Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Immunol Lett ; 261: 37-46, 2023 09.
Article in English | MEDLINE | ID: mdl-37459958

ABSTRACT

Immunity in a naïve organism is tightly controlled. Adequate proportions of the many immune cell subsets must be produced to mount efficient responses to eventual challenges. In addition, a functioning immune system is highly dynamic at steady state. Mature immune cells must be positioned properly and/or circulate to facilitate the detection of dangers. They must also be poised to promptly react to unusual encounters, while ignoring innocuous germs and self. Numerous regulatory mechanisms act at the molecular level to generate such an exquisite structure, including miRNA-mediated repression of protein synthesis. Notably, the miRNAs from the miR-142 locus are preferentially expressed in hematopoietic cells. Their importance is underscored by the deeply disturbed immune system seen upon inactivation of the locus in mice. In this review, we explore reported roles for the miR-142 miRNAs in the shaping of immunity in vertebrates, discussing in particular their contributions to the generation, migration and survival of hematopoietic cells.


Subject(s)
MicroRNAs , Animals , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Immune System/metabolism
2.
Front Immunol ; 13: 847415, 2022.
Article in English | MEDLINE | ID: mdl-36439112

ABSTRACT

B lymphocyte development proceeds through a well-ordered sequence of steps, leading to the formation of a sizeable mature B population recognizing a diversity of antigens. These latter cells are ultimately responsible for the production of antibodies upon immune challenges. The detection of threats to the organism is facilitated by the ability of naïve follicular B cells, the main subset of mature B cells in mice, to circulate between lymphoid tissues in search of their cognate antigens. miRNA-mediated fine-tuning of mRNA stability and translation participates in the optimal expression of genetic programs. This regulatory mechanism has been shown to contribute to B cell biology, although the role of individual miRNAs remains understudied. Here, we selectively inactivated the miR-142 locus in B cells. As a consequence, the mature B compartment was visibly perturbed, in agreement with work in miR-142 knockout mice. However, our strategy allowed us to identify roles for the miR-142 locus in B cell physiology obscured by the complexity of the immune phenotype in the null mutant mice. Thus, these miRNAs are necessary for the proper formation of the pre-B cell compartment during development. More remarkably, naïve follicular B cells demonstrated altered migratory properties upon conditional inactivation of the miR-142 locus. The latter mutant cells expressed reduced levels of the homing molecule CD62L. They also migrated more efficiently towards sphingosine-1-phosphate in vitro and displayed an increased abundance of the sphingosine-1-phosphate receptor 1, compatible with improved lymphocyte egress in vivo. In line with these observations, the ablation of the miR-142 locus in B cells caused a paucity of B cells in the lymph nodes. Mutant B cell accumulation in the latter tissues was also compromised upon transfer into a wild-type environment. These changes coincided with suboptimal levels of FOXO1, a positive regulator of CD62L transcription, in mutant B cells. Overall, our findings indicate contributions for the miR-142 locus in various aspects of the B cell life cycle. Notably, this locus appears to favor the establishment of the migratory behavior required for naïve follicular B cell patrolling activity.


Subject(s)
B-Lymphocytes , MicroRNAs , Mice , Animals , B-Lymphocytes/metabolism , Lymph Nodes , Lymphoid Tissue/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Lymphocytes/metabolism , Mice, Knockout
3.
Front Immunol ; 13: 845235, 2022.
Article in English | MEDLINE | ID: mdl-36052079

