1
|
Glynn RA, Hayer KE, Bassing CH. ATM-dependent Phosphorylation of Nemo SQ Motifs Is Dispensable for Nemo-mediated Gene Expression Changes in Response to DNA Double-Strand Breaks. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:628-640. [PMID: 39007641 PMCID: PMC11348802 DOI: 10.4049/jimmunol.2300139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 06/25/2024] [Indexed: 07/16/2024]
Abstract
In response to DNA double-strand breaks (DSBs), the ATM kinase activates NF-κB factors to stimulate gene expression changes that promote survival and allow time for cells to repair damage. In cell lines, ATM can activate NF-κB transcription factors via two independent, convergent mechanisms. One is ATM-mediated phosphorylation of nuclear NF-κB essential modulator (Nemo) protein, which leads to monoubiquitylation and export of Nemo to the cytoplasm where it engages the IκB kinase (IKK) complex to activate NF-κB. Another is DSB-triggered migration of ATM into the cytoplasm, where it promotes monoubiquitylation of Nemo and the resulting IKK-mediated activation of NF-κB. ATM has many other functions in the DSB response beyond activation of NF-κB, and Nemo activates NF-κB downstream of diverse stimuli, including developmental or proinflammatory stimuli such as LPSs. To elucidate the in vivo role of DSB-induced, ATM-dependent changes in expression of NF-κB-responsive genes, we generated mice expressing phosphomutant Nemo protein lacking consensus SQ sites for phosphorylation by ATM or related kinases. We demonstrate that these mice are viable/healthy and fertile and exhibit overall normal B and T lymphocyte development. Moreover, treatment of their B lineage cells with LPS induces normal NF-κB-regulated gene expression changes. Furthermore, in marked contrast to results from a pre-B cell line, primary B lineage cells expressing phosphomutant Nemo treated with the genotoxic drug etoposide induce normal ATM- and Nemo-dependent changes in expression of NF-κB-regulated genes. Our data demonstrate that ATM-dependent phosphorylation of Nemo SQ motifs in vivo is dispensable for DSB-signaled changes in expression of NF-κB-regulated genes.
Collapse
Affiliation(s)
- Rebecca A. Glynn
- Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Katharina E. Hayer
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
- Biomedical Engineering Doctoral Degree Program, School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104
- Department of Biomedical and Health Informatics, Children’s Hospital of Philadelphia, Perelman School of Medicine, Philadelphia, PA, 19104
| | - Craig H. Bassing
- Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
2
|
Rogges E, Pelliccia S, Savio C, Lopez G, Della Starza I, La Verde G, Di Napoli A. Molecular Features of HHV8 Monoclonal Microlymphoma Associated with Kaposi Sarcoma and Multicentric Castleman Disease in an HIV-Negative Patient. Int J Mol Sci 2024; 25:3775. [PMID: 38612584 PMCID: PMC11011749 DOI: 10.3390/ijms25073775] [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: 02/25/2024] [Revised: 03/24/2024] [Accepted: 03/24/2024] [Indexed: 04/14/2024] Open
Abstract
Human herpesvirus 8 (HHV8)-associated diseases include Kaposi sarcoma (KS), multicentric Castleman disease (MCD), germinotropic lymphoproliferative disorder (GLPD), Kaposi sarcoma inflammatory cytokine syndrome (KICS), HHV8-positive diffuse large B-cell lymphoma (HHV8+ DLBCL), primary effusion lymphoma (PEL), and extra-cavitary PEL (ECPEL). We report the case of a human immunodeficiency virus (HIV)-negative male treated for cutaneous KS, who developed generalized lymphadenopathy, hepatosplenomegaly, pleural and abdominal effusions, renal insufficiency, and pancytopenia. The excised lymph node showed features of concomitant involvement by micro-KS and MCD, with aggregates of HHV8+, Epstein Barr virus (EBV)-negative, IgM+, and lambda+ plasmablasts reminiscent of microlymphoma. Molecular investigations revealed a somatically hypermutated (SHM) monoclonal rearrangement of the immunoglobulin heavy chain (IGH), accounting for 4% of the B-cell population of the lymph node. Mutational analyses identified a pathogenic variant of KMT2D and variants of unknown significance in KMT2D, FOXO1, ARID1A, and KMT2A. The patient died shortly after surgery. The histological features (HHV8+, EBV-, IgM+, Lambda+, MCD+), integrated with the molecular findings (monoclonal IGH, SHM+, KMT2D mutated), supported the diagnosis of a monoclonal HHV8+ microlymphoma, with features intermediate between an incipient HHV8+ DLBCL and an EBV-negative ECPEL highlighting the challenges in the accurate classification of HHV8-driven lymphoid proliferations.
Collapse
Affiliation(s)
- Evelina Rogges
- Department of Medical and Surgical Sciences and Translational Medicine, Faculty of Medicine and Psychology, PhD School in Translational Medicine and Oncology, Sapienza University of Rome, 00189 Rome, Italy;
| | - Sabrina Pelliccia
- Hematology Unit, Department of Clinical and Molecular Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, 00189 Rome, Italy; (S.P.); (G.L.V.)
| | - Camilla Savio
- Medical Genetics Unit, Department of Diagnostic Sciences, Sant’Andrea University Hospital, 00189 Rome, Italy;
| | - Gianluca Lopez
- Pathology Unit, Department of Clinical and Molecular Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, 00189 Rome, Italy;
| | - Irene Della Starza
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, 00161 Rome, Italy;
| | - Giacinto La Verde
- Hematology Unit, Department of Clinical and Molecular Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, 00189 Rome, Italy; (S.P.); (G.L.V.)
| | - Arianna Di Napoli
- Pathology Unit, Department of Clinical and Molecular Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, 00189 Rome, Italy;
| |
Collapse
|
3
|
Schaffer AM, Fiala GJ, Hils M, Natali E, Babrak L, Herr LA, Romero-Mulero MC, Cabezas-Wallscheid N, Rizzi M, Miho E, Schamel WWA, Minguet S. Kidins220 regulates the development of B cells bearing the λ light chain. eLife 2024; 13:e83943. [PMID: 38271217 PMCID: PMC10810608 DOI: 10.7554/elife.83943] [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: 10/12/2022] [Accepted: 12/27/2023] [Indexed: 01/27/2024] Open
Abstract
The ratio between κ and λ light chain (LC)-expressing B cells varies considerably between species. We recently identified Kinase D-interacting substrate of 220 kDa (Kidins220) as an interaction partner of the BCR. In vivo ablation of Kidins220 in B cells resulted in a marked reduction of λLC-expressing B cells. Kidins220 knockout B cells fail to open and recombine the genes of the Igl locus, even in genetic scenarios where the Igk genes cannot be rearranged or where the κLC confers autoreactivity. Igk gene recombination and expression in Kidins220-deficient B cells is normal. Kidins220 regulates the development of λLC B cells by enhancing the survival of developing B cells and thereby extending the time-window in which the Igl locus opens and the genes are rearranged and transcribed. Further, our data suggest that Kidins220 guarantees optimal pre-BCR and BCR signaling to induce Igl locus opening and gene recombination during B cell development and receptor editing.
Collapse
Affiliation(s)
- Anna-Maria Schaffer
- Faculty of Biology, Albert-Ludwigs-University of FreiburgFreiburgGermany
- Signalling Research Centers BIOSS and CIBSS, University of FreiburgFreiburgGermany
- Center of Chronic Immunodeficiency CCI, University Clinics and Medical FacultyFreiburgGermany
| | - Gina Jasmin Fiala
- Faculty of Biology, Albert-Ludwigs-University of FreiburgFreiburgGermany
- Signalling Research Centers BIOSS and CIBSS, University of FreiburgFreiburgGermany
- Center of Chronic Immunodeficiency CCI, University Clinics and Medical FacultyFreiburgGermany
| | - Miriam Hils
- Faculty of Biology, Albert-Ludwigs-University of FreiburgFreiburgGermany
- Center of Chronic Immunodeficiency CCI, University Clinics and Medical FacultyFreiburgGermany
- Department of Dermatology and Allergy Biederstein, School of Medicine, Technical University of MunichMunichGermany
| | - Eriberto Natali
- Institute of Medical Engineering and Medical Informatics, School of Life Sciences, FHNW 15 University of Applied Sciences and Arts Northwestern SwitzerlandMuttenzSwitzerland
| | - Lmar Babrak
- Institute of Medical Engineering and Medical Informatics, School of Life Sciences, FHNW 15 University of Applied Sciences and Arts Northwestern SwitzerlandMuttenzSwitzerland
| | - Laurenz Alexander Herr
- Faculty of Biology, Albert-Ludwigs-University of FreiburgFreiburgGermany
- Signalling Research Centers BIOSS and CIBSS, University of FreiburgFreiburgGermany
- Center of Chronic Immunodeficiency CCI, University Clinics and Medical FacultyFreiburgGermany
| | - Mari Carmen Romero-Mulero
- Faculty of Biology, Albert-Ludwigs-University of FreiburgFreiburgGermany
- Max Planck Institute of Immunobiology and EpigeneticsFreiburgGermany
| | - Nina Cabezas-Wallscheid
- Max Planck Institute of Immunobiology and EpigeneticsFreiburgGermany
- CIBSS – Centre for Integrative Biological Signalling Studies, University of FreiburgFreiburgGermany
| | - Marta Rizzi
- Center of Chronic Immunodeficiency CCI, University Clinics and Medical FacultyFreiburgGermany
- CIBSS – Centre for Integrative Biological Signalling Studies, University of FreiburgFreiburgGermany
- Division of Clinical and Experimental Immunology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of ViennaViennaAustria
- Department of Rheumatology and Clinical Immunology, University Medical Center Freiburg, Faculty of Medicine, University of FreiburgFreiburgGermany
| | - Enkelejda Miho
- Institute of Medical Engineering and Medical Informatics, School of Life Sciences, FHNW 15 University of Applied Sciences and Arts Northwestern SwitzerlandMuttenzSwitzerland
- aiNET GmbHBaselSwitzerland
- SIB Swiss Institute of BioinformaticsLausanneSwitzerland
| | - Wolfgang WA Schamel
- Faculty of Biology, Albert-Ludwigs-University of FreiburgFreiburgGermany
- Signalling Research Centers BIOSS and CIBSS, University of FreiburgFreiburgGermany
- Center of Chronic Immunodeficiency CCI, University Clinics and Medical FacultyFreiburgGermany
| | - Susana Minguet
- Faculty of Biology, Albert-Ludwigs-University of FreiburgFreiburgGermany
- Signalling Research Centers BIOSS and CIBSS, University of FreiburgFreiburgGermany
- Center of Chronic Immunodeficiency CCI, University Clinics and Medical FacultyFreiburgGermany
| |
Collapse
|
4
|
Fathi N, Mojtahedi H, Nasiri M, Abolhassani H, Yousefpour Marzbali M, Esmaeili M, Salami F, Biglari F, Rezaei N. How do nuclear factor kappa B (NF-κB)1 and NF-κB2 defects lead to the incidence of clinical and immunological manifestations of inborn errors of immunity? Expert Rev Clin Immunol 2023; 19:329-339. [PMID: 36706462 DOI: 10.1080/1744666x.2023.2174105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
INTRODUCTION Genetic defects affect the manner of the immune system's development, activation, and function. Nuclear factor-kappa B subunit 1 (NF-κB1) and NF-κB2 are involved in different biological processes, and deficiency in these transcription factors may reveal clinical and immunological difficulties. AREAS COVERED This review article gathers the most frequent clinical and immunological remarkable characteristics of NF-κB1 and NF-κB2 deficiencies. Afterward, an effort is made to describe the biological mechanism, which is likely to be the cause of these clinical and immunological abnormalities. EXPERT OPINION The present review article has explained the mechanism of contributions of the NF-κB1 and NF-κB2 deficiency in revealing immunodeficiency symptoms, specifically immunological and clinical manifestations. These mechanisms demonstrate the importance of NF-κB1 and NF-κB2 signaling pathways for B and T cell development, activation, antibody production, and immunotolerance. The manifestation of a mutation can range from no symptoms to severe complications in a family.
Collapse
Affiliation(s)
- Nazanin Fathi
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Hanieh Mojtahedi
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Marzieh Nasiri
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Hassan Abolhassani
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Biosciences and Nutrition, Karolinska Institute, Stockholm, Sweden
| | - Mahsa Yousefpour Marzbali
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,International Network of Stem Cell (INSC), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Marzie Esmaeili
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Fereshte Salami
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Furozan Biglari
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Johnston R, Mathias B, Crowley SJ, Schmidt HA, White LS, Mosammaparast N, Green AM, Bednarski JJ. Nuclease-independent functions of RAG1 direct distinct DNA damage responses in B cells. EMBO Rep 2023; 24:e55429. [PMID: 36382770 PMCID: PMC9827558 DOI: 10.15252/embr.202255429] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 10/23/2022] [Accepted: 10/26/2022] [Indexed: 11/18/2022] Open
Abstract
Developing B cells generate DNA double-stranded breaks (DSBs) to assemble immunoglobulin receptor (Ig) genes necessary for the expression of a mature B cell receptor. These physiologic DSBs are made by the RAG endonuclease, which is comprised of the RAG1 and RAG2 proteins. In pre-B cells, RAG-mediated DSBs activate the ATM kinase to coordinate canonical and non-canonical DNA damage responses (DDR) that trigger DSB repair and B cell developmental signals, respectively. Whether this broad cellular response is distinctive to RAG DSBs is poorly understood. To delineate the factors that direct DDR signaling in B cells, we express a tetracycline-inducible Cas9 nuclease in Rag1-deficient pre-B cells. Both RAG- and Cas9-mediated DSBs at Ig genes activate canonical DDR. In contrast, RAG DSBs, but not Cas9 DSBs, induce the non-canonical DDR-dependent developmental program. This unique response to RAG DSBs is, in part, regulated by non-core regions of RAG1. Thus, B cells trigger distinct cellular responses to RAG DSBs through unique properties of the RAG endonuclease that promotes activation of B cell developmental programs.
