101
|
Min YN, Wang CY, Li XX, Hou Y, Qiu JH, Ma J, Shao LL, Zhang X, Wang YW, Peng J, Hou M, Shi Y. Participation of B-cell-activating factor receptors in the pathogenesis of immune thrombocytopenia. J Thromb Haemost 2016; 14:559-71. [PMID: 26749059 DOI: 10.1111/jth.13246] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 12/20/2015] [Indexed: 12/19/2022]
Abstract
UNLABELLED ESSENTIALS: Dysfunctional B-cell-activating factor (BAFF) system is related to many autoimmune diseases. The regulatory functions of BAFF/BAFF receptors were investigated in an in vitro coculture system. Different regulatory roles of BAFF were investigated via different receptors in immune thrombocytopenia. The upregulated BAFF receptors on autoreactive lymphocytes lead to their hypersensitivity to BAFF. SUMMARY BACKGROUND The pathogenesis of immune thrombocytopenia (ITP) remains enigmatic. B-cell-activating factor (BAFF) and its receptors (BAFF receptor [BAFF-R], transmembrane activator and calcium modulator and cyclophilin ligand interactor [TACI], and B-cell maturation antigen) play central roles in the integrated homeostatic regulation of lymphocytes. OBJECTIVES To investigate the pathologic roles of BAFF receptors in regulating the bioactivities of lymphocytes in ITP. METHODS An in vitro culture system was established by stimulating CD14(-) peripheral lymphocytes with platelet-preloaded dendritic cells in the presence of recombinant human BAFF (rhBAFF; 20 ng mL(-1)). The functions of BAFF receptors were specifically blocked with blocking antibodies. RESULTS BAFF-R, besides prolonging the survival of B cells in both patients and healthy controls, prominently promoted the survival of CD8(+) T cells and the proliferation of B cells in patients with ITP. TACI, as a positive regulator, not only promoted the proliferation of CD4(+) and CD8(+) T cells, but also significantly enhanced the secretion of interleukin-4 in patients with ITP, but not in controls. Besides revealing the pathologic roles of BAFF receptors, these results also indicate that lymphocytes of patients with ITP have enhanced antiapoptotic or proliferative capacity as compared with those from healthy controls when exposed under similar stimulation of rhBAFF. Further study demonstrated that activated autoreactive B cells and CD4(+) T cells from patients with ITP showed significantly higher expression of BAFF-R or TACI than those from healthy controls. CONCLUSIONS Both BAFF-R and TACI are pathogenic participants in ITP. Their dysregulated expression in patients with ITP may lead to hyperreactivity of activated autoreactive lymphocytes in response to rhBAFF, and thus is highly significant in the pathogenesis of ITP.
Collapse
Affiliation(s)
- Y-N Min
- Hematology Oncology Center, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - C-Y Wang
- Department of General Medicine, Second Hospital of Shandong University, Jinan, Shandong, China
| | - X-X Li
- Hematology Oncology Center, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Y Hou
- Hematology Oncology Center, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - J-H Qiu
- Hematology Oncology Center, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - J Ma
- Department of General Medicine, Medical Research Institute for Tumor Prevention and Cure, Shandong University, Jinan, Shandong, China
| | - L-L Shao
- Hematology Oncology Center, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - X Zhang
- Hematology Oncology Center, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Y-W Wang
- Hematology Oncology Center, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - J Peng
- Hematology Oncology Center, Qilu Hospital, Shandong University, Jinan, Shandong, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Jinan, Shandong, China
| | - M Hou
- Hematology Oncology Center, Qilu Hospital, Shandong University, Jinan, Shandong, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Jinan, Shandong, China
| | - Y Shi
- Hematology Oncology Center, Qilu Hospital, Shandong University, Jinan, Shandong, China
| |
Collapse
|
102
|
De Silva NS, Silva K, Anderson MM, Bhagat G, Klein U. Impairment of Mature B Cell Maintenance upon Combined Deletion of the Alternative NF-κB Transcription Factors RELB and NF-κB2 in B Cells. THE JOURNAL OF IMMUNOLOGY 2016; 196:2591-601. [PMID: 26851215 DOI: 10.4049/jimmunol.1501120] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 01/05/2016] [Indexed: 02/06/2023]
Abstract
BAFF is critical for the survival and maturation of mature B cells. BAFF, via BAFFR, activates multiple signaling pathways in B cells, including the alternative NF-κB pathway. The transcription factors RELB and NF-κB2 (p100/p52) are the downstream mediators of the alternative pathway; however, the B cell-intrinsic functions of these NF-κB subunits have not been studied in vivo using conditional alleles, either individually or in combination. We in this study report that B cell-specific deletion of relb led to only a slight decrease in the fraction of mature splenic B cells, whereas deletion of nfkb2 caused a marked reduction. This phenotype was further exacerbated upon combined deletion of relb and nfkb2 and most dramatically affected the maintenance of marginal zone B cells. BAFF stimulation, in contrast to CD40 activation, was unable to rescue relb/nfkb2-deleted B cells in vitro. RNA-sequencing analysis of BAFF-stimulated nfkb2-deleted versus normal B cells suggests that the alternative NF-κB pathway, in addition to its critical role in BAFF-mediated cell survival, may control the expression of genes involved in the positioning of B cells within the lymphoid microenvironment and in the establishment of T cell-B cell interactions. Thus, by ablating the downstream transcription factors of the alternative NF-κB pathway specifically in B cells, we identify in this study a critical role for the combined activity of the RELB and NF-κB2 subunits in B cell homeostasis that cannot be compensated for by the canonical NF-κB pathway under physiological conditions.
Collapse
Affiliation(s)
- Nilushi S De Silva
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10032; Department of Microbiology and Immunology, Columbia University, New York, NY 10032; and
| | - Kathryn Silva
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10032
| | - Michael M Anderson
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10032
| | - Govind Bhagat
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10032; Department of Pathology and Cell Biology, Columbia University, New York, NY 10032
| | - Ulf Klein
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10032; Department of Microbiology and Immunology, Columbia University, New York, NY 10032; and Department of Pathology and Cell Biology, Columbia University, New York, NY 10032
| |
Collapse
|
103
|
B-cell survival and development controlled by the coordination of NF-κB family members RelB and cRel. Blood 2016; 127:1276-86. [PMID: 26773039 PMCID: PMC4786837 DOI: 10.1182/blood-2014-10-606988] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2014] [Accepted: 12/29/2015] [Indexed: 11/20/2022] Open
Abstract
Targeted deletion of BAFF causes severe deficiency of splenic B cells. BAFF-R is commonly thought to signal to nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB)-inducing kinase dependent noncanonical NF-κB RelB. However, RelB-deficient mice have normal B-cell numbers. Recent studies showed that BAFF also signals to the canonical NF-κB pathway, and we found that both RelB and cRel are persistently activated, suggesting BAFF signaling coordinates both pathways to ensure robust B-cell development. Indeed, we report now that combined loss of these 2 NF-κB family members leads to impaired BAFF-mediated survival and development in vitro. Although single deletion of RelB and cRel was dispensable for normal B-cell development, double knockout mice displayed an early B-cell developmental blockade and decreased mature B cells. Despite disorganized splenic architecture in Relb(-/-)cRel(-/-) mice, generation of mixed-mouse chimeras established the developmental phenotype to be B-cell intrinsic. Together, our results indicate that BAFF signals coordinate both RelB and cRel activities to ensure survival during peripheral B-cell maturation.
Collapse
|
104
|
Kim J, Sturgill D, Tran AD, Sinclair DA, Oberdoerffer P. Controlled DNA double-strand break induction in mice reveals post-damage transcriptome stability. Nucleic Acids Res 2015; 44:e64. [PMID: 26687720 PMCID: PMC4838352 DOI: 10.1093/nar/gkv1482] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 12/07/2015] [Indexed: 02/06/2023] Open
Abstract
DNA double-strand breaks (DSBs) and their repair can cause extensive epigenetic changes. As a result, DSBs have been proposed to promote transcriptional and, ultimately, physiological dysfunction via both cell-intrinsic and cell-non-autonomous pathways. Studying the consequences of DSBs in higher organisms has, however, been hindered by a scarcity of tools for controlled DSB induction. Here, we describe a mouse model that allows for both tissue-specific and temporally controlled DSB formation at ∼140 defined genomic loci. Using this model, we show that DSBs promote a DNA damage signaling-dependent decrease in gene expression in primary cells specifically at break-bearing genes, which is reversed upon DSB repair. Importantly, we demonstrate that restoration of gene expression can occur independently of cell cycle progression, underlining its relevance for normal tissue maintenance. Consistent with this, we observe no evidence for persistent transcriptional repression in response to a multi-day course of continuous DSB formation and repair in mouse lymphocytes in vivo Together, our findings reveal an unexpected capacity of primary cells to maintain transcriptome integrity in response to DSBs, pointing to a limited role for DNA damage as a mediator of cell-autonomous epigenetic dysfunction.
Collapse
Affiliation(s)
- Jeongkyu Kim
- Laboratory for Receptor Biology and Gene Expression, National Cancer Institute, 41 Library Drive, Bethesda, MD 20892, USA
| | - David Sturgill
- Laboratory for Receptor Biology and Gene Expression, National Cancer Institute, 41 Library Drive, Bethesda, MD 20892, USA
| | - Andy D Tran
- Laboratory for Receptor Biology and Gene Expression, National Cancer Institute, 41 Library Drive, Bethesda, MD 20892, USA
| | - David A Sinclair
- The Paul F. Glenn Laboratories for the Biological Mechanisms of Aging, Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Philipp Oberdoerffer
- Laboratory for Receptor Biology and Gene Expression, National Cancer Institute, 41 Library Drive, Bethesda, MD 20892, USA
| |
Collapse
|
105
|
Polverino F, Cosio BG, Pons J, Laucho-Contreras M, Tejera P, Iglesias A, Rios A, Jahn A, Sauleda J, Divo M, Pinto-Plata V, Sholl L, Rosas IO, Agustí A, Celli BR, Owen CA. B Cell-Activating Factor. An Orchestrator of Lymphoid Follicles in Severe Chronic Obstructive Pulmonary Disease. Am J Respir Crit Care Med 2015; 192:695-705. [PMID: 26073875 DOI: 10.1164/rccm.201501-0107oc] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
RATIONALE Patients with chronic obstructive pulmonary disease (COPD) have increased pulmonary lymphoid follicle (LF) counts. B cell-activating factor of tumor necrosis factor family (BAFF) regulates B cells in health, but its role in COPD pathogenesis is unclear. OBJECTIVES To determine whether BAFF expression in pulmonary LFs correlates with COPD severity, LF size or number, and/or readouts of B-cell function in LFs. METHODS We correlated BAFF immunostaining in LFs in lung explants or biopsies from nonsmoking control subjects (NSC), smokers without COPD (SC), and patients with COPD with the number and size of LFs, and LF B-cell apoptosis, activation, and proliferation. We analyzed serum BAFF levels and BAFF expression in B cells in blood and bronchoalveolar lavage samples from the same subject groups. We assessed whether: (1) cigarette smoke extract (CSE) increases B-cell BAFF expression and (2) recombinant BAFF (rBAFF) rescues B cells from CSE-induced apoptosis by inhibiting activation of nuclear factor-κB (NF-κB). MEASUREMENTS AND MAIN RESULTS Patients with Global Initiative for Chronic Obstructive Lung Disease (GOLD) stage IV COPD had increased numbers and larger pulmonary LFs than patients with GOLD stages I-II COPD and SC. We identified two main types of pulmonary LFs: (1) type A, the predominant type in GOLD stages I-II COPD and SC, characterized by abundant apoptotic but few BAFF-positive cells (mostly B cells); and (2) type B, the main type in GOLD stage IV COPD, characterized by abundant BAFF-positive cells but few apoptotic cells (mostly B cells). BAFF levels were also higher in blood and bronchoalveolar lavage B cells in patients with COPD versus NSC and SC. Surprisingly, rBAFF blocked CSE-induced B-cell apoptosis by inhibiting CSE-induced NF-κB activation. CONCLUSIONS Our data support the hypothesis that B-cell BAFF expression creates a self-perpetuating loop contributing to COPD progression by promoting pulmonary B-cell survival and LF expansion.
