1
|
Zeng L, Yang K, Wu Y, Yu G, Yan Y, Hao M, Song T, Li Y, Chen J, Sun L. Telitacicept: A novel horizon in targeting autoimmunity and rheumatic diseases. J Autoimmun 2024; 148:103291. [PMID: 39146891 DOI: 10.1016/j.jaut.2024.103291] [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: 11/20/2023] [Revised: 06/19/2024] [Accepted: 07/15/2024] [Indexed: 08/17/2024]
Abstract
BLyS and APRIL have the capability to bind to B cells within the body, allowing these cells to evade elimination when they should naturally be removed. While BLyS primarily plays a role in B cell development and maturation, APRIL is linked to B cell activation and the secretion of antibodies. Thus, in theory, inhibiting BLyS or APRIL could diminish the population of aberrant B cells that contribute to SLE and reduce disease activity in patients. Telitacicept functions by binding to and neutralizing the activities of both BLyS and APRIL, thus hindering the maturation and survival of plasma cells and fully developed B cells. The design of telitacicept is distinctive; it is not a monoclonal antibody but a TACI-Fc fusion protein generated through recombinant DNA technology. This fusion involves merging gene segments of the TACI protein, which can target BLyS/APRIL simultaneously, with the Fc gene segment of the human IgG protein. The TACI-Fc fusion protein exhibits the combined characteristics of both proteins. Currently utilized for autoimmune disease treatment, telitacicept is undergoing clinical investigations globally to assess its efficacy in managing various autoimmune conditions. This review consolidates information on the mechanistic actions, dosing regimens, pharmacokinetics, efficacy, and safety profile of telitacicept-a dual-targeted biological agent. It integrates findings from prior experiments and pharmacokinetic analyses in the treatment of RA and SLE, striving to offer a comprehensive overview of telitacicept's research advancements.
Collapse
Affiliation(s)
- Liuting Zeng
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Graduate School of Peking Union Medical College, Nanjing, China.
| | - Kailin Yang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China; Psychosomatic laboratory, Department of Psychiatry, Daqing Hospital of Traditional Chinese Medicine, Daqing, China.
| | - Yang Wu
- Department of Rheumatology, National Clinical Research Center for Dermatologic and Immunologic Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Ganpeng Yu
- People's Hospital of Ningxiang City, Ningxiang, China
| | - Yexing Yan
- Psychosomatic laboratory, Department of Psychiatry, Daqing Hospital of Traditional Chinese Medicine, Daqing, China
| | - Moujia Hao
- Psychosomatic laboratory, Department of Psychiatry, Daqing Hospital of Traditional Chinese Medicine, Daqing, China
| | - Tian Song
- Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yuwei Li
- School of Mathematics and Computational Science, Hunan University of Science and Technology, Hunan, China
| | - Junpeng Chen
- Department of Physiology, School of Medicine, University of Louisville, Kentucky, USA; Psychosomatic laboratory, Department of Psychiatry, Daqing Hospital of Traditional Chinese Medicine, Daqing, China; Tong Jiecheng Studio, Hunan University of Science and Technology, Xiangtan, China.
| | - Lingyun Sun
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Graduate School of Peking Union Medical College, Nanjing, China; Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
2
|
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
|
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
|
Gil da Costa RM, Levesque C, Bianchi-Frias D, Chatterjee P, Lam HM, Santos C, Coleman IM, Ferreirinha P, Vilanova M, Pinto da Cunha N, Carvalho H, Moreira-Pais A, Faustino-Rocha A, Neto T, Batista da Costa J, Wright JL, Ferreira R, Oliveira PA, Mendes J, Bastos MMSM, Colaço B, Lopes C, Black PC, Sweeney CJ, Nelson PS. Pharmacological NF-κB inhibition decreases cisplatin chemoresistance in muscle-invasive bladder cancer and reduces cisplatin-induced toxicities. Mol Oncol 2023; 17:2709-2727. [PMID: 37533407 DOI: 10.1002/1878-0261.13504] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 03/28/2023] [Accepted: 08/01/2023] [Indexed: 08/04/2023] Open
Abstract
Most patients with muscle-invasive bladder cancer (MIBC) are not cured with platinum chemotherapy. Up-regulation of nuclear factor kappa light-chain enhancer of activated B cells (NF-κB) is a major mechanism underlying chemoresistance, suggesting that its pharmacological inhibition may increase platinum efficacy. NF-κB signaling was investigated in two patient cohorts. The Cancer Genome Atlas (TCGA) was used to correlate NF-κB signaling and patient survival. The efficacy of cisplatin plus the NF-κB inhibitor dimethylaminoparthenolide (DMAPT) versus cisplatin or DMAPT alone was tested in vitro. Xenografted and immunocompetent MIBC mouse models were studied in vivo. Platinum-naive claudin-low MIBC showed constitutive NF-κB signaling and this was associated with reduced disease-specific survival in TCGA patients. Chemotherapy up-regulated NF-κB signaling and chemoresistance-associated genes, including SPHK1, PLAUR, and SERPINE1. In mice, DMAPT significantly improved the efficacy of cisplatin in both models. The combination preserved body weight, renal function, and morphology, reduced muscle fatigue and IL-6 serum levels, and did not aggravate immuno-hematological toxicity compared with cisplatin alone. These data provide a rationale for combining NF-κB inhibition with platinum-based chemotherapy and conducting a clinical trial in MIBC patients.