Collapse
Affiliation(s)
- Rachel Johnston
- Department of PediatricsWashington University School of MedicineSt. LouisMOUSA
| | - Brendan Mathias
- Department of PediatricsWashington University School of MedicineSt. LouisMOUSA
| | - Stephanie J Crowley
- Department of PediatricsWashington University School of MedicineSt. LouisMOUSA
| | - Haley A Schmidt
- Department of PediatricsWashington University School of MedicineSt. LouisMOUSA
| | - Lynn S White
- Department of PediatricsWashington University School of MedicineSt. LouisMOUSA
| | - Nima Mosammaparast
- Department of Pathology and ImmunologyWashington University School of MedicineSt. LouisMOUSA
| | - Abby M Green
- Department of PediatricsWashington University School of MedicineSt. LouisMOUSA
| | - Jeffrey J Bednarski
- Department of PediatricsWashington University School of MedicineSt. LouisMOUSA
| |
Collapse
|
6
|
Liu P, Li Y, Wang W, Bai Y, Jia H, Yuan Z, Yang Z. Role and mechanisms of the NF-ĸB signaling pathway in various developmental processes. Biomed Pharmacother 2022; 153:113513. [DOI: 10.1016/j.biopha.2022.113513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/26/2022] [Accepted: 08/01/2022] [Indexed: 11/02/2022] Open
|
7
|
Okoreeh MK, Kennedy DE, Emmanuel AO, Veselits M, Moshin A, Ladd RH, Erickson S, McLean KC, Madrigal B, Nemazee D, Maienschein-Cline M, Mandal M, Clark MR. Asymmetrical forward and reverse developmental trajectories determine molecular programs of B cell antigen receptor editing. Sci Immunol 2022; 7:eabm1664. [PMID: 35930652 PMCID: PMC9636592 DOI: 10.1126/sciimmunol.abm1664] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
During B lymphopoiesis, B cell progenitors progress through alternating and mutually exclusive stages of clonal expansion and immunoglobulin (Ig) gene rearrangements. Great diversity is generated through the stochastic recombination of Ig gene segments encoding heavy and light chain variable domains. However, this commonly generates autoreactivity. Receptor editing is the predominant tolerance mechanism for self-reactive B cells in the bone marrow (BM). B cell receptor editing rescues autoreactive B cells from negative selection through renewed light chain recombination first at Igκ then Igλ loci. Receptor editing depends on BM microenvironment cues and key transcription factors such as NF-κB, FOXO, and E2A. The specific BM factor required for receptor editing is unknown. Furthermore, how transcription factors coordinate these developmental programs to promote usage of the λ chain remains poorly defined. Therefore, we used two mouse models that recapitulate pathways by which Igλ light chain-positive B cells develop. The first has deleted J kappa (Jκ) genes and hence models Igλ expression resulting from failed Igκ recombination (Igκdel). The second models autoreactivity by ubiquitous expression of a single-chain chimeric anti-Igκ antibody (κ-mac). Here, we demonstrated that autoreactive B cells transit asymmetric forward and reverse developmental trajectories. This imparted a unique epigenetic landscape on small pre-B cells, which opened chromatin to transcription factors essential for Igλ recombination. The consequences of this asymmetric developmental path were both amplified and complemented by CXCR4 signaling. These findings reveal how intrinsic molecular programs integrate with extrinsic signals to drive receptor editing.
Collapse
Affiliation(s)
- Michael K. Okoreeh
- Department of Medicine, Section of Rheumatology, University of Chicago, Chicago, IL, 60637, USA
- Gwen Knapp Center for Lupus and Immunology Research, University of Chicago, Chicago, IL, 60637, USA
- Growth, Development, Disabilities Training program (GDDTP), Pritzker School of Medicine, University of Chicago, IL, 60637, USA
| | - Domenick E. Kennedy
- Department of Medicine, Section of Rheumatology, University of Chicago, Chicago, IL, 60637, USA
- Gwen Knapp Center for Lupus and Immunology Research, University of Chicago, Chicago, IL, 60637, USA
- Present Address: Drug Discovery Science and Technology, Discovery Platform Technologies, Chemical Biology and Emerging Therapeutics, AbbVie, North Chicago, IL, United States
| | - Akinola Olumide Emmanuel
- Department of Medicine, Section of Rheumatology, University of Chicago, Chicago, IL, 60637, USA
- Gwen Knapp Center for Lupus and Immunology Research, University of Chicago, Chicago, IL, 60637, USA
| | - Margaret Veselits
- Department of Medicine, Section of Rheumatology, University of Chicago, Chicago, IL, 60637, USA
- Gwen Knapp Center for Lupus and Immunology Research, University of Chicago, Chicago, IL, 60637, USA
| | - Azam Moshin
- Department of Medicine, Section of Rheumatology, University of Chicago, Chicago, IL, 60637, USA
- Gwen Knapp Center for Lupus and Immunology Research, University of Chicago, Chicago, IL, 60637, USA
| | - Robert H. Ladd
- Cytometry and Antibody Technologies Facility, University of Chicago, Chicago, IL, 60637, USA
| | - Steven Erickson
- Department of Pathology, University of Chicago, Chicago, IL, 60637, USA
| | - Kaitlin C. McLean
- Department of Medicine, Section of Rheumatology, University of Chicago, Chicago, IL, 60637, USA
- Gwen Knapp Center for Lupus and Immunology Research, University of Chicago, Chicago, IL, 60637, USA
| | - Brianna Madrigal
- Department of Medicine, Section of Rheumatology, University of Chicago, Chicago, IL, 60637, USA
| | - David Nemazee
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | - Malay Mandal
- Department of Medicine, Section of Rheumatology, University of Chicago, Chicago, IL, 60637, USA
- Gwen Knapp Center for Lupus and Immunology Research, University of Chicago, Chicago, IL, 60637, USA
| | - Marcus R. Clark
- Department of Medicine, Section of Rheumatology, University of Chicago, Chicago, IL, 60637, USA
- Gwen Knapp Center for Lupus and Immunology Research, University of Chicago, Chicago, IL, 60637, USA
| |
Collapse
|
8
|
Glynn RA, Bassing CH. Nemo-Dependent, ATM-Mediated Signals from RAG DNA Breaks at Igk Feedback Inhibit V κ Recombination to Enforce Igκ Allelic Exclusion. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:371-383. [PMID: 34965965 PMCID: PMC8756740 DOI: 10.4049/jimmunol.2100696] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 11/01/2021] [Indexed: 01/17/2023]
Abstract
Monoallelic AgR gene expression underlies specific adaptive immune responses. AgR allelic exclusion is achieved by sequential initiation of V(D)J recombination between alleles and resultant protein from one allele signaling to prevent recombination of the other. The ATM kinase, a regulator of the DNA double-strand break (DSB) response, helps enforce allelic exclusion through undetermined mechanisms. ATM promotes repair of RAG1/RAG2 (RAG) endonuclease-induced DSBs and transduces signals from RAG DSBs during Igk gene rearrangement on one allele to transiently inhibit RAG1 protein expression, Igk accessibility, and RAG cleavage of the other allele. Yet, the relative contributions of ATM functions in DSB repair versus signaling to enforce AgR allelic exclusion remain undetermined. In this study, we demonstrate that inactivation in mouse pre-B cells of the NF-κB essential modulator (Nemo) protein, an effector of ATM signaling, diminishes RAG DSB-triggered repression of Rag1/Rag2 transcription and Igk accessibility but does not result in aberrant repair of RAG DSBs like ATM inactivation. We show that Nemo deficiency increases simultaneous biallelic Igk cleavage in pre-B cells and raises the frequency of B cells expressing Igκ proteins from both alleles. In contrast, the incidence of biallelic Igκ expression is not elevated by inactivation of the SpiC transcriptional repressor, which is induced by RAG DSBs in an ATM-dependent manner and suppresses Igk accessibility. Thus, we conclude that Nemo-dependent, ATM-mediated DNA damage signals enforce Igκ allelic exclusion by orchestrating transient repression of RAG expression and feedback inhibition of additional Igk rearrangements in response to RAG cleavage on one Igk allele.
Collapse
Affiliation(s)
- Rebecca A. Glynn
- Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104,Department of Pathology and Laboratory Medicine, Children’s Hospital of Pennsylvania, Philadelphia, PA 19104
| | - Craig H. Bassing
- Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104,Department of Pathology and Laboratory Medicine, Children’s Hospital of Pennsylvania, Philadelphia, PA 19104,Corresponding Author: Craig H. Bassing, Ph.D., Children’s Hospital of Philadelphia, 4054 Colket Translational Research Building, 3501 Civic Center Blvd., Philadelphia, PA 19104, 267-426-0311,
| |
Collapse
|
9
|
Zhou T, Wang HW, Pittaluga S, Jaffe ES. Multicentric Castleman disease and the evolution of the concept. Pathologica 2021; 113:339-353. [PMID: 34837092 PMCID: PMC8720411 DOI: 10.32074/1591-951x-351] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 08/25/2021] [Indexed: 12/29/2022] Open
Abstract
The term multicentric Castleman disease (MCD) encompasses a spectrum of conditions that share some overlapping clinicopathological manifestations. The fundamental pathogenetic mechanism involves dysregulated cytokine activity, causing systemic inflammatory symptoms as well as lymphadenopathy. Some of the histological changes in lymph nodes resemble the histology of unicentric Castleman disease (UCD). However, based on current knowledge, the use of this shared nomenclature is unfortunate, since these disorders differ in pathogenesis and prognosis. In Kaposi sarcoma-associated herpesvirus (KSHV)-associated MCD, cytokine overactivity is caused by viral products, which can also lead to atypical lymphoproliferations and potential progression to lymphoma. In idiopathic MCD, the hypercytokinemia can result from various mechanisms, which ultimately lead to different constellations of clinical presentations and varied pathology in lymphoid tissues. The authors review the evolving concepts and definitions of the various conditions under the eponym of multicentric Castleman disease.
Collapse
Affiliation(s)
- Ting Zhou
- Hematopathology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Hao-Wei Wang
- Hematopathology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Stefania Pittaluga
- Hematopathology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Elaine S Jaffe
- Hematopathology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
10
|
Beilinson HA, Glynn RA, Yadavalli AD, Xiao J, Corbett E, Saribasak H, Arya R, Miot C, Bhattacharyya A, Jones JM, Pongubala JM, Bassing CH, Schatz DG. The RAG1 N-terminal region regulates the efficiency and pathways of synapsis for V(D)J recombination. J Exp Med 2021; 218:e20210250. [PMID: 34402853 PMCID: PMC8374863 DOI: 10.1084/jem.20210250] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 06/30/2021] [Accepted: 07/30/2021] [Indexed: 11/29/2022] Open
Abstract
Immunoglobulin and T cell receptor gene assembly depends on V(D)J recombination initiated by the RAG1-RAG2 recombinase. The RAG1 N-terminal region (NTR; aa 1-383) has been implicated in regulatory functions whose influence on V(D)J recombination and lymphocyte development in vivo is poorly understood. We generated mice in which RAG1 lacks ubiquitin ligase activity (P326G), the major site of autoubiquitination (K233R), or its first 215 residues (Δ215). While few abnormalities were detected in R1.K233R mice, R1.P326G mice exhibit multiple features indicative of reduced recombination efficiency, including an increased Igκ+:Igλ+ B cell ratio and decreased recombination of Igh, Igκ, Igλ, and Tcrb loci. Previous studies indicate that synapsis of recombining partners during Igh recombination occurs through two pathways: long-range scanning and short-range collision. We find that R1Δ215 mice exhibit reduced short-range Igh and Tcrb D-to-J recombination. Our findings indicate that the RAG1 NTR regulates V(D)J recombination and lymphocyte development by multiple pathways, including control of the balance between short- and long-range recombination.
