cell differentiation, thus providing an additional layer of control in the establishment of humoral immunity.
Collapse
Affiliation(s)
- Eleni Kabrani
- Laboratory of Genome Diversification & Integrity, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125, Berlin, Germany
| | - Ali Rahjouei
- Laboratory of Genome Diversification & Integrity, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125, Berlin, Germany
- Department of Anesthesiology and Intensive Care Medicine, and Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin, 10117, Berlin, Germany
| | - Maria Berruezo-Llacuna
- Laboratory of Genome Diversification & Integrity, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125, Berlin, Germany
- Humboldt-Universität zu Berlin, 10117, Berlin, Germany
| | - Svenja Ebeling
- Laboratory of Genome Diversification & Integrity, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125, Berlin, Germany
- Freie Universität Berlin, 14195, Berlin, Germany
| | - Tannishtha Saha
- Laboratory of Genome Diversification & Integrity, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125, Berlin, Germany
- Freie Universität Berlin, 14195, Berlin, Germany
| | - Robert Altwasser
- Laboratory of Genome Diversification & Integrity, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125, Berlin, Germany
- Department of Hematology, Oncology, and Cancer Immunology, Charité-Universitätsmedizin Berlin, 10117, Berlin, Germany
| | - Veronica Delgado-Benito
- Laboratory of Genome Diversification & Integrity, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125, Berlin, Germany
| | - Rushad Pavri
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, London, SE1 9RT, UK
| | - Michela Di Virgilio
- Laboratory of Genome Diversification & Integrity, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125, Berlin, Germany.
- Charité-Universitätsmedizin Berlin, 10117, Berlin, Germany.
| |
Collapse
|
4
|
Kose H, Akalin A, Kilic SS. From agammaglobulinemia to neutropenia: 'The TCF-3 has different clinical presentations'. Scand J Immunol 2024; 100:e13423. [PMID: 39523462 DOI: 10.1111/sji.13423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 09/27/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
The clinical picture of the TCF3 deficiency may manifest differently from neutropenia to antibody production defects.
Collapse
Affiliation(s)
- Hulya Kose
- Department of Pediatric Immunology and Allergy, Diyarbakir Children's Hospital, Diyarbakir, Turkey
| | - Akcahan Akalin
- Department of Pediatric Genetics, Diyarbakir Children's Hospital, Diyarbakir, Turkey
| | - Sara Sebnem Kilic
- Department of Pediatric Immunology and Rheumatology, Bursa Uludag University Faculty of Medicine, Bursa, Turkey
| |
Collapse
|
5
|
Poveda-Garavito N, Orozco Castaño CA, Torres-Llanos Y, Cruz-Rodriguez N, Parra-Medina R, Quijano S, Zabaleta J, Combita AL. ID1 and ID3 functions in the modulation of the tumour immune microenvironment in adult patients with B-cell acute lymphoblastic leukaemia. Front Immunol 2024; 15:1473909. [PMID: 39676870 PMCID: PMC11638060 DOI: 10.3389/fimmu.2024.1473909] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/07/2024] [Indexed: 12/17/2024] Open
Abstract
Introduction B-cell acute lymphoblastic leukemia (B-ALL) in adults often presents a poor prognosis. ID1 and ID3 genes have been identified as predictors of poor response in Colombian adult B-ALL patients, contributing to cancer development. In various cancer models, these genes have been associated with immune regulatory populations within the tumor immune microenvironment (TIME). B-ALL progression alters immune cell composition and the bone marrow (BM) microenvironment, impacting disease progression and therapy response. This study investigates the relationship between ID1 and ID3 expression, TIME dynamics, and immune evasion mechanisms in adult B-ALL patients. Methods This exploratory study analysed BM samples from 10 B-ALL adult patients diagnosed at the National Cancer Institute of Colombia. First, RT-qPCR was used to assess ID1 and ID3 expression in BM tumour cells. Flow cytometry characterised immune populations in the TIME. RNA-seq evaluated immune genes associatedwith B-ALL immune response, while xCell and CytoSig analysed TIME cell profiles and cytokines. Pathway analysis, gene ontology, and differential gene expression (DEGs) were examined, with functional enrichment analysis performed using KEGG ontology. Results Patients were divided into two groups based on ID1 and ID3 expression, namely basal and overexpression. A total of 94 differentially expressed genes were identified between these groups, with top overexpressed genes associated with neutrophil pathways. Gene set enrichment analysis revealed increased expression of genes associated with neutrophil degranulation, immune response-related neutrophil activation, and neutrophil-mediated immunity. These findings correlated with xCell data. Overexpression group showed significant differences in neutrophils, monocytes and CD4+ naive T cells compared to basal group patients. Microenvironment and immune scores were also significantly different, consistent with the flow cytometry results. Elevated cytokine levels associated with neutrophil activation supported these findings. Validation was performed using the Therapeutically Applicable Research to Generate Effective Treatments (TARGET) TCGA B-ALL cohorts. Discussion These findings highlight significant differences in ID1 and ID3 expression levels and their impact on TIME populations, particularly neutrophil-related pathways. The results suggest a potential role for ID1 and ID3 in immune evasion in adult B-ALL, mediated through neutrophil activation and immune regulation.
Collapse
Affiliation(s)
- Nathaly Poveda-Garavito
- Grupo de Investigación en Biología del Cáncer - Instituto Nacional de Cancerología, Bogotá, Colombia
- Grupo de Investigación Traslacional en Oncología - Instituto Nacional de Cancerología, Bogotá, Colombia
- Maestría en Inmunología, Departamento de Microbiología - Universidad Nacional de Colombia, Bogotá, Colombia
| | - Carlos A Orozco Castaño
- Grupo de Investigación en Biología del Cáncer - Instituto Nacional de Cancerología, Bogotá, Colombia
- Grupo de Investigación Traslacional en Oncología - Instituto Nacional de Cancerología, Bogotá, Colombia
| | - Yulieth Torres-Llanos
- Grupo de Investigación en Biología del Cáncer - Instituto Nacional de Cancerología, Bogotá, Colombia
- Grupo de Investigación Traslacional en Oncología - Instituto Nacional de Cancerología, Bogotá, Colombia
- Laboratorio clínico, Hospital Universitario San Ignacio, Bogotá, Colombia
| | | | - Rafael Parra-Medina
- Departamento de Patología, Instituto Nacional de Cancerología, Bogotá, Colombia
- Research Institute, Fundación Universitaria de Ciencias de la Salud - FUCS, Bogotá, Colombia
| | - Sandra Quijano
- Grupo de Inmunobiología y Biología Celular, Departamento de Microbiología, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Jovanny Zabaleta
- Department of Interdisciplinary Oncology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Alba Lucia Combita
- Grupo de Investigación en Biología del Cáncer - Instituto Nacional de Cancerología, Bogotá, Colombia
- Grupo de Investigación Traslacional en Oncología - Instituto Nacional de Cancerología, Bogotá, Colombia
- Maestría en Inmunología, Departamento de Microbiología - Universidad Nacional de Colombia, Bogotá, Colombia
| |
Collapse
|
6
|
Etebar F, Whatmore P, Harkin DG, Dando SJ. Tissue-Specific Immune Transcriptional Signatures in the Bordering Tissues of the Mouse Retina and Brain. Invest Ophthalmol Vis Sci 2024; 65:42. [PMID: 39466230 PMCID: PMC11514940 DOI: 10.1167/iovs.65.12.42] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 09/25/2024] [Indexed: 10/29/2024] Open
Abstract
Purpose Bordering the central nervous system (CNS) parenchyma are the choroid (underlying the retina) and the leptomeninges (the inner layers of the meninges enveloping the brain). Although near the neural parenchyma, the choroid and leptomeninges are external to the immune privileged environment of the retina and brain and thus are distinct immune compartments. This study aimed to characterize the transcriptomic signatures of immune cells within the choroid and leptomeninges bordering the healthy adult mouse CNS. Methods Eyes and brains were obtained from 7-week-old C57Bl/6J mice. Choroid and leptomeninges were processed for isolation of CD45+ immune cells and single cell RNA-sequencing. Additionally, single cell RNA-sequencing was performed on immune cells isolated from choroid obtained from human donor eye tissue. Immunostaining and confocal microscopy of wholemount tissue were used to validate selected immune cell populations in situ. Results A total of 3606 cells were sequenced from mouse tissues, including 2125 CD45+ cells from choroid and 1481 CD45+ cells from leptomeninges. Clustering and differential gene expression analysis revealed heterogeneous subtypes of monocytes/macrophages, dendritic cells, T cells, and B cells. Whereas some clusters were common to both choroid and leptomeninges, others exhibited tissue-specific gene expression profiles and potential functional specializations. Analysis of 6501 CD45+ cells sequenced from human choroid identified similar immune cell populations to mouse choroid. Conclusions This study provides a detailed characterization of the molecular signatures of immune cells within the vascular connective tissues bordering the healthy retina and brain, and their potential roles in immune protection.
Collapse
Affiliation(s)
- Fazeleh Etebar
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia
| | - Paul Whatmore
- eResearch Office, Research Infrastructure, Queensland University of Technology, Brisbane, Australia
| | - Damien G. Harkin
- Centre for Vision and Eye Research, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia
| | - Samantha J. Dando
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia
- Centre for Vision and Eye Research, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia
| |
Collapse
|
7
|
Hillion S, Miranda A, Le Dantec C, Boudigou M, Le Pottier L, Cornec D, Torres RM, Pelanda R. Maf expression in B cells restricts reactive plasmablast and germinal center B cell expansion. Nat Commun 2024; 15:7982. [PMID: 39266537 PMCID: PMC11393457 DOI: 10.1038/s41467-024-52224-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 08/29/2024] [Indexed: 09/14/2024] Open
Abstract
Precise regulation of B cell differentiation is essential for an effective adaptive immune response. Here, we show that B cell development in mice with B cell-specific Maf deletion is unaffected, but marginal zone B cells, germinal centre B cells, and plasmablasts are significantly more frequent in the spleen of naive Maf-deficient mice compared to wild type controls. In the context of a T cell-dependent immunization, Maf deletion causes increased proliferation of germinal centre B cells and extrafollicular plasmablasts. This is accompanied by higher production of antigen-specific IgG1 antibodies with minimal modification of early memory B cells, but a reduction in plasma cell numbers. Single-cell RNA sequencing shows upregulation of genes associated with DNA replication and cell cycle progression, confirming the role of Maf in cell proliferation. Subsequent pathway analysis reveals that Maf influences cellular metabolism, transporter activity, and mitochondrial proteins, which have been implicated in controlling the germinal centre reaction. In summary, our findings demonstrate that Maf acts intrinsically in B cells as a negative regulator of late B cell differentiation, plasmablast proliferation and germinal centre B cell formation.
Collapse
Affiliation(s)
- Sophie Hillion
- LBAI, UMR1227, Univ Brest, Inserm, and CHU de Brest, Brest, France.