Collapse
Affiliation(s)
- Francesca Polverino
- 1 Division of Pulmonary and Critical Care Medicine and.,2 Lovelace Respiratory Research Institute, Albuquerque, New Mexico.,3 University of Parma, Parma, Italy
| | - Borja G Cosio
- 4 Research Unit, FISIB, IdISPa, and.,5 Department of Respiratory Medicine, Hospital Son Espases, Ciber de Enfermedades Respiratorias (CIBERES), Mallorca, Spain
| | - Jaime Pons
- 4 Research Unit, FISIB, IdISPa, and.,6 Immunology Department, Hospital Son Espases, Mallorca, Spain
| | | | - Paula Tejera
- 7 Harvard School of Public Health, Boston, Massachusetts; and
| | | | | | | | - Jaume Sauleda
- 4 Research Unit, FISIB, IdISPa, and.,5 Department of Respiratory Medicine, Hospital Son Espases, Ciber de Enfermedades Respiratorias (CIBERES), Mallorca, Spain
| | - Miguel Divo
- 1 Division of Pulmonary and Critical Care Medicine and.,2 Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| | - Victor Pinto-Plata
- 1 Division of Pulmonary and Critical Care Medicine and.,2 Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| | - Lynette Sholl
- 8 Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Ivan O Rosas
- 1 Division of Pulmonary and Critical Care Medicine and.,2 Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| | - Alvar Agustí
- 5 Department of Respiratory Medicine, Hospital Son Espases, Ciber de Enfermedades Respiratorias (CIBERES), Mallorca, Spain.,9 Thorax Institute, Hospital Clinic, IDIBAPS, University of Barcelona, and CIBERES, FISIB, Mallorca, Spain
| | - Bartolome R Celli
- 1 Division of Pulmonary and Critical Care Medicine and.,2 Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| | - Caroline A Owen
- 1 Division of Pulmonary and Critical Care Medicine and.,2 Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| |
Collapse
|
106
|
Lymphomagenic CARD11/BCL10/MALT1 signaling drives malignant B-cell proliferation via cooperative NF-κB and JNK activation. Proc Natl Acad Sci U S A 2015; 112:E7230-8. [PMID: 26668357 DOI: 10.1073/pnas.1507459112] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The aggressive activated B cell-like subtype of diffuse large B-cell lymphoma is characterized by aberrant B-cell receptor (BCR) signaling and constitutive nuclear factor kappa-B (NF-κB) activation, which is required for tumor cell survival. BCR-induced NF-κB activation requires caspase recruitment domain-containing protein 11 (CARD11), and CARD11 gain-of-function mutations are recurrently detected in human diffuse large B-cell lymphoma (DLBCL). To investigate the consequences of dysregulated CARD11 signaling in vivo, we generated mice that conditionally express the human DLBCL-derived CARD11(L225LI) mutant. Surprisingly, CARD11(L225LI) was sufficient to trigger aggressive B-cell lymphoproliferation, leading to early postnatal lethality. CARD11(L225LI) constitutively associated with B-cell CLL/lymphoma 10 (BCL10) and mucosa-associated lymphoid tissue lymphoma translocation gene 1 (MALT1) to simultaneously activate the NF-κB and c-Jun N-terminal kinase (JNK) signaling cascades. Genetic deficiencies of either BCL10 or MALT1 completely rescued the phenotype, and pharmacological inhibition of JNK was, similar to NF-κB blockage, toxic to autonomously proliferating CARD11(L225LI)-expressing B cells. Moreover, constitutive JNK activity was observed in primary human activated B cell-like (ABC)-DLBCL specimens, and human ABC-DLBCL cells were also sensitive to JNK inhibitors. Thus, our results demonstrate that enforced activation of CARD11/BCL10/MALT1 signaling is sufficient to drive transformed B-cell expansion in vivo and identify the JNK pathway as a therapeutic target for ABC-DLBCL.
Collapse
|
107
|
Finkin S, Yuan D, Stein I, Taniguchi K, Weber A, Unger K, Browning JL, Goossens N, Nakagawa S, Gunasekaran G, Schwartz ME, Kobayashi M, Kumada H, Berger M, Pappo O, Rajewsky K, Hoshida Y, Karin M, Heikenwalder M, Ben-Neriah Y, Pikarsky E. Ectopic lymphoid structures function as microniches for tumor progenitor cells in hepatocellular carcinoma. Nat Immunol 2015; 16:1235-44. [PMID: 26502405 PMCID: PMC4653079 DOI: 10.1038/ni.3290] [Citation(s) in RCA: 295] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 09/07/2015] [Indexed: 12/25/2022]
Abstract
Ectopic lymphoid-like structures (ELSs) are often observed in cancer, yet their function is obscure. Although ELSs signify good prognosis in certain malignancies, we found that hepatic ELSs indicated poor prognosis for hepatocellular carcinoma (HCC). We studied an HCC mouse model that displayed abundant ELSs and found that they constituted immunopathological microniches wherein malignant hepatocyte progenitor cells appeared and thrived in a complex cellular and cytokine milieu until gaining self-sufficiency. The egress of progenitor cells and tumor formation were associated with the autocrine production of cytokines previously provided by the niche. ELSs developed via cooperation between the innate immune system and adaptive immune system, an event facilitated by activation of the transcription factor NF-κB and abolished by depletion of T cells. Such aberrant immunological foci might represent new targets for cancer therapy.
Collapse
Affiliation(s)
- Shlomi Finkin
- Department of Immunology and Cancer Research, Institute for Medical Research Israel Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
- Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Detian Yuan
- Institute of Virology, Technische Universität München, Helmholtz Zentrum München, Munich, Germany
| | - Ilan Stein
- Department of Immunology and Cancer Research, Institute for Medical Research Israel Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
- Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Koji Taniguchi
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Achim Weber
- Institute of Surgical Pathology, University and University-Hospital Zurich, Zurich, Switzerland
| | - Kristian Unger
- Research Unit of Radiation Cytogenetics, Helmholtz-Zentrum München, Ingolstädter-Landstrasse, Neuherberg, Germany
| | - Jeffrey L Browning
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Nicolas Goossens
- Division of Liver Diseases, Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Division of Gastroenterology &Hepatology Geneva University Hospital, Geneva, Switzerland
| | - Shigeki Nakagawa
- Division of Liver Diseases, Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Ganesh Gunasekaran
- Recanati/Miller Transplantation Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Myron E Schwartz
- Recanati/Miller Transplantation Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | | | - Michael Berger
- Department of Immunology and Cancer Research, Institute for Medical Research Israel Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Orit Pappo
- Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Klaus Rajewsky
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Yujin Hoshida
- Division of Liver Diseases, Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Mathias Heikenwalder
- Institute of Virology, Technische Universität München, Helmholtz Zentrum München, Munich, Germany
- Division of Chronic Inflammation and Cancer, German Cancer Research Center, Heidelberg, Germany
| | - Yinon Ben-Neriah
- Department of Immunology and Cancer Research, Institute for Medical Research Israel Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Eli Pikarsky
- Department of Immunology and Cancer Research, Institute for Medical Research Israel Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
- Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
108
|
Kondylis V, Polykratis A, Ehlken H, Ochoa-Callejero L, Straub BK, Krishna-Subramanian S, Van TM, Curth HM, Heise N, Weih F, Klein U, Schirmacher P, Kelliher M, Pasparakis M. NEMO Prevents Steatohepatitis and Hepatocellular Carcinoma by Inhibiting RIPK1 Kinase Activity-Mediated Hepatocyte Apoptosis. Cancer Cell 2015; 28:582-598. [PMID: 26555174 PMCID: PMC4644221 DOI: 10.1016/j.ccell.2015.10.001] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 06/02/2015] [Accepted: 10/06/2015] [Indexed: 12/18/2022]
Abstract
IκB kinase/nuclear [corrected] factor κB (IKK/NF-κB) signaling exhibits important yet opposing functions in hepatocarcinogenesis. Mice lacking NEMO in liver parenchymal cells (LPC) spontaneously develop steatohepatitis and hepatocellular carcinoma (HCC) suggesting that NF-κB prevents liver disease and cancer. Here, we show that complete NF-κB inhibition by combined LPC-specific ablation of RelA, c-Rel, and RelB did not phenocopy NEMO deficiency, but constitutively active IKK2-mediated NF-κB activation prevented hepatocellular damage and HCC in NEMO(LPC-KO) mice. Knock-in expression of kinase inactive receptor-interacting protein kinase 1 (RIPK1) prevented hepatocyte apoptosis and HCC, while RIPK1 ablation induced TNFR1-associated death domain protein (TRADD)-dependent hepatocyte apoptosis and liver tumors in NEMO(LPC-KO) mice, revealing distinct kinase-dependent and scaffolding functions of RIPK1. Collectively, these results show that NEMO prevents hepatocarcinogenesis by inhibiting RIPK1 kinase activity-driven hepatocyte apoptosis through NF-κB-dependent and -independent functions.
Collapse
Affiliation(s)
- Vangelis Kondylis
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, 50931, Cologne, Germany
| | - Apostolos Polykratis
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, 50931, Cologne, Germany
| | - Hanno Ehlken
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, 50931, Cologne, Germany
| | - Laura Ochoa-Callejero
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, 50931, Cologne, Germany
| | - Beate Katharina Straub
- Institute of Pathology, University Hospital Heidelberg, INF 224, 69120 Heidelberg, Germany
| | - Santosh Krishna-Subramanian
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, 50931, Cologne, Germany
| | - Trieu-My Van
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, 50931, Cologne, Germany
| | - Harald-Morten Curth
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, 50931, Cologne, Germany
| | - Nicole Heise
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10032, USA
| | - Falk Weih
- Leibniz-Institute for Age Research-Fritz-Lipmann-Institute, 07745 Jena, Germany
| | - Ulf Klein
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10032, USA; Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA; Department of Microbiology and Immunology, Columbia University, New York, NY 10032, USA
| | - Peter Schirmacher
- Institute of Pathology, University Hospital Heidelberg, INF 224, 69120 Heidelberg, Germany
| | - Michelle Kelliher
- Department of Molecular, Cellular and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Manolis Pasparakis
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, 50931, Cologne, Germany.
| |
Collapse
|
109
|
Finkin S, Yuan D, Stein I, Taniguchi K, Weber A, Unger K, Browning JL, Goossens N, Nakagawa S, Gunasekaran G, Schwartz ME, Kobayashi M, Kumada H, Berger M, Pappo O, Rajewsky K, Hoshida Y, Karin M, Heikenwalder M, Ben-Neriah Y, Pikarsky E. Ectopic lymphoid structures function as microniches for tumor progenitor cells in hepatocellular carcinoma. Nat Immunol 2015. [PMID: 26502405 DOI: 10.1038/ni.3290.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Ectopic lymphoid-like structures (ELSs) are often observed in cancer, yet their function is obscure. Although ELSs signify good prognosis in certain malignancies, we found that hepatic ELSs indicated poor prognosis for hepatocellular carcinoma (HCC). We studied an HCC mouse model that displayed abundant ELSs and found that they constituted immunopathological microniches wherein malignant hepatocyte progenitor cells appeared and thrived in a complex cellular and cytokine milieu until gaining self-sufficiency. The egress of progenitor cells and tumor formation were associated with the autocrine production of cytokines previously provided by the niche. ELSs developed via cooperation between the innate immune system and adaptive immune system, an event facilitated by activation of the transcription factor NF-κB and abolished by depletion of T cells. Such aberrant immunological foci might represent new targets for cancer therapy.
Collapse
Affiliation(s)
- Shlomi Finkin
- Department of Immunology and Cancer Research, Institute for Medical Research Israel Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel.,Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Detian Yuan
- Institute of Virology, Technische Universität München, Helmholtz Zentrum München, Munich, Germany
| | - Ilan Stein
- Department of Immunology and Cancer Research, Institute for Medical Research Israel Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel.,Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Koji Taniguchi
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Achim Weber
- Institute of Surgical Pathology, University and University-Hospital Zurich, Zurich, Switzerland
| | - Kristian Unger
- Research Unit of Radiation Cytogenetics, Helmholtz-Zentrum München, Ingolstädter-Landstrasse, Neuherberg, Germany
| | - Jeffrey L Browning
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Nicolas Goossens
- Division of Liver Diseases, Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, USA.,Division of Gastroenterology &Hepatology Geneva University Hospital, Geneva, Switzerland
| | - Shigeki Nakagawa
- Division of Liver Diseases, Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Ganesh Gunasekaran
- Recanati/Miller Transplantation Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Myron E Schwartz
- Recanati/Miller Transplantation Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | | | - Michael Berger
- Department of Immunology and Cancer Research, Institute for Medical Research Israel Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Orit Pappo
- Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Klaus Rajewsky
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Yujin Hoshida
- Division of Liver Diseases, Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Mathias Heikenwalder
- Institute of Virology, Technische Universität München, Helmholtz Zentrum München, Munich, Germany.,Division of Chronic Inflammation and Cancer, German Cancer Research Center, Heidelberg, Germany
| | - Yinon Ben-Neriah
- Department of Immunology and Cancer Research, Institute for Medical Research Israel Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Eli Pikarsky
- Department of Immunology and Cancer Research, Institute for Medical Research Israel Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel.,Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
110
|
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
|
111
|
Lisak D, Schacht T, Gawlitza A, Albrecht P, Aktas O, Koop B, Gliem M, Hofstetter HH, Zanger K, Bultynck G, Parys JB, De Smedt H, Kindler T, Adams-Quack P, Hahn M, Waisman A, Reed JC, Hövelmeyer N, Methner A. BAX inhibitor-1 is a Ca(2+) channel critically important for immune cell function and survival. Cell Death Differ 2015; 23:358-68. [PMID: 26470731 DOI: 10.1038/cdd.2015.115] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 07/20/2015] [Accepted: 07/21/2015] [Indexed: 01/03/2023] Open
Abstract
The endoplasmic reticulum (ER) serves as the major intracellular Ca(2+) store and has a role in the synthesis and folding of proteins. BAX (BCL2-associated X protein) inhibitor-1 (BI-1) is a Ca(2+) leak channel also implicated in the response against protein misfolding, thereby connecting the Ca(2+) store and protein-folding functions of the ER. We found that BI-1-deficient mice suffer from leukopenia and erythrocytosis, have an increased number of splenic marginal zone B cells and higher abundance and nuclear translocation of NF-κB (nuclear factor-κ light-chain enhancer of activated B cells) proteins, correlating with increased cytosolic and ER Ca(2+) levels. When put into culture, purified knockout T cells and even more so B cells die spontaneously. This is preceded by increased activity of the mitochondrial initiator caspase-9 and correlated with a significant surge in mitochondrial Ca(2+) levels, suggesting an exhausted mitochondrial Ca(2+) buffer capacity as the underlying cause for cell death in vitro. In vivo, T-cell-dependent experimental autoimmune encephalomyelitis and B-cell-dependent antibody production are attenuated, corroborating the ex vivo results. These results suggest that BI-1 has a major role in the functioning of the adaptive immune system by regulating intracellular Ca(2+) homeostasis in lymphocytes.