Collapse
Affiliation(s)
- Helen A. Beilinson
- Department of Immunobiology, Yale School of Medicine, Yale University, New Haven, CT
| | - Rebecca A. Glynn
- Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Anurupa Devi Yadavalli
- Department of Immunobiology, Yale School of Medicine, Yale University, New Haven, CT
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Jianxiong Xiao
- Department of Immunobiology, Yale School of Medicine, Yale University, New Haven, CT
| | - Elizabeth Corbett
- Department of Immunobiology, Yale School of Medicine, Yale University, New Haven, CT
| | - Huseyin Saribasak
- Department of Immunobiology, Yale School of Medicine, Yale University, New Haven, CT
| | - Rahul Arya
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Charline Miot
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Anamika Bhattacharyya
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC
| | - Jessica M. Jones
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC
| | - Jagan M.R. Pongubala
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Craig H. Bassing
- Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - David G. Schatz
- Department of Immunobiology, Yale School of Medicine, Yale University, New Haven, CT
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT
| |
Collapse
|
11
|
Barnabei L, Laplantine E, Mbongo W, Rieux-Laucat F, Weil R. NF-κB: At the Borders of Autoimmunity and Inflammation. Front Immunol 2021; 12:716469. [PMID: 34434197 PMCID: PMC8381650 DOI: 10.3389/fimmu.2021.716469] [Citation(s) in RCA: 340] [Impact Index Per Article: 85.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 07/22/2021] [Indexed: 12/18/2022] Open
Abstract
The transcription factor NF-κB regulates multiple aspects of innate and adaptive immune functions and serves as a pivotal mediator of inflammatory response. In the first part of this review, we discuss the NF-κB inducers, signaling pathways, and regulators involved in immune homeostasis as well as detail the importance of post-translational regulation by ubiquitination in NF-κB function. We also indicate the stages of central and peripheral tolerance where NF-κB plays a fundamental role. With respect to central tolerance, we detail how NF-κB regulates medullary thymic epithelial cell (mTEC) development, homeostasis, and function. Moreover, we elaborate on its role in the migration of double-positive (DP) thymocytes from the thymic cortex to the medulla. With respect to peripheral tolerance, we outline how NF-κB contributes to the inactivation and destruction of autoreactive T and B lymphocytes as well as the differentiation of CD4+-T cell subsets that are implicated in immune tolerance. In the latter half of the review, we describe the contribution of NF-κB to the pathogenesis of autoimmunity and autoinflammation. The recent discovery of mutations involving components of the pathway has both deepened our understanding of autoimmune disease and informed new therapeutic approaches to treat these illnesses.
Collapse
Affiliation(s)
- Laura Barnabei
- INSERM UMR 1163, Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute Paris Descartes Sorbonne Paris Cité University, Paris, France
| | - Emmanuel Laplantine
- Sorbonne Universités, Institut National de la Santé et de la Recherche Médicale (INSERM, UMR1135), Centre National de la Recherche Scientifique (CNRS, ERL8255), Centre d'Immunologie et des Maladies Infectieuses CMI, Paris, France
| | - William Mbongo
- Sorbonne Universités, Institut National de la Santé et de la Recherche Médicale (INSERM, UMR1135), Centre National de la Recherche Scientifique (CNRS, ERL8255), Centre d'Immunologie et des Maladies Infectieuses CMI, Paris, France
| | - Frédéric Rieux-Laucat
- INSERM UMR 1163, Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute Paris Descartes Sorbonne Paris Cité University, Paris, France
| | - Robert Weil
- Sorbonne Universités, Institut National de la Santé et de la Recherche Médicale (INSERM, UMR1135), Centre National de la Recherche Scientifique (CNRS, ERL8255), Centre d'Immunologie et des Maladies Infectieuses CMI, Paris, France
| |
Collapse
|
12
|
Constitutive activation of NF-κB during early bone marrow development results in loss of B cells at the pro-B-cell stage. Blood Adv 2021; 5:745-755. [PMID: 33560391 DOI: 10.1182/bloodadvances.2020002932] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 12/21/2020] [Indexed: 11/20/2022] Open
Abstract
There is a considerable body of work exploring the role of NF-κB family of transcription factors in the maturation and functions of later stage B cells; however, their role in the earlier bone marrow stages of development is less well understood despite the demonstration that NF-κB activity is present at all early stages of B-cell development. To explore the consequences of early, B cell-targeted constitutive activation of both NF-κB pathways on B-cell development, we generated mice that have either or both. NF-κB pathways constitutively activated beginning in early pro-B cells. In marked contrast to activating a single pathway, we found mice with both pathways constitutively activated displayed a profound loss of B cells, starting with early pro-B cells and peaking at the late pro-B-cell stage, at least in part as a result of increased apoptosis. This effect was found to be cell autonomous and to have striking phenotypic consequences on the secondary lymphoid organs and circulating antibody levels. This effect was also found to be temporal in nature as similar activation under a Cre expressed later in development did not result in generation of a similar phenotype. Taken together, these findings help to shed further light on the need for tight regulation of the NF-κB family of transcription factors during the various stages of B-cell development in the bone marrow.
Collapse
|
13
|
Zhang T, Sun J, Cheng J, Yin W, Li J, Miller H, Herrada AA, Gu H, Song H, Chen Y, Gong Q, Liu C. The role of ubiquitinase in B cell development and function. J Leukoc Biol 2020; 109:395-405. [PMID: 32816356 DOI: 10.1002/jlb.1mr0720-185rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 07/16/2020] [Accepted: 07/18/2020] [Indexed: 11/10/2022] Open
Abstract
Ubiquitinases are a select group of enzymes that modify target proteins through ubiquitination, which plays a crucial role in the regulation of protein degradation, location, and function. B lymphocytes that originated from bone marrow hematopoietic stem cells (HSC), exert humoral immune functions by differentiating into plasma cells and producing antibodies. Previous studies have shown that ubiquitination is involved in the regulation of the cell cycle and signal transduction important for B lymphocyte development and function. In this review, how ubiquitinases regulate B cell development, activation, apoptosis, and proliferation is discussed, which could help in understanding the physiological processes and diseases related to B cells and also provides potential new targets for further studies.
Collapse
Affiliation(s)
- Tong Zhang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianxuan Sun
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiali Cheng
- Department of hematology, Tongji Hospital, Tongji Medical college, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Yin
- Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingwen Li
- Department of hematology, Wuhan Union Hospital, Tongji Medical college, Huazhong University of Science and Technology, Wuhan, China
| | - Heather Miller
- Department of Intracellular Pathogens, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, USA
| | - Andrés A Herrada
- Lymphatic and Inflammation Research Laboratory, Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Talca, Chile
| | - Heng Gu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongmei Song
- Department of Pediatrics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No 1, Shuaifuyuan, Dongcheng District, Beijing, China
| | - Yan Chen
- The Second Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Quan Gong
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China.,Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, China
| | - Chaohong Liu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
14
|
McLean KC, Mandal M. It Takes Three Receptors to Raise a B Cell. Trends Immunol 2020; 41:629-642. [PMID: 32451219 DOI: 10.1016/j.it.2020.05.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/03/2020] [Accepted: 05/04/2020] [Indexed: 12/14/2022]
Abstract
As the unique source of diverse immunoglobulin repertoires, B lymphocytes are an indispensable part of humoral immunity. B cell progenitors progress through sequential and mutually exclusive states of proliferation and recombination, coordinated by cytokines and chemokines. Mutations affecting the crucial pre-B cell checkpoint result in immunodeficiency, autoimmunity, and leukemia. This checkpoint was previously modeled by the signaling of two opposing receptors, IL-7R and the pre-BCR. We provide an update to this model in which three receptors, IL-7R, pre-BCR, and CXCR4, work in concert to coordinate both the proper positioning of B cell progenitors in the bone marrow (BM) microenvironment and their progression through the pre-B checkpoint. Furthermore, signaling initiated by all three receptors directly instructs cell fate and developmental progression.
Collapse
Affiliation(s)
- Kaitlin C McLean
- Section of Rheumatology, and Gwen Knapp Center for Lupus and Immunology Research, Department of Medicine, University of Chicago, IL 60637, USA
| | - Malay Mandal
- Section of Rheumatology, and Gwen Knapp Center for Lupus and Immunology Research, Department of Medicine, University of Chicago, IL 60637, USA.
| |
Collapse
|
15
|
Mandal M, Okoreeh MK, Kennedy DE, Maienschein-Cline M, Ai J, McLean KC, Kaverina N, Veselits M, Aifantis I, Gounari F, Clark MR. CXCR4 signaling directs Igk recombination and the molecular mechanisms of late B lymphopoiesis. Nat Immunol 2019; 20:1393-1403. [PMID: 31477919 PMCID: PMC6754289 DOI: 10.1038/s41590-019-0468-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 07/16/2019] [Indexed: 12/17/2022]
Abstract
In B lymphopoiesis, activation of the pre-B cell antigen receptor (pre-BCR) is associated with both cell cycle exit and Igk recombination. Yet how the pre-BCR mediates these functions remains unclear. Here, we demonstrate that the pre-BCR initiates a feed-forward amplification loop mediated by the transcription factor interferon regulatory factor 4 and the chemokine receptor C-X-C motif chemokine receptor 4 (CXCR4). CXCR4 ligation by C-X-C motif chemokine ligand 12 activates the mitogen-activated protein kinase extracellular-signal-regulated kinase, which then directs the development of small pre- and immature B cells, including orchestrating cell cycle exit, pre-BCR repression, Igk recombination and BCR expression. In contrast, pre-BCR expression and escape from interleukin-7 have only modest effects on B cell developmental transcriptional and epigenetic programs. These data show a direct and central role for CXCR4 in orchestrating late B cell lymphopoiesis. Furthermore, in the context of previous findings, our data provide a three-receptor system sufficient to recapitulate the essential features of B lymphopoiesis in vitro.
Collapse
Affiliation(s)
- Malay Mandal
- Department of Medicine, Section of Rheumatology, University of Chicago, Chicago, IL, USA.
- Gwen Knapp Center for Lupus and Immunology Research, University of Chicago, Chicago, IL, USA.
| | - Michael K Okoreeh
- Department of Medicine, Section of Rheumatology, University of Chicago, Chicago, IL, USA
- Gwen Knapp Center for Lupus and Immunology Research, University of Chicago, Chicago, IL, USA
| | - Domenick E Kennedy
- Department of Medicine, Section of Rheumatology, University of Chicago, Chicago, IL, USA
- Gwen Knapp Center for Lupus and Immunology Research, University of Chicago, Chicago, IL, USA
| | | | - Junting Ai
- Department of Medicine, Section of Rheumatology, University of Chicago, Chicago, IL, USA
- Gwen Knapp Center for Lupus and Immunology Research, University of Chicago, Chicago, IL, USA
| | - Kaitlin C McLean
- Department of Medicine, Section of Rheumatology, University of Chicago, Chicago, IL, USA
- Gwen Knapp Center for Lupus and Immunology Research, University of Chicago, Chicago, IL, USA
| | - Natalya Kaverina
- Division of Nephrology, University of Washington, Seattle, WA, USA
| | - Margaret Veselits
- Department of Medicine, Section of Rheumatology, University of Chicago, Chicago, IL, USA
- Gwen Knapp Center for Lupus and Immunology Research, University of Chicago, Chicago, IL, USA
| | - Iannis Aifantis
- Department of Pathology, NYU School of Medicine, New York, NY, USA
- Laura and Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY, USA
| | - Fotini Gounari
- Department of Medicine, Section of Rheumatology, University of Chicago, Chicago, IL, USA
- Gwen Knapp Center for Lupus and Immunology Research, University of Chicago, Chicago, IL, USA
| | - Marcus R Clark
- Department of Medicine, Section of Rheumatology, University of Chicago, Chicago, IL, USA.
- Gwen Knapp Center for Lupus and Immunology Research, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
16
|
Transcription factors IRF8 and PU.1 are required for follicular B cell development and BCL6-driven germinal center responses. Proc Natl Acad Sci U S A 2019; 116:9511-9520. [PMID: 31000603 DOI: 10.1073/pnas.1901258116] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The IRF and Ets families of transcription factors regulate the expression of a range of genes involved in immune cell development and function. However, the understanding of the molecular mechanisms of each family member has been limited due to their redundancy and broad effects on multiple lineages of cells. Here, we report that double deletion of floxed Irf8 and Spi1 (encoding PU.1) by Mb1-Cre (designated DKO mice) in the B cell lineage resulted in severe defects in the development of follicular and germinal center (GC) B cells. Class-switch recombination and antibody affinity maturation were also compromised in DKO mice. RNA-seq (sequencing) and ChIP-seq analyses revealed distinct IRF8 and PU.1 target genes in follicular and activated B cells. DKO B cells had diminished expression of target genes vital for maintaining follicular B cell identity and GC development. Moreover, our findings reveal that expression of B-cell lymphoma protein 6 (BCL6), which is critical for development of germinal center B cells, is dependent on IRF8 and PU.1 in vivo, providing a mechanism for the critical role for IRF8 and PU.1 in the development of GC B cells.
Collapse
|
17
|
Abstract
DNA damage occurs on exposure to genotoxic agents and during physiological DNA transactions. DNA double-strand breaks (DSBs) are particularly dangerous lesions that activate DNA damage response (DDR) kinases, leading to initiation of a canonical DDR (cDDR). This response includes activation of cell cycle checkpoints and engagement of pathways that repair the DNA DSBs to maintain genomic integrity. In adaptive immune cells, programmed DNA DSBs are generated at precise genomic locations during the assembly and diversification of lymphocyte antigen receptor genes. In innate immune cells, the production of genotoxic agents, such as reactive nitrogen molecules, in response to pathogens can also cause genomic DNA DSBs. These DSBs in adaptive and innate immune cells activate the cDDR. However, recent studies have demonstrated that they also activate non-canonical DDRs (ncDDRs) that regulate cell type-specific processes that are important for innate and adaptive immune responses. Here, we review these ncDDRs and discuss how they integrate with other signals during immune system development and function.