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, 80045, USA.
| | - Anjelica Miranda
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | | | | | | | - Divi Cornec
- LBAI, UMR1227, Univ Brest, Inserm, and CHU de Brest, Brest, France
| | - Raul M Torres
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Roberta Pelanda
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, 80045, USA
| |
Collapse
|
8
|
Wang X, Liu H, Fei Y, Song Z, Meng X, Yu J, Liu X, Li L, Qiu L, Qian Z, Zhou S, Wang X, Zhang H. Metabolic pathway-based subtyping reveals distinct microenvironmental states associated with diffuse large B-cell lymphoma outcomes. Hematol Oncol 2024; 42:e3279. [PMID: 38819002 DOI: 10.1002/hon.3279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 03/22/2024] [Accepted: 05/01/2024] [Indexed: 06/01/2024]
Abstract
Diffuse large B-cell lymphoma (DLBCL) is a biologically and clinically heterogeneous disease that requires personalized clinical treatment. Assigning patients to different risk categories and cytogenetic abnormality and genetic mutation groups has been widely applied for prognostic stratification of DLBCL. Increasing evidence has demonstrated that dysregulated metabolic processes contribute to the initiation and progression of DLBCL. Metabolic competition within the tumor microenvironment is also known to influence immune cell metabolism. However, metabolism- and immune-related stratification has not been established. Here, 1660 genes involved in 84 metabolic pathways were selected and tested to establish metabolic clusters (MECs) of DLBCL. MECs established based on independent lymphoma datasets distinguished different survival outcomes. The CIBERSORT algorithm and EcoTyper were applied to quantify the relative abundance of immune cell types and identify variation in cell states for 13 lineages comprising the tumor micro environment among different MECs, respectively. Functional characterization showed that MECs were an indicator of the immune microenvironment and correlated with distinctive mutational characteristics and oncogenic signaling pathways. The novel immune-related MECs exhibited promising clinical prognostic value and potential for informing DLBCL treatment decisions.
Collapse
Affiliation(s)
- Xiaohui Wang
- National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine and Department of Lymphoma, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, The Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| | - Hengqi Liu
- National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine and Department of Lymphoma, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, The Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| | - Yue Fei
- National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine and Department of Lymphoma, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, The Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| | - Zheng Song
- National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine and Department of Lymphoma, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, The Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| | - Xiangrui Meng
- National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine and Department of Lymphoma, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, The Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| | - Jingwei Yu
- National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine and Department of Lymphoma, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, The Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| | - Xia Liu
- National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine and Department of Lymphoma, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, The Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| | - Lanfang Li
- National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine and Department of Lymphoma, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, The Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| | - Lihua Qiu
- National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine and Department of Lymphoma, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, The Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| | - Zhengzi Qian
- National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine and Department of Lymphoma, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, The Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| | - Shiyong Zhou
- National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine and Department of Lymphoma, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, The Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| | - Xianhuo Wang
- National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine and Department of Lymphoma, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, The Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| | - Huilai Zhang
- National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine and Department of Lymphoma, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, The Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| |
Collapse
|
9
|
Elsner RA, Smita S, Shlomchik MJ. IL-12 induces a B cell-intrinsic IL-12/IFNγ feed-forward loop promoting extrafollicular B cell responses. Nat Immunol 2024; 25:1283-1295. [PMID: 38862796 PMCID: PMC11992614 DOI: 10.1038/s41590-024-01858-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 04/26/2024] [Indexed: 06/13/2024]
Abstract
While some infections elicit germinal centers, others produce only extrafollicular responses. The mechanisms controlling these dichotomous fates are poorly understood. We identify IL-12 as a cytokine switch, acting directly on B cells to promote extrafollicular and suppress germinal center responses. IL-12 initiates a B cell-intrinsic feed-forward loop between IL-12 and IFNγ, amplifying IFNγ production, which promotes proliferation and plasmablast differentiation from mouse and human B cells, in synergy with IL-12. IL-12 sustains the expression of a portion of IFNγ-inducible genes. Together, they also induce unique gene changes, reflecting both IFNγ amplification and cooperative effects between both cytokines. In vivo, cells lacking both IL-12 and IFNγ receptors are more impaired in plasmablast production than those lacking either receptor alone. Further, B cell-derived IL-12 enhances both plasmablast responses and T helper 1 cell commitment. Thus, B cell-derived IL-12, acting on T and B cells, determines the immune response mode, with implications for vaccines, pathogen protection and autoimmunity.
Collapse
Affiliation(s)
- Rebecca A Elsner
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Shuchi Smita
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mark J Shlomchik
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
10
|
Steinmetz T, Thomas J, Reimann L, Himmelreich AK, Schulz SR, Golombek F, Castiglione K, Jäck HM, Brodesser S, Warscheid B, Mielenz D. Identification of TFG- and Autophagy-Regulated Proteins and Glycerophospholipids in B Cells. J Proteome Res 2024; 23:1615-1633. [PMID: 38649144 PMCID: PMC11077586 DOI: 10.1021/acs.jproteome.3c00713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 03/01/2024] [Accepted: 03/06/2024] [Indexed: 04/25/2024]
Abstract
Autophagy supervises the proteostasis and survival of B lymphocytic cells. Trk-fused gene (TFG) promotes autophagosome-lysosome flux in murine CH12 B cells, as well as their survival. Hence, quantitative proteomics of CH12tfgKO and WT B cells in combination with lysosomal inhibition should identify proteins that are prone to lysosomal degradation and contribute to autophagy and B cell survival. Lysosome inhibition via NH4Cl unexpectedly reduced a number of proteins but increased a large cluster of translational, ribosomal, and mitochondrial proteins, independent of TFG. Hence, we propose a role for lysosomes in ribophagy in B cells. TFG-regulated proteins include CD74, BCL10, or the immunoglobulin JCHAIN. Gene ontology (GO) analysis reveals that proteins regulated by TFG alone, or in concert with lysosomes, localize to mitochondria and membrane-bound organelles. Likewise, TFG regulates the abundance of metabolic enzymes, such as ALDOC and the fatty acid-activating enzyme ACOT9. To test consequently for a function of TFG in lipid metabolism, we performed shotgun lipidomics of glycerophospholipids. Total phosphatidylglycerol is more abundant in CH12tfgKO B cells. Several glycerophospholipid species with similar acyl side chains, such as 36:2 phosphatidylethanolamine and 36:2 phosphatidylinositol, show a dysequilibrium. We suggest a role for TFG in lipid homeostasis, mitochondrial functions, translation, and metabolism in B cells.
Collapse
Affiliation(s)
- Tobit
D. Steinmetz
- Division
of Molecular Immunology, Department of Internal Medicine 3, Nikolaus-Fiebiger-Zentrum, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | - Jana Thomas
- Division
of Molecular Immunology, Department of Internal Medicine 3, Nikolaus-Fiebiger-Zentrum, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | - Lena Reimann
- CIBSS
Centre for Integrative Biological Signalling Studies, University of Freiburg, D-79104 Freiburg, Germany
| | - Ann-Kathrin Himmelreich
- Division
of Molecular Immunology, Department of Internal Medicine 3, Nikolaus-Fiebiger-Zentrum, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | - Sebastian R. Schulz
- Division
of Molecular Immunology, Department of Internal Medicine 3, Nikolaus-Fiebiger-Zentrum, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | - Florian Golombek
- Chair
of Bioprocess Engineering, Technical Faculty, FAU Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | - Kathrin Castiglione
- Chair
of Bioprocess Engineering, Technical Faculty, FAU Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | - Hans-Martin Jäck
- Division
of Molecular Immunology, Department of Internal Medicine 3, Nikolaus-Fiebiger-Zentrum, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, D-91054 Erlangen, Germany
- FAU
Profile Center Immunomedicine (FAU I-MED), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, D-91054 Erlangen, Germany
| | - Susanne Brodesser
- Cologne
Excellence Cluster on Cellular Stress Responses in Aging-associated
Diseases (CECAD), University of Köln, D-50931 Köln, Germany
| | - Bettina Warscheid
- CIBSS
Centre for Integrative Biological Signalling Studies, University of Freiburg, D-79104 Freiburg, Germany
- Department
of Biochemistry, Theodor Boveri-Institute, Biocenter, University of Würzburg, D-97074 Würzburg, Germany
| | - Dirk Mielenz
- Division
of Molecular Immunology, Department of Internal Medicine 3, Nikolaus-Fiebiger-Zentrum, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, D-91054 Erlangen, Germany
- FAU
Profile Center Immunomedicine (FAU I-MED), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, D-91054 Erlangen, Germany
| |
Collapse
|
11
|
Boast B, Goel S, González-Granado LI, Niemela J, Stoddard J, Edwards ESJ, Seneviratne S, Spensberger D, Quesada-Espinosa JF, Allende LM, McDonnell J, Haseley A, Lesmana H, Walkiewicz MA, Muhammad E, Bosco JJ, Fleisher TA, Cohen S, Holland SM, van Zelm MC, Enders A, Kuehn HS, Rosenzweig SD. TCF3 haploinsufficiency defined by immune, clinical, gene-dosage, and murine studies. J Allergy Clin Immunol 2023; 152:736-747. [PMID: 37277074 PMCID: PMC10527523 DOI: 10.1016/j.jaci.2023.05.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/01/2023] [Accepted: 05/08/2023] [Indexed: 06/07/2023]
Abstract
BACKGROUND TCF3 is a transcription factor contributing to early lymphocyte differentiation. Germline monoallelic dominant negative and biallelic loss-of-function (LOF) null TCF3 mutations cause a fully penetrant severe immunodeficiency. We identified 8 individuals from 7 unrelated families with monoallelic LOF TCF3 variants presenting with immunodeficiency with incomplete clinical penetrance. OBJECTIVE We sought to define TCF3 haploinsufficiency (HI) biology and its association with immunodeficiency. METHODS Patient clinical data and blood samples were analyzed. Flow cytometry, Western blot analysis, plasmablast differentiation, immunoglobulin secretion, and transcriptional activity studies were conducted on individuals carrying TCF3 variants. Mice with a heterozygous Tcf3 deletion were analyzed for lymphocyte development and phenotyping. RESULTS Individuals carrying monoallelic LOF TCF3 variants showed B-cell defects (eg, reduced total, class-switched memory, and/or plasmablasts) and reduced serum immunoglobulin levels; most but not all presented with recurrent but nonsevere infections. These TCF3 LOF variants were either not transcribed or translated, resulting in reduced wild-type TCF3 protein expression, strongly suggesting HI pathophysiology for the disease. Targeted RNA sequencing analysis of T-cell blasts from TCF3-null, dominant negative, or HI individuals clustered away from healthy donors, implying that 2 WT copies of TCF3 are needed to sustain a tightly regulated TCF3 gene-dosage effect. Murine TCF3 HI resulted in a reduction of circulating B cells but overall normal humoral immune responses. CONCLUSION Monoallelic LOF TCF3 mutations cause a gene-dosage-dependent reduction in wild-type protein expression, B-cell defects, and a dysregulated transcriptome, resulting in immunodeficiency. Tcf3+/- mice partially recapitulate the human phenotype, underscoring the differences between TCF3 in humans and mice.