Collapse
Affiliation(s)
- Rui M Gil da Costa
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Portugal
- ALiCE-Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Portugal
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), Porto, Portugal
- Center for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal
| | - Christine Levesque
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | - Payel Chatterjee
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Hung-Ming Lam
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Carlos Santos
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), Porto, Portugal
| | - Ilsa M Coleman
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | | | | | | | | | - Ana Faustino-Rocha
- Center for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal
| | - Tiago Neto
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), Porto, Portugal
| | | | | | | | - Paula A Oliveira
- Center for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal
- Department of Animal Science, UTAD, Vila Real, Portugal
| | | | - Margarida M S M Bastos
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Portugal
| | - Bruno Colaço
- Center for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal
- Department of Animal Science, UTAD, Vila Real, Portugal
| | | | - Peter C Black
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | - Christopher J Sweeney
- Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Peter S Nelson
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| |
Collapse
|
5
|
Khoenkhoen S, Ádori M, Solís-Sayago D, Soulier J, Russell J, Beutler B, Pedersen GK, Karlsson Hedestam GB. IκBNS expression in B cells is dispensable for IgG responses to T cell-dependent antigens. Front Immunol 2022; 13:1000755. [DOI: 10.3389/fimmu.2022.1000755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 10/03/2022] [Indexed: 11/13/2022] Open
Abstract
Mice lacking the atypical inhibitory kappa B (IκB) protein, IκBNS, a regulator of the NF-κB pathway encoded by the nfkbid gene, display impaired antibody responses to both T cell-independent (TI) and T cell-dependent (TD) antigens. To better understand the basis of these defects, we crossed mice carrying floxed nfkbid alleles with mice expressing Cre under the transcriptional control of the Cd79a gene to create mice that lacked IκBNS expression only in B cells. Analyses of these conditional knock-out mice revealed intact CD4+ and CD8+ T cell populations, including preserved frequencies of FoxP3+ regulatory T cells, which are known to be reduced in IκBNS knock-out mice. Like IκBNS knock-out mice, mice with conditional IκBNS ablation in B cells displayed defective IgM responses to TI antigens and a severe reduction in peritoneal B-1a cells. However, in contrast to mice lacking IκBNS altogether, the conditional IκBNS knock-out mice responded well to TD antigens compared to the control mice, with potent IgG responses following immunization with the viral antigen, rSFV-βGal or the widely used hapten-protein model antigen, NP-CGG. Furthermore, B cell intrinsic IκBNS expression was dispensable for germinal center (GC) formation and T follicular helper cell responses to NP-CGG immunization. The results presented here suggest that the defect in antibody responses to TD antigens observed in IκBNS knock-out mice results from a B cell extrinsic defect.