Collapse
Affiliation(s)
- Jeffrey J Bednarski
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Barry P Sleckman
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
18
|
STIM- and Orai-mediated calcium entry controls NF-κB activity and function in lymphocytes. Cell Calcium 2018; 74:131-143. [PMID: 30048879 DOI: 10.1016/j.ceca.2018.07.003] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 07/09/2018] [Accepted: 07/09/2018] [Indexed: 12/24/2022]
Abstract
The central role of Ca2+ signaling in the development of functional immunity and tolerance is well established. These signals are initiated by antigen binding to cognate receptors on lymphocytes that trigger store operated Ca2+ entry (SOCE). The underlying mechanism of SOCE in lymphocytes involves TCR and BCR mediated activation of Stromal Interaction Molecule 1 and 2 (STIM1/2) molecules embedded in the ER membrane leading to their activation of Orai channels in the plasma membrane. STIM/Orai dependent Ca2+ signals guide key antigen induced lymphocyte development and function principally through direct regulation of Ca2+ dependent transcription factors. The role of Ca2+ signaling in NFAT activation and signaling is well known and has been studied extensively, but a wide appreciation and mechanistic understanding of how Ca2+ signals also shape the activation and specificity of NF-κB dependent gene expression has lagged. Here we discuss and interpret what is known about Ca2+ dependent mechanisms of NF-kB activation, including what is known and the gaps in our understanding of how these signals control lymphocyte development and function.
Collapse
|
19
|
Bashford-Rogers RJM, Smith KGC, Thomas DC. Antibody repertoire analysis in polygenic autoimmune diseases. Immunology 2018; 155:3-17. [PMID: 29574826 PMCID: PMC6099162 DOI: 10.1111/imm.12927] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 03/01/2018] [Accepted: 03/06/2018] [Indexed: 12/18/2022] Open
Abstract
High-throughput sequencing of the DNA/RNA encoding antibody heavy- and light-chains is rapidly transforming the field of adaptive immunity. It can address key questions, including: (i) how the B-cell repertoire differs in health and disease; and (ii) if it does differ, the point(s) in B-cell development at which this occurs. The advent of technologies, such as whole-genome sequencing, offers the chance to link abnormalities in the B-cell antibody repertoire to specific genomic variants and polymorphisms. Here, we discuss the current research using B-cell antibody repertoire sequencing in three polygenic autoimmune diseases where there is good evidence for a pathological role for B-cells, namely systemic lupus erythematosus, multiple sclerosis and rheumatoid arthritis. These autoimmune diseases exhibit significantly skewed B-cell receptor repertoires compared with healthy controls. Interestingly, some common repertoire defects are shared between diseases, such as elevated IGHV4-34 gene usage. B-cell clones have effectively been characterized and tracked between different tissues and blood in autoimmune disease. It has been hypothesized that these differences may signify differences in B-cell tolerance; however, the mechanisms and implications of these defects are not clear.
Collapse
Affiliation(s)
| | | | - David C Thomas
- Department of Medicine, University of Cambridge, Cambridge, UK
| |
Collapse
|
20
|
Ádori M, Pedersen GK, Ádori C, Erikson E, Khoenkhoen S, Stark JM, Choi JH, Dosenovic P, Karlsson MCI, Beutler B, Karlsson Hedestam GB. Altered Marginal Zone B Cell Selection in the Absence of IκBNS. THE JOURNAL OF IMMUNOLOGY 2018; 200:775-787. [PMID: 29222168 DOI: 10.4049/jimmunol.1700791] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 11/07/2017] [Indexed: 12/20/2022]
Abstract
Marginal zone (MZ) B cells reside in the splenic MZ and play important roles in T cell-independent humoral immune responses against blood-borne pathogens. IκBNS-deficient bumble mice exhibit a severe reduction in the MZ B compartment but regain an MZ B population with age and, thus, represent a valuable model to examine the biology of MZ B cells. In this article, we characterized the MZ B cell defect in further detail and investigated the nature of the B cells that appear in the MZ of aged bumble mice. Flow cytometry analysis of the splenic transitional B cell subsets demonstrated that MZ B cell development was blocked at the transitional-1 to transitional-2-MZ precursor stage in the absence of functional IκBNS. Immunohistochemical analysis of spleen sections from wild-type and bumble mice revealed no alteration in the cellular MZ microenvironment, and analysis of bone marrow chimeras indicated that the MZ B cell development defect in bumble mice was B cell intrinsic. Further, we demonstrate that the B cells that repopulate the MZ in aged bumble mice were distinct from age-matched wild-type MZ B cells. Specifically, the expression of surface markers characteristic for MZ B cells was altered and the L chain Igλ+ repertoire was reduced in bumble mice. Finally, plasma cell differentiation of sorted LPS-stimulated MZ B cells was impaired, and aged bumble mice were unable to respond to NP-Ficoll immunization. These results demonstrate that IκBNS is required for an intact MZ B cell compartment in C57BL/6 mice.
Collapse
Affiliation(s)
- Monika Ádori
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, S-171 77 Stockholm, Sweden
| | - Gabriel K Pedersen
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, S-171 77 Stockholm, Sweden
| | - Csaba Ádori
- Department of Neuroscience, Karolinska Institutet, S-171 77 Stockholm, Sweden; and
| | - Elina Erikson
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, S-171 77 Stockholm, Sweden
| | - Sharesta Khoenkhoen
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, S-171 77 Stockholm, Sweden
| | - Julian M Stark
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, S-171 77 Stockholm, Sweden
| | - Jin Huk Choi
- Center for the Genetics of Host Defense, UT Southwestern Medical Center, Dallas, TX 75390-8505
| | - Pia Dosenovic
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, S-171 77 Stockholm, Sweden
| | - Mikael C I Karlsson
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, S-171 77 Stockholm, Sweden
| | - Bruce Beutler
- Center for the Genetics of Host Defense, UT Southwestern Medical Center, Dallas, TX 75390-8505
| | | |
Collapse
|
21
|
PTIP chromatin regulator controls development and activation of B cell subsets to license humoral immunity in mice. Proc Natl Acad Sci U S A 2017; 114:E9328-E9337. [PMID: 29078319 PMCID: PMC5676899 DOI: 10.1073/pnas.1707938114] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
To provide optimal host defense, the full spectrum of antibody-based immunity requires natural antibodies and immunization-induced antigen-specific antibodies. Here we show that the PTIP (Pax transactivation domain-interacting protein) chromatin regulator is induced by B cell activation to potentiate the establishment of steady-state and postimmune serum antibody levels. It does so by promoting activation-associated proliferation and differentiation of all the major B cell subsets, at least in part, through regulating the NF-κB pathway. With the genetic basis still unknown for a majority of patients with common variable immunodeficiency, further work investigating how PTIP controls cell signaling may generate valuable new insight for human health and disease. B cell receptor signaling and downstream NF-κB activity are crucial for the maturation and functionality of all major B cell subsets, yet the molecular players in these signaling events are not fully understood. Here we use several genetically modified mouse models to demonstrate that expression of the multifunctional BRCT (BRCA1 C-terminal) domain-containing PTIP (Pax transactivation domain-interacting protein) chromatin regulator is controlled by B cell activation and potentiates steady-state and postimmune antibody production in vivo. By examining the effects of PTIP deficiency in mice at various ages during ontogeny, we demonstrate that PTIP promotes bone marrow B cell development as well as the neonatal establishment and subsequent long-term maintenance of self-reactive B-1 B cells. Furthermore, we find that PTIP is required for B cell receptor- and T:B interaction-induced proliferation, differentiation of follicular B cells during germinal center formation, and normal signaling through the classical NF-κB pathway. Together with the previously identified role for PTIP in promoting sterile transcription at the Igh locus, the present results establish PTIP as a licensing factor for humoral immunity that acts at several junctures of B lineage maturation and effector cell differentiation by controlling B cell activation.
Collapse
|
22
|
Bahjat M, Guikema JEJ. The Complex Interplay between DNA Injury and Repair in Enzymatically Induced Mutagenesis and DNA Damage in B Lymphocytes. Int J Mol Sci 2017; 18:ijms18091876. [PMID: 28867784 PMCID: PMC5618525 DOI: 10.3390/ijms18091876] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 08/24/2017] [Accepted: 08/29/2017] [Indexed: 11/25/2022] Open
Abstract
Lymphocytes are endowed with unique and specialized enzymatic mutagenic properties that allow them to diversify their antigen receptors, which are crucial sensors for pathogens and mediators of adaptive immunity. During lymphocyte development, the antigen receptors expressed by B and T lymphocytes are assembled in an antigen-independent fashion by ordered variable gene segment recombinations (V(D)J recombination), which is a highly ordered and regulated process that requires the recombination activating gene products 1 & 2 (RAG1, RAG2). Upon activation by antigen, B lymphocytes undergo additional diversifications of their immunoglobulin B-cell receptors. Enzymatically induced somatic hypermutation (SHM) and immunoglobulin class switch recombination (CSR) improves the affinity for antigen and shape the effector function of the humoral immune response, respectively. The activation-induced cytidine deaminase (AID) enzyme is crucial for both SHM and CSR. These processes have evolved to both utilize as well as evade different DNA repair and DNA damage response pathways. The delicate balance between enzymatic mutagenesis and DNA repair is crucial for effective immune responses and the maintenance of genomic integrity. Not surprisingly, disturbances in this balance are at the basis of lymphoid malignancies by provoking the formation of oncogenic mutations and chromosomal aberrations. In this review, we discuss recent mechanistic insight into the regulation of RAG1/2 and AID expression and activity in lymphocytes and the complex interplay between these mutagenic enzymes and DNA repair and DNA damage response pathways, focusing on the base excision repair and mismatch repair pathways. We discuss how disturbances of this interplay induce genomic instability and contribute to oncogenesis.
Collapse
Affiliation(s)
- Mahnoush Bahjat
- Department of Pathology, Academic Medical Center, University of Amsterdam; Lymphoma and Myeloma Center Amsterdam (LYMMCARE), Amsterdam 1105 AZ, The Netherlands.
| | - Jeroen E J Guikema
- Department of Pathology, Academic Medical Center, University of Amsterdam; Lymphoma and Myeloma Center Amsterdam (LYMMCARE), Amsterdam 1105 AZ, The Netherlands.
| |
Collapse
|
23
|
Iacoangeli A, Lui A, Haines A, Ohta Y, Flajnik M, Hsu E. Evidence for Ig Light Chain Isotype Exclusion in Shark B Lymphocytes Suggests Ordered Mechanisms. THE JOURNAL OF IMMUNOLOGY 2017; 199:1875-1885. [PMID: 28760881 DOI: 10.4049/jimmunol.1700762] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 06/27/2017] [Indexed: 01/06/2023]
Abstract
Unlike most vertebrates, the shark IgL gene organization precludes secondary rearrangements that delete self-reactive VJ rearranged genes. Nurse sharks express four L chain isotypes, κ, λ, σ, and σ-2, encoded by 35 functional minigenes or clusters. The sequence of gene activation/expression and receptor editing of these isotypes have not been studied. We therefore investigated the extent of isotypic exclusion in separated B cell subpopulations. Surface Ig (sIg)κ-expressing cells, isolated with mAb LK14 that recognizes Cκ, carry predominantly nonproductive rearrangements of other L chain isotypes. Conversely, after depletion with LK14, sIgM+ cells contained largely nonproductive κ and enrichment for in-frame VJ of the others. Because some isotypic inclusion was observed at the mRNA level, expression in the BCR was examined. Functional λ mRNA was obtained, as expected, from the LK14-depleted population, but was also in sIgκ+ splenocytes. Whereas λ somatic mutants from the depleted sample displayed evidence of positive selection, the λ genes in sIgκ+ cells accumulated bystander mutations indicating a failure to express their products at the cell surface in association with the BCR H chain. In conclusion, a shark B cell expresses one L chain isotype at the surface and other isotypes as nonproductive VJ, sterile transcripts, or in-frame VJ whose products may not associate with the H chain. Based on the mRNA content found in the B cell subpopulations, an order of L chain gene activation is suggested as: σ-2 followed by κ, then σ and λ.
Collapse
Affiliation(s)
- Anna Iacoangeli
- Tisch Multiple Sclerosis Research Center of New York, New York, NY 10019
| | - Anita Lui
- Department of Physiology and Pharmacology, State University of New York Health Science Center at Brooklyn, Brooklyn, NY 11203; and
| | - Ashley Haines
- Department of Microbiology and Immunology, University of Maryland Baltimore, Baltimore, MD 21201
| | - Yuko Ohta
- Department of Microbiology and Immunology, University of Maryland Baltimore, Baltimore, MD 21201
| | - Martin Flajnik
- Department of Microbiology and Immunology, University of Maryland Baltimore, Baltimore, MD 21201
| | - Ellen Hsu
- Department of Physiology and Pharmacology, State University of New York Health Science Center at Brooklyn, Brooklyn, NY 11203; and
| |
Collapse
|
24
|
Arya R, Bassing CH. V(D)J Recombination Exploits DNA Damage Responses to Promote Immunity. Trends Genet 2017; 33:479-489. [PMID: 28532625 PMCID: PMC5499712 DOI: 10.1016/j.tig.2017.04.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Revised: 04/25/2017] [Accepted: 04/26/2017] [Indexed: 11/16/2022]
Abstract
It has been recognized for 40 years that the variable (diversity) joining [V(D)J] recombination-mediated assembly of diverse B and T lymphocyte antigen receptor (AgR) genes is not only essential for adaptive immunity, but also a risk for autoimmunity and lymphoid malignancies. Over the past few years, several studies have revealed that recombination-activating gene (RAG) endonuclease-induced DNA double-strand breaks (DSBs) transcend hazardous intermediates during antigen receptor gene assembly. RAG cleavage within the genomes of lymphocyte progenitors and immature lymphocytes regulates the expression of ubiquitous and lymphocyte-specific gene transcripts to control the differentiation and function of both adaptive and innate immune cell lineages. These unexpected discoveries raise important new questions that have broad implications for basic immunology research and the screening, diagnosis, and treatment of human immunological disease.