Collapse
Affiliation(s)
- Brigette Boast
- Immunology Service, Department of Laboratory Medicine, National Institutes of Health Clinical Center, Bethesda, Md
| | - Shubham Goel
- Immunology Service, Department of Laboratory Medicine, National Institutes of Health Clinical Center, Bethesda, Md
| | - Luis I González-Granado
- Department of Pediatrics, Hospital 12 de Octubre, Research Institute Hospital 12 de Octubre (i+12), School of Medicine, Complutense University, Madrid, Spain
| | - Julie Niemela
- Immunology Service, Department of Laboratory Medicine, National Institutes of Health Clinical Center, Bethesda, Md
| | - Jennifer Stoddard
- Immunology Service, Department of Laboratory Medicine, National Institutes of Health Clinical Center, Bethesda, Md
| | - Emily S J Edwards
- Department of Immunology, Monash University, and The Jeffrey Modell Diagnostic and Research Centre for Primary Immunodeficiencies, Melbourne, Australia
| | - Sandali Seneviratne
- Centre for Personalised Immunology and Division of Immunology and Infectious Disease, John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Dominik Spensberger
- ANU Gene Targeting Facility, Australian Phenomics Facility, John Curtin School of Medical Research, Australian National University, Canberra, Australia
| | | | - Luis M Allende
- Department of Immunology, Hospital 12 de Octubre, Research Institute Hospital 12 de Octubre (i+12), Madrid, Spain
| | - John McDonnell
- Department of Pediatric Allergy and Immunology, Cleveland Clinic, Cleveland, Ohio
| | - Alexandria Haseley
- Center for Personalized Genetic Healthcare, Genomic Medicine Institute, Cleveland Clinic, Cleveland, Ohio
| | - Harry Lesmana
- Center for Personalized Genetic Healthcare, Genomic Medicine Institute, Cleveland Clinic, Cleveland, Ohio; Department of Pediatric Hematology, Oncology and Bone Marrow Transplantation, Cleveland Clinic, Cleveland, Ohio
| | - Magdalena A Walkiewicz
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Emad Muhammad
- Hematology Laboratory, Carmel Medical Center, Haifa, Spain
| | - Julian J Bosco
- Allergy, Asthma and Clinical Immunology Service, Alfred Hospital, Melbourne, Australia
| | - Thomas A Fleisher
- Immunology Service, Department of Laboratory Medicine, National Institutes of Health Clinical Center, Bethesda, Md
| | - Shai Cohen
- Allergy and Clinical Immunology Service, Department of Internal Medicine B, Lin and Carmel Medical Center, The Technion, Israel Institute of Technology, Haifa, Israel
| | - Steven M Holland
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Menno C van Zelm
- Department of Immunology, Monash University, and The Jeffrey Modell Diagnostic and Research Centre for Primary Immunodeficiencies, Melbourne, Australia; Allergy, Asthma and Clinical Immunology Service, Alfred Hospital, Melbourne, Australia
| | - Anselm Enders
- Centre for Personalised Immunology and Division of Immunology and Infectious Disease, John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Hye Sun Kuehn
- Immunology Service, Department of Laboratory Medicine, National Institutes of Health Clinical Center, Bethesda, Md
| | - Sergio D Rosenzweig
- Immunology Service, Department of Laboratory Medicine, National Institutes of Health Clinical Center, Bethesda, Md.
| |
Collapse
|
12
|
Oras A, Kallionpää H, Suomi T, Koskinen S, Laiho A, Elo LL, Knip M, Lahesmaa R, Aints A, Uibo R, DIABIMMUNE Study Group. Profiling of peripheral blood B-cell transcriptome in children who developed coeliac disease in a prospective study. Heliyon 2023; 9:e13147. [PMID: 36718152 PMCID: PMC9883278 DOI: 10.1016/j.heliyon.2023.e13147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 12/20/2022] [Accepted: 01/17/2023] [Indexed: 01/22/2023] Open
Abstract
Background In coeliac disease (CoD), the role of B-cells has mainly been considered to be production of antibodies. The functional role of B-cells has not been analysed extensively in CoD. Methods We conducted a study to characterize gene expression in B-cells from children developing CoD early in life using samples collected before and at the diagnosis of the disease. Blood samples were collected from children at risk at 12, 18, 24 and 36 months of age. RNA from peripheral blood CD19+ cells was sequenced and differential gene expression was analysed using R package Limma. Findings Overall, we found one gene, HNRNPL, modestly downregulated in all patients (logFC -0·7; q = 0·09), and several others downregulated in those diagnosed with CoD already by the age of 2 years. Interpretation The data highlight the role of B-cells in CoD development. The role of HNRPL in suppressing enteroviral replication suggests that the predisposing factor for both CoD and enteroviral infections is the low level of HNRNPL expression. Funding EU FP7 grant no. 202063, EU Regional Developmental Fund and research grant PRG712, The Academy of Finland Centre of Excellence in Molecular Systems Immunology and Physiology Research (SyMMyS) 2012-2017, grant no. 250114) and, AoF Personalized Medicine Program (grant no. 292482), AoF grants 292335, 294337, 319280, 31444, 319280, 329277, 331790) and grants from the Sigrid Jusélius Foundation (SJF).
Collapse
Affiliation(s)
- Astrid Oras
- Department of Immunology, Institute of Biomedicine and Translational Medicine, University of Tartu, Estonia
| | - Henna Kallionpää
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Finland
| | - Tomi Suomi
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Finland,InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Satu Koskinen
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Finland
| | - Asta Laiho
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Finland,InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Laura L. Elo
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Finland,InFLAMES Research Flagship Center, University of Turku, Turku, Finland,Institute of Biomedicine, University of Turku, Finland
| | - Mikael Knip
- Pediatric Research Center, Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland,Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland,Center for Child Health Research, Tampere University Hospital, Tampere, Finland
| | - Riitta Lahesmaa
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Finland,InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Alar Aints
- Department of Immunology, Institute of Biomedicine and Translational Medicine, University of Tartu, Estonia,Corresponding author. Department of Immunology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, EE50411, Tartu, Estonia.
| | - Raivo Uibo
- Department of Immunology, Institute of Biomedicine and Translational Medicine, University of Tartu, Estonia
| | | |
Collapse
|
13
|
A p38α-BLIMP1 signalling pathway is essential for plasma cell differentiation. Nat Commun 2022; 13:7321. [PMID: 36443297 PMCID: PMC9703440 DOI: 10.1038/s41467-022-34969-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 11/11/2022] [Indexed: 11/29/2022] Open
Abstract
Plasma cells (PC) are antibody-secreting cells and terminal effectors in humoral responses. PCs differentiate directly from activated B cells in response to T cell-independent (TI) antigens or from germinal center B (GCB) cells in T cell-dependent (TD) antigen-induced humoral responses, both of which pathways are essentially regulated by the transcription factor BLIMP1. The p38 mitogen-activated protein kinase isoforms have already been implicated in B cell development, but the precise role of p38α in B cell differentiation is still largely unknown. Here we show that PC differentiation and antibody responses are severely impaired in mice with B cell-specific deletion of p38α, while B cell development and the GCB cell response are spared. By utilizing a Blimp1 reporter mouse model, we show that p38α-deficiency results in decreased BLIMP1 expression. p38α-driven BLIMP1 up-regulation is required for both TI and TD PCs differentiation. By combining CRISPR/Cas9 screening and other approaches, we identify TCF3, TCF4 and IRF4 as downstream effectors of p38α to control PC differentiation via Blimp1 transcription. This study thus identifies an important signalling pathway underpinning PC differentiation upstream of BLIMP1, and points to a highly specialized and non-redundant role for p38α among p38 isoforms.
Collapse
|
14
|
Asashima H, Axisa PP, Pham THG, Longbrake EE, Ruff WE, Lele N, Cohen I, Raddassi K, Sumida TS, Hafler DA. Impaired TIGIT expression on B cells drives circulating follicular helper T cell expansion in multiple sclerosis. J Clin Invest 2022; 132:156254. [PMID: 36250467 PMCID: PMC9566906 DOI: 10.1172/jci156254] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 08/25/2022] [Indexed: 11/17/2022] Open
Abstract
B cell depletion in patients with relapsing-remitting multiple sclerosis (RRMS) markedly prevents new MRI-detected lesions and disease activity, suggesting the hypothesis that altered B cell function leads to the activation of T cells driving disease pathogenesis. Here, we performed comprehensive analyses of CD40 ligand- (CD40L-) and IL-21-stimulated memory B cells from patients with MS and healthy age-matched controls, modeling the help of follicular helper T cells (Tfh cells), and found a differential gene expression signature in multiple B cell pathways. Most striking was the impaired TIGIT expression on MS-derived B cells mediated by dysregulation of the transcription factor TCF4. Activated circulating Tfh cells (cTfh cells) expressed CD155, the ligand of TIGIT, and TIGIT on B cells revealed their capacity to suppress the proliferation of IL-17-producing cTfh cells via the TIGIT/CD155 axis. Finally, CCR6+ cTfh cells were significantly increased in patients with MS, and their frequency was inversely correlated with that of TIGIT+ B cells. Together, these data suggest that the dysregulation of negative feedback loops between TIGIT+ memory B cells and cTfh cells in MS drives the activated immune system in this disease.
Collapse
|
15
|
Gupta S, Fink MK, Kempuraj D, Sinha NR, Martin LM, Keele LM, Sinha PR, Giuliano EA, Hesemann NP, Raikwar SP, Chaurasia SS, Mohan RR. Corneal fibrosis abrogation by a localized AAV-mediated inhibitor of differentiation 3 (Id3) gene therapy in rabbit eyes in vivo. Mol Ther 2022; 30:3257-3269. [PMID: 35780298 PMCID: PMC9552811 DOI: 10.1016/j.ymthe.2022.06.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 05/18/2022] [Accepted: 06/29/2022] [Indexed: 11/28/2022] Open
Abstract
Previously we found that inhibitor of differentiation 3 (Id3) gene, a transcriptional repressor, efficiently inhibits corneal keratocyte differentiation to myofibroblasts in vitro. This study evaluated the potential of adeno-associated virus 5 (AAV5)-mediated Id3 gene therapy to treat corneal scarring using an established rabbit in vivo disease model. Corneal scarring/fibrosis in rabbit eyes was induced by alkali trauma, and 24 h thereafter corneas were administered with either balanced salt solution AAV5-naked vector, or AAV5-Id3 vector (n = 6/group) via an optimized reported method. Therapeutic effects of AAV5-Id3 gene therapy on corneal pathology and ocular health were evaluated with clinical, histological, and molecular techniques. Localized AAV5-Id3 gene therapy significantly inhibited corneal fibrosis/haze clinically from 2.7 to 0.7 on the Fantes scale in live animals (AAV5-naked versus AAV5-Id3; p < 0.001). Furthermore, AAV5-Id3 treatment significantly reduced profibrotic gene mRNA levels: α-smooth muscle actin (α-SMA) (2.8-fold; p < 0.001), fibronectin (3.2-fold; p < 0.001), collagen I (0.8-fold; p < 0.001), and collagen III (1.4-fold; p < 0.001), as well as protein levels of α-SMA (23.8%; p < 0.001) and collagens (1.8-fold; p < 0.001). The anti-fibrotic activity of AAV5-Id3 is attributed to reduced myofibroblast formation by disrupting the binding of E-box proteins to the promoter of α-SMA, a transforming growth factor-β signaling downstream target gene. In conclusion, these results indicate that localized AAV5-Id3 delivery in stroma caused no clinically relevant ocular symptoms or corneal cellular toxicity in the rabbit eyes.