Collapse
|
6
|
Ren Y, Cui G, Gao Y. Research progress on inflammatory mechanism of primary Sjögren syndrome. Zhejiang Da Xue Xue Bao Yi Xue Ban 2021; 50:783-794. [PMID: 35347914 PMCID: PMC8931614 DOI: 10.3724/zdxbyxb-2021-0072] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 06/30/2021] [Indexed: 06/14/2023]
Abstract
Primary Sjögren syndrome is an autoimmune disease, in which a large number of lymphocytes infiltrate the exocrine glands and cause gland dysfunction. Its pathogenesis is related to the chronic inflammation of the exocrine glands caused by genetic factors, immunodeficiency or viral infection. Long-term inflammation leads to accelerated apoptosis of epithelial cells, disordered gland structure, increased expression of proinflammatory cytokine such as CXC subfamily ligand (CXCL) 12, CXCL13, B cell-activating factor (BAF), interleukin (IL)-6, interferon (IFN)-γ and tumor necrosis factor (TNF)-α in submandibular gland. With the action of antigen-presenting cells such as dendritic cells and macrophages, lymphocytes (mainly B cells) are induced to mature in secondary lymphoid organs and migrate to the submandibular gland to promotes the formation of germinal centers and the synthesis of autoantibodies. Meanwhile, innate lymphocytes, vascular endothelial cells and mucosa-associated constant T cells as important immune cells, also participated in the inflammatory response of the submandibular gland in primary Sjögren syndrome through different mechanisms. This process involves the activation of multiple signal pathways such as JAK/STAT, MAPK/ERK, PI3K/AKT/mTOR, PD-1/PD-L1, TLR/MyD88/NF-κB, BAF/BAF-R and IFN. These signaling pathways interact with each other and are intricately complex, causing lymphocytes to continuously activate and invade the submandibular glands. This article reviews the latest literature to clarify the mechanism of submandibular gland inflammation in primary Sjögren syndrome, and to provide insights for further research.
Collapse
|
7
|
Liu R, King A, Tarlinton D, Heierhorst J. The ASCIZ-DYNLL1 Axis Is Essential for TLR4-Mediated Antibody Responses and NF-κB Pathway Activation. Mol Cell Biol 2021; 41:e0025121. [PMID: 34543116 PMCID: PMC8608018 DOI: 10.1128/mcb.00251-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/25/2021] [Accepted: 09/09/2021] [Indexed: 11/20/2022] Open
Abstract
Toll-like receptors (TLRs) and interleukin-1 (IL-1) receptors regulate immune and inflammatory responses by activating the NF-κB pathway. Here, we report that B-cell-specific loss of dynein light chain 1 (DYNLL1, LC8) or its designated transcription factor ASCIZ (ATMIN) leads to severely reduced in vivo antibody responses to TLR4-dependent but not T-cell-dependent antigens in mice. This defect was independent of DYNLL1's established roles in modulating BIM-dependent apoptosis and 53BP1-dependent antibody class-switch recombination. In B cells and fibroblasts, the ASCIZ-DYNLL1 axis was required for TLR4-, IL-1-, and CD40-mediated NF-κB pathway activation but dispensable for antigen receptor and tumor necrosis factor α (TNF-α) signaling. In contrast to previous reports that overexpressed DYNLL1 directly inhibits the phosphorylation and degradation of the NF-κB inhibitor IκBα, we found here that under physiological conditions, DYNLL1 is required for signal-specific activation of the NF-κB pathway upstream of IκBα. Our data identify DYNLL1 as a signal-specific regulator of the NF-κB pathway and indicate that it may act as a universal modulator of TLR4 (and IL-1) signaling with wide-ranging roles in inflammation and immunity.