Collapse
Affiliation(s)
- D Lisak
- Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn) and Department of Neurology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - T Schacht
- Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn) and Department of Neurology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - A Gawlitza
- Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn) and Department of Neurology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - P Albrecht
- Heinrich Heine Universität Düsseldorf, Department of Neurology, Düsseldorf, Germany
| | - O Aktas
- Heinrich Heine Universität Düsseldorf, Department of Neurology, Düsseldorf, Germany
| | - B Koop
- Heinrich Heine Universität Düsseldorf, Department of Neurology, Düsseldorf, Germany
| | - M Gliem
- Heinrich Heine Universität Düsseldorf, Department of Neurology, Düsseldorf, Germany
| | - H H Hofstetter
- Heinrich Heine Universität Düsseldorf, Department of Neurology, Düsseldorf, Germany
| | - K Zanger
- Center for Anatomy and Brain Research, Düsseldorf, Germany
| | - G Bultynck
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, KU Leuven, Belgium
| | - J B Parys
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, KU Leuven, Belgium
| | - H De Smedt
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, KU Leuven, Belgium
| | - T Kindler
- III Medical Clinic, University Medical Center of the Johannes Gutenberg-University of Mainz, Mainz, Germany
| | - P Adams-Quack
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University of Mainz, Mainz, Germany
| | - M Hahn
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University of Mainz, Mainz, Germany
| | - A Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University of Mainz, Mainz, Germany
| | - J C Reed
- Sanford Burnham Institute, La Jolla, CA, USA
| | - N Hövelmeyer
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University of Mainz, Mainz, Germany
| | - A Methner
- Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn) and Department of Neurology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
112
|
Ridder DA, Wenzel J, Müller K, Töllner K, Tong XK, Assmann JC, Stroobants S, Weber T, Niturad C, Fischer L, Lembrich B, Wolburg H, Grand'Maison M, Papadopoulos P, Korpos E, Truchetet F, Rades D, Sorokin LM, Schmidt-Supprian M, Bedell BJ, Pasparakis M, Balschun D, D'Hooge R, Löscher W, Hamel E, Schwaninger M. Brain endothelial TAK1 and NEMO safeguard the neurovascular unit. ACTA ACUST UNITED AC 2015; 212:1529-49. [PMID: 26347470 PMCID: PMC4577837 DOI: 10.1084/jem.20150165] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 08/07/2015] [Indexed: 12/25/2022]
Abstract
Ridder et al. show that deletion of NEMO, a component of NF-kB signaling, in brain endothelial cells results in increased cerebral vascular permeability and endothelial cell death, and recapitulates the neurological symptoms observed in the genetic disease incontinentia pigmenti. Inactivating mutations of the NF-κB essential modulator (NEMO), a key component of NF-κB signaling, cause the genetic disease incontinentia pigmenti (IP). This leads to severe neurological symptoms, but the mechanisms underlying brain involvement were unclear. Here, we show that selectively deleting Nemo or the upstream kinase Tak1 in brain endothelial cells resulted in death of endothelial cells, a rarefaction of brain microvessels, cerebral hypoperfusion, a disrupted blood–brain barrier (BBB), and epileptic seizures. TAK1 and NEMO protected the BBB by activating the transcription factor NF-κB and stabilizing the tight junction protein occludin. They also prevented brain endothelial cell death in a NF-κB–independent manner by reducing oxidative damage. Our data identify crucial functions of inflammatory TAK1–NEMO signaling in protecting the brain endothelium and maintaining normal brain function, thus explaining the neurological symptoms associated with IP.
Collapse
Affiliation(s)
- Dirk A Ridder
- Institute of Experimental and Clinical Pharmacology and Toxicology and Department of Radiation Oncology, University of Lübeck, 23562 Lübeck, Germany
| | - Jan Wenzel
- Institute of Experimental and Clinical Pharmacology and Toxicology and Department of Radiation Oncology, University of Lübeck, 23562 Lübeck, Germany German Research Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Lübeck/Kiel, 23562 Lübeck, Germany
| | - Kristin Müller
- Institute of Experimental and Clinical Pharmacology and Toxicology and Department of Radiation Oncology, University of Lübeck, 23562 Lübeck, Germany
| | - Kathrin Töllner
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany Center for Systems Neuroscience, 30559 Hannover, Germany
| | - Xin-Kang Tong
- Montreal Neurological Institute, McGill University, Montreal QC H3A 0G4, Canada
| | - Julian C Assmann
- Institute of Experimental and Clinical Pharmacology and Toxicology and Department of Radiation Oncology, University of Lübeck, 23562 Lübeck, Germany
| | - Stijn Stroobants
- Laboratory of Biological Psychology, KU Leuven, 3000 Leuven, Belgium
| | - Tobias Weber
- Institute of Experimental and Clinical Pharmacology and Toxicology and Department of Radiation Oncology, University of Lübeck, 23562 Lübeck, Germany
| | - Cristina Niturad
- Institute of Experimental and Clinical Pharmacology and Toxicology and Department of Radiation Oncology, University of Lübeck, 23562 Lübeck, Germany
| | - Lisanne Fischer
- Institute of Experimental and Clinical Pharmacology and Toxicology and Department of Radiation Oncology, University of Lübeck, 23562 Lübeck, Germany
| | - Beate Lembrich
- Institute of Experimental and Clinical Pharmacology and Toxicology and Department of Radiation Oncology, University of Lübeck, 23562 Lübeck, Germany
| | - Hartwig Wolburg
- Institute of Pathology and Neuropathology, University Hospital Tübingen, 72076 Tübingen, Germany
| | | | | | - Eva Korpos
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, 48149 Münster, Germany
| | | | - Dirk Rades
- Institute of Experimental and Clinical Pharmacology and Toxicology and Department of Radiation Oncology, University of Lübeck, 23562 Lübeck, Germany
| | - Lydia M Sorokin
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, 48149 Münster, Germany
| | - Marc Schmidt-Supprian
- Department of Hematology and Oncology, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany
| | - Barry J Bedell
- Montreal Neurological Institute, McGill University, Montreal QC H3A 0G4, Canada
| | | | - Detlef Balschun
- Laboratory of Biological Psychology, KU Leuven, 3000 Leuven, Belgium
| | - Rudi D'Hooge
- Laboratory of Biological Psychology, KU Leuven, 3000 Leuven, Belgium
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany Center for Systems Neuroscience, 30559 Hannover, Germany
| | - Edith Hamel
- Montreal Neurological Institute, McGill University, Montreal QC H3A 0G4, Canada
| | - Markus Schwaninger
- Institute of Experimental and Clinical Pharmacology and Toxicology and Department of Radiation Oncology, University of Lübeck, 23562 Lübeck, Germany German Research Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Lübeck/Kiel, 23562 Lübeck, Germany
| |
Collapse
|
113
|
B-Cell Activating Factor as a Cancer Biomarker and Its Implications in Cancer-Related Cachexia. BIOMED RESEARCH INTERNATIONAL 2015; 2015:792187. [PMID: 26339644 PMCID: PMC4538579 DOI: 10.1155/2015/792187] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 04/28/2015] [Indexed: 01/21/2023]
Abstract
B-cell activating factor (BAFF) is a cytokine and adipokine of the TNF ligand superfamily. The main biological function of BAFF in maintaining the maturation of B-cells to plasma cells has recently made it a target of the first FDA-approved selective BAFF antibody, belimumab, for the therapy of systemic lupus erythematosus. Concomitantly, the role of BAFF in cancer has been a subject of research since its discovery. Here we review BAFF as a biomarker of malignant disease activity and prognostic factor in B-cell derived malignancies such as multiple myeloma. Moreover, anti-BAFF therapy seems to be a promising approach in treatment of B-cell derived leukemias/lymphomas. In nonhematologic solid tumors, BAFF may contribute to cancer progression by mechanisms both dependent on and independent of BAFF's proinflammatory role. We also describe ongoing research into the pathophysiological link between BAFF and cancer-related cachexia. BAFF has been shown to contribute to inflammation and insulin resistance which are known to worsen cancer cachexia syndrome. Taking all the above together, BAFF is emerging as a biomarker of several malignancies and a possible hallmark of cancer cachexia.
Collapse
|
114
|
Lu H, Lei X, Zhang Q. Moderate activation of IKK2-NF-kB in unstressed adult mouse liver induces cytoprotective genes and lipogenesis without apparent signs of inflammation or fibrosis. BMC Gastroenterol 2015. [PMID: 26219821 PMCID: PMC4518658 DOI: 10.1186/s12876-015-0325-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background The NF-kB signaling, regulated by IKK1-p52/RelB and IKK2-p65, is activated by various stresses to protect or damage the liver, in context-specific manners. Two previous studies of liver-specific expression of constitutive active IKK2 (IKK2ca) showed that strong activation of IKK2-NF-kB in mouse livers caused inflammation, insulin resistance, and/or fibrosis. The purpose of this study was to understand how moderate activation of IKK2-NF-kB in adult mouse livers alters hepatic gene expression and pathophysiology. Method We generated mice with adult hepatocyte-specific activation of Ikk2 (Liv-Ikk2ca) using Alb-cre mice and Ikk2ca Rosa26 knockin mice in which a moderate expression of Ikk2ca transgene was driven by the endogenous Rosa26 promoter. Results Surprisingly, compared to wild-type mice, adult male Liv-Ikk2ca mice had higher hepatic mRNA expression of Ikk2 and classical NF-kB targets (e.g. Lcn2 and A20), as well as IKK1, NIK, and RelB, but no changes in markers of inflammation or fibrosis. Blood levels of IL-6 and MCP-1 remained unchanged, and histology analysis showed a lack of injury or infiltration of inflammatory cells in livers of Liv-Ikk2ca mice. Moreover, Liv-Ikk2ca mice had lower mRNA expression of prooxidative enzymes Cyp2e1 and Cyp4a14, higher expression of antioxidative enzymes Sod2, Gpx1, and Nqo1, without changes in key enzymes for fatty acid oxidation, glucose utilization, or gluconeogenesis. In parallel, Liv-Ikk2ca mice and wild-type mice had similar levels of hepatic reduced glutathione, endogenous reactive oxygen species, and lipid peroxidation. Additionally, Liv-Ikk2ca mice had higher Cyp3a11 without down-regulation of most drug processing genes. Regarding nuclear proteins of NF-kB subunits, Liv-Ikk2ca mice had moderately higher p65 and p50 but much higher RelB. Results of ChIP-qPCR showed that the binding of p50 to multiple NF-kB-target genes was markedly increased in Liv-Ikk2ca mice. Additionally, Liv-Ikk2ca mice had moderate increase in triglycerides in liver, which was associated with higher lipogenic factors Pparγ, Lxr, Fasn, Scd1, and CD36. Conclusion In summary, moderate activation of IKK2-NF-kB in unstressed adult mouse hepatocytes produces a cytoprotective gene expression profile and induces lipogenesis without apparent signs of inflammation or fibrosis, likely due to strong activation of the anti-inflammatory IKK1-RelB alternative NF-kB pathway as well as the Lxr. Electronic supplementary material The online version of this article (doi:10.1186/s12876-015-0325-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hong Lu
- Department of Pharmacology, SUNY Upstate Medical University, 750 E Adams ST, Syracuse, NY, 13210, USA.
| | - Xiaohong Lei
- Department of Pharmacology, SUNY Upstate Medical University, 750 E Adams ST, Syracuse, NY, 13210, USA.
| | - Qinghao Zhang
- Department of Pharmacology, SUNY Upstate Medical University, 750 E Adams ST, Syracuse, NY, 13210, USA.
| |
Collapse
|
115
|
Wang S, Charbonnier LM, Noval Rivas M, Georgiev P, Li N, Gerber G, Bry L, Chatila TA. MyD88 Adaptor-Dependent Microbial Sensing by Regulatory T Cells Promotes Mucosal Tolerance and Enforces Commensalism. Immunity 2015; 43:289-303. [PMID: 26231118 DOI: 10.1016/j.immuni.2015.06.014] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 04/20/2015] [Accepted: 06/18/2015] [Indexed: 02/07/2023]
Abstract
Commensal microbiota promote mucosal tolerance in part by engaging regulatory T (Treg) cells via Toll-like receptors (TLRs). We report that Treg-cell-specific deletion of the TLR adaptor MyD88 resulted in deficiency of intestinal Treg cells, a reciprocal increase in T helper 17 (Th17) cells and heightened interleukin-17 (IL-17)-dependent inflammation in experimental colitis. It also precipitated dysbiosis with overgrowth of segmented filamentous bacteria (SFB) and increased microbial loads in deep tissues. The Th17 cell dysregulation and bacterial dysbiosis were linked to impaired anti-microbial intestinal IgA responses, related to defective MyD88 adaptor- and Stat3 transcription factor-dependent T follicular regulatory and helper cell differentiation in the Peyer's patches. These findings establish an essential role for MyD88-dependent microbial sensing by Treg cells in enforcing mucosal tolerance and maintaining commensalism by promoting intestinal Treg cell formation and anti-commensal IgA responses.
Collapse
Affiliation(s)
- Sen Wang
- Division of Immunology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Louis-Marie Charbonnier
- Division of Immunology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Magali Noval Rivas
- Division of Immunology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Peter Georgiev
- Division of Immunology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Ning Li
- Center for Clinical and Translational Metagenomics, Department of Pathology, Brigham & Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Georg Gerber
- Center for Clinical and Translational Metagenomics, Department of Pathology, Brigham & Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Lynn Bry
- Center for Clinical and Translational Metagenomics, Department of Pathology, Brigham & Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Talal A Chatila
- Division of Immunology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
116
|
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
|
117
|
Zeng Q, Zhang H, Qin J, Xu Z, Gui L, Liu B, Liu C, Xu C, Liu W, Zhang S, Huang S, Chen L. Rapamycin inhibits BAFF-stimulated cell proliferation and survival by suppressing mTOR-mediated PP2A-Erk1/2 signaling pathway in normal and neoplastic B-lymphoid cells. Cell Mol Life Sci 2015; 72:4867-84. [PMID: 26118661 DOI: 10.1007/s00018-015-1976-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Revised: 05/18/2015] [Accepted: 06/22/2015] [Indexed: 10/23/2022]
Abstract
B-cell activating factor (BAFF) is involved in not only physiology of normal B cells, but also pathophysiology of aggressive B cells related to malignant and autoimmune diseases. Rapamycin, a lipophilic macrolide antibiotic, has recently shown to be effective in the treatment of human lupus erythematosus. However, how rapamycin inhibits BAFF-stimulated B-cell proliferation and survival has not been fully elucidated. Here, we show that rapamycin inhibited human soluble BAFF (hsBAFF)-induced cell proliferation and survival in normal and B-lymphoid (Raji and Daudi) cells by activation of PP2A and inactivation of Erk1/2. Pretreatment with PD98059, down-regulation of Erk1/2, expression of dominant negative MKK1, or overexpression of wild-type PP2A potentiated rapamycin's suppression of hsBAFF-activated Erk1/2 and B-cell proliferation/viability, whereas expression of constitutively active MKK1, inhibition of PP2A by okadaic acid, or expression of dominant negative PP2A attenuated the inhibitory effects of rapamycin. Furthermore, expression of a rapamycin-resistant and kinase-active mTOR (mTOR-T), but not a rapamycin-resistant and kinase-dead mTOR-T (mTOR-TE), conferred resistance to rapamycin's effects on PP2A, Erk1/2 and B-cell proliferation/viability, implying mTOR-dependent mechanism involved. The findings indicate that rapamycin inhibits BAFF-stimulated cell proliferation/survival by targeting mTOR-mediated PP2A-Erk1/2 signaling pathway in normal and neoplastic B-lymphoid cells. Our data highlight that rapamycin may be exploited for preventing excessive BAFF-induced aggressive B-cell malignancies and autoimmune diseases.