Collapse
Affiliation(s)
- Rahul Arya
- Division of Cancer Pathobiology, Department of Pathology and Laboratory Medicine, Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Craig H Bassing
- Division of Cancer Pathobiology, Department of Pathology and Laboratory Medicine, Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
25
|
Abstract
Immune tolerance hinders the potentially destructive responses of lymphocytes to host tissues. Tolerance is regulated at the stage of immature B cell development (central tolerance) by clonal deletion, involving apoptosis, and by receptor editing, which reprogrammes the specificity of B cells through secondary recombination of antibody genes. Recent mechanistic studies have begun to elucidate how these divergent mechanisms are controlled. Single-cell antibody cloning has revealed defects of B cell central tolerance in human autoimmune diseases and in several human immunodeficiency diseases caused by single gene mutations, which indicates the relevance of B cell tolerance to disease and suggests possible genetic pathways that regulate tolerance.
Collapse
|
26
|
Fisher MR, Rivera-Reyes A, Bloch NB, Schatz DG, Bassing CH. Immature Lymphocytes Inhibit Rag1 and Rag2 Transcription and V(D)J Recombination in Response to DNA Double-Strand Breaks. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2017; 198:2943-2956. [PMID: 28213501 PMCID: PMC5360515 DOI: 10.4049/jimmunol.1601639] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 01/16/2017] [Indexed: 12/26/2022]
Abstract
Mammalian cells have evolved a common DNA damage response (DDR) that sustains cellular function, maintains genomic integrity, and suppresses malignant transformation. In pre-B cells, DNA double-strand breaks (DSBs) induced at Igκ loci by the Rag1/Rag2 (RAG) endonuclease engage this DDR to modulate transcription of genes that regulate lymphocyte-specific processes. We previously reported that RAG DSBs induced at one Igκ allele signal through the ataxia telangiectasia mutated (ATM) kinase to feedback-inhibit RAG expression and RAG cleavage of the other Igκ allele. In this article, we show that DSBs induced by ionizing radiation, etoposide, or bleomycin suppress Rag1 and Rag2 mRNA levels in primary pre-B cells, pro-B cells, and pro-T cells, indicating that inhibition of Rag1 and Rag2 expression is a prevalent DSB response among immature lymphocytes. DSBs induced in pre-B cells signal rapid transcriptional repression of Rag1 and Rag2, causing downregulation of both Rag1 and Rag2 mRNA, but only Rag1 protein. This transcriptional inhibition requires the ATM kinase and the NF-κB essential modulator protein, implicating a role for ATM-mediated activation of canonical NF-κB transcription factors. Finally, we demonstrate that DSBs induced in pre-B cells by etoposide or bleomycin inhibit recombination of Igκ loci and a chromosomally integrated substrate. Our data indicate that immature lymphocytes exploit a common DDR signaling pathway to limit DSBs at multiple genomic locations within developmental stages wherein monoallelic Ag receptor locus recombination is enforced. We discuss the implications of our findings for mechanisms that orchestrate the differentiation of monospecific lymphocytes while suppressing oncogenic Ag receptor locus translocations.
Collapse
Affiliation(s)
- Megan R Fisher
- Division of Cancer Pathobiology, Department of Pathology and Laboratory Medicine, Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA 19104
- Immunology Graduate Group, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104
- Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104
| | - Adrian Rivera-Reyes
- Division of Cancer Pathobiology, Department of Pathology and Laboratory Medicine, Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA 19104
- Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104
- Cancer Biology Program of the Cell and Molecular Biology Graduate Group, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104; and
| | - Noah B Bloch
- Division of Cancer Pathobiology, Department of Pathology and Laboratory Medicine, Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA 19104
| | - David G Schatz
- Department of Immunobiology, Yale University School of Medicine, Howard Hughes Medical Institute, New Haven, CT 06520
| | - Craig H Bassing
- Division of Cancer Pathobiology, Department of Pathology and Laboratory Medicine, Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA 19104;
- Immunology Graduate Group, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104
- Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104
- Cancer Biology Program of the Cell and Molecular Biology Graduate Group, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104; and
| |
Collapse
|
27
|
Yu L, Li L, Medeiros LJ, Young KH. NF-κB signaling pathway and its potential as a target for therapy in lymphoid neoplasms. Blood Rev 2017; 31:77-92. [PMID: 27773462 PMCID: PMC5382109 DOI: 10.1016/j.blre.2016.10.001] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 10/07/2016] [Accepted: 10/07/2016] [Indexed: 01/01/2023]
Abstract
The NF-κB pathway, a critical regulator of apoptosis, plays a key role in many normal cellular functions. Genetic alterations and other mechanisms leading to constitutive activation of the NF-κB pathway contribute to cancer development, progression and therapy resistance by activation of downstream anti-apoptotic pathways, unfavorable microenvironment interactions, and gene dysregulation. Not surprisingly, given its importance to normal and cancer cell function, the NF-κB pathway has emerged as a target for therapy. In the review, we present the physiologic role of the NF-κB pathway and recent advances in better understanding of the pathologic roles of the NF-κB pathway in major types of lymphoid neoplasms. We also provide an update of clinical trials that use NF-κB pathway inhibitors. These trials are exploring the clinical efficiency of combining NF-κB pathway inhibitors with various agents that target diverse mechanisms of action with the goal being to optimize novel therapeutic opportunities for targeting oncogenic pathways to eradicate cancer cells.
Collapse
Affiliation(s)
- Li Yu
- Department of Hematopathology, The University of Texas MD, Anderson Cancer Center, 6 Houston, TX, USA
- Department of Hematology, The Second Affiliate Hospital of Nanchang University, Nanchang, China
| | - Ling Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - L. Jeffrey Medeiros
- Department of Hematopathology, The University of Texas MD, Anderson Cancer Center, 6 Houston, TX, USA
| | - Ken H. Young
- Department of Hematopathology, The University of Texas MD, Anderson Cancer Center, 6 Houston, TX, USA
- The University of Texas Graduate School of Biomedical Science, Houston, TX, USA
| |
Collapse
|
28
|
Shinohara H, Nagashima T, Cascalho MI, Kurosaki T. TAK1 maintains the survival of immunoglobulin λ-chain-positive B cells. Genes Cells 2016; 21:1233-1243. [PMID: 27696624 DOI: 10.1111/gtc.12442] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Accepted: 09/08/2016] [Indexed: 12/13/2022]
Abstract
TAK1 (MAP3K7) mediation of the IκB kinase (IKK) complex-nuclear factor-κB (NF-κB) pathway is crucial for the activation of immune response and to perpetuate inflammation. Although progress has been made to understand TAK1 function in the B-cell receptor (BCR) signaling, the physiological roles of TAK1 in B-cell development, particularly in the bone marrow (BM), remain elusive. Previous studies suggested that the IKK complex is required for the development of immunoglobulin light chain λ-positive B cells, but not for receptor editing. In contrast, NF-κB activity is suggested to be involved in the regulation of receptor editing. Thus, NF-κB signaling in early B-cell development is yet to be fully characterized. Therefore, we addressed the role of TAK1 in early B-cell development. TAK1-deficient mice showed significant reduction of BM Igλ-positive B-cell numbers without any alteration in the BCR editing. Furthermore, the expression of survival factor Bcl-2 was reduced in TAK1-deficient BM B cells as assessed by microarray and quantitative PCR analyses. Ex vivo over-expression of exogenous Bcl-2 enhanced the survival of TAK1-deficient Igλ-positive B cells. TAK1-IKK-NF-κB signaling contributes to the survival of λ-chain-positive B cells through NF-κB-dependent anti-apoptotic Bcl-2 expression.
Collapse
Affiliation(s)
- Hisaaki Shinohara
- Laboratory for Integrated Cellular Systems, RIKEN Center for Integrative Medical Sciences (IMS-RCAI), Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
| | - Takeshi Nagashima
- Division of Cell Proliferation, ART, Graduate School of Medicine, Tohoku University, Sendai, Miyagi, 980-8575, Japan
| | - Marilia I Cascalho
- Department of Surgery, University of Michigan, Ann Arbor, MI, 48109, USA.,Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Tomohiro Kurosaki
- Laboratory for Lymphocyte Differentiation, RIKEN Center for Integrative Medical Sciences (IMS-RCAI), Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan.,Laboratory for Lymphocyte Differentiation, Immunology Frontier Research Center, Osaka University, 3-1 Yamada-oka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
29
|
Differential requirements for the canonical NF-κB transcription factors c-REL and RELA during the generation and activation of mature B cells. Immunol Cell Biol 2016; 95:261-271. [PMID: 27649781 PMCID: PMC5360551 DOI: 10.1038/icb.2016.95] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 08/29/2016] [Accepted: 09/13/2016] [Indexed: 12/13/2022]
Abstract
Signaling through the canonical NF-κB pathway is critical for the generation and maintenance of mature B-cells and for antigen-dependent B-cell activation. c-REL (rel) and RELA (rela) are the downstream transcriptional activators of the canonical NF-κB pathway. Studies of B-cells derived from constitutional rel knockout mice and chimeric mice repopulated with rela−/− fetal liver cells provided evidence that the subunits can have distinct roles during B-cell development. However, the B-cell-intrinsic functions of c-REL and RELA during B-cell generation and antigen-dependent B-cell activation have not been determined in vivo. To clarify this issue, we crossed mice with conditional rel and rela alleles individually or in combination to mice that express Cre-recombinase in B-cells. We here report that, whereas single deletion of rel or rela did not impair mature B-cell generation and maintenance, their simultaneous deletion led to a dramatic reduction of follicular and marginal zone B-cells. Upon T-cell-dependent immunization, B-cell-specific deletion of the c-REL subunit alone abrogated the formation of germinal centers (GC), whereas rela deletion did not affect GC formation. T-independent responses were strongly impaired in mice with B-cell-specific deletion of rel, and only modestly in mice with RELA-deficient B-cells. Our findings identify differential requirements for the canonical NF-κB subunits c-REL and RELA at distinct stages of mature B-cell development. The subunits are jointly required for the generation of mature B-cells. During antigen-dependent B-cell activation, c-REL is the critical subunit required for the initiation of the GC-reaction and for optimal T-independent antibody responses, with RELA being largely dispensable at this stage.
Collapse
|
30
|
Abstract
Multicentric Castleman disease (MCD) encompasses a spectrum of conditions that give rise to overlapping clinicopathological manifestations. The fundamental pathogenetic mechanism involves dysregulated cytokine activity that causes systemic inflammatory symptoms as well as lymphadenopathy. The histological changes in lymph nodes resemble in part the findings originally described in the unicentric forms Castleman disease, both hyaline vascular and plasma cell variants. In MCD caused by Kaposi sarcoma-associated herpesvirus/human herpesvirus-8 (KSHV/HHV8), the cytokine over activity is caused by viral products, which can also lead to atypical lymphoproliferations and potential progression to lymphoma. In cases negative for KSHV/HHV8, so-called idiopathic MCD, the hypercytokinemia can result from various mechanisms, which ultimately lead to different constellations of clinical presentations and varied pathology in lymphoid tissues. In this article, we review the evolving concepts and definitions of the various conditions under the eponym of Castleman disease, and summarize current knowledge regarding the histopathology and pathogenesis of lesions within the MCD spectrum.
Collapse
Affiliation(s)
- Hao-Wei Wang
- Hematopathology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Stefania Pittaluga
- Hematopathology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Elaine S Jaffe
- Hematopathology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD.
| |
Collapse
|
31
|
Antigen nature and complexity influence human antibody light chain usage and specificity. Vaccine 2016; 34:2813-20. [PMID: 27113164 DOI: 10.1016/j.vaccine.2016.04.040] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 04/05/2016] [Accepted: 04/14/2016] [Indexed: 01/03/2023]
Abstract
Human antibodies consist of a heavy chain and one of two possible light chains, kappa (κ) or lambda (λ). Here we tested how these two possible light chains influence the overall antibody response to polysaccharide and protein antigens by measuring light chain usage in human monoclonal antibodies from antibody secreting cells obtained following vaccination with Pneumovax23. Remarkably, we found that individuals displayed restricted light chain usage to certain serotypes and that lambda antibodies have different specificities and modes of cross-reactivity than kappa antibodies. Thus, at both the monoclonal (7 kappa, no lambda) and serum levels (145μg/mL kappa, 2.82μg/mL lambda), antibodies to cell wall polysaccharide were nearly always kappa. The pneumococcal reference serum 007sp was analyzed for light chain usage to 12 pneumococcal serotypes for which it is well characterized. Similar to results at the monoclonal level, certain serotypes tended to favor one of the light chains (14 and 19A, lambda; 6A and 23F, kappa). We also explored differences in light chain usage at the serum level to a variety of antigens. We examined serum antibodies to diphtheria toxin mutant CRM197 and Epstein-Barr virus protein EBNA-1. These responses tended to be kappa dominant (average kappa-to-lambda ratios of 4.52 and 9.72 respectively). Responses to the influenza vaccine were more balanced with kappa-to-lambda ratio averages having slight strain variations: seasonal H1N1, 1.1; H3N2, 0.96; B, 0.91. We conclude that antigens with limited epitopes tend to produce antibodies with restricted light chain usage and that in most individuals, antibodies with lambda light chains have specificities different and complementary to kappa-containing antibodies.