Collapse
Affiliation(s)
- Rui Liu
- St. Vincent’s Institute of Medical Research, Fitzroy, Victoria, Australia
| | - Ashleigh King
- St. Vincent’s Institute of Medical Research, Fitzroy, Victoria, Australia
| | - David Tarlinton
- Department of Immunology and Pathology, Monash University, Melbourne, Victoria, Australia
| | - Jörg Heierhorst
- St. Vincent’s Institute of Medical Research, Fitzroy, Victoria, Australia
- Department of Medicine at St. Vincent’s Hospital, University of Melbourne Medical School, Fitzroy, Victoria, Australia
| |
Collapse
|
8
|
Zhan T, Wang B, Fu J, Shao Y, Ye L, Shi H, Zheng L. Artesunate inhibits Sjögren's syndrome-like autoimmune responses and BAFF-induced B cell hyperactivation via TRAF6-mediated NF-κB signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 80:153381. [PMID: 33086170 DOI: 10.1016/j.phymed.2020.153381] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 10/07/2020] [Accepted: 10/12/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Hyperactivation of B cells by activators has been demonstrated to play a central role in the pathogenesis of Sjögren's syndrome (SS). In this study, we found that artesunate (ART) can attenuate BAFF-induced B cell hyperactivation and SS-like symptoms in NOD/Ltj mice. PURPOSE To determine the efficacy of ART in attenuating SS-like symptoms in vivo and explore the underlying mechanism in vitro. STUDY DESIGN ART was intragastrically injected into SS-like NOD/Ltj mice. The cytokine hsBAFF was used to activate Raji and Daudi B cells to mimic B cell hyperactivation in vitro. METHODS The efficacy of ART in inhibiting SS progression was studied in NOD/Ltj mice. Salivary flow rate, the number of lymphocytic infiltration foci, the level of autoantibodies and the extent of B cell infiltration were measured in the indicated groups. CCK-8 assays, flow cytometry-based EdU staining and Annexin V/PI staining were also used to detect the effect of ART on the survival and proliferation mechanism in BAFF-induced Raji and Daudi cells. Further studies determined that TRAF6 degradation is a potential mechanism by which ART determines B cell fate. RESULTS Treatment with ART inhibited lymphocytic foci formation, B cell infiltration and autoantibody secretion in SS-like NOD/Ltj mice. In vitro assay results indicated that ART effectively inhibited BAFF-induced viability, survival and proliferation of neoplastic B cells. Mechanistically, ART targeted BAFF-activated NFκB by regulating the proteasome-mediated degradation of TRAF6 in Raji and Daudi cells. CONCLUSION ART ameliorated murine SS-like symptoms and regulated TRAF6-NFκB signaling, thus determining survival and proliferation of B cells.
Collapse
Affiliation(s)
- Tianle Zhan
- Department of Oral Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center of Oral Disease, Shanghai, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Baoli Wang
- Department of Oral Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center of Oral Disease, Shanghai, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Jiayao Fu
- Department of Oral Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center of Oral Disease, Shanghai, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Yanxiong Shao
- Department of Oral Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center of Oral Disease, Shanghai, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Lei Ye
- Department of Oral Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center of Oral Disease, Shanghai, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Huan Shi
- Department of Oral Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center of Oral Disease, Shanghai, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China.
| | - Lingyan Zheng
- Department of Oral Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center of Oral Disease, Shanghai, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China.
| |
Collapse
|
9
|
Michaud E, Mastrandrea C, Rochereau N, Paul S. Human Secretory IgM: An Elusive Player in Mucosal Immunity. Trends Immunol 2020; 41:141-156. [PMID: 31928913 DOI: 10.1016/j.it.2019.12.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 12/06/2019] [Accepted: 12/10/2019] [Indexed: 12/29/2022]
Abstract
Secretory IgMs (SIgMs) were amongst the first identified immunoglobulins. However, their importance was not fully understood and recent advances have shown they play a key role in establishing and promoting commensal gut tolerance in mice and humans. The true interactions between SIgMs and the microbiota remain controversial and we aim to consolidate current knowledge in this review. Through comprehensive examination of SIgMs and their corresponding B cell secretors in several different pathological immunological contexts, we review the presumed role of these molecules in gut tolerance, inflammatory bowel diseases, and lung immunity. As SIgMs harbor a mostly tolerogenic function, we posit that their inclusion in further immunological research is paramount.
Collapse
Affiliation(s)
- Eva Michaud
- GIMAP/EA3064, Université de Lyon, CIC 1408 Vaccinology, Saint-Etienne, France
| | | | - Nicolas Rochereau
- GIMAP/EA3064, Université de Lyon, CIC 1408 Vaccinology, Saint-Etienne, France
| | - Stéphane Paul
- GIMAP/EA3064, Université de Lyon, CIC 1408 Vaccinology, Saint-Etienne, France.
| |
Collapse
|
10
|
A Role for NF-κB in Organ Specific Cancer and Cancer Stem Cells. Cancers (Basel) 2019; 11:cancers11050655. [PMID: 31083587 PMCID: PMC6563002 DOI: 10.3390/cancers11050655] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 05/02/2019] [Accepted: 05/08/2019] [Indexed: 02/07/2023] Open
Abstract
Cancer stem cells (CSCs) account for tumor initiation, invasiveness, metastasis, and recurrence in a broad range of human cancers. Although being a key player in cancer development and progression by stimulating proliferation and metastasis and preventing apoptosis, the role of the transcription factor NF-κB in cancer stem cells is still underestimated. In the present review, we will evaluate the role of NF-κB in CSCs of glioblastoma multiforme, ovarian cancer, multiple myeloma, lung cancer, colon cancer, prostate cancer, as well as cancer of the bone. Next to summarizing current knowledge regarding the presence and contribution of CSCs to the respective types of cancer, we will emphasize NF-κB-mediated signaling pathways directly involved in maintaining characteristics of cancer stem cells associated to tumor progression. Here, we will also focus on the status of NF-κB-activity predominantly in CSC populations and the tumor mass. Genetic alterations leading to NF-κB activity in glioblastoma, ependymoma, and multiple myeloma will be discussed.