Collapse
Affiliation(s)
- Qingyu Zeng
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Jiangsu Key Laboratory for Microbes and Functional Genomics, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Chixia District, Nanjing, 210023, Jiangsu, People's Republic of China
| | - Hai Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Jiangsu Key Laboratory for Microbes and Functional Genomics, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Chixia District, Nanjing, 210023, Jiangsu, People's Republic of China
| | - Jiamin Qin
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Jiangsu Key Laboratory for Microbes and Functional Genomics, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Chixia District, Nanjing, 210023, Jiangsu, People's Republic of China
| | - Zhigang Xu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Jiangsu Key Laboratory for Microbes and Functional Genomics, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Chixia District, Nanjing, 210023, Jiangsu, People's Republic of China
| | - Lin Gui
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Jiangsu Key Laboratory for Microbes and Functional Genomics, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Chixia District, Nanjing, 210023, Jiangsu, People's Republic of China
| | - Beibei Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Jiangsu Key Laboratory for Microbes and Functional Genomics, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Chixia District, Nanjing, 210023, Jiangsu, People's Republic of China
| | - Chunxiao Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Jiangsu Key Laboratory for Microbes and Functional Genomics, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Chixia District, Nanjing, 210023, Jiangsu, People's Republic of China
| | - Chong Xu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Jiangsu Key Laboratory for Microbes and Functional Genomics, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Chixia District, Nanjing, 210023, Jiangsu, People's Republic of China
| | - Wen Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Jiangsu Key Laboratory for Microbes and Functional Genomics, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Chixia District, Nanjing, 210023, Jiangsu, People's Republic of China
| | - Shuangquan Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Jiangsu Key Laboratory for Microbes and Functional Genomics, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Chixia District, Nanjing, 210023, Jiangsu, People's Republic of China
| | - Shile Huang
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71130-3932, USA. .,Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA, 71130-3932, USA.
| | - Long Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Jiangsu Key Laboratory for Microbes and Functional Genomics, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Chixia District, Nanjing, 210023, Jiangsu, People's Republic of China.
| |
Collapse
|
118
|
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
|
119
|
Walsh NC, Waters LR, Fowler JA, Lin M, Cunningham CR, Brooks DG, Rehg JE, Morse HC, Teitell MA. LKB1 inhibition of NF-κB in B cells prevents T follicular helper cell differentiation and germinal center formation. EMBO Rep 2015; 16:753-68. [PMID: 25916856 PMCID: PMC4467859 DOI: 10.15252/embr.201439505] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 03/06/2015] [Accepted: 03/16/2015] [Indexed: 11/09/2022] Open
Abstract
T-cell-dependent antigenic stimulation drives the differentiation of B cells into antibody-secreting plasma cells and memory B cells, but how B cells regulate this process is unclear. We show that LKB1 expression in B cells maintains B-cell quiescence and prevents the premature formation of germinal centers (GCs). Lkb1-deficient B cells (BKO) undergo spontaneous B-cell activation and secretion of multiple inflammatory cytokines, which leads to splenomegaly caused by an unexpected expansion of T cells. Within this cytokine response, increased IL-6 production results from heightened activation of NF-κB, which is suppressed by active LKB1. Secreted IL-6 drives T-cell activation and IL-21 production, promoting T follicular helper (TFH ) cell differentiation and expansion to support a ~100-fold increase in steady-state GC B cells. Blockade of IL-6 secretion by BKO B cells inhibits IL-21 expression, a known inducer of TFH -cell differentiation and expansion. Together, these data reveal cell intrinsic and surprising cell extrinsic roles for LKB1 in B cells that control TFH -cell differentiation and GC formation, and place LKB1 as a central regulator of T-cell-dependent humoral immunity.
Collapse
Affiliation(s)
- Nicole C Walsh
- Department of Pathology & Laboratory Medicine, University of California, Los Angeles, CA, USA
| | - Lynnea R Waters
- Molecular Biology Institute, University of California, Los Angeles, CA, USA
| | - Jessica A Fowler
- Department of Pathology & Laboratory Medicine, University of California, Los Angeles, CA, USA
| | - Mark Lin
- Department of Pathology & Laboratory Medicine, University of California, Los Angeles, CA, USA
| | - Cameron R Cunningham
- Department of Microbiology, Immunology and Molecular Genetics and UCLA AIDS Institute University of California, Los Angeles, CA, USA
| | - David G Brooks
- Department of Microbiology, Immunology and Molecular Genetics and UCLA AIDS Institute University of California, Los Angeles, CA, USA
| | - Jerold E Rehg
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Herbert C Morse
- Virology and Cellular Immunology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases National Institutes of Health, Rockville, MD, USA
| | - Michael A Teitell
- Department of Pathology & Laboratory Medicine, University of California, Los Angeles, CA, USA Molecular Biology Institute, University of California, Los Angeles, CA, USA Broad Stem Cell Research Center, Departments of Pediatrics and Bioengineering, California NanoSystems Institute, and Jonsson Comprehensive Cancer Center University of California, Los Angeles, CA, USA
| |
Collapse
|
120
|
Jacque E, Schweighoffer E, Tybulewicz VLJ, Ley SC. BAFF activation of the ERK5 MAP kinase pathway regulates B cell survival. ACTA ACUST UNITED AC 2015; 212:883-92. [PMID: 25987726 PMCID: PMC4451135 DOI: 10.1084/jem.20142127] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 04/23/2015] [Indexed: 01/01/2023]
Abstract
B cell activating factor (BAFF) stimulation of the BAFF receptor (BAFF-R) is essential for the homeostatic survival of mature B cells. Earlier in vitro experiments with inhibitors that block MEK 1 and 2 suggested that activation of ERK 1 and 2 MAP kinases is required for BAFF-R to promote B cell survival. However, these inhibitors are now known to also inhibit MEK5, which activates the related MAP kinase ERK5. In the present study, we demonstrated that BAFF-induced B cell survival was actually independent of ERK1/2 activation but required ERK5 activation. Consistent with this, we showed that conditional deletion of ERK5 in B cells led to a pronounced global reduction in mature B2 B cell numbers, which correlated with impaired survival of ERK5-deficient B cells after BAFF stimulation. ERK5 was required for optimal BAFF up-regulation of Mcl1 and Bcl2a1, which are prosurvival members of the Bcl-2 family. However, ERK5 deficiency did not alter BAFF activation of the PI3-kinase-Akt or NF-κB signaling pathways, which are also important for BAFF to promote mature B cell survival. Our study reveals a critical role for the MEK5-ERK5 MAP kinase signaling pathway in BAFF-induced mature B cell survival and homeostatic maintenance of B2 cell numbers.
Collapse
Affiliation(s)
- Emilie Jacque
- Division of Immune Cell Biology, MRC National Institute for Medical Research London, London NW7 1AA, England, UK
| | - Edina Schweighoffer
- Division of Immune Cell Biology, MRC National Institute for Medical Research London, London NW7 1AA, England, UK
| | - Victor L J Tybulewicz
- Division of Immune Cell Biology, MRC National Institute for Medical Research London, London NW7 1AA, England, UK
| | - Steven C Ley
- Division of Immune Cell Biology, MRC National Institute for Medical Research London, London NW7 1AA, England, UK
| |
Collapse
|
121
|
Zhang B, Calado DP, Wang Z, Fröhler S, Köchert K, Qian Y, Koralov SB, Schmidt-Supprian M, Sasaki Y, Unitt C, Rodig S, Chen W, Dalla-Favera R, Alt FW, Pasqualucci L, Rajewsky K. An oncogenic role for alternative NF-κB signaling in DLBCL revealed upon deregulated BCL6 expression. Cell Rep 2015; 11:715-26. [PMID: 25921526 PMCID: PMC4426003 DOI: 10.1016/j.celrep.2015.03.059] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 01/29/2015] [Accepted: 03/24/2015] [Indexed: 12/31/2022] Open
Abstract
Diffuse large B cell lymphoma (DLBCL) is a complex disease comprising diverse subtypes and genetic profiles. Possibly because of the prevalence of genetic alterations activating canonical NF-κB activity, a role for oncogenic lesions that activate the alternative NF-κB pathway in DLBCL has remained elusive. Here, we show that deletion/mutation of TRAF3, a negative regulator of the alternative NF-κB pathway, occurs in ∼15% of DLBCLs and that it often coexists with BCL6 translocation, which prevents terminal B cell differentiation. Accordingly, in a mouse model constitutive activation of the alternative NF-κB pathway cooperates with BCL6 deregulation in DLBCL development. This work demonstrates a key oncogenic role for the alternative NF-κB pathway in DLBCL development.
Collapse
Affiliation(s)
- Baochun Zhang
- Program of Cellular and Molecular Medicine, Children's Hospital, and Immune Disease Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA.
| | - Dinis Pedro Calado
- Program of Cellular and Molecular Medicine, Children's Hospital, and Immune Disease Institute, Harvard Medical School, Boston, MA 02115, USA; Max Delbrück Center for Molecular Medicine, Robert-Rössle-Str 10, Berlin 13125, Germany; Cancer Research UK, London Research Institute, London WC2A 3LY, UK; Peter Gorer Department of Immunobiology, Kings College London, London SE1 9RT, UK.
| | - Zhe Wang
- Program of Cellular and Molecular Medicine, Children's Hospital, and Immune Disease Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Sebastian Fröhler
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Str 10, Berlin 13125, Germany
| | - Karl Köchert
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Str 10, Berlin 13125, Germany
| | - Yu Qian
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Sergei B Koralov
- Program of Cellular and Molecular Medicine, Children's Hospital, and Immune Disease Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Marc Schmidt-Supprian
- Program of Cellular and Molecular Medicine, Children's Hospital, and Immune Disease Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Hematology and Oncology, Klinikum rechts der Isar, Technische Universität München, Ismaninger Strasse 22, Munich 81675, Germany
| | - Yoshiteru Sasaki
- Program of Cellular and Molecular Medicine, Children's Hospital, and Immune Disease Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Molecular and Cellular Physiology, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Christine Unitt
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Scott Rodig
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Wei Chen
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Str 10, Berlin 13125, Germany
| | - Riccardo Dalla-Favera
- Institute for Cancer Genetics and the Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10032, USA; Department of Pathology & Cell Biology, Columbia University, New York, NY 10032, USA
| | - Frederick W Alt
- Howard Hughes Medical Institute, Program in Cellular and Molecular Medicine, Boston Children's Hospital, and Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Laura Pasqualucci
- Institute for Cancer Genetics and the Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10032, USA; Department of Pathology & Cell Biology, Columbia University, New York, NY 10032, USA
| | - Klaus Rajewsky
- Program of Cellular and Molecular Medicine, Children's Hospital, and Immune Disease Institute, Harvard Medical School, Boston, MA 02115, USA; Max Delbrück Center for Molecular Medicine, Robert-Rössle-Str 10, Berlin 13125, Germany.
| |
Collapse
|
122
|
Xu Y, Evaristo C, Alegre ML, Gurbuxani S, Kee BL. Analysis of GzmbCre as a Model System for Gene Deletion in the Natural Killer Cell Lineage. PLoS One 2015; 10:e0125211. [PMID: 25923440 PMCID: PMC4414598 DOI: 10.1371/journal.pone.0125211] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Accepted: 03/23/2015] [Indexed: 12/15/2022] Open
Abstract
The analysis of gene function in mature and activated natural killer cells has been hampered by the lack of model systems for Cre-mediated recombination in these cells. Here we have investigated the utility of GzmbCre for recombination of loxp sequences in these cells predicated on the observation that Gzmb mRNA is highly expressed in mature and activated natural killer cells. Using two different reporter strains we determined that gene function could be investigated in mature natural killer cells after GzmbCre mediated recombination in vitro in conditions that lead to natural killer cell activation such as in the cytokine combination of interleukin 2 and interleukin 12. We demonstrated the utility of this model by creating GzmbCre;Rosa26IKKbca mice in which Cre-mediated recombination resulted in expression of constitutively active IKKβ, which results in activation of the NFκB transcription factor. In vivo and in vitro activation of IKKβ in natural killer cells revealed that constitutive activation of this pathway leads to natural killer cell hyper-activation and altered morphology. As a caveat to the use of GzmbCre we found that this transgene can lead to recombination in all hematopoietic cells the extent of which varies with the particular loxp flanked allele under investigation. We conclude that GzmbCre can be used under some conditions to investigate gene function in mature and activated natural killer cells.