Collapse
Affiliation(s)
- Suneel Gupta
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO 65201, USA; Departments of Veterinary Medicine & Surgery and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, 1600 East Rollins Street, Columbia, MO 65211, USA
| | - Michael K Fink
- Department of Pathology, School of Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Duraisamy Kempuraj
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO 65201, USA; Departments of Veterinary Medicine & Surgery and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, 1600 East Rollins Street, Columbia, MO 65211, USA
| | - Nishant R Sinha
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO 65201, USA; Departments of Veterinary Medicine & Surgery and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, 1600 East Rollins Street, Columbia, MO 65211, USA
| | - Lynn M Martin
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO 65201, USA; Departments of Veterinary Medicine & Surgery and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, 1600 East Rollins Street, Columbia, MO 65211, USA
| | - Landon M Keele
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO 65201, USA; Departments of Veterinary Medicine & Surgery and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, 1600 East Rollins Street, Columbia, MO 65211, USA
| | - Prashant R Sinha
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO 65201, USA; Departments of Veterinary Medicine & Surgery and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, 1600 East Rollins Street, Columbia, MO 65211, USA
| | - Elizabeth A Giuliano
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO 65201, USA; Departments of Veterinary Medicine & Surgery and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, 1600 East Rollins Street, Columbia, MO 65211, USA
| | - Nathan P Hesemann
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO 65201, USA; Department of Pathology, School of Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Sudhanshu P Raikwar
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO 65201, USA; Departments of Veterinary Medicine & Surgery and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, 1600 East Rollins Street, Columbia, MO 65211, USA
| | - Shyam S Chaurasia
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO 65201, USA; Departments of Veterinary Medicine & Surgery and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, 1600 East Rollins Street, Columbia, MO 65211, USA; Department of Ophthalmology & Visual Sciences, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Rajiv R Mohan
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO 65201, USA; Departments of Veterinary Medicine & Surgery and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, 1600 East Rollins Street, Columbia, MO 65211, USA; Mason Eye Institute, School of Medicine, University of Missouri, 1600 East Rollins Street, Columbia, MO 65212, USA.
| |
Collapse
|
16
|
Aubrey M, Warburg ZJ, Murre C. Helix-Loop-Helix Proteins in Adaptive Immune Development. Front Immunol 2022; 13:881656. [PMID: 35634342 PMCID: PMC9134016 DOI: 10.3389/fimmu.2022.881656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/04/2022] [Indexed: 11/13/2022] Open
Abstract
The E/ID protein axis is instrumental for defining the developmental progression and functions of hematopoietic cells. The E proteins are dimeric transcription factors that activate gene expression programs and coordinate changes in chromatin organization. Id proteins are antagonists of E protein activity. Relative levels of E/Id proteins are modulated throughout hematopoietic development to enable the progression of hematopoietic stem cells into multiple adaptive and innate immune lineages including natural killer cells, B cells and T cells. In early progenitors, the E proteins promote commitment to the T and B cell lineages by orchestrating lineage specific programs of gene expression and regulating VDJ recombination of antigen receptor loci. In mature B cells, the E/Id protein axis functions to promote class switch recombination and somatic hypermutation. E protein activity further regulates differentiation into distinct CD4+ and CD8+ T cells subsets and instructs mature T cell immune responses. In this review, we discuss how the E/Id proteins define the adaptive immune system lineages, focusing on their role in directing developmental gene programs.
Collapse
Affiliation(s)
| | | | - Cornelis Murre
- Division of Biological Sciences, Section of Molecular Biology, University of California, San Diego, San Diego, CA, United States
| |
Collapse
|
17
|
Bonduelle O, Chaudesaigues C, Tolazzi M, Suleiman E, de Bernard S, Alves K, Nourikyan J, Bohec M, Baudrin LG, Katinger D, Debré P, Scarlatti G, Vieillard V, Combadière B. Dichotomy in Neutralizing Antibody Induction to Peptide-Conjugated Vaccine in Squalene Emulsion Contrast With Aluminum Hydroxide Formulation. Front Immunol 2022; 13:848571. [PMID: 35464449 PMCID: PMC9021396 DOI: 10.3389/fimmu.2022.848571] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 02/22/2022] [Indexed: 01/28/2023] Open
Abstract
W614A-3S peptide is a modified 3S motif of the HIV-gp41 (mutation W614A). We previously detected the presence of natural neutralizing antibodies directed against W614A-3S peptide (NAbs) in long-term non-progressor HIV+ patients. Here, we compared the efficacy of W614A-3S peptide formulated in either squalene emulsion (SQE) or in aluminum hydroxide (Alum) in inducing broadly-NAbs (bNAbs). Rabbit and mouse models were used to screen the induction of bNAbs following 4 immunizations. SQE was more efficient than Alum formulation in inducing W614A-3S-specific bNAbs with up to 67%-93% of HIV strains neutralized. We then analyzed the quality of peptide-specific murine B cells by single-cell gene expression by quantitative reverse transcription-PCR and single-cell V(D)J sequencing. We found more frequent germinal center B cells in SQE than in Alum, albeit with a different gene expression profile. The V(D)J sequencing of W614A-3S-specific BCR showed significant differences in BCR sequences and validates the dichotomy between adjuvant formulations. All sixteen BCR sequences which were cloned were specific of peptide. Adjuvant formulations of W614A-3S-peptide-conjugated immunogen impact the quantity and quality of B cell immune responses at both the gene expression level and BCR sequence.
Collapse
Affiliation(s)
- Olivia Bonduelle
- Sorbonne Université, Institut national de la santé et de la recherche médicale (Inserm) U1135, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Chloé Chaudesaigues
- Sorbonne Université, Institut national de la santé et de la recherche médicale (Inserm) U1135, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Monica Tolazzi
- Viral Evolution and Transmission Unit, Division of Immunology, Transplantation and Infectious Diseases, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Ehsan Suleiman
- Polymun Scientific Immunbiologische Forschung GmbH, Klosterneuburg, Austria
| | | | | | | | - Mylene Bohec
- Institut Curie, Genomics of Excellence (ICGex) Platform, Paris Science et Lettres (PSL) Research University, Paris, France
| | - Laura G Baudrin
- Institut Curie, Genomics of Excellence (ICGex) Platform, Paris Science et Lettres (PSL) Research University, Paris, France
| | - Dietmar Katinger
- Polymun Scientific Immunbiologische Forschung GmbH, Klosterneuburg, Austria
| | - Patrice Debré
- Sorbonne Université, Institut national de la santé et de la recherche médicale (Inserm) U1135, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Gabriella Scarlatti
- Viral Evolution and Transmission Unit, Division of Immunology, Transplantation and Infectious Diseases, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Vincent Vieillard
- Sorbonne Université, Institut national de la santé et de la recherche médicale (Inserm) U1135, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Behazine Combadière
- Sorbonne Université, Institut national de la santé et de la recherche médicale (Inserm) U1135, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| |
Collapse
|
18
|
Zhou Y, Murre C. Bursty gene expression and mRNA decay pathways orchestrate B cell activation. SCIENCE ADVANCES 2021; 7:eabm0819. [PMID: 34860551 PMCID: PMC8641932 DOI: 10.1126/sciadv.abm0819] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 10/14/2021] [Indexed: 06/13/2023]
Abstract
It is well established that the helix-loop-helix proteins, E2A and E2-2, promote B cell activation. Here, we examined how during the course of B cell activation E2A and E2-2 gene expression is regulated. We found that E2A and E2-2 mRNA abundance concomitantly increased in activated B cells. The increase in E2A and E2-2 mRNA abundance correlated with increased cell growth. Elevated E2A and E2-2 mRNA abundance was instructed by increased transcriptional bursting frequencies and elevated E2A and E2-2 mRNA half-lives. The increase in E2A and E2-2 bursting frequencies often occurred at shared interchromosomal transcriptional hubs. We suggest that in naïve B cells low E2A and E2-2 bursting frequencies and high E2A and E2-2 mRNA decay rates instruct noisy gene expression that allows a clonal and swift response to invading pathogens whereas in activated B cells increased transcriptional bursting and low mRNA decay rates dictate an activated B lineage gene program.
Collapse
Affiliation(s)
- Yi Zhou
- Division of Biological Sciences, Section of Molecular Biology, University of California, San Diego, La Jolla, CA 92039, USA
| | - Cornelis Murre
- Division of Biological Sciences, Section of Molecular Biology, University of California, San Diego, La Jolla, CA 92039, USA
| |
Collapse
|
19
|
Loss of synergistic transcriptional feedback loops drives diverse B-cell cancers. EBioMedicine 2021; 71:103559. [PMID: 34461601 PMCID: PMC8403728 DOI: 10.1016/j.ebiom.2021.103559] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 08/10/2021] [Accepted: 08/13/2021] [Indexed: 12/30/2022] Open
Abstract
Background The most common B-cell cancers, chronic lymphocytic leukemia/lymphoma (CLL), follicular and diffuse large B-cell (FL, DLBCL) lymphomas, have distinct clinical courses, yet overlapping “cell-of-origin”. Dynamic changes to the epigenome are essential regulators of B-cell differentiation. Therefore, we reasoned that these distinct cancers may be driven by shared mechanisms of disruption in transcriptional circuitry. Methods We compared purified malignant B-cells from 52 patients with normal B-cell subsets (germinal center centrocytes and centroblasts, naïve and memory B-cells) from 36 donor tonsils using >325 high-resolution molecular profiling assays for histone modifications, open chromatin (ChIP-, FAIRE-seq), transcriptome (RNA-seq), transcription factor (TF) binding, and genome copy number (microarrays). Findings From the resulting data, we identified gains in active chromatin in enhancers/super-enhancers that likely promote unchecked B-cell receptor signaling, including one we validated near the immunoglobulin superfamily receptors FCMR and PIGR. More striking and pervasive was the profound loss of key B-cell identity TFs, tumor suppressors and their super-enhancers, including EBF1, OCT2(POU2F2), and RUNX3. Using a novel approach to identify transcriptional feedback, we showed that these core transcriptional circuitries are self-regulating. Their selective gain and loss form a complex, iterative, and interactive process that likely curbs B-cell maturation and spurs proliferation. Interpretation Our study is the first to map the transcriptional circuitry of the most common blood cancers. We demonstrate that a critical subset of B-cell TFs and their cognate enhancers form self-regulatory transcriptional feedback loops whose disruption is a shared mechanism underlying these diverse subtypes of B-cell lymphoma. Funding National Institute of Health, Siteman Cancer Center, Barnes-Jewish Hospital Foundation, Doris Duke Foundation.
Collapse
|
20
|
Sun L, Zhao X, Liu X, Zhong B, Tang H, Jin W, Clevers H, Wang H, Wang X, Dong C. Transcription factor Ascl2 promotes germinal center B cell responses by directly regulating AID transcription. Cell Rep 2021; 35:109188. [PMID: 34077723 DOI: 10.1016/j.celrep.2021.109188] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 08/14/2020] [Accepted: 05/07/2021] [Indexed: 11/24/2022] Open
Abstract
During germinal center (GC) reactions, activated B cells undergo clonal expansion and functional maturation to produce high-affinity antibodies and differentiate into plasma and memory cells, accompanied with class-switching recombination (CSR) and somatic hypermutation (SHM). Activation-induced cytidine deaminase (AID) is responsible for both CSR and SHM in GC B cells. Transcriptional mechanisms underlying AID regulation and GC B cell reactions are still not well understood. Here, we show that expression of Ascl2 transcription factor is upregulated in GC B cells. Ectopic expression of Ascl2 promotes GC B cell development and enhances antibody production and affinity maturation. Conversely, deletion of Ascl2 in B cells impairs the GC response. Genome-wide analysis reveals that Ascl2 directly regulates GC B cell-related genes, including AID; ectopic expression of AID in Ascl2-deficient B cells rescues their antibody defects. Thus, Ascl2 regulates AID transcription and promotes GC B cell responses.
Collapse
Affiliation(s)
- Lin Sun
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China; Tsinghua University-Peking University Joint Center for Life Science, Beijing 100084, China
| | - Xiaohong Zhao
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Xindong Liu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Bo Zhong
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
| | - Hong Tang
- CAS Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Beijing, China
| | - Wei Jin
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Hans Clevers
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Centre (UMC) Utrecht, 3584 CT Utrecht, the Netherlands
| | - Hui Wang
- Department of Immunology and Center for Inflammation and Cancer, MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Xiaohu Wang
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Chen Dong
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China; Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing 100084, China.
| |
Collapse
|
21
|
Yoshikawa G, Miyazaki K, Ogata H, Miyazaki M. The Evolution of Rag Gene Enhancers and Transcription Factor E and Id Proteins in the Adaptive Immune System. Int J Mol Sci 2021; 22:ijms22115888. [PMID: 34072618 PMCID: PMC8199221 DOI: 10.3390/ijms22115888] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 05/23/2021] [Accepted: 05/24/2021] [Indexed: 11/17/2022] Open
Abstract
Adaptive immunity relies on the V(D)J DNA recombination of immunoglobulin (Ig) and T cell receptor (TCR) genes, which enables the recognition of highly diverse antigens and the elicitation of antigen-specific immune responses. This process is mediated by recombination-activating gene (Rag) 1 and Rag2 (Rag1/2), whose expression is strictly controlled in a cell type-specific manner; the expression of Rag1/2 genes represents a hallmark of lymphoid lineage commitment. Although Rag genes are known to be evolutionally conserved among jawed vertebrates, how Rag genes are regulated by lineage-specific transcription factors (TFs) and how their regulatory system evolved among vertebrates have not been fully elucidated. Here, we reviewed the current body of knowledge concerning the cis-regulatory elements (CREs) of Rag genes and the evolution of the basic helix-loop-helix TF E protein regulating Rag gene CREs, as well as the evolution of the antagonist of this protein, the Id protein. This may help to understand how the adaptive immune system develops along with the evolution of responsible TFs and enhancers.