Collapse
|
11
|
Abstract
In this review, Boothby et al. summarize some salient advances toward elucidation of the molecular programming of the fate choices and function of B cells in the periphery. They also note unanswered questions that pertain to differences among subsets of B lymphocytes and plasma cells. Mature B lymphocytes are crucial components of adaptive immunity, a system essential for the evolutionary fitness of mammals. Adaptive lymphocyte function requires an initially naïve cell to proliferate extensively and its progeny to have the capacity to assume a variety of fates. These include either terminal differentiation (the long-lived plasma cell) or metastable transcriptional reprogramming (germinal center and memory B cells). In this review, we focus principally on the regulation of differentiation and functional diversification of the “B2” subset. An overview is combined with an account of more recent advances, including initial work on mechanisms that eliminate DNA methylation and potential links between intracellular metabolites and chromatin editing.
Collapse
|
12
|
Khoenkhoen S, Erikson E, Ádori M, Stark JM, Scholz JL, Cancro MP, Pedersen GK, Karlsson Hedestam GB. TACI expression and plasma cell differentiation are impaired in the absence of functional IκBNS. Immunol Cell Biol 2019; 97:485-497. [PMID: 30597621 PMCID: PMC6850186 DOI: 10.1111/imcb.12228] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 12/17/2018] [Accepted: 12/27/2018] [Indexed: 12/15/2022]
Abstract
Impaired classical NF‐κB pathway signaling causes reduced antibody responses to T‐independent (TI) antigens. We investigated the potential reasons for defective TI responses in mice lacking the atypical inhibitory kappa B (IκB) protein of the NF‐κB pathway, IκBNS. Analyses of the plasma cell compartment in vitro and in vivo after challenge with lipopolysaccharide (LPS) showed significant decreases in the frequencies of plasma cells in the absence of IκBNS. In vitro activation of B cells via the B cell receptor or via Toll‐like receptor 4 revealed that early activation events were unaffected in IκBNS‐deficient B cells, while proliferation was reduced compared to in similarly stimulated wildtype (wt) B cells. IκBNS‐deficient B cells also displayed impaired upregulation of the transmembrane activator and calcium modulator cyclophilin ligand interactor (TACI), which is essential for TI responses, and decreased sensitivity to TACI ligands upon stimulation. Furthermore, IκBNS‐deficient B cells, in contrast to wt B cells, displayed altered expression of IRF4, Blimp‐1 and Pax5 upon LPS‐induced differentiation, indicating impaired transcriptional regulation of plasma cell generation.
Collapse
Affiliation(s)
- Sharesta Khoenkhoen
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Elina Erikson
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Monika Ádori
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Julian M Stark
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Jean L Scholz
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael P Cancro
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gabriel K Pedersen
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
13
|
Critical influences on the pathogenesis of follicular lymphoma. Blood 2018; 131:2297-2306. [PMID: 29666116 DOI: 10.1182/blood-2017-11-764365] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 12/28/2017] [Indexed: 12/15/2022] Open
Abstract
The development of follicular lymphoma (FL) from a founder B cell with an upregulation of B-cell lymphoma 2 (BCL2), via the t(14;18) translocation, to a proliferating clone, poised to undergo further transformation to an aggressive lymphoma, illustrates the opportunistic Darwinian process of tumorigenesis. Protection against apoptosis allows an innocent cell to persist and divide, with dangerous accumulation of further mutational changes, commonly involving inactivation of chromatin-modifying genes. But this is not all. FL cells reflect normal B cells in relying on expression of surface immunoglobulin. In doing so, they add another supportive mechanism by exploiting the natural process of somatic hypermutation of the IGV genes. Positive selection of motifs for addition of glycan into the antigen-binding sites of virtually all cases, and the placement of unusual mannoses in those sites, reveals a posttranslational strategy to engage the microenvironment. A bridge between mannosylated surface immunoglobulin of FL cells and macrophage-expressed dendritic cell-specific ICAM-3-grabbing nonintegrin produces a persistent low-level signal that appears essential for life in the hostile germinal center. Early-stage FL therefore requires a triad of changes: protection from apoptosis, mutations in chromatin modifiers, and an ability to interact with lectin-expressing macrophages. These changes are common and persistent. Genetic/epigenetic analysis is providing important data but investigation of the posttranslational landscape is the next challenge. We have one glimpse of its operation via the influence of added glycan on the B-cell receptor of FL. The consequential interaction with environmental lectins illustrates how posttranslational modifications can be exploited by tumor cells, and could lead to new approaches to therapy.