Collapse
Affiliation(s)
- Yiying Xu
- Committee on Molecular Pathogenesis and Molecular Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - Cesar Evaristo
- Committee on Immunology, University of Chicago, Chicago, Illinois, United States of America
- Department of Medicine, Section of Rhuematology, University of Chicago, Chicago, Illinois, United States of America
| | - Maria-Luisa Alegre
- Committee on Molecular Pathogenesis and Molecular Medicine, University of Chicago, Chicago, Illinois, United States of America
- Committee on Immunology, University of Chicago, Chicago, Illinois, United States of America
- Department of Medicine, Section of Rhuematology, University of Chicago, Chicago, Illinois, United States of America
| | - Sandeep Gurbuxani
- Department of Pathology, University of Chicago, Chicago, Illinois, United States of America
| | - Barbara L. Kee
- Committee on Molecular Pathogenesis and Molecular Medicine, University of Chicago, Chicago, Illinois, United States of America
- Committee on Immunology, University of Chicago, Chicago, Illinois, United States of America
- Committee on Cancer Biology, University of Chicago, Chicago, Illinois, United States of America
- Department of Pathology, University of Chicago, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
123
|
Heger K, Kober M, Rieß D, Drees C, de Vries I, Bertossi A, Roers A, Sixt M, Schmidt-Supprian M. A novel Cre recombinase reporter mouse strain facilitates selective and efficient infection of primary immune cells with adenoviral vectors. Eur J Immunol 2015; 45:1614-20. [DOI: 10.1002/eji.201545457] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 02/16/2015] [Accepted: 03/13/2015] [Indexed: 12/21/2022]
Affiliation(s)
- Klaus Heger
- Hematology and Oncology; Klinikum rechts der Isar, Technische Universität München; Munich Germany
- Max Planck Institute of Biochemistry; Martinsried Germany
| | - Maike Kober
- Hematology and Oncology; Klinikum rechts der Isar, Technische Universität München; Munich Germany
- Max Planck Institute of Biochemistry; Martinsried Germany
| | - David Rieß
- Hematology and Oncology; Klinikum rechts der Isar, Technische Universität München; Munich Germany
- Max Planck Institute of Biochemistry; Martinsried Germany
| | - Christoph Drees
- Hematology and Oncology; Klinikum rechts der Isar, Technische Universität München; Munich Germany
| | - Ingrid de Vries
- Institute of Science and Technology Austria; Klosterneuburg Austria
| | | | - Axel Roers
- Institute for Immunology; Technische Universität Dresden; Dresden Germany
| | - Michael Sixt
- Institute of Science and Technology Austria; Klosterneuburg Austria
| | - Marc Schmidt-Supprian
- Hematology and Oncology; Klinikum rechts der Isar, Technische Universität München; Munich Germany
- Max Planck Institute of Biochemistry; Martinsried Germany
| |
Collapse
|
124
|
Deficiency of the B cell-activating factor receptor results in limited CD169+ macrophage function during viral infection. J Virol 2015; 89:4748-59. [PMID: 25673724 DOI: 10.1128/jvi.02976-14] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2014] [Accepted: 02/04/2015] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED The B cell-activating factor (BAFF) is critical for B cell development and humoral immunity in mice and humans. While the role of BAFF in B cells has been widely described, its role in innate immunity remains unknown. Using BAFF receptor (BAFFR)-deficient mice, we characterized BAFFR-related innate and adaptive immune functions following infection with vesicular stomatitis virus (VSV) and lymphocytic choriomeningitis virus (LCMV). We identified a critical role for BAFFR signaling in the generation and maintenance of the CD169(+) macrophage compartment. Consequently, Baffr(-) (/) (-) mice exhibited limited induction of innate type I interferon production after viral infection. Lack of BAFFR signaling reduced virus amplification and presentation following viral infection, resulting in highly reduced antiviral adaptive immune responses. As a consequence, BAFFR-deficient mice showed exacerbated and fatal disease after viral infection. Mechanistically, transient lack of B cells in Baffr(-) (/) (-) animals resulted in limited lymphotoxin expression, which is critical for maintenance of CD169(+) cells. In conclusion, BAFFR signaling affects both innate and adaptive immune activation during viral infections. IMPORTANCE Viruses cause acute and chronic infections in humans resulting in millions of deaths every year. Innate immunity is critical for the outcome of a viral infection. Innate type I interferon production can limit viral replication, while adaptive immune priming by innate immune cells induces pathogen-specific immunity with long-term protection. Here, we show that BAFFR deficiency not only perturbed B cells, but also resulted in limited CD169(+) macrophages. These macrophages are critical in amplifying viral particles to trigger type I interferon production and initiate adaptive immune priming. Consequently, BAFFR deficiency resulted in reduced enforced viral replication, limited type I interferon production, and reduced adaptive immunity compared to BAFFR-competent controls. As a result, BAFFR-deficient mice were predisposed to fatal viral infections. Thus, BAFFR expression is critical for innate immune activation and antiviral immunity.
Collapse
|
125
|
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
|
126
|
Control of the inheritance of regulatory T cell identity by a cis element in the Foxp3 locus. Cell 2014; 158:749-763. [PMID: 25126783 DOI: 10.1016/j.cell.2014.07.031] [Citation(s) in RCA: 316] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 06/30/2014] [Accepted: 07/25/2014] [Indexed: 01/09/2023]
Abstract
In multicellular organisms, specialized functions are delegated to distinct cell types whose identity and functional integrity are maintained upon challenge. However, little is known about the mechanisms enabling lineage inheritance and their biological implications. Regulatory T (Treg) cells, which express the transcription factor Foxp3, suppress fatal autoimmunity throughout the lifespan of animals. Here, we show that a dedicated Foxp3 intronic element CNS2 maintains Treg cell lineage identity by acting as a sensor of the essential Treg cell growth factor IL-2 and its downstream target STAT5. CNS2 sustains Foxp3 expression during division of mature Treg cells when IL-2 is limiting and counteracts proinflammatory cytokine signaling that leads to the loss of Foxp3. CNS2-mediated stable inheritance of Foxp3 expression is critical for adequate suppression of diverse types of chronic inflammation by Treg cells and prevents their differentiation into inflammatory effector cells. The described mechanism may represent a general principle of the inheritance of differentiated cell states.
Collapse
|
127
|
Tsaousidou E, Paeger L, Belgardt BF, Pal M, Wunderlich CM, Brönneke H, Collienne U, Hampel B, Wunderlich FT, Schmidt-Supprian M, Kloppenburg P, Brüning JC. Distinct Roles for JNK and IKK Activation in Agouti-Related Peptide Neurons in the Development of Obesity and Insulin Resistance. Cell Rep 2014; 9:1495-506. [PMID: 25456138 DOI: 10.1016/j.celrep.2014.10.045] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 07/10/2014] [Accepted: 10/16/2014] [Indexed: 01/01/2023] Open
Abstract
Activation of c-Jun N-terminal kinase 1 (JNK1)- and inhibitor of nuclear factor kappa-B kinase 2 (IKK2)-dependent signaling plays a crucial role in the development of obesity-associated insulin and leptin resistance not only in peripheral tissues but also in the CNS. Here, we demonstrate that constitutive JNK activation in agouti-related peptide (AgRP)-expressing neurons of the hypothalamus is sufficient to induce weight gain and adiposity in mice as a consequence of hyperphagia. JNK activation increases spontaneous action potential firing of AgRP cells and causes both neuronal and systemic leptin resistance. Similarly, activation of IKK2 signaling in AgRP neurons also increases firing of these cells but fails to cause obesity and leptin resistance. In contrast to JNK activation, IKK2 activation blunts insulin signaling in AgRP neurons and impairs systemic glucose homeostasis. Collectively, these experiments reveal both overlapping and nonredundant effects of JNK- and IKK-dependent signaling in AgRP neurons, which cooperate in the manifestation of the metabolic syndrome.
Collapse
Affiliation(s)
- Eva Tsaousidou
- Department of Mouse Genetics and Metabolism, Institute for Genetics, Cologne 50674, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), Cologne 50931, Germany; Center for Molecular Medicine (CMMC), University of Cologne, Cologne 50931, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Cologne 50937, Germany; Max Planck Institute for Metabolism Research, Cologne 50931, Germany
| | - Lars Paeger
- Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), Cologne 50931, Germany; Institute for Zoology, University of Cologne, Cologne 50674, Germany
| | - Bengt F Belgardt
- Department of Mouse Genetics and Metabolism, Institute for Genetics, Cologne 50674, Germany
| | - Martin Pal
- Baker IDI Heart and Diabetes Institute, Cellular and Molecular Metabolism Laboratory, Melbourne, VIC 3004, Australia
| | - Claudia M Wunderlich
- Department of Mouse Genetics and Metabolism, Institute for Genetics, Cologne 50674, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), Cologne 50931, Germany; Center for Molecular Medicine (CMMC), University of Cologne, Cologne 50931, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Cologne 50937, Germany; Max Planck Institute for Metabolism Research, Cologne 50931, Germany
| | - Hella Brönneke
- Mouse Phenotyping Core Facility, Cologne Excellence Cluster on Cellular Stress Responses, Cologne 50931, Germany
| | - Ursel Collienne
- Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), Cologne 50931, Germany; Institute for Zoology, University of Cologne, Cologne 50674, Germany
| | - Brigitte Hampel
- Department of Mouse Genetics and Metabolism, Institute for Genetics, Cologne 50674, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), Cologne 50931, Germany; Center for Molecular Medicine (CMMC), University of Cologne, Cologne 50931, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Cologne 50937, Germany; Max Planck Institute for Metabolism Research, Cologne 50931, Germany
| | - F Thomas Wunderlich
- Department of Mouse Genetics and Metabolism, Institute for Genetics, Cologne 50674, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), Cologne 50931, Germany; Center for Molecular Medicine (CMMC), University of Cologne, Cologne 50931, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Cologne 50937, Germany; Max Planck Institute for Metabolism Research, Cologne 50931, Germany
| | - Marc Schmidt-Supprian
- Department of Hematology and Oncology, Klinikum Rechts der Isar, Technical University of Munich, Munich 81675, Germany
| | - Peter Kloppenburg
- Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), Cologne 50931, Germany; Institute for Zoology, University of Cologne, Cologne 50674, Germany
| | - Jens C Brüning
- Department of Mouse Genetics and Metabolism, Institute for Genetics, Cologne 50674, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), Cologne 50931, Germany; Center for Molecular Medicine (CMMC), University of Cologne, Cologne 50931, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Cologne 50937, Germany; Max Planck Institute for Metabolism Research, Cologne 50931, Germany.
| |
Collapse
|
128
|
FOXP1 directly represses transcription of proapoptotic genes and cooperates with NF-κB to promote survival of human B cells. Blood 2014; 124:3431-40. [PMID: 25267198 DOI: 10.1182/blood-2014-01-553412] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The forkhead transcription factor FOXP1 is involved in B-cell development and function and is generally regarded as an oncogene in activated B-cell-like subtype of diffuse large B-cell lymphoma (DLBCL) and mucosa-associated lymphoid tissue lymphoma, lymphomas relying on constitutive nuclear factor κB (NF-κB) activity for survival. However, the mechanism underlying its putative oncogenic activity has not been established. By gene expression microarray, upon overexpression or silencing of FOXP1 in primary human B cells and DLBCL cell lines, combined with chromatin immunoprecipitation followed by next-generation sequencing, we established that FOXP1 directly represses a set of 7 proapoptotic genes. Low expression of these genes, encoding the BH3-only proteins BIK and Harakiri, the p53-regulatory proteins TP63, RASSF6, and TP53INP1, and AIM2 and EAF2, is associated with poor survival in DLBCL patients. In line with these findings, we demonstrated that FOXP1 promotes the expansion of primary mature human B cells by inhibiting caspase-dependent apoptosis, without affecting B-cell proliferation. Furthermore, FOXP1 is dependent upon, and cooperates with, NF-κB signaling to promote B-cell expansion and survival. Taken together, our data indicate that, through direct repression of proapoptotic genes, (aberrant) expression of FOXP1 complements (constitutive) NF-κB activity to promote B-cell survival and can thereby contribute to B-cell homeostasis and lymphomagenesis.
Collapse
|
129
|
IκB kinase 2 is essential for IgE-induced mast cell de novo cytokine production but not for degranulation. Cell Rep 2014; 8:1300-7. [PMID: 25176657 DOI: 10.1016/j.celrep.2014.07.046] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 06/06/2014] [Accepted: 07/25/2014] [Indexed: 01/12/2023] Open
Abstract
The immunoglobulin E (IgE)-mediated mast cell (MC) response is central to the pathogenesis of type I allergy and asthma. IκB kinase 2 (IKK2) was reported to couple IgE-induced signals to MC degranulation by phosphorylating the SNARE protein SNAP23. We investigated MC responses in mice with MC-specific inactivation of IKK2 or NF-κB essential modulator (NEMO), or animals with MC-specific expression of a mutant, constitutively active IKK2. We show that the IgE-induced late-phase cytokine response is reduced in mice lacking IKK2 or NEMO in MCs. However, anaphylactic in vivo responses of these animals are not different from those of control mice, and in vitro IKK2-deficient MCs readily phosphorylate SNAP23 and degranulate similarly to control cells in response to allergen or calcium ionophore. Constitutive overactivation of the NF-κB pathway has only slight effects on allergen-triggered MC responses. Thus, IKK2 is dispensable for MC degranulation, and the important question how IgE-induced signals trigger MC vesicle fusion remains open.