Collapse
Affiliation(s)
- Genki Yoshikawa
- Bioinformatics Center, Institute for Chemical Research, Kyoto University, Uji 611-0011, Japan;
| | - Kazuko Miyazaki
- Laboratory of Immunology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan;
| | - Hiroyuki Ogata
- Bioinformatics Center, Institute for Chemical Research, Kyoto University, Uji 611-0011, Japan;
- Correspondence: (H.O.); (M.M.)
| | - Masaki Miyazaki
- Laboratory of Immunology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan;
- Correspondence: (H.O.); (M.M.)
| |
Collapse
|
22
|
Abstract
Memory B cells (MBCs) are critical for the rapid development of protective immunity following re-infection. MBCs capable of neutralizing distinct subclasses of pathogens, such as influenza and HIV, have been identified in humans. However, efforts to develop vaccines that induce broadly protective MBCs to rapidly mutating pathogens have not yet been successful. Better understanding of the signals regulating MBC development and function are essential to overcome current challenges hindering successful vaccine development. Here, we discuss recent advancements regarding the signals and transcription factors regulating germinal centre-derived MBC development and function.
Collapse
Affiliation(s)
- Brian J Laidlaw
- Division of Allergy and Immunology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
| | - Jason G Cyster
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
- Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
23
|
Liu GJ, Jaritz M, Wöhner M, Agerer B, Bergthaler A, Malin SG, Busslinger M. Repression of the B cell identity factor Pax5 is not required for plasma cell development. J Exp Med 2021; 217:152012. [PMID: 32780801 PMCID: PMC7596824 DOI: 10.1084/jem.20200147] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 05/20/2020] [Accepted: 07/01/2020] [Indexed: 01/05/2023] Open
Abstract
B cell and plasma cell fates are controlled by different transcriptional networks, as exemplified by the mutually exclusive expression and cross-antagonism of the B cell identity factor Pax5 and the plasma cell regulator Blimp1. It has been postulated that repression of Pax5 by Blimp1 is essential for plasma cell development. Here, we challenged this hypothesis by analyzing the IghPax5/+ mouse, which expressed a Pax5 minigene from the immunoglobulin heavy-chain locus. Despite high Pax5 expression, plasma cells efficiently developed in young IghPax5/+ mice at steady state and upon immunization, while their number moderately declined in older mice. Although Pax5 significantly deregulated the plasma cell expression program, key plasma cell regulators were normally expressed in IghPax5/+ plasma cells. While IgM and IgA secretion by IghPax5/+ plasma cells was normal, IgG secretion was modestly decreased. Hence, Pax5 repression is not essential for robust plasma cell development and antibody secretion, although it is required for optimal IgG production and accumulation of long-lived plasma cells.
Collapse
Affiliation(s)
- Grace J Liu
- Research Institute of Molecular Pathology, Vienna BioCenter, Vienna, Austria
| | - Markus Jaritz
- Research Institute of Molecular Pathology, Vienna BioCenter, Vienna, Austria
| | - Miriam Wöhner
- Research Institute of Molecular Pathology, Vienna BioCenter, Vienna, Austria
| | - Benedikt Agerer
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Andreas Bergthaler
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Stephen G Malin
- Research Institute of Molecular Pathology, Vienna BioCenter, Vienna, Austria
| | - Meinrad Busslinger
- Research Institute of Molecular Pathology, Vienna BioCenter, Vienna, Austria
| |
Collapse
|
24
|
King HW, Orban N, Riches JC, Clear AJ, Warnes G, Teichmann SA, James LK. Single-cell analysis of human B cell maturation predicts how antibody class switching shapes selection dynamics. Sci Immunol 2021; 6:6/56/eabe6291. [PMID: 33579751 DOI: 10.1126/sciimmunol.abe6291] [Citation(s) in RCA: 157] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 01/14/2021] [Indexed: 12/21/2022]
Abstract
Protective humoral memory forms in secondary lymphoid organs where B cells undergo affinity maturation and differentiation into memory or plasma cells. Here, we provide a comprehensive roadmap of human B cell maturation with single-cell transcriptomics matched with bulk and single-cell antibody repertoires to define gene expression, antibody repertoires, and clonal sharing of B cell states at single-cell resolution, including memory B cell heterogeneity that reflects diverse functional and signaling states. We reconstruct gene expression dynamics during B cell activation to reveal a pre-germinal center state primed to undergo class switch recombination and dissect how antibody class-dependent gene expression in germinal center and memory B cells is linked with a distinct transcriptional wiring with potential to influence their fate and function. Our analyses reveal the dynamic cellular states that shape human B cell-mediated immunity and highlight how antibody isotype may play a role during their antibody-based selection.
Collapse
Affiliation(s)
- Hamish W King
- Centre for Immunobiology, Blizard Institute, Queen Mary University of London, London E1 2AT, UK. .,Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Nara Orban
- Barts Health Ear, Nose and Throat Service, Royal London Hospital, London E1 1BB, UK
| | - John C Riches
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK.,Francis Crick Institute, London NW1 1AT, UK
| | - Andrew J Clear
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Gary Warnes
- Flow Cytometry Core Facility, Blizard Institute, Queen Mary University of London, London E1 2AT, UK
| | - Sarah A Teichmann
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK.,Theory of Condensed Matter, Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge CB3 0EH, UK
| | - Louisa K James
- Centre for Immunobiology, Blizard Institute, Queen Mary University of London, London E1 2AT, UK.
| |
Collapse
|
25
|
Lipid Metabolism in Tumor-Associated B Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1316:133-147. [PMID: 33740248 DOI: 10.1007/978-981-33-6785-2_9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Breakthroughs have been made in the cancer immunotherapy field focusing on utilizing T cells' antitumor immunity, and the lipid metabolism of tumor-associated B cells is not well studied compared to T cells. Accumulating evidence suggested that B cells also play important roles in tumor biology and antitumor immunity, especially the germinal center B cells that present in the tumor-related tertiary lymphoid structures. Due to scarce studies on lipid metabolisms of tumor-associated B cells, this chapter mainly summarized findings on B cell lipid metabolism and discussed B cell development and major transcription factors, tumor-associated B cell populations and their potential functions in antitumor immunity, fatty acid oxidation in germinal center B cells, and tumor microenvironment factors that potentially affect B cell lipid metabolism, focusing on hypoxia and nutrients competition, as well as lipid metabolites that affect B cell function, including cholesterol, geranylgeranyl pyrophosphate, oxysterols, and short-chain fatty acids.
Collapse
|
26
|
Ge C, Zhu X, Niu X, Zhang B, Chen L. A transcriptome profile in gallbladder cancer based on annotation analysis of microarray studies. Mol Med Rep 2020; 23:25. [PMID: 33179115 PMCID: PMC7673323 DOI: 10.3892/mmr.2020.11663] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 06/25/2020] [Indexed: 01/12/2023] Open
Abstract
The purpose of the present study was to identify aberrantly expressed genes for gallbladder cancer based on the annotation analysis of microarray studies and to explore their potential functions. Differential gene expression was investigated in cholesterol polyps, gallbladder adenoma and gallbladder cancer using microarrays. Subsequently, microarray results were comprehensively analyzed. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were performed to determine the affected biological processes or pathways. Differentially expressed genes (DEGs) of cholesterol polyps, gallbladder adenoma and gallbladder cancer were identified. Following comprehensive analysis, 14 genes were found to be differentially expressed in the gallbladder wall of both gallbladder cancer and gallbladder adenoma. The 20 most significantly upregulated genes were only upregulated in the gallbladder wall of gallbladder cancer, but not in the gallbladder wall of cholesterol polyps and gallbladder adenoma. In addition, 182 DEGs were upregulated in the gallbladder wall of gallbladder adenoma compared with the gallbladder wall of cholesterol polyps. A total of 20 most significant DEGs were found in both the tumor and gallbladder wall of gallbladder cancer. In addition, the most significant DEGs that were identified were only upregulated in the tumor of gallbladder cancer. GO and KEGG analysis indicated that the aforementioned DEGs could participate in numerous biological processes or pathways associated with the development of gallbladder cancer. The present findings will help improve the current understanding of tumorigenesis and the development of gallbladder cancer.
Collapse
Affiliation(s)
- Chunlin Ge
- Department of General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Xuan Zhu
- Department of General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Xing Niu
- Department of Second Clinical College, Shengjing Hospital Affiliated to China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Bingye Zhang
- Department of General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Lijie Chen
- Department of Second Clinical College, Shengjing Hospital Affiliated to China Medical University, Shenyang, Liaoning 110004, P.R. China
| |
Collapse
|
27
|
Jain N, Hartert K, Tadros S, Fiskus W, Havranek O, Ma MCJ, Bouska A, Heavican T, Kumar D, Deng Q, Moore D, Pak C, Liu CL, Gentles AJ, Hartmann E, Kridel R, Smedby KE, Juliusson G, Rosenquist R, Gascoyne RD, Rosenwald A, Giancotti F, Neelapu SS, Westin J, Vose JM, Lunning MA, Greiner T, Rodig S, Iqbal J, Alizadeh AA, Davis RE, Bhalla K, Green MR. Targetable genetic alterations of TCF4 ( E2-2) drive immunoglobulin expression in diffuse large B cell lymphoma. Sci Transl Med 2020; 11:11/497/eaav5599. [PMID: 31217338 DOI: 10.1126/scitranslmed.aav5599] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 01/31/2019] [Accepted: 05/15/2019] [Indexed: 12/12/2022]
Abstract
The activated B cell (ABC-like) subtype of diffuse large B cell lymphoma (DLBCL) is characterized by chronic activation of signaling initiated by immunoglobulin μ (IgM). By analyzing the DNA copy number profiles of 1000 DLBCL tumors, we identified gains of 18q21.2 as the most frequent genetic alteration in ABC-like DLBCL. Using integrative analysis of matched gene expression profiling data, we found that the TCF4 (E2-2) transcription factor gene was the target of these alterations. Overexpression of TCF4 in ABC-like DLBCL cell lines led to its occupancy on immunoglobulin (IGHM) and MYC gene enhancers and increased expression of these genes at the transcript and protein levels. Inhibition of TCF4 activity with dominant-negative constructs was synthetically lethal to ABC-like DLBCL cell lines harboring TCF4 DNA copy gains, highlighting these gains as an attractive potential therapeutic target. Furthermore, the TCF4 gene was one of the top BRD4-regulated genes in DLBCL cell lines. BET proteolysis-targeting chimera (PROTAC) ARV771 extinguished TCF4, MYC, and IgM expression and killed ABC-like DLBCL cells in vitro. In DLBCL xenograft models, ARV771 treatment reduced tumor growth and prolonged survival. This work highlights a genetic mechanism for promoting immunoglobulin signaling in ABC-like DLBCL and provides a functional rationale for the use of BET inhibitors in this disease.