ABSTRACT

B cells are key mediators of humoral immunity. Mature B cells fall into various sub-classes that can be separated by their ontogeny, expression of cell surface markers, anatomical location, and function. B1 subsets play important roles in natural immunity and constitute the majority of B cells in newborns. In the adult, B1 cells predominate in the pleural and peritoneal cavities, while the mature B2 follicular subset makes up the major fraction of B cells in lymphoid tissue, although important subsets of antibody-secreting B1 cells are also present at these sites. B1 cells are the main producers of natural IgM but can also contribute to elimination of some pathogens, while B2 cells primarily mediate response to foreign antigens. The differential molecular underpinning of the B1 and B2 subsets remains incompletely understood. Here we demonstrate that germline-deficiency of the orphan nuclear receptor NR2F6 causes a partial loss of B1b and B2 B cells in the peritoneum while leaving peritoneal B1a cells unaltered. A competitive bone marrow chimera in Nr2f6+/+ host mice produced similar numbers of Nr2f6+/+ and Nr2f6-/- peritoneal B1b and B2 cells. The proliferation of Nr2f6-/- peritoneal B cells was not altered, while the migration marker CXCR5 was reduced on all subsets but Beta7-integrin was reduced only on peritoneal B1b and B2 cells. Similarly, B1b and B2 but not B1a cells, exhibited significantly reduced survival.


Subject(s)
B-Lymphocytes , Peritoneum , Repressor Proteins/metabolism , Animals , Homeostasis , Mice , Peritoneal Cavity , Receptors, Cytoplasmic and Nuclear
4.
Exp Gerontol ; 138: 110985, 2020 09.
Article in English | MEDLINE | ID: mdl-32504658

ABSTRACT

The uniqueness of each B cell lies in the structural diversity of the B-cell antigen receptor allowing the virtually limitless recognition of antigens, a necessity to protect individuals against a range of challenges. B-cell development and response to stimulation are exquisitely regulated by a group of cell surface receptors modulating various signaling cascades and their associated genetic programs. The effects of these signaling pathways in optimal antibody-mediated immunity or the aberrant promotion of immune pathologies have been intensely researched in the past in young individuals. In contrast, we are only beginning to understand the contribution of these pathways to the changes in B cells of old organisms. Thus, critical transcription factors such as E2A and STAT5 show differential expression or activity between young and old B cells. As a result, B-cell physiology appears altered, and antibody production is impaired. Here, we discuss selected phenotypic changes during B-cell aging and attempt to relate them to alterations of molecular mechanisms.


Subject(s)
B-Lymphocytes , Receptors, Antigen, B-Cell , Aging , Humans , Lymphocyte Activation , Signal Transduction
5.
Gerontology ; 66(2): 105-113, 2020.
Article in English | MEDLINE | ID: mdl-31553969

ABSTRACT

Aging results for the immune system in a departure from the optimal homeostatic state seen in young organisms. This divergence regrettably contributes to a higher frequency of compromised responses to infections and inefficient classical vaccination in aged populations. In B cells, the cornerstone of humoral immunity, the development and distribution of the various mature B cell subsets are impacted by aging in both humans and mice. In addition, aged mature B cells demonstrate limited capacity to mount efficient antibody responses. An expected culprit for the decline in effective immunity is the rise of the systemic levels of pro-inflammatory molecules during aging, establishing a chronic low-grade inflammation. Indeed, numerous alterations affecting directly or indirectly B cells in old people and mice are reminiscent of various effects of acute inflammation on this cell type in young adults. The present mini-review will highlight the possible adverse contributions of the persistent low-level inflammation observed in susceptible older organisms to the inadequate B-cell physiology.


Subject(s)
Aging/physiology , B-Lymphocytes/physiology , Inflammation/physiopathology , Aged , Animals , Cell Physiological Phenomena , Humans , Immune System/physiology , Mice
6.
Genes Dev ; 33(23-24): 1673-1687, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31699777

ABSTRACT

Knockout of the ubiquitously expressed miRNA-17∼92 cluster in mice produces a lethal developmental lung defect, skeletal abnormalities, and blocked B lymphopoiesis. A shared target of miR-17∼92 miRNAs is the pro-apoptotic protein BIM, central to life-death decisions in mammalian cells. To clarify the contribution of miR-17∼92:Bim interactions to the complex miR-17∼92 knockout phenotype, we used a system of conditional mutagenesis of the nine Bim 3' UTR miR-17∼92 seed matches. Blocking miR-17∼92:Bim interactions early in development phenocopied the lethal lung phenotype of miR-17∼92 ablation and generated a skeletal kinky tail. In the hematopoietic system, instead of causing the predicted B cell developmental block, it produced a selective inability of B cells to resist cellular stress; and prevented B and T cell hyperplasia caused by Bim haploinsufficiency. Thus, the interaction of miR-17∼92 with a single target is essential for life, and BIM regulation by miRNAs serves as a rheostat controlling cell survival in specific physiological contexts.