Collapse
|
130
|
Dannappel M, Vlantis K, Kumari S, Polykratis A, Kim C, Wachsmuth L, Eftychi C, Lin J, Corona T, Hermance N, Zelic M, Kirsch P, Basic M, Bleich A, Kelliher M, Pasparakis M. RIPK1 maintains epithelial homeostasis by inhibiting apoptosis and necroptosis. Nature 2014; 513:90-4. [PMID: 25132550 DOI: 10.1038/nature13608] [Citation(s) in RCA: 539] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 06/25/2014] [Indexed: 11/09/2022]
Abstract
Necroptosis has emerged as an important pathway of programmed cell death in embryonic development, tissue homeostasis, immunity and inflammation. RIPK1 is implicated in inflammatory and cell death signalling and its kinase activity is believed to drive RIPK3-mediated necroptosis. Here we show that kinase-independent scaffolding RIPK1 functions regulate homeostasis and prevent inflammation in barrier tissues by inhibiting epithelial cell apoptosis and necroptosis. Intestinal epithelial cell (IEC)-specific RIPK1 knockout caused IEC apoptosis, villus atrophy, loss of goblet and Paneth cells and premature death in mice. This pathology developed independently of the microbiota and of MyD88 signalling but was partly rescued by TNFR1 (also known as TNFRSF1A) deficiency. Epithelial FADD ablation inhibited IEC apoptosis and prevented the premature death of mice with IEC-specific RIPK1 knockout. However, mice lacking both RIPK1 and FADD in IECs displayed RIPK3-dependent IEC necroptosis, Paneth cell loss and focal erosive inflammatory lesions in the colon. Moreover, a RIPK1 kinase inactive knock-in delayed but did not prevent inflammation caused by FADD deficiency in IECs or keratinocytes, showing that RIPK3-dependent necroptosis of FADD-deficient epithelial cells only partly requires RIPK1 kinase activity. Epidermis-specific RIPK1 knockout triggered keratinocyte apoptosis and necroptosis and caused severe skin inflammation that was prevented by RIPK3 but not FADD deficiency. These findings revealed that RIPK1 inhibits RIPK3-mediated necroptosis in keratinocytes in vivo and identified necroptosis as a more potent trigger of inflammation compared with apoptosis. Therefore, RIPK1 is a master regulator of epithelial cell survival, homeostasis and inflammation in the intestine and the skin.
Collapse
Affiliation(s)
- Marius Dannappel
- 1] Institute for Genetics, Centre for Molecular Medicine (CMMC), and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany [2]
| | - Katerina Vlantis
- 1] Institute for Genetics, Centre for Molecular Medicine (CMMC), and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany [2]
| | - Snehlata Kumari
- 1] Institute for Genetics, Centre for Molecular Medicine (CMMC), and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany [2]
| | - Apostolos Polykratis
- 1] Institute for Genetics, Centre for Molecular Medicine (CMMC), and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany [2]
| | - Chun Kim
- Institute for Genetics, Centre for Molecular Medicine (CMMC), and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany
| | - Laurens Wachsmuth
- Institute for Genetics, Centre for Molecular Medicine (CMMC), and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany
| | - Christina Eftychi
- Institute for Genetics, Centre for Molecular Medicine (CMMC), and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany
| | - Juan Lin
- Institute for Genetics, Centre for Molecular Medicine (CMMC), and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany
| | - Teresa Corona
- Institute for Genetics, Centre for Molecular Medicine (CMMC), and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany
| | - Nicole Hermance
- Department of Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | - Matija Zelic
- Department of Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | - Petra Kirsch
- Tierforschungszentrum, University of Ulm, Albert-Einstein-Allee 11, D-89081 Ulm, Germany
| | - Marijana Basic
- Institute for Laboratory Animal Science, Hannover Medical School, D-30625 Hannover, Germany
| | - Andre Bleich
- Institute for Laboratory Animal Science, Hannover Medical School, D-30625 Hannover, Germany
| | - Michelle Kelliher
- 1] Department of Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA [2]
| | - Manolis Pasparakis
- 1] Institute for Genetics, Centre for Molecular Medicine (CMMC), and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany [2]
| |
Collapse
|
131
|
Kai X, Chellappa V, Donado C, Reyon D, Sekigami Y, Ataca D, Louissaint A, Mattoo H, Joung JK, Pillai S. IκB kinase β (IKBKB) mutations in lymphomas that constitutively activate canonical nuclear factor κB (NFκB) signaling. J Biol Chem 2014; 289:26960-26972. [PMID: 25107905 DOI: 10.1074/jbc.m114.598763] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Somatic mutations altering lysine 171 of the IKBKB gene that encodes (IKKβ), the critical activating kinase in canonical (NFκB) signaling, have been described in splenic marginal zone lymphomas and multiple myeloma. Lysine 171 forms part of a cationic pocket that interacts with the activation loop phosphate in the activated wild type kinase. We show here that K171E IKKβ and K171T IKKβ represent kinases that are constitutively active even in the absence of activation loop phosphorylation. Predictive modeling and biochemical studies establish why mutations in a positively charged residue in the cationic pocket of an activation loop phosphorylation-dependent kinase result in constitutive activation. Transcription activator-like effector nuclease-based knock-in mutagenesis provides evidence from a B lymphoid context that K171E IKKβ contributes to lymphomagenesis.
Collapse
Affiliation(s)
- Xin Kai
- Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02129
| | - Vasant Chellappa
- Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02129
| | - Carlos Donado
- Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02129
| | - Deepak Reyon
- Molecular Pathology Unit, Massachusetts General Hospital, Boston, Massachusetts 02129, and
| | - Yurie Sekigami
- Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02129
| | - Dalya Ataca
- Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02129
| | - Abner Louissaint
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114
| | - Hamid Mattoo
- Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02129
| | - J Keith Joung
- Molecular Pathology Unit, Massachusetts General Hospital, Boston, Massachusetts 02129, and; Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114
| | - Shiv Pillai
- Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02129,.
| |
Collapse
|
132
|
Luo W, Mayeux J, Gutierrez T, Russell L, Getahun A, Müller J, Tedder T, Parnes J, Rickert R, Nitschke L, Cambier J, Satterthwaite AB, Garrett-Sinha LA. A balance between B cell receptor and inhibitory receptor signaling controls plasma cell differentiation by maintaining optimal Ets1 levels. THE JOURNAL OF IMMUNOLOGY 2014; 193:909-920. [PMID: 24929000 DOI: 10.4049/jimmunol.1400666] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Signaling through the BCR can drive B cell activation and contribute to B cell differentiation into Ab-secreting plasma cells. The positive BCR signal is counterbalanced by a number of membrane-localized inhibitory receptors that limit B cell activation and plasma cell differentiation. Deficiencies in these negative signaling pathways may cause autoantibody generation and autoimmune disease in both animal models and human patients. We have previously shown that the transcription factor Ets1 can restrain B cell differentiation into plasma cells. In this study, we tested the roles of the BCR and inhibitory receptors in controlling the expression of Ets1 in mouse B cells. We found that Ets1 is downregulated in B cells by BCR or TLR signaling through a pathway dependent on PI3K, Btk, IKK2, and JNK. Deficiencies in inhibitory pathways, such as a loss of the tyrosine kinase Lyn, the phosphatase Src homology region 2 domain-containing phosphatase 1 (SHP1) or membrane receptors CD22 and/or Siglec-G, result in enhanced BCR signaling and decreased Ets1 expression. Restoring Ets1 expression in Lyn- or SHP1-deficient B cells inhibits their enhanced plasma cell differentiation. Our findings indicate that downregulation of Ets1 occurs in response to B cell activation via either BCR or TLR signaling, thereby allowing B cell differentiation and that the maintenance of Ets1 expression is an important function of the inhibitory Lyn → CD22/SiglecG → SHP1 pathway in B cells.
Collapse
Affiliation(s)
- Wei Luo
- Department of Biochemistry, Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, NY 14203
| | - Jessica Mayeux
- Department of Biochemistry, Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, NY 14203
| | - Toni Gutierrez
- Department of Biochemistry, Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, NY 14203
| | - Lisa Russell
- Department of Biochemistry, Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, NY 14203
| | - Andrew Getahun
- Department of Biochemistry, Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, NY 14203
| | - Jennifer Müller
- Department of Biochemistry, Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, NY 14203
| | - Thomas Tedder
- Department of Biochemistry, Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, NY 14203
| | - Jane Parnes
- Department of Biochemistry, Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, NY 14203
| | - Robert Rickert
- Department of Biochemistry, Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, NY 14203
| | - Lars Nitschke
- Department of Biochemistry, Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, NY 14203
| | - John Cambier
- Department of Biochemistry, Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, NY 14203
| | - Anne B Satterthwaite
- Department of Biochemistry, Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, NY 14203
| | - Lee Ann Garrett-Sinha
- Department of Biochemistry, Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, NY 14203
| |
Collapse
|
133
|
Abstract
T cells are essential for immune defenses against pathogens, such that viability of naïve T cells before antigen encounter is critical to preserve a polyclonal repertoire and prevent immunodeficiencies. The viability of naïve T cells before antigen recognition is ensured by IL-7, which drives expression of the prosurvival factor Bcl-2. Quiescent naïve T cells have low basal activity of the transcription factor NF-κB, which was assumed to have no functional consequences. In contrast to this postulate, our data show that basal nuclear NF-κB activity plays an important role in the transcription of IL-7 receptor α-subunit (CD127), enabling responsiveness of naïve T cells to the prosurvival effects of IL-7 and allowing T-cell persistence in vivo. Moreover, we show that this property of basal NF-κB activity is shared by mouse and human naïve T cells. Thus, NF-κB drives a distinct transcriptional program in T cells before antigen encounter by controlling susceptibility to IL-7. Our results reveal an evolutionarily conserved role of NF-κB in T cells before antigenic stimulation and identify a novel molecular pathway that controls T-cell homeostasis.
Collapse
|
134
|
B cell transcription factors: Potential new therapeutic targets for SLE. Clin Immunol 2014; 152:140-51. [DOI: 10.1016/j.clim.2014.03.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 03/14/2014] [Accepted: 03/18/2014] [Indexed: 02/06/2023]
|
135
|
Hematopoietic overexpression of FOG1 does not affect B-cells but reduces the number of circulating eosinophils. PLoS One 2014; 9:e92836. [PMID: 24747299 PMCID: PMC3991581 DOI: 10.1371/journal.pone.0092836] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2012] [Accepted: 02/26/2014] [Indexed: 12/31/2022] Open
Abstract
We have identified expression of the gene encoding the transcriptional coactivator FOG-1 (Friend of GATA-1; Zfpm1, Zinc finger protein multitype 1) in B lymphocytes. We found that FOG-1 expression is directly or indirectly dependent on the B cell-specific coactivator OBF-1 and that it is modulated during B cell development: expression is observed in early but not in late stages of B cell development. To directly test in vivo the role of FOG-1 in B lymphocytes, we developed a novel embryonic stem cell recombination system. For this, we combined homologous recombination with the FLP recombinase activity to rapidly generate embryonic stem cell lines carrying a Cre-inducible transgene at the Rosa26 locus. Using this system, we successfully generated transgenic mice where FOG-1 is conditionally overexpressed in mature B-cells or in the entire hematopoietic system. While overexpression of FOG-1 in B cells did not significantly affect B cell development or function, we found that enforced expression of FOG-1 throughout all hematopoietic lineages led to a reduction in the number of circulating eosinophils, confirming and extending to mammals the known function of FOG-1 in this lineage.
Collapse
|
136
|
Uslu K, Coleman AS, Allman WR, Katsenelson N, Bram RJ, Alugupalli KR, Akkoyunlu M. Impaired B Cell Receptor Signaling Is Responsible for Reduced TACI Expression and Function in X-Linked Immunodeficient Mice. THE JOURNAL OF IMMUNOLOGY 2014; 192:3582-95. [DOI: 10.4049/jimmunol.1203468] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
137
|
Constitutive activation of IKK2/NF-κB impairs osteogenesis and skeletal development. PLoS One 2014; 9:e91421. [PMID: 24618907 PMCID: PMC3949987 DOI: 10.1371/journal.pone.0091421] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 02/11/2014] [Indexed: 12/17/2022] Open
Abstract
Pathologic conditions impair bone homeostasis. The transcription factor NF-κB regulates bone homeostasis and is central to bone pathologies. Whereas contribution of NF-κB to heightened osteoclast activity is well-documented, the mechanisms underlying NF-κB impact on chondrocytes and osteoblasts are scarce. In this study, we examined the effect of constitutively active IKK2 (IKK2ca) on chondrogenic and osteogenic differentiation. We show that retroviral IKK2ca but not GFP, IKK2WT, or the inactive IKK2 forms IKK2KM and IKK2SSAA, strongly suppressed osteogenesis and chondrogenesis, in vitro. In order to explore the effect of constitutive NF-κB activation on bone formation in vivo, we activated this pathway in a conditional fashion. Specifically, we crossed the R26StopIKK2ca mice with mice carrying the Col2-cre in order to express IKK2ca in osteoblasts and chondrocytes. Both chondrocytes and osteoblasts derived from Col2Cre/IKK2ca expressed IKK2ca. Mice were born alive yet died shortly thereafter. Histologically, newborn Col2Cre+/RosaIKK2ca heterozygotes (Cre+IKK2ca_w/f (het)) and homozygotes (Cre+IKK2ca_f/f (KI)) showed smaller skeleton, deformed vertebrate and reduced or missing digit ossification. The width of neural arches, as well as ossification in vertebral bodies of Cre+IKK2ca_w/f and Cre+IKK2ca_f/f, was reduced or diminished. H&E staining of proximal tibia from new born pups revealed that Cre+IKK2ca_f/f displayed disorganized hypertrophic zones within the smaller epiphysis. Micro-CT analysis indicated that 4-wk old Cre+IKK2ca_w/f has abnormal trabecular bone in proximal tibia compared to WT littermates. Mechanistically, ex-vivo experiments showed that expression of differentiation markers in calvarial osteoblasts derived from newborn IKK2ca knock-in mice was diminished compared to WT-derived cells. In situ hybridization studies demonstrated that the hypertrophic chondrocyte marker type-X collagen, the pre-hypertrophic chondrocyte markers Indian hedgehog and alkaline phosphatase, and the early markers Aggrecan and type-II collagen were reduced in Cre+IKK2ca_w/f and Cre+IKK2ca_f/f mice. Altogether, the in-vitro, in vivo and ex-vivo evidence suggest that IKK2ca perturbs osteoblast and chondrocyte maturation and impairs skeletal development.