Collapse
Affiliation(s)
- Neeraj Jain
- Department of Lymphoma/Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Keenan Hartert
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Saber Tadros
- Department of Lymphoma/Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Warren Fiskus
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ondrej Havranek
- Department of Lymphoma/Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Man Chun John Ma
- Department of Lymphoma/Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Alyssa Bouska
- Department of Pathology and Immunology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Tayla Heavican
- Department of Pathology and Immunology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Dhiraj Kumar
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Qing Deng
- Department of Lymphoma/Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Dalia Moore
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Christine Pak
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Chih Long Liu
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Andrew J Gentles
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Elena Hartmann
- Institute of Pathology, University of Würzburg, Würzburg 97080, Germany.,Comprehensive Cancer Center Mainfranken, Wurzburg 97080, Germany
| | - Robert Kridel
- Princess Margaret Cancer Center, University of Toronto, Toronto, ON M5G 2C4, Canada
| | - Karin Ekstrom Smedby
- Department of Medicine, Solna, Clinical Epidemiology Unit, Karolinska Institutet, and Hematology Center, Karolinska University Hospital, Stockholm SE-171 76, Sweden
| | - Gunnar Juliusson
- Department of Laboratory Medicine, Stem Cell Center, Lund University, Lund SE-221 00, Sweden
| | - Richard Rosenquist
- Department of Molecular Medicine and Surgery, Karolinska Universitetssjukhuset, Stockholm SE-171 76, Sweden
| | - Randy D Gascoyne
- Center for Lymphoid Cancer, British Columbia Cancer Agency, Vancouver, BC V5Z 4E6, Canada
| | - Andreas Rosenwald
- Institute of Pathology, University of Würzburg, Würzburg 97080, Germany.,Comprehensive Cancer Center Mainfranken, Wurzburg 97080, Germany
| | - Filippo Giancotti
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sattva S Neelapu
- Department of Lymphoma/Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jason Westin
- Department of Lymphoma/Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Julie M Vose
- Division of Hematology and Oncology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Matthew A Lunning
- Division of Hematology and Oncology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Timothy Greiner
- Department of Pathology and Immunology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Scott Rodig
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Javeed Iqbal
- Department of Pathology and Immunology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Ash A Alizadeh
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - R Eric Davis
- Department of Lymphoma/Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kapil Bhalla
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Center for Cancer Epigenetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Michael R Green
- Department of Lymphoma/Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA. .,Center for Cancer Epigenetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
28
|
Bortnick A, He Z, Aubrey M, Chandra V, Denholtz M, Chen K, Lin YC, Murre C. Plasma Cell Fate Is Orchestrated by Elaborate Changes in Genome Compartmentalization and Inter-chromosomal Hubs. Cell Rep 2020; 31:107470. [PMID: 32268089 PMCID: PMC10871151 DOI: 10.1016/j.celrep.2020.03.034] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 01/31/2020] [Accepted: 03/12/2020] [Indexed: 12/27/2022] Open
Abstract
The transition from the follicular B to the plasma cell stage is associated with large-scale changes in cell morphology. Here, we examine whether plasma cell development is also associated with changes in nuclear architecture. We find that the onset of plasma cell development is concomitant with a decline in remote genomic interactions; a gain in euchromatic character at loci encoding for factors that specify plasma cell fate, including Prdm1 and Atf4; and establishment of de novo inter-chromosomal hubs. We find that, in developing plasma cells and concurrent with transcriptional silencing, the Ebf1 locus repositions from an euchromatic to peri-centromeric heterochromatic environment. Finally, we find that inter-chromosomal hubs are enriched for the deposition of either H3K27Ac or H3K27me3. These data indicate that plasma cell fate is orchestrated by elaborate changes in genome topology and that epigenetic marks, linked with super-enhancers or transcriptionally repressed regions, are enriched at inter-chromosomal hubs.
Collapse
Affiliation(s)
- Alexandra Bortnick
- Department of Molecular Biology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Zhaoren He
- Department of Molecular Biology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Megan Aubrey
- Department of Molecular Biology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Vivek Chandra
- Department of Molecular Biology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Matthew Denholtz
- Department of Molecular Biology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Kenian Chen
- Baylor Institute for Immunology Research, Baylor Research Institute, Dallas, TX 75246, USA
| | - Yin C Lin
- Baylor Institute for Immunology Research, Baylor Research Institute, Dallas, TX 75246, USA
| | - Cornelis Murre
- Department of Molecular Biology, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
29
|
Transcription Factor T-bet in B Cells Modulates Germinal Center Polarization and Antibody Affinity Maturation in Response to Malaria. Cell Rep 2019; 29:2257-2269.e6. [DOI: 10.1016/j.celrep.2019.10.087] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 06/06/2019] [Accepted: 10/22/2019] [Indexed: 12/14/2022] Open
|
30
|
Waters LR, Ahsan FM, Ten Hoeve J, Hong JS, Kim DNH, Minasyan A, Braas D, Graeber TG, Zangle TA, Teitell MA. Ampk regulates IgD expression but not energy stress with B cell activation. Sci Rep 2019; 9:8176. [PMID: 31160601 PMCID: PMC6546716 DOI: 10.1038/s41598-019-43985-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 04/28/2019] [Indexed: 12/25/2022] Open
Abstract
Ampk is an energy gatekeeper that responds to decreases in ATP by inhibiting energy-consuming anabolic processes and promoting energy-generating catabolic processes. Recently, we showed that Lkb1, an understudied kinase in B lymphocytes and a major upstream kinase for Ampk, had critical and unexpected roles in activating naïve B cells and in germinal center formation. Therefore, we examined whether Lkb1 activities during B cell activation depend on Ampk and report surprising Ampk activation with in vitro B cell stimulation in the absence of energy stress, coupled to rapid biomass accumulation. Despite Ampk activation and a controlling role for Lkb1 in B cell activation, Ampk knockout did not significantly affect B cell activation, differentiation, nutrient dynamics, gene expression, or humoral immune responses. Instead, Ampk loss specifically repressed the transcriptional expression of IgD and its regulator, Zfp318. Results also reveal that early activation of Ampk by phenformin treatment impairs germinal center formation but does not significantly alter antibody responses. Combined, the data show an unexpectedly specific role for Ampk in the regulation of IgD expression during B cell activation.
Collapse
Affiliation(s)
- Lynnea R Waters
- Molecular Biology Interdepartmental Program, UCLA, Los Angeles, CA, 90095, USA
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, CA, 90095, USA
| | - Fasih M Ahsan
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, CA, 90095, USA
| | - Johanna Ten Hoeve
- Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, CA, 90095, USA
- UCLA Metabolomics Center, UCLA, Los Angeles, CA, 90095, USA
- Crump Institute for Molecular Imaging, UCLA, Los Angeles, CA, 90095, USA
| | - Jason S Hong
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, CA, 90095, USA
| | - Diane N H Kim
- Department of Bioengineering, UCLA, Los Angeles, CA, 90095, USA
| | - Aspram Minasyan
- Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, CA, 90095, USA
- Crump Institute for Molecular Imaging, UCLA, Los Angeles, CA, 90095, USA
| | - Daniel Braas
- Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, CA, 90095, USA
- UCLA Metabolomics Center, UCLA, Los Angeles, CA, 90095, USA
- Crump Institute for Molecular Imaging, UCLA, Los Angeles, CA, 90095, USA
| | - Thomas G Graeber
- Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, CA, 90095, USA
- UCLA Metabolomics Center, UCLA, Los Angeles, CA, 90095, USA
- Crump Institute for Molecular Imaging, UCLA, Los Angeles, CA, 90095, USA
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, 90095, USA
- California NanoSystems Institute, UCLA, Los Angeles, CA, 90095, USA
- Broad Stem Cell Research Center, UCLA, Los Angeles, CA, 90095, USA
| | - Thomas A Zangle
- Department of Chemical Engineering, University of Utah, Salt Lake City, UT, 84112, USA
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, 84112, USA
| | - Michael A Teitell
- Molecular Biology Interdepartmental Program, UCLA, Los Angeles, CA, 90095, USA.
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, CA, 90095, USA.
- Department of Bioengineering, UCLA, Los Angeles, CA, 90095, USA.
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, 90095, USA.
- California NanoSystems Institute, UCLA, Los Angeles, CA, 90095, USA.
- Broad Stem Cell Research Center, UCLA, Los Angeles, CA, 90095, USA.
- Department of Pediatrics, UCLA, Los Angeles, CA, 90095, USA.
| |
Collapse
|
31
|
Willis SN, Nutt SL. New players in the gene regulatory network controlling late B cell differentiation. Curr Opin Immunol 2019; 58:68-74. [DOI: 10.1016/j.coi.2019.04.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 04/19/2019] [Indexed: 02/07/2023]
|
32
|
Lio CWJ, Shukla V, Samaniego-Castruita D, González-Avalos E, Chakraborty A, Yue X, Schatz DG, Ay F, Rao A. TET enzymes augment activation-induced deaminase (AID) expression via 5-hydroxymethylcytosine modifications at the Aicda superenhancer. Sci Immunol 2019; 4:eaau7523. [PMID: 31028100 PMCID: PMC6599614 DOI: 10.1126/sciimmunol.aau7523] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 03/19/2019] [Indexed: 12/15/2022]
Abstract
TET enzymes are dioxygenases that promote DNA demethylation by oxidizing the methyl group of 5-methylcytosine to 5-hydroxymethylcytosine (5hmC). Here, we report a close correspondence between 5hmC-marked regions, chromatin accessibility and enhancer activity in B cells, and a strong enrichment for consensus binding motifs for basic region-leucine zipper (bZIP) transcription factors at TET-responsive genomic regions. Functionally, Tet2 and Tet3 regulate class switch recombination (CSR) in murine B cells by enhancing expression of Aicda, which encodes the activation-induced cytidine deaminase (AID) enzyme essential for CSR. TET enzymes deposit 5hmC, facilitate DNA demethylation, and maintain chromatin accessibility at two TET-responsive enhancer elements, TetE1 and TetE2, located within a superenhancer in the Aicda locus. Our data identify the bZIP transcription factor, ATF-like (BATF) as a key transcription factor involved in TET-dependent Aicda expression. 5hmC is not deposited at TetE1 in activated Batf-deficient B cells, indicating that BATF facilitates TET recruitment to this Aicda enhancer. Our study emphasizes the importance of TET enzymes for bolstering AID expression and highlights 5hmC as an epigenetic mark that captures enhancer dynamics during cell activation.
Collapse
Affiliation(s)
- Chan-Wang J Lio
- Division of Signaling and Gene Expression, La Jolla Institute, San Diego, CA, USA.
| | - Vipul Shukla
- Division of Signaling and Gene Expression, La Jolla Institute, San Diego, CA, USA
| | | | | | | | - Xiaojing Yue
- Division of Signaling and Gene Expression, La Jolla Institute, San Diego, CA, USA
| | - David G Schatz
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Ferhat Ay
- Division of Vaccine Discovery, La Jolla Institute, San Diego, CA, USA
| | - Anjana Rao
- Division of Signaling and Gene Expression, La Jolla Institute, San Diego, CA, USA.