Collapse
|
14
|
Abstract
B cell receptor (BCR) signalling is crucial for normal B cell development and adaptive immunity. BCR signalling also supports the survival and growth of malignant B cells in patients with B cell leukaemias or lymphomas. The mechanism of BCR pathway activation in these diseases includes continuous BCR stimulation by microbial antigens or autoantigens present in the tissue microenvironment, activating mutations within the BCR complex or downstream signalling components and ligand-independent tonic BCR signalling. The most established agents targeting BCR signalling are Bruton tyrosine kinase (BTK) inhibitors and PI3K isoform-specific inhibitors, and their introduction into the clinic is rapidly changing how B cell malignancies are treated. B cells and BCR-related kinases, such as BTK, also play a role in the microenvironment of solid tumours, such as squamous cell carcinoma and pancreatic cancer, and therefore targeting B cells or BCR-related kinases may have anticancer activity beyond B cell malignancies.
Collapse
MESH Headings
- Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors
- Antineoplastic Agents/pharmacology
- B-Lymphocytes/metabolism
- B-Lymphocytes/pathology
- Drug Resistance, Neoplasm
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Lymphoma, Mantle-Cell/drug therapy
- Lymphoma, Mantle-Cell/metabolism
- Molecular Targeted Therapy/methods
- Neoplasms/drug therapy
- Neoplasms/metabolism
- Protein Kinase Inhibitors/pharmacology
- Receptors, Antigen, B-Cell/antagonists & inhibitors
- Receptors, Antigen, B-Cell/immunology
- Receptors, Antigen, B-Cell/metabolism
- Signal Transduction/drug effects
- Tumor Microenvironment
Collapse
Affiliation(s)
- Jan A Burger
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Adrian Wiestner
- Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
15
|
Schweighoffer E, Tybulewicz VL. Signalling for B cell survival. Curr Opin Cell Biol 2017; 51:8-14. [PMID: 29149682 DOI: 10.1016/j.ceb.2017.10.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 10/10/2017] [Indexed: 12/21/2022]
Abstract
The number of mature B cells is carefully controlled by signalling from receptors that support B cell survival. The best studied of these are the B cell antigen receptor (BCR) and BAFFR. Recent work has shown that signalling from these receptors is closely linked, involves the CD19 co-receptor, and leads to activation of canonical and non-canonical NF-κB pathways, ERK1, ERK2 and ERK5 MAP kinases, and PI-3 kinases. Importantly, studies show that investigation of the importance of signalling molecules in cell survival requires the use of inducible gene deletions within mature B cells. This overcomes the limitations of many earlier studies using constitutive gene deletions which were unable to distinguish between requirements for a protein in development versus survival.
Collapse
Affiliation(s)
| | - Victor Lj Tybulewicz
- The Francis Crick Institute, London NW1 1AT, UK; Imperial College, London W12 0NN, UK.