Collapse
|
32
|
Canonical NF-κB signaling is uniquely required for the long-term persistence of functional mature B cells. Proc Natl Acad Sci U S A 2016; 113:5065-70. [PMID: 27099294 DOI: 10.1073/pnas.1604529113] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Although canonical NF-κB signaling is crucial to generate a normal mature B-cell compartment, its role in the persistence of resting mature B cells is controversial. To resolve this conflict, we ablated NF-κB essential modulator (NEMO) and IκB kinase 2 (IKK2), two essential mediators of the canonical pathway, either early on in B-cell development or specifically in mature B cells. Early ablation severely inhibited the generation of all mature B-cell subsets, but follicular B-cell numbers could be largely rescued by ectopic expression of B-cell lymphoma 2 (Bcl2), despite a persisting block at the transitional stage. Marginal zone (MZ) B and B1 cells were not rescued, indicating a possible role of canonical NF-κB signals beyond the control of cell survival in these subsets. When canonical NF-κB signaling was ablated specifically in mature B cells, the differentiation and/or persistence of MZ B cells was still abrogated, but follicular B-cell numbers were only mildly affected. However, the mutant cells exhibited increased turnover as well as functional deficiencies upon activation, suggesting that canonical NF-κB signals contribute to their long-term persistence and functional fitness.
Collapse
|
33
|
Kilzheimer M, Quandt J, Langhans J, Weihrich P, Wirth T, Brunner C. NF-κB-dependent signals control BOB.1/OBF.1 and Oct2 transcriptional activity in B cells. Eur J Immunol 2015; 45:3441-53. [DOI: 10.1002/eji.201545475] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 07/30/2015] [Accepted: 09/12/2015] [Indexed: 12/18/2022]
Affiliation(s)
| | - Jasmin Quandt
- Institute of Physiological Chemistry; Ulm University; Ulm Germany
| | - Julia Langhans
- Department of Otorhinolaryngology; Ulm University; Ulm Germany
| | - Petra Weihrich
- Institute of Physiological Chemistry; Ulm University; Ulm Germany
- Department of Otorhinolaryngology; Ulm University; Ulm Germany
| | - Thomas Wirth
- Institute of Physiological Chemistry; Ulm University; Ulm Germany
| | - Cornelia Brunner
- Institute of Physiological Chemistry; Ulm University; Ulm Germany
- Department of Otorhinolaryngology; Ulm University; Ulm Germany
| |
Collapse
|
34
|
Iacoangeli A, Lui A, Naik U, Ohta Y, Flajnik M, Hsu E. Biased Immunoglobulin Light Chain Gene Usage in the Shark. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2015; 195:3992-4000. [PMID: 26342033 PMCID: PMC4592821 DOI: 10.4049/jimmunol.1501426] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 08/04/2015] [Indexed: 12/13/2022]
Abstract
This study of a large family of κ L chain clusters in nurse shark completes the characterization of its classical Ig gene content (two H chain isotypes, μ and ω, and four L chain isotypes, κ, λ, σ, and σ-2). The shark κ clusters are minigenes consisting of a simple VL-JL-CL array, where V to J recombination occurs over an ~500-bp interval, and functional clusters are widely separated by at least 100 kb. Six out of ~39 κ clusters are prerearranged in the germline (germline joined). Unlike the complex gene organization and multistep assembly process of Ig in mammals, each shark Ig rearrangement, somatic or in the germline, appears to be an independent event localized to the minigene. This study examined the expression of functional, nonproductive, and sterile transcripts of the κ clusters compared with the other three L chain isotypes. κ cluster usage was investigated in young sharks, and a skewed pattern of split gene expression was observed, one similar in functional and nonproductive rearrangements. These results show that the individual activation of the spatially distant κ clusters is nonrandom. Although both split and germline-joined κ genes are expressed, the latter are prominent in young animals and wane with age. We speculate that, in the shark, the differential activation of the multiple isotypes can be advantageously used in receptor editing.
Collapse
Affiliation(s)
- Anna Iacoangeli
- Department of Physiology and Pharmacology, State University of New York Health Science Center at Brooklyn, Brooklyn, NY 11203; and
| | - Anita Lui
- Department of Physiology and Pharmacology, State University of New York Health Science Center at Brooklyn, Brooklyn, NY 11203; and
| | - Ushma Naik
- Department of Physiology and Pharmacology, State University of New York Health Science Center at Brooklyn, Brooklyn, NY 11203; and
| | - Yuko Ohta
- Department of Microbiology and Immunology, University of Maryland, Baltimore, MD 21201
| | - Martin Flajnik
- Department of Microbiology and Immunology, University of Maryland, Baltimore, MD 21201
| | - Ellen Hsu
- Department of Physiology and Pharmacology, State University of New York Health Science Center at Brooklyn, Brooklyn, NY 11203; and
| |
Collapse
|
35
|
Fiala GJ, Janowska I, Prutek F, Hobeika E, Satapathy A, Sprenger A, Plum T, Seidl M, Dengjel J, Reth M, Cesca F, Brummer T, Minguet S, Schamel WWA. Kidins220/ARMS binds to the B cell antigen receptor and regulates B cell development and activation. ACTA ACUST UNITED AC 2015; 212:1693-708. [PMID: 26324445 PMCID: PMC4577850 DOI: 10.1084/jem.20141271] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 08/14/2015] [Indexed: 01/04/2023]
Abstract
Fiala et al. report that Kidins220/ARMS is a novel interactor of the B cell antigen receptor (BCR) and its deletion impairs B cell development and B cell functioning. B cell antigen receptor (BCR) signaling is critical for B cell development and activation. Using mass spectrometry, we identified a protein kinase D–interacting substrate of 220 kD (Kidins220)/ankyrin repeat–rich membrane-spanning protein (ARMS) as a novel interaction partner of resting and stimulated BCR. Upon BCR stimulation, the interaction increases in a Src kinase–independent manner. By knocking down Kidins220 in a B cell line and generating a conditional B cell–specific Kidins220 knockout (B-KO) mouse strain, we show that Kidins220 couples the BCR to PLCγ2, Ca2+, and extracellular signal-regulated kinase (Erk) signaling. Consequently, BCR-mediated B cell activation was reduced in vitro and in vivo upon Kidins220 deletion. Furthermore, B cell development was impaired at stages where pre-BCR or BCR signaling is required. Most strikingly, λ light chain–positive B cells were reduced sixfold in the B-KO mice, genetically placing Kidins220 in the PLCγ2 pathway. Thus, our data indicate that Kidins220 positively regulates pre-BCR and BCR functioning.
Collapse
Affiliation(s)
- Gina J Fiala
- Department of Molecular Immunology, BioIII, Faculty of Biology, University of Freiburg and Max Planck Institute of Immunobiology and Epigenetics, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany
| | - Iga Janowska
- Department of Molecular Immunology, BioIII, Faculty of Biology, University of Freiburg and Max Planck Institute of Immunobiology and Epigenetics, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany
| | - Fabiola Prutek
- Department of Molecular Immunology, BioIII, Faculty of Biology, University of Freiburg and Max Planck Institute of Immunobiology and Epigenetics, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany
| | - Elias Hobeika
- Department of Molecular Immunology, BioIII, Faculty of Biology, University of Freiburg and Max Planck Institute of Immunobiology and Epigenetics, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany Institute of Immunology, University Hospital Ulm, 89081 Ulm, Germany
| | - Annyesha Satapathy
- Center of Synaptic Neuroscience, Italian Institute of Technology, 16163 Genova, Italy
| | - Adrian Sprenger
- Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany
| | - Thomas Plum
- Department of Molecular Immunology, BioIII, Faculty of Biology, University of Freiburg and Max Planck Institute of Immunobiology and Epigenetics, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany
| | - Maximilian Seidl
- Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany
| | - Jörn Dengjel
- Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany
| | - Michael Reth
- Department of Molecular Immunology, BioIII, Faculty of Biology, University of Freiburg and Max Planck Institute of Immunobiology and Epigenetics, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany
| | - Fabrizia Cesca
- Center of Synaptic Neuroscience, Italian Institute of Technology, 16163 Genova, Italy
| | - Tilman Brummer
- Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany
| | - Susana Minguet
- Department of Molecular Immunology, BioIII, Faculty of Biology, University of Freiburg and Max Planck Institute of Immunobiology and Epigenetics, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany
| | - Wolfgang W A Schamel
- Department of Molecular Immunology, BioIII, Faculty of Biology, University of Freiburg and Max Planck Institute of Immunobiology and Epigenetics, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany
| |
Collapse
|
36
|
NF-κB and AKT signaling prevent DNA damage in transformed pre-B cells by suppressing RAG1/2 expression and activity. Blood 2015; 126:1324-35. [PMID: 26153519 DOI: 10.1182/blood-2015-01-621623] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 07/01/2015] [Indexed: 01/16/2023] Open
Abstract
In developing lymphocytes, expression and activity of the recombination activation gene protein 1 (RAG1) and RAG2 endonuclease complex is tightly regulated to ensure ordered recombination of the immunoglobulin genes and to avoid genomic instability. Aberrant RAG activity has been implicated in the generation of secondary genetic events in human B-cell acute lymphoblastic leukemias (B-ALLs), illustrating the oncogenic potential of the RAG complex. Several layers of regulation prevent collateral genomic DNA damage by restricting RAG activity to the G1 phase of the cell cycle. In this study, we show a novel pathway that suppresses RAG expression in cycling-transformed mouse pre-B cells and human pre-B B-ALL cells that involves the negative regulation of FOXO1 by nuclear factor κB (NF-κB). Inhibition of NF-κB in cycling pre-B cells resulted in upregulation of RAG expression and recombination activity, which provoked RAG-dependent DNA damage. In agreement, we observe a negative correlation between NF-κB activity and the expression of RAG1, RAG2, and TdT in B-ALL patients. Our data suggest that targeting NF-κB in B-ALL increases the risk of RAG-dependent genomic instability.
Collapse
|
37
|
Palmer VL, Aziz-Seible R, Kassmeier MD, Rothermund M, Perry GA, Swanson PC. VprBP Is Required for Efficient Editing and Selection of Igκ+ B Cells, but Is Dispensable for Igλ+ and Marginal Zone B Cell Maturation and Selection. THE JOURNAL OF IMMUNOLOGY 2015; 195:1524-37. [PMID: 26150531 DOI: 10.4049/jimmunol.1500952] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 06/02/2015] [Indexed: 01/01/2023]
Abstract
B cell development past the pro-B cell stage in mice requires the Cul4-Roc1-DDB1 E3 ubiquitin ligase substrate recognition subunit VprBP. Enforced Bcl2 expression overcomes defects in distal VH-DJH and secondary Vκ-Jκ rearrangement associated with VprBP insufficiency in B cells and substantially rescues maturation of marginal zone and Igλ(+) B cells, but not Igκ(+) B cells. In this background, expression of a site-directed Igκ L chain transgene increases Igκ(+) B cell frequency, suggesting VprBP does not regulate L chain expression from a productively rearranged Igk allele. In site-directed anti-dsDNA H chain transgenic mice, loss of VprBP function in B cells impairs selection of Igκ editor L chains typically arising through secondary Igk rearrangement, but not selection of Igλ editor L chains. Both H and L chain site-directed transgenic mice show increased B cell anergy when VprBP is inactivated in B cells. Taken together, these data argue that VprBP is required for the efficient receptor editing and selection of Igκ(+) B cells, but is largely dispensable for Igλ(+) B cell development and selection, and that VprBP is necessary to rescue autoreactive B cells from anergy induction.
Collapse
Affiliation(s)
- Victoria L Palmer
- Department of Medical Microbiology and Immunology, Creighton University, Omaha, NE 68178
| | - Razia Aziz-Seible
- Department of Medical Microbiology and Immunology, Creighton University, Omaha, NE 68178
| | - Michele D Kassmeier
- Department of Medical Microbiology and Immunology, Creighton University, Omaha, NE 68178
| | - Mary Rothermund
- Department of Medical Microbiology and Immunology, Creighton University, Omaha, NE 68178
| | - Greg A Perry
- Department of Medical Microbiology and Immunology, Creighton University, Omaha, NE 68178
| | - Patrick C Swanson
- Department of Medical Microbiology and Immunology, Creighton University, Omaha, NE 68178
| |
Collapse
|
38
|
Pedersen GK, Ádori M, Karlsson Hedestam GB. NF-κB signaling in B-1 cell development. Ann N Y Acad Sci 2015; 1362:39-47. [PMID: 26096766 DOI: 10.1111/nyas.12800] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
NF-κB transcription factors play essential roles in hematopoiesis. In this review, we summarize the requirements of different components of the NF-κB pathway for B-1 cell development and maintenance. The B-1 cell developmental steps are also reviewed, with particular emphasis on stages where NF-κB signaling may be critical.