Collapse
|
138
|
He WA, Berardi E, Cardillo VM, Acharyya S, Aulino P, Thomas-Ahner J, Wang J, Bloomston M, Muscarella P, Nau P, Shah N, Butchbach MER, Ladner K, Adamo S, Rudnicki MA, Keller C, Coletti D, Montanaro F, Guttridge DC. NF-κB-mediated Pax7 dysregulation in the muscle microenvironment promotes cancer cachexia. J Clin Invest 2014; 123:4821-35. [PMID: 24084740 DOI: 10.1172/jci68523] [Citation(s) in RCA: 271] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2012] [Accepted: 08/06/2013] [Indexed: 01/09/2023] Open
Abstract
Cachexia is a debilitating condition characterized by extreme skeletal muscle wasting that contributes significantly to morbidity and mortality. Efforts to elucidate the underlying mechanisms of muscle loss have predominantly focused on events intrinsic to the myofiber. In contrast, less regard has been given to potential contributory factors outside the fiber within the muscle microenvironment. In tumor-bearing mice and patients with pancreatic cancer, we found that cachexia was associated with a type of muscle damage resulting in activation of both satellite and nonsatellite muscle progenitor cells. These muscle progenitors committed to a myogenic program, but were inhibited from completing differentiation by an event linked with persistent expression of the self-renewing factor Pax7. Overexpression of Pax7 was sufficient to induce atrophy in normal muscle, while under tumor conditions, the reduction of Pax7 or exogenous addition of its downstream target, MyoD, reversed wasting by restoring cell differentiation and fusion with injured fibers. Furthermore, Pax7 was induced by serum factors from cachectic mice and patients, in an NF-κB-dependent manner, both in vitro and in vivo. Together, these results suggest that Pax7 responds to NF-κB by impairing the regenerative capacity of myogenic cells in the muscle microenvironment to drive muscle wasting in cancer.
Collapse
MESH Headings
- Adolescent
- Adult
- Aged
- Aged, 80 and over
- Animals
- Cachexia/etiology
- Cachexia/metabolism
- Cachexia/pathology
- Case-Control Studies
- Cell Line, Tumor
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Male
- Mice
- Mice, Inbred mdx
- Mice, Nude
- Mice, Transgenic
- Microscopy, Electron, Transmission
- Middle Aged
- Muscle Development
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Myoblasts, Skeletal/metabolism
- Myoblasts, Skeletal/pathology
- NF-kappa B/metabolism
- PAX7 Transcription Factor/genetics
- PAX7 Transcription Factor/metabolism
- Pancreatic Neoplasms/complications
- Pancreatic Neoplasms/metabolism
- Satellite Cells, Skeletal Muscle/metabolism
- Satellite Cells, Skeletal Muscle/pathology
- Tumor Microenvironment
- Young Adult
Collapse
|
139
|
Gardam S, Brink R. Non-Canonical NF-κB Signaling Initiated by BAFF Influences B Cell Biology at Multiple Junctures. Front Immunol 2014; 4:509. [PMID: 24432023 PMCID: PMC3880999 DOI: 10.3389/fimmu.2013.00509] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 12/24/2013] [Indexed: 01/13/2023] Open
Abstract
It has been more than a decade since it was recognized that the nuclear factor of kappa light polypeptide gene enhancer in B cells (NF-κB) transcription factor family was activated by two distinct pathways: the canonical pathway involving NF-κB1 and the non-canonical pathway involving NF-κB2. During this time a great deal of evidence has been amassed on the ligands and receptors that activate these pathways, the cytoplasmic adapter molecules involved in transducing the signals from receptors to nucleus, and the resulting physiological outcomes within body tissues. In contrast to NF-κB1 signaling, which can be activated by a wide variety of receptors, the NF-κB2 pathway is typically only activated by a subset of receptor and ligand pairs belonging to the tumor necrosis factor (TNF) family. Amongst these is B cell activating factor of the TNF family (BAFF) and its receptor BAFFR. Whilst BAFF is produced by many cell types throughout the body, BAFFR expression appears to be restricted to the hematopoietic lineage and B cells in particular. For this reason, the main physiological outcomes of BAFF mediated NF-κB2 activation are confined to B cells. Indeed BAFF mediated NF-κB2 signaling contributes to peripheral B cell survival and maturation as well as playing a role in antibody responses and long term maintenance plasma cells. Thus the importance BAFF and NF-κB2 permeates the entire B cell lifespan and impacts on this important component of the immune system in a variety of ways.
Collapse
Affiliation(s)
- Sandra Gardam
- Immunology Division, Garvan Institute of Medical Research , Darlinghurst, NSW , Australia
| | - Robert Brink
- Immunology Division, Garvan Institute of Medical Research , Darlinghurst, NSW , Australia ; St. Vincent's Clinical School, University of New South Wales , Darlinghurst, NSW , Australia
| |
Collapse
|
140
|
Tan CL, Plotkin JL, Venø MT, von Schimmelmann M, Feinberg P, Mann S, Handler A, Kjems J, Surmeier DJ, O'Carroll D, Greengard P, Schaefer A. MicroRNA-128 governs neuronal excitability and motor behavior in mice. Science 2013; 342:1254-8. [PMID: 24311694 DOI: 10.1126/science.1244193] [Citation(s) in RCA: 227] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The control of motor behavior in animals and humans requires constant adaptation of neuronal networks to signals of various types and strengths. We found that microRNA-128 (miR-128), which is expressed in adult neurons, regulates motor behavior by modulating neuronal signaling networks and excitability. miR-128 governs motor activity by suppressing the expression of various ion channels and signaling components of the extracellular signal-regulated kinase ERK2 network that regulate neuronal excitability. In mice, a reduction of miR-128 expression in postnatal neurons causes increased motor activity and fatal epilepsy. Overexpression of miR-128 attenuates neuronal responsiveness, suppresses motor activity, and alleviates motor abnormalities associated with Parkinson's-like disease and seizures in mice. These data suggest a therapeutic potential for miR-128 in the treatment of epilepsy and movement disorders.
Collapse
Affiliation(s)
- Chan Lek Tan
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY 10065, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
141
|
Munitic I, Torchia MLG, Meena NP, Zhu G, Li CC, Ashwell JD. Optineurin insufficiency impairs IRF3 but not NF-κB activation in immune cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2013; 191:6231-40. [PMID: 24244017 PMCID: PMC3886234 DOI: 10.4049/jimmunol.1301696] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Optineurin is a widely expressed polyubiquitin-binding protein that has been implicated in regulating cell signaling via its NF-κB essential modulator-homologous C-terminal ubiquitin (Ub)-binding region. Its functions are controversial, with in vitro studies finding that optineurin suppressed TNF-mediated NF-κB activation and virus-induced activation of IFN regulatory factor 3 (IRF3), whereas bone marrow-derived macrophages (BMDMs) from mice carrying an optineurin Ub-binding point mutation had normal TLR-mediated NF-κB activation and diminished IRF3 activation. We have generated a mouse model in which the entire Ub-binding C-terminal region is deleted (Optn(470T)). Akin to C-terminal optineurin mutations found in patients with certain neurodegenerative diseases, Optn(470T) was expressed at substantially lower levels than the native protein, allowing assessment not only of the lack of Ub binding, but also of protein insufficiency. Embryonic lethality with incomplete penetrance was observed for 129 × C57BL/6 Optn(470T/470T) mice, but after further backcrossing to C57BL/6, offspring viability was restored. Moreover, the mice that survived were indistinguishable from wild type littermates and had normal immune cell distributions. Activation of NF-κB in Optn(470T) BMDM and BM-derived dendritic cells with TNF or via TLR4, T cells via the TCR, and B cells with LPS or anti-CD40 was normal. In contrast, optineurin and/or its Ub-binding function was necessary for optimal TANK binding kinase 1 and IRF3 activation, and both Optn(470T) BMDMs and bone marrow-derived dendritic cells had diminished IFN-β production upon LPS stimulation. Importantly, Optn(470T) mice produced less IFN-β upon LPS challenge. Therefore, endogenous optineurin is dispensable for NF-κB activation but necessary for optimal IRF3 activation in immune cells.
Collapse
Affiliation(s)
- Ivana Munitic
- Laboratory of Immune Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892
| | | | - Netra Pal Meena
- Laboratory of Immune Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892
| | - Guozhi Zhu
- Laboratory of Immune Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892
| | - Caiyi C. Li
- Laboratory of Immune Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892
| | - Jonathan D. Ashwell
- Laboratory of Immune Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|
142
|
Jellusova J, Miletic AV, Cato MH, Lin WW, Hu Y, Bishop GA, Shlomchik MJ, Rickert RC. Context-specific BAFF-R signaling by the NF-κB and PI3K pathways. Cell Rep 2013; 5:1022-35. [PMID: 24239354 PMCID: PMC3887557 DOI: 10.1016/j.celrep.2013.10.022] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Revised: 08/10/2013] [Accepted: 10/10/2013] [Indexed: 11/24/2022] Open
Abstract
BAFF is a soluble factor required for B cell maturation and survival. BAFF-R signals via the noncanonical NF-κB pathway regulated by the TRAF3/NIK/IKK1 axis. We show that deletion of Ikk1 during early B cell development causes a partial impairment in B cell maturation and BAFF-dependent survival, but inactivation of Ikk1 in mature B cells does not affect survival. We further show that BAFF-R employs CD19 to promote survival via phosphatidylinositol 3-kinase (PI3K), and that coinactivation of Cd19 and Ikk1 causes a profound block in B cell maturation at the transitional stage. Consistent with a role for PI3K in BAFF-R function, inactivation of PTEN mediates a partial rescue of B cell maturation and function in Baff(-/-) animals. Elevated PI3K signaling also circumvents BAFF-dependent survival in a spontaneous B cell lymphoma model. These findings indicate that the combined activities of PI3K and IKK1 drive peripheral B cell differentiation and survival in a context-dependent manner.
Collapse
Affiliation(s)
- Julia Jellusova
- Program on Inflammatory Diseases, Infectious and Inflammatory Diseases Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037 USA
| | - Ana V. Miletic
- Program on Inflammatory Diseases, Infectious and Inflammatory Diseases Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037 USA
| | - Matthew H. Cato
- Program on Inflammatory Diseases, Infectious and Inflammatory Diseases Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037 USA
| | - Wai-Wai Lin
- Graduate Program in Immunology, The University of Iowa and the VA Medical Center, Iowa City, IA 52242, USA
| | - Yinling Hu
- Cancer & Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21701, USA
| | - Gail A. Bishop
- Graduate Program in Immunology, The University of Iowa and the VA Medical Center, Iowa City, IA 52242, USA
- Departments of Microbiology and Internal Medicine, The University of Iowa and the VA Medical Center, Iowa City, IA 52242, USA
| | - Mark J. Shlomchik
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Robert C. Rickert
- Program on Inflammatory Diseases, Infectious and Inflammatory Diseases Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037 USA
| |
Collapse
|
143
|
Souma T, Yamazaki S, Moriguchi T, Suzuki N, Hirano I, Pan X, Minegishi N, Abe M, Kiyomoto H, Ito S, Yamamoto M. Plasticity of renal erythropoietin-producing cells governs fibrosis. J Am Soc Nephrol 2013; 24:1599-616. [PMID: 23833259 PMCID: PMC3785278 DOI: 10.1681/asn.2013010030] [Citation(s) in RCA: 156] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Accepted: 04/24/2013] [Indexed: 12/31/2022] Open
Abstract
CKD progresses with fibrosis and erythropoietin (Epo)-dependent anemia, leading to increased cardiovascular complications, but the mechanisms linking Epo-dependent anemia and fibrosis remain unclear. Here, we show that the cellular phenotype of renal Epo-producing cells (REPs) alternates between a physiologic Epo-producing state and a pathologic fibrogenic state in response to microenvironmental signals. In a novel mouse model, unilateral ureteral obstruction-induced inflammatory milieu activated NFκB and Smad signaling pathways in REPs, rapidly repressed the Epo-producing potential of REPs, and led to myofibroblast transformation of these cells. Moreover, we developed a unique Cre-based cell-fate tracing method that marked current and/or previous Epo-producing cells and revealed that the majority of myofibroblasts are derived from REPs. Genetic induction of NFκB activity selectively in REPs resulted in myofibroblastic transformation, indicating that NFκB signaling elicits a phenotypic switch. Reversing the unilateral ureteral obstruction-induced inflammatory microenvironment restored the Epo-producing potential and the physiologic phenotype of REPs. This phenotypic reversion was accelerated by anti-inflammatory therapy. These findings demonstrate that REPs possess cellular plasticity, and suggest that the phenotypic transition of REPs to myofibroblasts, modulated by inflammatory molecules, underlies the connection between anemia and renal fibrosis in CKD.
Collapse
Affiliation(s)
- Tomokazu Souma
- Department of Medical Biochemistry
- Division of Nephrology, Endocrinology, and Vascular Medicine, Department of Medicine
| | | | | | - Norio Suzuki
- Division of Interdisciplinary Medical Science, United Centers for Advanced Research and Translational Medicine, and
| | | | - Xiaoqing Pan
- Department of Medical Biochemistry
- Tohoku Medical Megabank Organization, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan; and
| | - Naoko Minegishi
- Department of Medical Biochemistry
- Tohoku Medical Megabank Organization, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan; and
| | - Michiaki Abe
- Division of Nephrology, Endocrinology, and Vascular Medicine, Department of Medicine
- Tohoku Medical Megabank Organization, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan; and
| | - Hideyasu Kiyomoto
- Division of Nephrology, Endocrinology, and Vascular Medicine, Department of Medicine
- Tohoku Medical Megabank Organization, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan; and
| | - Sadayoshi Ito
- Division of Nephrology, Endocrinology, and Vascular Medicine, Department of Medicine
| | - Masayuki Yamamoto
- Department of Medical Biochemistry
- Tohoku Medical Megabank Organization, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan; and
- JST, CREST, Sendai, Miyagi, Japan
| |
Collapse
|
144
|
Grund LZ, Lopes-Ferreira M, Lima C. The hierarchical process of differentiation of long-lived antibody-secreting cells is dependent on integrated signals derived from antigen and IL-17A. PLoS One 2013; 8:e74566. [PMID: 24058589 PMCID: PMC3776816 DOI: 10.1371/journal.pone.0074566] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Accepted: 08/06/2013] [Indexed: 12/02/2022] Open
Abstract
Switched CD19-positive memory B cells purified from mice with chronic immune response against Thalassophrynenattereri venom proteins were cultured with venom or cytokines. Our results confirm the existence of a hierarchic process of differentiation: activated memory B cells progressively acquire increasing levels of CD138 and decreasing levels of CD45R/B220 to finally arrive at ASC with B220neg phenotype, which are IgG1-secreting cells. Only Bmem from peritoneal cavity or bone marrow of VTn immunized mice presented the capacity to generate ASC functionally active. IL-17A or IL-21/IL-23/IL-33 improves the ability of venom to induce intracellular IgG of peritoneal derived-ASC. Cognate stimulation with venom and IL-17A is sufficient to down-regulate the expression of CD45R/B220. BAFF-R is up-regulated in splenic or medullar derived-ASC stimulated by venom, CpG or cytokines. Only splenic derived-ASC up-regulate Bcl-2 expression after CpG or the combination of IL-21/IL-23/IL-33 stimulation. Finally, the activation of ASC for IgG1 secretion is triggered by venom proteins in peritoneal cavity and by IL-17A in medullar niche. These results show the importance of the integration of signals downstream of BCR and IL17-A receptors in modulating ASC differentiation, focusing in the microenvironment niche of their generation.