| |
Collapse
|
16
|
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
|
17
|
Smith CIE. From identification of the BTK kinase to effective management of leukemia. Oncogene 2017; 36:2045-2053. [PMID: 27669440 PMCID: PMC5395699 DOI: 10.1038/onc.2016.343] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 08/02/2016] [Accepted: 08/03/2016] [Indexed: 12/11/2022]
Abstract
BTK is a cytoplasmic protein-tyrosine kinase, whose corresponding gene was isolated in the early 1990s. BTK was initially identified by positional cloning of the gene causing X-linked agammaglobulinemia and independently in a search for new kinases. Given the phenotype of affected patients, namely lack of B-lymphocytes and plasma cells with the ensuing inability to mount humoral immune responses, BTK inhibitors were anticipated to have beneficial effects on antibody-mediated pathologies, such as autoimmunity. In contrast to, for example, the SRC-family of cytoplasmic kinases, there was no obvious way in which structural alterations would yield constitutively active forms of BTK, and such mutations were also not found in leukemias or lymphomas. In 2007, the first efficient inhibitor, ibrutinib, was reported and soon became approved both in the United States and in Europe for the treatment of three B-cell malignancies, mantle cell lymphoma, chronic lymphocytic leukemia and Waldenström's macroglobulinemia. Over the past few years, additional inhibitors have been developed, with acalabrutinib being more selective, and recently demonstrating fewer clinical adverse effects. The antitumor mechanism is also not related to mutations in BTK. Instead tumor residency in lymphoid organs is inhibited, making these drugs highly versatile. BTK is one of the only 10 human kinases that carry a cysteine in the adenosine triphosphate-binding cleft. As this allows for covalent, irreversible inhibitor binding, it provides these compounds with a highly advantageous character. This quality may be crucial and bodes well for the future of BTK-modifying medicines, which have been estimated to reach annual multi-billion dollar sales in the future.
Collapse
Affiliation(s)
- C I E Smith
- Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital, Huddinge, Sweden
| |
Collapse
|
18
|
Oleksyn D, Zhao J, Vosoughi A, Zhao JC, Misra R, Pentland AP, Ryan D, Anolik J, Ritchlin C, Looney J, Anandarajah AP, Schwartz G, Calvi LM, Georger M, Mohan C, Sanz I, Chen L. PKK deficiency in B cells prevents lupus development in Sle lupus mice. Immunol Lett 2017; 185:1-11. [PMID: 28274793 DOI: 10.1016/j.imlet.2017.03.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Revised: 02/27/2017] [Accepted: 03/01/2017] [Indexed: 12/25/2022]
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease characterized by the production of autoantibodies that can result in damage to multiple organs. It is well documented that B cells play a critical role in the development of the disease. We previously showed that protein kinase C associated kinase (PKK) is required for B1 cell development as well as for the survival of recirculating mature B cells and B-lymphoma cells. Here, we investigated the role of PKK in lupus development in a lupus mouse model. We demonstrate that the conditional deletion of PKK in B cells prevents lupus development in Sle1Sle3 mice. The loss of PKK in Sle mice resulted in the amelioration of multiple classical lupus-associated phenotypes and histologic features of lupus nephritis, including marked reduction in the levels of serum autoantibodies, proteinuria, spleen size, peritoneal B-1 cell population and the number of activated CD4 T cells. In addition, the abundance of autoreactive plasma cells normally seen in Sle lupus mice was also significantly decreased in the PKK-deficient Sle mice. Sle B cells deficient in PKK display defective proliferation responses to BCR and LPS stimulation. Consistently, B cell receptor-mediated NF-κB activation, which is required for the survival of activated B cells, was impaired in the PKK-deficient B cells. Taken together, our work uncovers a critical role of PKK in lupus development and suggests that targeting the PKK-mediated pathway may represent a promising therapeutic strategy for lupus treatment.
Collapse
Affiliation(s)
- D Oleksyn
- Division of Allergy/Immunology and Rheumatology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - J Zhao
- Department of Biomedical Genetics, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - A Vosoughi
- Division of Allergy/Immunology and Rheumatology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - J C Zhao
- Department of Biology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - R Misra
- Department of Pediatrics, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - A P Pentland
- Department of Dermatology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - D Ryan
- Department of Pathology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - J Anolik
- Division of Allergy/Immunology and Rheumatology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - C Ritchlin
- Division of Allergy/Immunology and Rheumatology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - J Looney
- Division of Allergy/Immunology and Rheumatology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - A P Anandarajah
- Division of Allergy/Immunology and Rheumatology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - G Schwartz
- Department of Pediatrics, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - L M Calvi
- Department of Medicine, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - M Georger
- Department of Medicine, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - C Mohan
- Department Biomedical Engineering, University of Houston, Houston, TX 77204, United States
| | - I Sanz
- Division of Allergy/Immunology and Rheumatology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - L Chen
- Division of Allergy/Immunology and Rheumatology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States; Department of Dermatology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States.
| |
Collapse
|
19
|
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
|