- Sanford Consortium for Regenerative Medicine, San Diego, CA, USA
- Department of Pharmacology, University of California, San Diego, San Diego, CA, USA
- Moores Cancer Center, University of California, San Diego, San Diego, CA, USA
| |
Collapse
|
33
|
Mlynarczyk C, Fontán L, Melnick A. Germinal center-derived lymphomas: The darkest side of humoral immunity. Immunol Rev 2019; 288:214-239. [PMID: 30874354 PMCID: PMC6518944 DOI: 10.1111/imr.12755] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 02/11/2019] [Accepted: 02/11/2019] [Indexed: 02/06/2023]
Abstract
One of the unusual features of germinal center (GC) B cells is that they manifest many hallmarks of cancer cells. Accordingly, most B-cell neoplasms originate from the GC reaction, and characteristically display abundant point mutations, structural genomic lesions, and clonal diversity from the genetic and epigenetic standpoints. The dominant biological theme of GC-derived lymphomas is mutation of genes involved in epigenetic regulation and immune receptor signaling, which come into play at critical transitional stages of the GC reaction. Hence, mechanistic studies of these mutations reveal fundamental insight into the biology of the normal and malignant GC B cell. The BCL6 transcription factor plays a central role in establishing the GC phenotype in B cells, and most lymphomas are dependent on BCL6 to maintain survival, proliferation, and perhaps immune evasion. Many lymphoma mutations have the commonality of enhancing the oncogenic functions of BCL6, or overcoming some of its tumor suppressive effects. Herein, we discuss how unique features of the GC reaction create vulnerabilities that select for particular lymphoma mutations. We examine the interplay between epigenetic programming, metabolism, signaling, and immune regulatory mechanisms in lymphoma, and discuss how these are leading to novel precision therapy strategies to treat lymphoma patients.
Collapse
Affiliation(s)
- Coraline Mlynarczyk
- Department of MedicineDivision of Hematology & Medical OncologyWeill Cornell MedicineNew York CityNew York
| | - Lorena Fontán
- Department of MedicineDivision of Hematology & Medical OncologyWeill Cornell MedicineNew York CityNew York
| | - Ari Melnick
- Department of MedicineDivision of Hematology & Medical OncologyWeill Cornell MedicineNew York CityNew York
| |
Collapse
|
34
|
Abstract
In this review from Murre, the evolution of HLH genes, the structures of HLH domains, and the elaborate activities of HLH proteins in multicellular life are discussed. Helix–loop–helix (HLH) proteins are dimeric transcription factors that control lineage- and developmental-specific gene programs. Genes encoding for HLH proteins arose in unicellular organisms >600 million years ago and then duplicated and diversified from ancestral genes across the metazoan and plant kingdoms to establish multicellularity. Hundreds of HLH proteins have been identified with diverse functions in a wide variety of cell types. HLH proteins orchestrate lineage specification, commitment, self-renewal, proliferation, differentiation, and homing. HLH proteins also regulate circadian clocks, protect against hypoxic stress, promote antigen receptor locus assembly, and program transdifferentiation. HLH proteins deposit or erase epigenetic marks, activate noncoding transcription, and sequester chromatin remodelers across the chromatin landscape to dictate enhancer–promoter communication and somatic recombination. Here the evolution of HLH genes, the structures of HLH domains, and the elaborate activities of HLH proteins in multicellular life are discussed.
Collapse
Affiliation(s)
- Cornelis Murre
- Division of Biological Sciences, University of California at San Diego, La Jolla, California 92903, USA
| |
Collapse
|
35
|
Abstract
In this review, Boothby et al. summarize some salient advances toward elucidation of the molecular programming of the fate choices and function of B cells in the periphery. They also note unanswered questions that pertain to differences among subsets of B lymphocytes and plasma cells. Mature B lymphocytes are crucial components of adaptive immunity, a system essential for the evolutionary fitness of mammals. Adaptive lymphocyte function requires an initially naïve cell to proliferate extensively and its progeny to have the capacity to assume a variety of fates. These include either terminal differentiation (the long-lived plasma cell) or metastable transcriptional reprogramming (germinal center and memory B cells). In this review, we focus principally on the regulation of differentiation and functional diversification of the “B2” subset. An overview is combined with an account of more recent advances, including initial work on mechanisms that eliminate DNA methylation and potential links between intracellular metabolites and chromatin editing.
Collapse
|
36
|
Bönelt P, Wöhner M, Minnich M, Tagoh H, Fischer M, Jaritz M, Kavirayani A, Garimella M, Karlsson MC, Busslinger M. Precocious expression of Blimp1 in B cells causes autoimmune disease with increased self-reactive plasma cells. EMBO J 2018; 38:embj.2018100010. [PMID: 30498131 PMCID: PMC6331720 DOI: 10.15252/embj.2018100010] [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: 06/08/2018] [Revised: 10/29/2018] [Accepted: 11/02/2018] [Indexed: 11/25/2022] Open
Abstract
The transcription factor Blimp1 is not only an essential regulator of plasma cells, but also a risk factor for the development of autoimmune disease in humans. Here, we demonstrate in the mouse that the Prdm1 (Blimp1) gene was partially activated at the chromatin and transcription level in early B cell development, although mature Prdm1 mRNA did not accumulate due to posttranscriptional regulation. By analyzing a mouse model that facilitated ectopic Blimp1 protein expression throughout B lymphopoiesis, we could demonstrate that Blimp1 impaired B cell development by interfering with the B cell gene expression program, while leading to an increased abundance of plasma cells by promoting premature plasmablast differentiation of immature and mature B cells. With progressing age, these mice developed an autoimmune disease characterized by the presence of autoantibodies and glomerulonephritis. Hence, these data identified ectopic Blimp1 expression as a novel mechanism, through which Blimp1 can act as a risk factor in the development of autoimmune disease.
Collapse
Affiliation(s)
- Peter Bönelt
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Vienna, Austria
| | - Miriam Wöhner
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Vienna, Austria
| | - Martina Minnich
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Vienna, Austria
| | - Hiromi Tagoh
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Vienna, Austria
| | - Maria Fischer
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Vienna, Austria
| | - Markus Jaritz
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Vienna, Austria
| | - Anoop Kavirayani
- Vienna Biocenter Core Facilities (VBCF), Vienna Biocenter (VBC), Vienna, Austria
| | - Manasa Garimella
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | - Mikael Ci Karlsson
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | - Meinrad Busslinger
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Vienna, Austria
| |
Collapse
|
37
|
Sawai CM, Serpas L, Neto AG, Jang G, Rashidfarrokhi A, Kolbeck R, Sanjuan MA, Reizis B, Sisirak V. Plasmacytoid Dendritic Cells Are Largely Dispensable for the Pathogenesis of Experimental Inflammatory Bowel Disease. Front Immunol 2018; 9:2475. [PMID: 30410494 PMCID: PMC6209677 DOI: 10.3389/fimmu.2018.02475] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 10/08/2018] [Indexed: 12/31/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory condition caused by an aberrant immune response to microbial components of the gastrointestinal tract. Plasmacytoid dendritic cells (pDCs) are innate immune cells specialized in the production of type I interferons and were recently implicated in the pathogenesis of autoimmune disorders such as lupus and scleroderma. While pDCs were shown to infiltrate intestinal mucosa of IBD patients and proposed to participate in intestinal inflammation, their net contribution to the disease remains unclear. We addressed this question by targeting the pDC-specific transcription factor TCF4 (E2-2) in experimental IBD caused by deficiency of Wiskott-Aldrich syndrome protein (WASP) or of interleukin-10 (IL-10). Monoallelic Tcf4 deletion, which was previously shown to abrogate experimental lupus, did not affect autoimmunity manifestations or colitis in WASP-deficient animals. Furthermore, conditional biallelic Tcf4 targeting resulted in a near-complete pDC ablation, yet had no effect on the development of colitis in IL-10-deficient mice. Our results suggest that, in contrast to other inflammatory and autoimmune diseases, pDCs do not play a major role in the pathogenesis of intestinal inflammation during IBD.
Collapse
Affiliation(s)
- Catherine M Sawai
- Department of Pathology, New York University School of Medicine, New York, NY, United States.,INSERM, ACTION Laboratory, University of Bordeaux, Bordeaux, France
| | - Lee Serpas
- Department of Pathology, New York University School of Medicine, New York, NY, United States
| | - Antonio Galvao Neto
- Department of Pathology, New York University School of Medicine, New York, NY, United States
| | - Geunhyo Jang
- Department of Pathology, New York University School of Medicine, New York, NY, United States
| | - Ali Rashidfarrokhi
- Department of Pathology, New York University School of Medicine, New York, NY, United States
| | - Roland Kolbeck
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune LLC, Gaithersburg, MD, United States
| | - Miguel A Sanjuan
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune LLC, Gaithersburg, MD, United States
| | - Boris Reizis
- Department of Pathology, New York University School of Medicine, New York, NY, United States.,Department of Medicine, New York University School of Medicine, New York, NY, United States
| | - Vanja Sisirak
- Department of Pathology, New York University School of Medicine, New York, NY, United States.,CNRS-UMR, Immunoconcept, Université de Bordeaux, Bordeaux, France
| |
Collapse
|
38
|
Kowalchuk AM, Maurer KA, Shoja-Taheri F, Brown NL. Requirements for Neurogenin2 during mouse postnatal retinal neurogenesis. Dev Biol 2018; 442:220-235. [PMID: 30048641 PMCID: PMC6143394 DOI: 10.1016/j.ydbio.2018.07.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 07/22/2018] [Accepted: 07/23/2018] [Indexed: 02/02/2023]
Abstract
During embryonic retinal development, the bHLH factor Neurog2 regulates the temporal progression of neurogenesis, but no role has been assigned for this gene in the postnatal retina. Using Neurog2 conditional mutants, we found that Neurog2 is necessary for the development of an early, embryonic cohort of rod photoreceptors, but also required by both a subset of cone bipolar subtypes, and rod bipolars. Using transcriptomics, we identified a subset of downregulated genes in P2 Neurog2 mutants, which act during rod differentiation, outer segment morphogenesis or visual processing. We also uncovered defects in neuronal cell culling, which suggests that the rod and bipolar cell phenotypes may arise via more complex mechanisms rather than a simple cell fate shift. However, given an overall phenotypic resemblance between Neurog2 and Blimp1 mutants, we explored the relationship between these two factors. We found that Blimp1 is downregulated between E12-birth in Neurog2 mutants, which probably reflects a dependence on Neurog2 in embryonic progenitor cells. Overall, we conclude that the Neurog2 gene is expressed and active prior to birth, but also exerts an influence on postnatal retinal neuron differentiation.
Collapse
Affiliation(s)
- Angelica M Kowalchuk
- Department of Cell Biology and Human Anatomy, University of California-Davis, Davis, CA 95616, USA
| | - Kate A Maurer
- Division of Developmental Biology, Cincinnati Children's Hospital Research Foundation, Cincinnati, OH 45229, USA
| | - Farnaz Shoja-Taheri
- Department of Cell Biology and Human Anatomy, University of California-Davis, Davis, CA 95616, USA
| | - Nadean L Brown
- Department of Cell Biology and Human Anatomy, University of California-Davis, Davis, CA 95616, USA; Division of Developmental Biology, Cincinnati Children's Hospital Research Foundation, Cincinnati, OH 45229, USA.
| |
Collapse
|
39
|
Paradoxical role of Id proteins in regulating tumorigenic potential of lymphoid cells. Front Med 2018; 12:374-386. [PMID: 30043222 DOI: 10.1007/s11684-018-0652-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Accepted: 06/26/2018] [Indexed: 12/11/2022]
Abstract
A family of transcription factors known as Id proteins, or inhibitor of DNA binding and differentiation, is capable of regulating cell proliferation, survival and differentiation, and is often upregulated in multiple types of tumors. Due to their ability to promote self-renewal, Id proteins have been considered as oncogenes, and potential therapeutic targets in cancer models. On the contrary, certain Id proteins are reported to act as tumor suppressors in the development of Burkitt's lymphoma in humans, and hepatosplenic and innate-like T cell lymphomas in mice. The contexts and mechanisms by which Id proteins can serve in such contradictory roles to determine tumor outcomes are still not well understood. In this review, we explore the roles of Id proteins in lymphocyte development and tumorigenesis, particularly with respect to inhibition of their canonical DNA binding partners known as E proteins. Transcriptional regulation by E proteins, and their antagonism by Id proteins, act as gatekeepers to ensure appropriate lymphocyte development at key checkpoints. We re-examine the derailment of these regulatory mechanisms in lymphocytes that facilitate tumor development. These mechanistic insights can allow better appreciation of the context-dependent roles of Id proteins in cancers and improve considerations for therapy.