Collapse
Affiliation(s)
- Lidiane Zito Grund
- Immunoregulation Unit, Special Laboratory of Applied Toxinology (CEPID/FAPESP), Butantan Institute and Department of Immunology, University of São Paulo, São Paulo, Brazil
| | - Monica Lopes-Ferreira
- Immunoregulation Unit, Special Laboratory of Applied Toxinology (CEPID/FAPESP), Butantan Institute and Department of Immunology, University of São Paulo, São Paulo, Brazil
| | - Carla Lima
- Immunoregulation Unit, Special Laboratory of Applied Toxinology (CEPID/FAPESP), Butantan Institute and Department of Immunology, University of São Paulo, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
145
|
Uhde M, Kuehl S, Richardt U, Fleischer B, Osterloh A. Differential regulation of marginal zone and follicular B cell responses by CD83. Int Immunol 2013; 25:507-20. [PMID: 23728778 DOI: 10.1093/intimm/dxt021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2023] Open
Abstract
Transgenic over-expression of CD83 on B cells leads to a reduced response to BCR engagement but to an enhanced secretion of IL-10 upon LPS stimulation. In this study, we analyzed the differential influence of CD83 on the stimulation of different B cell subsets via the BCR or TLR4. Neither wild type nor CD83 transgenic (CD83tg) B cells produced any IL-10 in response to BCR stimulation. BCR engagement led to reduced activation of LYN, SYK and ERK1/2 resulting in reduced numbers of proliferating cells in all CD83tg B cell subsets. Moreover, CD83tg follicular (FO) but not marginal zone (MZ) or transitional (TN) B cells showed significantly enhanced cell death. In contrast, LPS stimulation led to normal frequencies of proliferating CD83tg FO, MZ and TN B cells although TLR4 engagement did not rescue FO B cells from apoptosis. Furthermore, LPS stimulation led to high IL-10 production derived from CD83tg MZ B cells that reacted to LPS stimulation with enhanced ERK1/2 activation. Finally, we show that CD83 co-localizes with the BCR complex as well as with the LPS receptor complex suggesting that CD83 interacts with components of both signaling complexes. Taken together, the results of this study show that CD83 already inhibits the initiation of BCR signaling leading to insufficient activation signals in all B cells and reduced survival especially of FO B cells. On the other hand, CD83 supports TLR4-mediated IL-10 release exclusively in MZ B cells. Thus, CD83 differentially modulates FO and MZ B cell responses.
Collapse
Affiliation(s)
- Melanie Uhde
- Department of Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg 20359, Germany
| | | | | | | | | |
Collapse
|
146
|
Abstract
The mammalian target of rapamycin (mTOR), an essential serine/threonine kinase, functions in biochemically distinct multiprotein complexes, but little is known about roles of the complexes in B cells. The acutely rapamycin-sensitive mTOR complex 1 (mTORC1) is defined by a core subunit Raptor, whereas mTORC2 lacks Raptor and, instead, has Rictor and SIN1 as distinct essential components. We now show that homeostasis and function of B cells require Rictor. Conditional deletion of Rictor before lymphoid specification impaired generation of mature follicular, marginal zone, and B1a B lymphocytes. Induced inactivation in adult mice caused cell-autonomous defects in B lymphoid homeostasis and antibody responses in vivo, along with affecting plasma cells in bone marrow. Survival of B lymphocytes depended on Rictor, which was vital for normal induction of prosurvival genes, suppression of proapoptotic genes, nuclear factor κB induction after B-cell receptor stimulation, and B-cell activating factor-induced nuclear factor κB2/p52 generation. Collectively, the findings provide evidence that mTOR signaling affects survival and proliferation of mature B lymphocytes, and establish Rictor as an important signal relay in B-cell homeostasis, fate, and functions.
Collapse
|
147
|
Matharu K, Zarember KA, Marciano BE, Kuhns DB, Spalding C, Garofalo M, Dimaggio T, Estwick T, Huang CY, Fink D, Priel DL, Fleisher TA, Holland SM, Malech HL, Gallin JI. B-cell activating factor (BAFF) is elevated in chronic granulomatous disease. Clin Immunol 2013; 148:258-64. [PMID: 23773925 PMCID: PMC3774275 DOI: 10.1016/j.clim.2013.05.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Revised: 05/09/2013] [Accepted: 05/13/2013] [Indexed: 01/05/2023]
Abstract
Chronic Granulomatous Disease (CGD) is an inherited defect in superoxide production leading to life-threatening infections, granulomas, and, possibly, abnormal immunoglobulin concentrations. We investigated whether factors controlling antibody production, such as B-cell activating factor (BAFF), were altered in CGD. CGD subjects had significantly increased mean (2.3-fold, p < 0.0001) plasma concentrations of BAFF compared to healthy donors. Patients on IFN-γ treatment had significantly higher BAFF concentrations compared with CGD patients not taking IFN-γ (1.6-fold, p < 0.005). Leukocytes from CGD subjects produced normal amounts of BAFF in response to IFN-γ or G-CSF in vitro. Expression of BAFF-R and TACI was significantly reduced on CGD B cells. Elevated BAFF in CGD correlated with CRP (R = 0.44), ESR (R = 0.49), and IgM (R = 0.47) and increased rapidly in healthy subjects following intravenous endotoxin administration. These findings suggest that elevated BAFF in CGD subjects and healthy donors is a consequence of acute and chronic inflammation.
Collapse
Affiliation(s)
- Kabir Matharu
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
- Howard Hughes Medical Institute, Chevy Chase, MD
| | - Kol A. Zarember
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | | | - Douglas B. Kuhns
- Clinical Services Program, Applied/Development Research Directorate, SAIC-Frederick Inc., Frederick National Laboratory for Cancer Research, Frederick, MD
| | | | - Mary Garofalo
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Thomas Dimaggio
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Tyra Estwick
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | | | - Danielle Fink
- Clinical Services Program, Applied/Development Research Directorate, SAIC-Frederick Inc., Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Debra L. Priel
- Clinical Services Program, Applied/Development Research Directorate, SAIC-Frederick Inc., Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Thomas A. Fleisher
- Department of Laboratory Medicine, NIH Clinical Center, NIH, Bethesda, MD
| | - Steven M. Holland
- Laboratory of Clinical Infectious Diseases, NIAID, NIH, Bethesda, MD
| | - Harry L. Malech
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - John I. Gallin
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| |
Collapse
|
148
|
Stepensky P, Keller B, Buchta M, Kienzler AK, Elpeleg O, Somech R, Cohen S, Shachar I, Miosge LA, Schlesier M, Fuchs I, Enders A, Eibel H, Grimbacher B, Warnatz K. Deficiency of caspase recruitment domain family, member 11 (CARD11), causes profound combined immunodeficiency in human subjects. J Allergy Clin Immunol 2013; 131:477-85.e1. [PMID: 23374270 DOI: 10.1016/j.jaci.2012.11.050] [Citation(s) in RCA: 139] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2012] [Revised: 11/26/2012] [Accepted: 11/29/2012] [Indexed: 12/21/2022]
Abstract
BACKGROUND Profound combined immunodeficiency can present with normal numbers of T and B cells, and therefore the functional defect of the cellular and humoral immune response is often not recognized until the first severe clinical manifestation. Here we report a patient of consanguineous descent presenting at 13 months of age with hypogammaglobulinemia, Pneumocystis jirovecii pneumonia, and a suggestive family history. OBJECTIVE We sought to identify the genetic alteration in a patient with combined immunodeficiency and characterize human caspase recruitment domain family, member 11 (CARD11), deficiency. METHODS Molecular, immunologic, and functional assays were performed. RESULTS The immunologic characterization revealed only subtle changes in the T-cell and natural killer cell compartment, whereas B-cell differentiation, although normal in number, was distinctively blocked at the transitional stage. Genetic evaluation revealed a homozygous deletion of exon 21 in CARD11 as the underlying defect. This deletion abrogated protein expression and activation of the canonical nuclear factor κB (NF-κB) pathway in lymphocytes after antigen receptor or phorbol 12-myristate 13-acetate stimulation, whereas CD40 signaling in B cells was preserved. The abrogated activation of the canonical NF-κB pathway was associated with severely impaired upregulation of inducible T-cell costimulator, OX40, cytokine production, proliferation of T cells, and B cell-activating factor receptor expression on B cells. CONCLUSION Thus in patients with CARD11 deficiency, the combination of impaired activation and especially upregulation of inducible T-cell costimulator on T cells, together with severely disturbed peripheral B-cell differentiation, apparently leads to a defective T-cell/B-cell cooperation and probably germinal center formation and clinically results in severe immunodeficiency. This report discloses the crucial and nonredundant role of canonical NF-κB activation and specifically CARD11 in the antigen-specific immune response in human subjects.
Collapse
Affiliation(s)
- Polina Stepensky
- Pediatric Hematology-Oncology and Bone Marrow Transplantation, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
149
|
Myant K, Cammareri P, McGhee E, Ridgway R, Huels D, Cordero J, Schwitalla S, Kalna G, Ogg EL, Athineos D, Timpson P, Vidal M, Murray G, Greten F, Anderson K, Sansom O. ROS production and NF-κB activation triggered by RAC1 facilitate WNT-driven intestinal stem cell proliferation and colorectal cancer initiation. Cell Stem Cell 2013; 12:761-73. [PMID: 23665120 PMCID: PMC3690525 DOI: 10.1016/j.stem.2013.04.006] [Citation(s) in RCA: 313] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Revised: 11/27/2012] [Accepted: 04/08/2013] [Indexed: 12/18/2022]
Abstract
The Adenomatous Polyposis Coli (APC) gene is mutated in the majority of colorectal cancers (CRCs). Loss of APC leads to constitutively active WNT signaling, hyperproliferation, and tumorigenesis. Identification of pathways that facilitate tumorigenesis after APC loss is important for therapeutic development. Here, we show that RAC1 is a critical mediator of tumorigenesis after APC loss. We find that RAC1 is required for expansion of the LGR5 intestinal stem cell (ISC) signature, progenitor hyperproliferation, and transformation. Mechanistically, RAC1-driven ROS and NF-κB signaling mediate these processes. Together, these data highlight that ROS production and NF-κB activation triggered by RAC1 are critical events in CRC initiation.
Collapse
Affiliation(s)
- Kevin B. Myant
- Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Patrizia Cammareri
- Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Ewan J. McGhee
- Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Rachel A. Ridgway
- Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - David J. Huels
- Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Julia B. Cordero
- Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Sarah Schwitalla
- Institute of Molecular Immunology, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany
| | - Gabriela Kalna
- Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Erinn-Lee Ogg
- Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Dimitris Athineos
- Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Paul Timpson
- Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Marcos Vidal
- Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Graeme I. Murray
- Department of Pathology, Division of Applied Medicine, School of Medicine and Dentistry, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Florian R. Greten
- Institute of Molecular Immunology, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany
| | - Kurt I. Anderson
- Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Owen J. Sansom
- Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| |
Collapse
|
150
|
Stolp J, Mariño E, Batten M, Sierro F, Cox SL, Grey ST, Silveira PA. Intrinsic molecular factors cause aberrant expansion of the splenic marginal zone B cell population in nonobese diabetic mice. THE JOURNAL OF IMMUNOLOGY 2013; 191:97-109. [PMID: 23740954 DOI: 10.4049/jimmunol.1203252] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Marginal zone (MZ) B cells are an innate-like population that oscillates between MZ and follicular areas of the splenic white pulp. Differentiation of B cells into the MZ subset is governed by BCR signal strength and specificity, NF-κB activation through the B cell-activating factor belonging to the TNF family (BAFF) receptor, Notch2 signaling, and migration signals mediated by chemokine, integrin, and sphingosine-1-phosphate receptors. An imbalance in splenic B cell development resulting in expansion of the MZ subset has been associated with autoimmune pathogenesis in various murine models. One example is the NOD inbred mouse strain, in which MZ B cell expansion has been linked to development of type 1 diabetes and Sjögren's syndrome. However, the cause of MZ B cell expansion in this strain remains poorly understood. We have determined that increased MZ B cell development in NOD mice is independent of T cell autoimmunity, BCR specificity, BCR signal strength, and increased exposure to BAFF. Rather, mixed bone marrow chimeras showed that the factor(s) responsible for expansion of the NOD MZ subset is B cell intrinsic. Analysis of microarray expression data indicated that NOD MZ and precursor transitional 2-MZ subsets were particularly dysregulated for genes controlling cellular trafficking, including Apoe, Ccbp2, Cxcr7, Lgals1, Pla2g7, Rgs13, S1pr3, Spn, Bid, Cd55, Prf1, and Tlr3. Furthermore, these B cell subsets exhibited an increased steady state dwell time within splenic MZ areas. Our data therefore reveal that precursors of mature B cells in NOD mice exhibit an altered migration set point, allowing increased occupation of the MZ, a niche favoring MZ B cell differentiation.
Collapse
Affiliation(s)
- Jessica Stolp
- Garvan Institute of Medical Research, Immunology Program, Darlinghurst, New South Wales 2010, Australia
| | | | | | | | | | | | | |
Collapse
|