Subject(s)
B-Lymphocytes/cytology , Bcl-2-Like Protein 11/metabolism , Cell Survival/genetics , Gene Expression Regulation, Developmental/genetics , Hematopoiesis/genetics , MicroRNAs/metabolism , 3' Untranslated Regions/genetics , Animals , B-Lymphocytes/pathology , Bcl-2-Like Protein 11/genetics , Gene Knockout Techniques , Lung/embryology , Mice , MicroRNAs/genetics , Mutation , Stress, Physiological
7.
FEBS J ; 286(18): 3566-3581, 2019 09.
Article in English | MEDLINE | ID: mdl-31120187

ABSTRACT

Upon activation by antigen, B cells form germinal centres where they clonally expand and introduce affinity-enhancing mutations into their B-cell receptor genes. Somatic mutagenesis and class switch recombination (CSR) in germinal centre B cells are initiated by the activation-induced cytidine deaminase (AID). Upon germinal centre exit, B cells differentiate into antibody-secreting plasma cells. Germinal centre maintenance and terminal fate choice require transcriptional reprogramming that associates with a substantial reconfiguration of DNA methylation patterns. Here we examine the role of ten-eleven-translocation (TET) proteins, enzymes that facilitate DNA demethylation and promote a permissive chromatin state by oxidizing 5-methylcytosine, in antibody-mediated immunity. Using a conditional gene ablation strategy, we show that TET2 and TET3 guide the transition of germinal centre B cells to antibody-secreting plasma cells. Optimal AID expression requires TET function, and TET2 and TET3 double-deficient germinal centre B cells show defects in CSR. However, TET2/TET3 double-deficiency does not prevent the generation and selection of high-affinity germinal centre B cells. Rather, combined TET2 and TET3 loss-of-function in germinal centre B cells favours C-to-T and G-to-A transition mutagenesis, a finding that may be of significance for understanding the aetiology of B-cell lymphomas evolving in conditions of reduced TET function.


Subject(s)
Antibody-Producing Cells/immunology , B-Lymphocytes/immunology , DNA-Binding Proteins/genetics , Dioxygenases/genetics , Proto-Oncogene Proteins/genetics , 5-Methylcytosine/metabolism , Animals , Antibody Formation/genetics , Antibody Formation/immunology , B-Lymphocytes/metabolism , Cell Differentiation/genetics , Chromatin/genetics , Cytidine Deaminase/genetics , Cytidine Deaminase/immunology , DNA Demethylation , DNA Methylation/genetics , DNA-Binding Proteins/immunology , Dioxygenases/immunology , Gene Expression Regulation/immunology , Germinal Center/immunology , Humans , Immunoglobulin Class Switching/genetics , Immunoglobulin Class Switching/immunology , Mice , Mutation/genetics , Mutation/immunology , Proto-Oncogene Proteins/immunology
8.
Proc Natl Acad Sci U S A ; 113(18): 5065-70, 2016 May 03.
Article in English | MEDLINE | ID: mdl-27099294

ABSTRACT

Although canonical NF-κB signaling is crucial to generate a normal mature B-cell compartment, its role in the persistence of resting mature B cells is controversial. To resolve this conflict, we ablated NF-κB essential modulator (NEMO) and IκB kinase 2 (IKK2), two essential mediators of the canonical pathway, either early on in B-cell development or specifically in mature B cells. Early ablation severely inhibited the generation of all mature B-cell subsets, but follicular B-cell numbers could be largely rescued by ectopic expression of B-cell lymphoma 2 (Bcl2), despite a persisting block at the transitional stage. Marginal zone (MZ) B and B1 cells were not rescued, indicating a possible role of canonical NF-κB signals beyond the control of cell survival in these subsets. When canonical NF-κB signaling was ablated specifically in mature B cells, the differentiation and/or persistence of MZ B cells was still abrogated, but follicular B-cell numbers were only mildly affected. However, the mutant cells exhibited increased turnover as well as functional deficiencies upon activation, suggesting that canonical NF-κB signals contribute to their long-term persistence and functional fitness.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cell Survival/immunology , NF-kappa B/immunology , Signal Transduction/immunology , Animals , Cells, Cultured , Mice , Mice, Inbred C57BL
9.
Proc Natl Acad Sci U S A ; 112(39): 12145-50, 2015 Sep 29.
Article in English | MEDLINE | ID: mdl-26371314