Collapse
Affiliation(s)
- Gabriel K Pedersen
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Monika Ádori
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
39
|
Kluin PM, Langerak AW, Beverdam-Vincent J, Geurts-Giele WRR, Visser L, Rutgers B, Schuuring E, Van Baarlen J, Lam KH, Seldenrijk K, Kibbelaar RE, de Wit P, Diepstra A, Rosati S, van Noesel MM, Zwaan CM, Hunting JCB, Hoogendoorn M, van der Gaag EJ, van Esser JWJ, de Bont E, Kluin-Nelemans HC, Winter RH, Lo ten Foe JR, van der Zanden AGM. Paediatric nodal marginal zone B-cell lymphadenopathy of the neck: a Haemophilus influenzae
-driven immune disorder? J Pathol 2015; 236:302-14. [DOI: 10.1002/path.4524] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 02/10/2015] [Accepted: 02/23/2015] [Indexed: 01/06/2023]
Affiliation(s)
- Philip M Kluin
- Department of Pathology and Medical Biology; University Medical Centre Groningen, University of Groningen; Groningen The Netherlands
| | - Anton W Langerak
- Department of Immunology, Erasmus MC; University Medical Centre Rotterdam; Rotterdam The Netherlands
| | - Jannetta Beverdam-Vincent
- Microbiology and Infection Control, Zorggroep Twente; Hengelo The Netherlands
- Laboratory for Microbiology; Twente Achterhoek Hengelo The Netherlands
| | | | - Lydia Visser
- Department of Pathology and Medical Biology; University Medical Centre Groningen, University of Groningen; Groningen The Netherlands
| | - Bea Rutgers
- Department of Pathology and Medical Biology; University Medical Centre Groningen, University of Groningen; Groningen The Netherlands
| | - Ed Schuuring
- Department of Pathology and Medical Biology; University Medical Centre Groningen, University of Groningen; Groningen The Netherlands
| | | | - King H Lam
- Department of Pathology, Erasmus Medical Centre Rotterdam; EMCR; Rotterdam The Netherlands
| | - Kees Seldenrijk
- Department of Pathology; St Antonius Hospital; Nieuwegein The Netherlands
| | - Robby E Kibbelaar
- Department of Pathology; Pathologie Friesland; Leeuwarden The Netherlands
| | - Peter de Wit
- Department of Pathology; Amphia Hospital; Breda The Netherlands
| | - Arjan Diepstra
- Department of Pathology and Medical Biology; University Medical Centre Groningen, University of Groningen; Groningen The Netherlands
| | - Stefano Rosati
- Department of Pathology and Medical Biology; University Medical Centre Groningen, University of Groningen; Groningen The Netherlands
| | - Max M van Noesel
- Department of Oncology and Hematology; Sophia Children Hospital; Rotterdam The Netherlands
| | - C Michel Zwaan
- Department of Oncology and Hematology; Sophia Children Hospital; Rotterdam The Netherlands
| | - Jarmo CB Hunting
- Department of Internal Medicine; St Antonius Ziekenhuis; Nieuwegein The Netherlands
| | - Mels Hoogendoorn
- Department of Internal Medicine; Medisch Centrum Leeuwarden; The Netherlands
| | | | | | - Eveline de Bont
- Department of Pediatric Oncology & Hematology; University Medical Centre Groningen, University of Groningen; Groningen The Netherlands
| | - Hanneke C Kluin-Nelemans
- Department of Hematology; University Medical Centre Groningen, University of Groningen; Groningen The Netherlands
| | - Rik H Winter
- Department of Medical Microbiology; University Medical Centre Groningen, University of Groningen; Groningen The Netherlands
| | - Jerome R Lo ten Foe
- Department of Medical Microbiology; University Medical Centre Groningen, University of Groningen; Groningen The Netherlands
| | - Adri GM van der Zanden
- Microbiology and Infection Control, Zorggroep Twente; Hengelo The Netherlands
- Laboratory for Microbiology; Twente Achterhoek Hengelo The Netherlands
| |
Collapse
|
40
|
Ren W, Grimsholm O, Bernardi AI, Höök N, Stern A, Cavallini N, Mårtensson IL. Surrogate light chain is required for central and peripheral B-cell tolerance and inhibits anti-DNA antibody production by marginal zone B cells. Eur J Immunol 2015; 45:1228-37. [PMID: 25546233 DOI: 10.1002/eji.201444917] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 12/08/2014] [Accepted: 12/23/2014] [Indexed: 12/15/2022]
Abstract
Selection of the primary antibody repertoire takes place in pro-/pre-B cells, and subsequently in immature and transitional B cells. At the first checkpoint, μ heavy (μH) chains assemble with surrogate light (SL) chain into a precursor B-cell receptor. In mice lacking SL chain, μH chain selection is impaired, and serum autoantibody levels are elevated. However, whether the development of autoantibody-producing cells is due to an inability of the resultant B-cell receptors to induce central and/or peripheral B-cell tolerance or other factors is unknown. Here, we show that receptor editing is defective, and that a higher proportion of BM immature B cells are prone to undergoing apoptosis. Furthermore, transitional B cells are also more prone to undergoing apoptosis, with a stronger selection pressure to enter the follicular B-cell pool. Those that enter the marginal zone (MZ) B-cell pool escape selection and survive, possibly due to the B-lymphopenia and elevated levels of B-cell activating factor. Moreover, the MZ B cells are responsible for the elevated IgM anti-dsDNA antibody levels detected in these mice. Thus, the SL chain is required for central and peripheral B-cell tolerance and inhibits anti-DNA antibody production by MZ B cells.
Collapse
Affiliation(s)
- Weicheng Ren
- Department of Rheumatology and Inflammation Research, University of Gothenburg, Gothenburg, Sweden
| | | | | | | | | | | | | |
Collapse
|
41
|
Sasaki Y, Iwai K. Roles of the NF-κB Pathway in B-Lymphocyte Biology. Curr Top Microbiol Immunol 2015; 393:177-209. [PMID: 26275874 DOI: 10.1007/82_2015_479] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
NF-κB was originally identified as a family of transcription factors that bind the enhancer of the immunoglobulin κ light-chain gene. Although its function in the regulation of immunoglobulin κ light-chain gene remains unclear, NF-κB plays critical roles in development, survival, and activation of B lymphocytes. In B cells, many receptors, including B-cell antigen receptor (BCR), activate NF-κB pathway, and the molecular mechanism of receptor-mediated activation of IκB kinase (IKK) complex has been partially revealed. In addition to normal B lymphocytes, NF-κB is also involved in the growth of some types of B-cell lymphomas, and many oncogenic mutations involved in constitutive activation of the NF-κB pathway were recently identified in such cancers. In this review, we first summarize the function of NF-κB in B-cell development and activation, and then describe recent progress in understanding the molecular mechanism of receptor-mediated activation of the IKK complex, focusing on the roles of the ubiquitin system. In the last section, we describe oncogenic mutations that induce NF-κB activation in B-cell lymphoma.
Collapse
Affiliation(s)
- Yoshiteru Sasaki
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Kyoto University, Kyoto, 606-8501, Japan.
| | - Kazuhiro Iwai
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Kyoto University, Kyoto, 606-8501, Japan
| |
Collapse
|
42
|
de Almeida CR, Hendriks RW, Stadhouders R. Dynamic Control of Long-Range Genomic Interactions at the Immunoglobulin κ Light-Chain Locus. Adv Immunol 2015; 128:183-271. [DOI: 10.1016/bs.ai.2015.07.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
43
|
Ramachandiran S, Adon A, Guo X, Wang Y, Wang H, Chen Z, Kowalski J, Sunay UR, Young AN, Brown T, Mar JC, Du Y, Fu H, Mann KP, Natkunam Y, Boise LH, Saavedra HI, Lossos IS, Bernal-Mizrachi L. Chromosome instability in diffuse large B cell lymphomas is suppressed by activation of the noncanonical NF-κB pathway. Int J Cancer 2014; 136:2341-51. [PMID: 25359525 DOI: 10.1002/ijc.29301] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 10/16/2014] [Indexed: 12/12/2022]
Abstract
Diffuse large B cell lymphoma (DLBCL) is the most common form of lymphoma in the United States. DLBCL comprises biologically distinct subtypes including germinal center-like (GCB) and activated-B-cell-like DLBCL (ABC). The most aggressive type, ABC-DLBCL, displays dysregulation of both canonical and noncanonical NF-κB pathway as well as genomic instability. Although, much is known about the tumorigenic roles of the canonical NF-kB pathway, the precise role of the noncanonical NF-kB pathway remains unknown. Here we show that activation of the noncanonical NF-κB pathway regulates chromosome stability, DNA damage response and centrosome duplication in DLBCL. Analysis of 92 DLBCL samples revealed that activation of the noncanonical NF-κB pathway is associated with low levels of DNA damage and centrosome amplification. Inhibiting the noncanonical pathway in lymphoma cells uncovered baseline DNA damage and prevented doxorubicin-induced DNA damage repair. In addition, it triggered centrosome amplification and chromosome instability, indicated by anaphase bridges, multipolar spindles and chromosome missegregation. We determined that the noncanonical NF-κB pathway execute these functions through the regulation of GADD45α and REDD1 in a p53-independent manner, while it collaborates with p53 to regulate cyclin G2 expression. Furthermore, this pathway regulates GADD45α, REDD1 and cyclin G2 through direct binding of NF-κB sites to their promoter region. Overall, these results indicate that the noncanonical NF-κB pathway plays a central role in maintaining genome integrity in DLBCL. Our data suggests that inhibition of the noncanonical NF-kB pathway should be considered as an important component in DLBCL therapeutic approach.
Collapse
Affiliation(s)
- Sampath Ramachandiran
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Jacque E, Schweighoffer E, Visekruna A, Papoutsopoulou S, Janzen J, Zillwood R, Tarlinton DM, Tybulewicz VLJ, Ley SC. IKK-induced NF-κB1 p105 proteolysis is critical for B cell antibody responses to T cell-dependent antigen. ACTA ACUST UNITED AC 2014; 211:2085-101. [PMID: 25225457 PMCID: PMC4172221 DOI: 10.1084/jem.20132019] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Jacque et al. investigate the functions of NF-κB1 p105 and its associated NF-κB–binding partners in B cells, using a mutant mouse strain that carries a form of the NF-κB1 precursor that is resistant to IKK-induced proteolysis. They identify a critical B cell–intrinsic role for this IKK signaling pathway in the antigen-induced survival and differentiation of follicular mature B cells. The importance of IκB kinase (IKK)–induced proteolysis of NF-κB1 p105 in B cells was investigated using Nfkb1SSAA/SSAA mice, in which this NF-κB signaling pathway is blocked. Nfkb1SSAA mutation had no effect on the development and homeostasis of follicular mature (FM) B cells. However, analysis of mixed bone marrow chimeras revealed that Nfkb1SSAA/SSAA FM B cells were completely unable to mediate T cell–dependent antibody responses. Nfkb1SSAA mutation decreased B cell antigen receptor (BCR) activation of NF-κB in FM B cells, which selectively blocked BCR stimulation of cell survival and antigen-induced differentiation into plasmablasts and germinal center B cells due to reduced expression of Bcl-2 family proteins and IRF4, respectively. In contrast, the antigen-presenting function of FM B cells and their BCR-induced migration to the follicle T cell zone border, as well as their growth and proliferation after BCR stimulation, were not affected. All of the inhibitory effects of Nfkb1SSAA mutation on B cell functions were rescued by normalizing NF-κB activation genetically. Our study identifies critical B cell-intrinsic functions for IKK-induced NF-κB1 p105 proteolysis in the antigen-induced survival and differentiation of FM B cells, which are essential for T-dependent antibody responses.
Collapse
Affiliation(s)
- Emilie Jacque
- Division of Immune Cell Biology, MRC National Institute for Medical Research, London NW7 1AA, England, UK
| | - Edina Schweighoffer
- Division of Immune Cell Biology, MRC National Institute for Medical Research, London NW7 1AA, England, UK
| | - Alexander Visekruna
- Division of Immune Cell Biology, MRC National Institute for Medical Research, London NW7 1AA, England, UK
| | - Stamatia Papoutsopoulou
- Division of Immune Cell Biology, MRC National Institute for Medical Research, London NW7 1AA, England, UK
| | - Julia Janzen
- Division of Immune Cell Biology, MRC National Institute for Medical Research, London NW7 1AA, England, UK
| | - Rachel Zillwood
- Division of Immune Cell Biology, MRC National Institute for Medical Research, London NW7 1AA, England, UK
| | - David M Tarlinton
- The Walter and Eliza Hall Institute for Medical Research, Parkville, Victoria 3052, Australia
| | - Victor L J Tybulewicz
- Division of Immune Cell Biology, MRC National Institute for Medical Research, London NW7 1AA, England, UK
| | - Steven C Ley
- Division of Immune Cell Biology, MRC National Institute for Medical Research, London NW7 1AA, England, UK
| |
Collapse
|
45
|
Riddell J, Gazit R, Garrison BS, Guo G, Saadatpour A, Mandal PK, Ebina W, Volchkov P, Yuan GC, Orkin SH, Rossi DJ. Reprogramming committed murine blood cells to induced hematopoietic stem cells with defined factors. Cell 2014; 157:549-64. [PMID: 24766805 DOI: 10.1016/j.cell.2014.04.006] [Citation(s) in RCA: 259] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 03/13/2014] [Accepted: 04/03/2014] [Indexed: 01/17/2023]
Abstract
Hematopoietic stem cells (HSCs) sustain blood formation throughout life and are the functional units of bone marrow transplantation. We show that transient expression of six transcription factors Run1t1, Hlf, Lmo2, Prdm5, Pbx1, and Zfp37 imparts multilineage transplantation potential onto otherwise committed lymphoid and myeloid progenitors and myeloid effector cells. Inclusion of Mycn and Meis1 and use of polycistronic viruses increase reprogramming efficacy. The reprogrammed cells, designated induced-HSCs (iHSCs), possess clonal multilineage differentiation potential, reconstitute stem/progenitor compartments, and are serially transplantable. Single-cell analysis revealed that iHSCs derived under optimal conditions exhibit a gene expression profile that is highly similar to endogenous HSCs. These findings demonstrate that expression of a set of defined factors is sufficient to activate the gene networks governing HSC functional identity in committed blood cells. Our results raise the prospect that blood cell reprogramming may be a strategy for derivation of transplantable stem cells for clinical application.