Collapse
|
40
|
Plasma cell differentiation is controlled by multiple cell division-coupled epigenetic programs. Nat Commun 2018; 9:1698. [PMID: 29703886 PMCID: PMC5923265 DOI: 10.1038/s41467-018-04125-8] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 04/05/2018] [Indexed: 12/13/2022] Open
Abstract
The genomic loci associated with B cell differentiation that are subject to transcriptional and epigenetic regulation in vivo are not well defined, leaving a gap in our understanding of the development of humoral immune responses. Here, using an in vivo T cell independent B cell differentiation model, we define a cellular division-dependent cis-regulatory element road map using ATAC-seq. Chromatin accessibility changes correlate with gene expression and reveal the reprogramming of transcriptional networks and the genes they regulate at specific cell divisions. A subset of genes in naive B cells display accessible promoters in the absence of transcription and are marked by H3K27me3, an EZH2 catalyzed repressive modification. Such genes encode regulators of cell division and metabolism and include the essential plasma cell transcription factor Blimp-1. Chemical inhibition of EZH2 results in enhanced plasma cell formation, increased expression of the above gene set, and premature expression of Blimp-1 ex vivo. These data provide insights into cell-division coupled epigenetic and transcriptional processes that program plasma cells. During B cell differentiation, the role of different genomic loci in transcriptional and epigenetic regulation in vivo is not well defined. Here the authors use an in vivo B cell differentiation model to map cellular division-dependent cis-regulatory element road map with ATAC-seq.
Collapse
|
41
|
T Follicular Helper Cell-Germinal Center B Cell Interaction Strength Regulates Entry into Plasma Cell or Recycling Germinal Center Cell Fate. Immunity 2018; 48:702-715.e4. [DOI: 10.1016/j.immuni.2018.03.027] [Citation(s) in RCA: 226] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 01/15/2018] [Accepted: 03/23/2018] [Indexed: 12/17/2022]
|
42
|
Budzyńska PM, Kyläniemi MK, Lassila O, Nera KP, Alinikula J. BLIMP-1 is insufficient to induce antibody secretion in the absence of IRF4 in DT40 cells. Scand J Immunol 2018; 87. [PMID: 29430664 DOI: 10.1111/sji.12646] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 02/01/2018] [Indexed: 01/18/2023]
Abstract
Differentiation of B cells into antibody-secreting cells (ASCs), plasmablasts and plasma cells is regulated by a network of transcription factors. Within this network, factors including PAX5 and BCL6 prevent ASC differentiation and maintain the B cell phenotype. In contrast, BLIMP-1 and high IRF4 expression promote plasma cell differentiation. BLIMP-1 is thought to induce immunoglobulin secretion, whereas IRF4 is needed for the survival of ASCs. The role of IRF4 in the regulation of antibody secretion has remained controversial. To study the role of IRF4 in the regulation of antibody secretion, we have created a double knockout (DKO) DT40 B cell line deficient in both IRF4 and BCL6. Although BCL6-deficient DT40 B cell line had upregulated BLIMP-1 expression and secreted antibodies, the DKO cell line did not. Even enforced BLIMP-1 expression in DKO cells or IRF4-deficient cells could not induce IgM secretion while in WT DT40 cells, it could. However, enforced IRF4 expression in DKO cells induced strong IgM secretion. Our findings support a model where IRF4 expression in addition to BLIMP-1 expression is required to induce robust antibody secretion.
Collapse
Affiliation(s)
- P M Budzyńska
- Department of Medical Microbiology and Immunology, Institute of Biomedicine, University of Turku, Turku, Finland.,Turku Doctoral Programme of Biomedical Sciences and Turku Doctoral Programme of Molecular Medicine, University of Turku, Turku, Finland
| | - M K Kyläniemi
- Department of Medical Microbiology and Immunology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - O Lassila
- Department of Medical Microbiology and Immunology, Institute of Biomedicine, University of Turku, Turku, Finland.,Department of Clinical Microbiology and Immunology, Turku University Hospital, Turku, Finland
| | - K-P Nera
- Department of Medical Microbiology and Immunology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - J Alinikula
- Department of Medical Microbiology and Immunology, Institute of Biomedicine, University of Turku, Turku, Finland
| |
Collapse
|
43
|
De S, Zhang B, Shih T, Singh S, Winkler A, Donnelly R, Barnes BJ. B Cell-Intrinsic Role for IRF5 in TLR9/BCR-Induced Human B Cell Activation, Proliferation, and Plasmablast Differentiation. Front Immunol 2018; 8:1938. [PMID: 29367853 PMCID: PMC5768180 DOI: 10.3389/fimmu.2017.01938] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 12/15/2017] [Indexed: 12/25/2022] Open
Abstract
Upon recognition of antigen, B cells undergo rapid proliferation followed by differentiation to specialized antibody secreting cells (ASCs). During this transition, B cells are reliant upon a multilayer transcription factor network to achieve a dramatic remodeling of the B cell transcriptional landscape. Increased levels of ASCs are often seen in autoimmune diseases and it is believed that altered expression of regulatory transcription factors play a role in this imbalance. The transcription factor interferon regulatory factor 5 (IRF5) is one such candidate as polymorphisms in IRF5 associate with risk of numerous autoimmune diseases and correlate with elevated IRF5 expression. IRF5 genetic risk has been widely replicated in systemic lupus erythematosus (SLE), and loss of Irf5 ameliorates disease in murine lupus models, in part, through the lack of pathogenic autoantibody secretion. It remains unclear, however, whether IRF5 is contributing to autoantibody production through a B cell-intrinsic function. To date, IRF5 function in healthy human B cells has not been characterized. Using human primary naive B cells, we define a critical intrinsic role for IRF5 in B cell activation, proliferation, and plasmablast differentiation. Targeted IRF5 knockdown resulted in significant immunoglobulin (Ig) D retention, reduced proliferation, plasmablast differentiation, and IgG secretion. The observed decreases were due to impaired B cell activation and clonal expansion. Distinct from murine studies, we identify and confirm new IRF5 target genes, IRF4, ERK1, and MYC, and pathways that mediate IRF5 B cell-intrinsic function. Together, these results identify IRF5 as an early regulator of human B cell activation and provide the first dataset in human primary B cells to map IRF5 dysfunction in SLE.
Collapse
Affiliation(s)
- Saurav De
- Rutgers Graduate School of Biomedical Sciences, Newark, NJ, United States.,Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institute for Medical Research, Manhasset, NY, United States
| | - Baohong Zhang
- Clinical Genetics and Bioinformatics, Pfizer Inc., Cambridge, MA, United States
| | - Tiffany Shih
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institute for Medical Research, Manhasset, NY, United States
| | - Sukhwinder Singh
- Department of Pathology and Laboratory Medicine, Rutgers Biomedical and Health Sciences, New Jersey Medical School, Newark, NJ, United States
| | - Aaron Winkler
- Department of Inflammation and Immunology, Pfizer Inc., Cambridge, MA, United States
| | - Robert Donnelly
- Department of Pathology and Laboratory Medicine, Rutgers Biomedical and Health Sciences, New Jersey Medical School, Newark, NJ, United States
| | - Betsy J Barnes
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institute for Medical Research, Manhasset, NY, United States.,Rutgers Biomedical and Health Sciences, New Jersey Medical School-Cancer Center, Newark, NJ, United States
| |
Collapse
|
44
|
Boothby M, Rickert RC. Metabolic Regulation of the Immune Humoral Response. Immunity 2017; 46:743-755. [PMID: 28514675 DOI: 10.1016/j.immuni.2017.04.009] [Citation(s) in RCA: 187] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 04/15/2017] [Accepted: 04/21/2017] [Indexed: 12/13/2022]
Abstract
Productive humoral responses require that naive B cells and their differentiated progeny move among distinct micro-environments. In this review, we discuss how studies are beginning to address the nature of these niches as well as the interplay between cellular signaling, metabolic programming, and adaptation to the locale. Recent work adds evidence to the expectation that B cells at distinct stages of development or functional subsets are influenced by the altered profiles of nutrients and metabolic by-products that distinguish these sites. Moreover, emerging findings reveal a cross-talk among the external milieu, signal transduction pathways, and transcription factors that direct B cell fate in the periphery.
Collapse
Affiliation(s)
- Mark Boothby
- Department of Pathology, Microbiology and Immunology, School of Medicine, Vanderbilt University, and Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Medicine, School of Medicine, Vanderbilt University, and Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| | - Robert C Rickert
- Tumor Microenvironment and Cancer Immunology Program, Sanford Burnham Prebys Medical Discovery Institute (SBP), La Jolla, CA 92037, USA; NCI-designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
45
|
Bannard O, Cyster JG. Germinal centers: programmed for affinity maturation and antibody diversification. Curr Opin Immunol 2017; 45:21-30. [DOI: 10.1016/j.coi.2016.12.004] [Citation(s) in RCA: 144] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 12/14/2016] [Indexed: 12/17/2022]
|
46
|
Grajkowska LT, Ceribelli M, Lau CM, Warren ME, Tiniakou I, Nakandakari Higa S, Bunin A, Haecker H, Mirny LA, Staudt LM, Reizis B. Isoform-Specific Expression and Feedback Regulation of E Protein TCF4 Control Dendritic Cell Lineage Specification. Immunity 2017; 46:65-77. [PMID: 27986456 PMCID: PMC5243153 DOI: 10.1016/j.immuni.2016.11.006] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 09/22/2016] [Accepted: 10/24/2016] [Indexed: 12/24/2022]
Abstract
The cell fate decision between interferon-producing plasmacytoid DC (pDC) and antigen-presenting classical DC (cDC) is controlled by the E protein transcription factor TCF4 (E2-2). We report that TCF4 comprises two transcriptional isoforms, both of which are required for optimal pDC development in vitro. The long Tcf4 isoform is expressed specifically in pDCs, and its deletion in mice impaired pDCs development and led to the expansion of non-canonical CD8+ cDCs. The expression of Tcf4 commenced in progenitors and was further upregulated in pDCs, correlating with stage-specific activity of multiple enhancer elements. A conserved enhancer downstream of Tcf4 was required for its upregulation during pDC differentiation, revealing a positive feedback loop. The expression of Tcf4 and the resulting pDC differentiation were selectively sensitive to the inhibition of enhancer-binding BET protein activity. Thus, lineage-specifying function of E proteins is facilitated by lineage-specific isoform expression and by BET-dependent feedback regulation through distal regulatory elements.
Collapse
Affiliation(s)
- Lucja T Grajkowska
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA; Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032, USA
| | - Michele Ceribelli
- Lymphoid Malignancies Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA; Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Bethesda, MD 20892, USA
| | - Colleen M Lau
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Margaret E Warren
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA; Graduate Program in Genetics and Development, Columbia University Medical Center, New York, NY 10032, USA
| | - Ioanna Tiniakou
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | | | - Anna Bunin
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032, USA
| | - Hans Haecker
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Leonid A Mirny
- Department of Physics and Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Louis M Staudt
- Lymphoid Malignancies Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Boris Reizis
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA; Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032, USA.
| |
Collapse
|
47
|
Gloury R, Zotos D, Zuidscherwoude M, Masson F, Liao Y, Hasbold J, Corcoran LM, Hodgkin PD, Belz GT, Shi W, Nutt SL, Tarlinton DM, Kallies A. Dynamic changes in Id3 and E-protein activity orchestrate germinal center and plasma cell development. J Biophys Biochem Cytol 2016. [DOI: 10.1083/jcb.2135oia110] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|