Collapse
Affiliation(s)
- Jonah Riddell
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Program in Cellular and Molecular Medicine, Division of Hematology/Oncology, Boston Children's Hospital, MA 02116, USA
| | - Roi Gazit
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Program in Cellular and Molecular Medicine, Division of Hematology/Oncology, Boston Children's Hospital, MA 02116, USA
| | - Brian S Garrison
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Program in Cellular and Molecular Medicine, Division of Hematology/Oncology, Boston Children's Hospital, MA 02116, USA
| | - Guoji Guo
- Dana Farber/Boston Children's Hospital Cancer and Blood Disorders Center, Boston, MA 02116, USA
| | - Assieh Saadatpour
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biostatistics, Harvard School of Public Health, Boston, MA 02115, USA
| | - Pankaj K Mandal
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Program in Cellular and Molecular Medicine, Division of Hematology/Oncology, Boston Children's Hospital, MA 02116, USA
| | - Wataru Ebina
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Program in Cellular and Molecular Medicine, Division of Hematology/Oncology, Boston Children's Hospital, MA 02116, USA
| | - Pavel Volchkov
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Program in Cellular and Molecular Medicine, Division of Hematology/Oncology, Boston Children's Hospital, MA 02116, USA
| | - Guo-Cheng Yuan
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biostatistics, Harvard School of Public Health, Boston, MA 02115, USA
| | - Stuart H Orkin
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Dana Farber/Boston Children's Hospital Cancer and Blood Disorders Center, Boston, MA 02116, USA; Howard Hughes Medical Institute; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Derrick J Rossi
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Program in Cellular and Molecular Medicine, Division of Hematology/Oncology, Boston Children's Hospital, MA 02116, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA.
| |
Collapse
|
46
|
Gasparini C, Celeghini C, Monasta L, Zauli G. NF-κB pathways in hematological malignancies. Cell Mol Life Sci 2014; 71:2083-102. [PMID: 24419302 PMCID: PMC11113378 DOI: 10.1007/s00018-013-1545-4] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 12/13/2013] [Accepted: 12/17/2013] [Indexed: 12/22/2022]
Abstract
The nuclear factor κB or NF-κB transcription factor family plays a key role in several cellular functions, i.e. inflammation, apoptosis, cell survival, proliferation, angiogenesis, and innate and acquired immunity. The constitutive activation of NF-κB is typical of most malignancies and plays a major role in tumorigenesis. In this review, we describe NF-κB and its two pathways: the canonical pathway (RelA/p50) and the non-canonical pathway (RelB/p50 or RelB/p52). We then consider the role of the NF-κB subunits in the development and functional activity of B cells, T cells, macrophages and dendritic cells, which are the targets of hematological malignancies. The relevance of the two pathways is described in normal B and T cells and in hematological malignancies, acute and chronic leukemias (ALL, AML, CLL, CML), B lymphomas (DLBCLs, Hodgkin's lymphoma), T lymphomas (ATLL, ALCL) and multiple myeloma. We describe the interaction of NF-κB with the apoptotic pathways induced by TRAIL and the transcription factor p53. Finally, we discuss therapeutic anti-tumoral approaches as mono-therapies or combination therapies aimed to block NF-κB activity and to induce apoptosis (PARAs and Nutlin-3).
Collapse
Affiliation(s)
- Chiara Gasparini
- Institute for Maternal and Child Health-IRCCS "Burlo Garofolo", Via dell'Istria 65/1, 34137, Trieste, Italy,
| | | | | | | |
Collapse
|
47
|
Anbazhagan K, Rabbind Singh A, Isabelle P, Stella I, Céline ADM, Bissac E, Bertrand B, Rémy N, Naomi T, Vincent F, Rochette J, Lassoued K. Human pre-B cell receptor signal transduction: evidence for distinct roles of PI3kinase and MAP-kinase signalling pathways. IMMUNITY INFLAMMATION AND DISEASE 2013; 1:26-36. [PMID: 25400915 PMCID: PMC4217539 DOI: 10.1002/iid3.4] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 07/08/2013] [Accepted: 07/11/2013] [Indexed: 12/16/2022]
Abstract
Pre-BCR acts as a critical checkpoint in B cell development. However, its signalling cascade still remains indistinctly characterised in human. We investigated pre-BCR signalling pathway to examine its regulation in normal primary pre-B lymphocytes and pre-B cell lines. In cell lines, early signalling events occurring after pre-BCR stimulation include phosphorylation of Lyn, Blk and Syk together with ZAP70, Btk, Vav, PLC-γ2 and various adaptor proteins, such as BLNK, LAB, LAT and SLP-76. Further downstream, these molecules induced activation of the PI3K/AKT and MAP-kinase resulting in an augmentation of canonical NF-κB pathways and cFos/AP1 activation. PI3K and MAPK exerted opposing effects on the pre-BCR-induced activation of the canonical NF-κB and c-Fos/AP1 pathways. Immediate nuclear export of FoxO3A and delayed import of IRF4 were additional events observed after pre-BCR crosslinking in primary cells. Pre-BCR-induced down-regulation of Rag1, Rag2, E2A and Pax5 transcripts occurred in a PI3K-dependent manner. Finally we bring evidence that pre-BCR stimulation or co stimulation with CD19 enhances cell cycle signal.
Collapse
Affiliation(s)
- Kolandaswamy Anbazhagan
- Inserm/UMR925, Université Picardie Jules Verne, Laboratoire d'Immunologie, UFR de Médecine 3, rue des Louvels, 80036, Amiens, France
| | - Amrathlal Rabbind Singh
- Inserm/UMR925, Université Picardie Jules Verne, Laboratoire d'Immunologie, UFR de Médecine 3, rue des Louvels, 80036, Amiens, France
| | - Piec Isabelle
- Inserm/UMR925, Université Picardie Jules Verne, Laboratoire d'Immunologie, UFR de Médecine 3, rue des Louvels, 80036, Amiens, France
| | - Ibata Stella
- Inserm/UMR925, Université Picardie Jules Verne, Laboratoire d'Immunologie, UFR de Médecine 3, rue des Louvels, 80036, Amiens, France
| | - Alleaume-De Martel Céline
- Inserm/UMR925, Université Picardie Jules Verne, Laboratoire d'Immunologie, UFR de Médecine 3, rue des Louvels, 80036, Amiens, France
| | - Eliane Bissac
- Inserm/UMR925, Université Picardie Jules Verne, Laboratoire d'Immunologie, UFR de Médecine 3, rue des Louvels, 80036, Amiens, France
| | - Brassart Bertrand
- Inserm/UMR925, Université Picardie Jules Verne, Laboratoire d'Immunologie, UFR de Médecine 3, rue des Louvels, 80036, Amiens, France
| | - Nyga Rémy
- Inserm/UMR925, Université Picardie Jules Verne, Laboratoire d'Immunologie, UFR de Médecine 3, rue des Louvels, 80036, Amiens, France
| | - Taylor Naomi
- CNRS/UMR 5535, Institut de Génétique Moléculaire de Montpellier 1919 Route de Mende, 34293, Montpellier, Cedex 5, France
| | - Fuentes Vincent
- Inserm/UMR925, Université Picardie Jules Verne, Laboratoire d'Immunologie, UFR de Médecine 3, rue des Louvels, 80036, Amiens, France
| | - Jacques Rochette
- Inserm/UMR925, Université Picardie Jules Verne, Laboratoire d'Immunologie, UFR de Médecine 3, rue des Louvels, 80036, Amiens, France
| | - Kaïss Lassoued
- Inserm/UMR925, Université Picardie Jules Verne, Laboratoire d'Immunologie, UFR de Médecine 3, rue des Louvels, 80036, Amiens, France
| |
Collapse
|
48
|
B lymphocytes: development, tolerance, and their role in autoimmunity-focus on systemic lupus erythematosus. Autoimmune Dis 2013; 2013:827254. [PMID: 24187614 PMCID: PMC3804284 DOI: 10.1155/2013/827254] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Accepted: 08/06/2013] [Indexed: 01/10/2023] Open
Abstract
B lymphocytes are the effectors of humoral immunity, providing defense against pathogens through different functions including antibody production. B cells constitute approximately 15% of peripheral blood leukocytes and arise from hemopoietic stem cells in the bone marrow. It is here that their antigen receptors (surface immunoglobulin) are assembled. In the context of autoimmune diseases defined by B and/or T cell autoreactive that upon activation lead to chronic tissue inflammation and often irreversible structural and functional damage, B lymphocytes play an essential role by not only producing autoantibodies but also functioning as antigen-presenting cells (APC) and as a source of cytokines. In this paper, we describe B lymphocyte functions in autoimmunity and autoimmune diseases with a special focus on their abnormalities in systemic lupus erythematosus.
Collapse
|
49
|
Innate immune-directed NF-κB signaling requires site-specific NEMO ubiquitination. Cell Rep 2013; 4:352-61. [PMID: 23871670 DOI: 10.1016/j.celrep.2013.06.036] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Revised: 06/05/2013] [Accepted: 06/25/2013] [Indexed: 11/23/2022] Open
Abstract
While the I kappa kinase (IKK) scaffolding protein NF-κB essential modulator (NEMO) binds to polyubiquitin chains to transmit inflammatory signals, NEMO itself is also ubiquitinated in response to a variety of inflammatory agonists. Although there have been hints that polyubiquitination of NEMO is essential for avoiding inflammatory disorders, the in vivo physiologic role of NEMO ubiquitination is unknown. In this work, we knock in a NEMO allele in which two major inflammatory agonist-induced ubiquitination sites cannot be ubiquitinated. We show that mice with a nonubiquitinatable NEMO allele display embryonic lethality. Heterozygous females develop inflammatory skin lesions, decreased B cell numbers, and hypercellular spleens. Embryonic lethality can be complemented by mating onto a TNFR1(-/-) background, at the cost of severe steatohepatitis and early mortality, and we also show that NEMO ubiquitination is required for optimal innate immune signaling responses. These findings suggest that NEMO ubiquitination is crucial for NF-κB activity in response to innate immune agonists.
Collapse
|
50
|
Bastian L, Hof J, Pfau M, Fichtner I, Eckert C, Henze G, Prada J, von Stackelberg A, Seeger K, Shalapour S. Synergistic activity of bortezomib and HDACi in preclinical models of B-cell precursor acute lymphoblastic leukemia via modulation of p53, PI3K/AKT, and NF-κB. Clin Cancer Res 2013; 19:1445-57. [PMID: 23357978 DOI: 10.1158/1078-0432.ccr-12-1511] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Relapse of disease and subsequent resistance to established therapies remains a major challenge in the treatment of childhood B-cell precursor acute lymphoblastic leukemia (BCP-ALL). New therapeutic options, such as proteasome and histone deacetylase inhibitors (HDACi) with a toxicity profile differing from that of conventional cytotoxic agents, are needed for these extensively pretreated patients. EXPERIMENTAL DESIGN Antiproliferative and proapoptotic effects of combined HDACi/proteasome inhibitor treatments were analyzed using BCP-ALL monocultures, cocultures with primary mesenchymal stroma cells from patients with ALL, and xenograft mouse models. The underlying molecular mechanisms associated with combined treatment were determined by gene expression profiling and protein validation. RESULTS We identified the proteasome inhibitor bortezomib as a promising combination partner for HDACi due to the substantial synergistic antileukemic activity in BCP-ALL cells after concomitant application. This effect was maintained or even increased in the presence of chemotherapeutic agents. The synergistic effect of combined HDACi/BTZ treatment was associated with the regulation of genes involved in cell cycle, JUN/MAPK, PI3K/AKT, p53, ubiquitin/proteasome, and NF-κB pathways. We observed an activation of NF-κB after bortezomib treatment and the induction of apoptosis-related NF-κB target genes such as TNFαRs after concomitant treatment, indicating a possible involvement of NF-κB as proapoptotic mediator. In this context, significantly lower NF-κB subunits gene expression was detected in leukemia cells from patients who developed a relapse during frontline chemotherapy, compared with those who relapsed after cessation of frontline therapy. CONCLUSION These results provide a rationale for the integration of HDACi/BTZ combinations into current childhood BCP-ALL treatment protocols.
Collapse
Affiliation(s)
- Lorenz Bastian
- Department of Pediatric Oncology/Hematology, Charité-Universitätsmedizin Berlin, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|