ABSTRACT

B cells respond to antigens by engagement of their B-cell antigen receptor (BCR) and of coreceptors through which signals from helper T cells or pathogen-associated molecular patterns are delivered. We show that the proliferative response of B cells to the latter stimuli is controlled by BCR-dependent activation of phosphoinositidyl 3-kinase (PI-3K) signaling. Glycogen synthase kinase 3ß and Foxo1 are two PI-3K-regulated targets that play important roles, but to different extents, depending on the specific mitogen. These results suggest a model for integrating signals from the innate and the adaptive immune systems in the control of the B-cell immune response.


Subject(s)
Adaptive Immunity/immunology , Cell Proliferation/physiology , Immunity, Innate/immunology , Models, Immunological , Receptors, Antigen, B-Cell/immunology , Signal Transduction/immunology , Animals , Cell Survival/immunology , Cells, Cultured , DNA Primers/genetics , Electrophoretic Mobility Shift Assay , Flow Cytometry , Immunoblotting , Mice , Phosphatidylinositol 3-Kinases/metabolism , Real-Time Polymerase Chain Reaction
10.
Proc Natl Acad Sci U S A ; 111(41): 14794-9, 2014 Oct 14.
Article in English | MEDLINE | ID: mdl-25267645

ABSTRACT

TNFα is a potent cytokine that plays a critical role in numerous cellular processes, particularly immune and inflammatory responses, programmed cell death, angiogenesis, and cell migration. Thus, understanding the molecular mechanisms that mediate TNFα-induced cellular responses is a crucial issue. It is generally accepted that global DNA binding activity of the NF-κB avian reticuloendotheliosis viral (v-rel) oncogene related B (RelB) subunit is not induced upon TNFα treatment in fibroblasts, despite its TNFα-induced nuclear accumulation. Here, we demonstrate that RelB plays a critical role in promoting fibroblast migration upon prolonged TNFα treatment. We identified the two kinases IκB kinase α (IKKα) and IκB kinase ß (IKKß) as RelB interacting partners whose activation by TNFα promotes RelB phosphorylation at serine 472. Once phosphorylated on serine 472, nuclear RelB dissociates from its interaction with the inhibitory protein IκBα and binds to the promoter of critical migration-associated genes, such as the matrix metallopeptidase 3 (MMP3). Further, we show that RelB serine 472 phosphorylation status controls MMP3 expression and promigration activity downstream of TNF receptors. Our findings provide new insights into the regulation of RelB activity and reveal a novel link between selective NF-κB target gene expression and cellular response in response to TNFα.


Subject(s)
Cell Movement , Fibroblasts/cytology , Fibroblasts/metabolism , I-kappa B Kinase/metabolism , Promoter Regions, Genetic/genetics , Receptors, Tumor Necrosis Factor/metabolism , Transcription Factor RelB/metabolism , Animals , Cell Movement/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Fibroblasts/drug effects , HEK293 Cells , Humans , I-kappa B Proteins/metabolism , Matrix Metalloproteinase 3/metabolism , Mice , NF-KappaB Inhibitor alpha , Phosphorylation/drug effects , Phosphoserine/metabolism , Tumor Necrosis Factor-alpha/pharmacology
11.
J Immunol ; 191(6): 3100-11, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23966625

ABSTRACT

Th17 cells are a proinflammatory subset of effector T cells that have been implicated in the pathogenesis of asthma. Their production of the cytokine IL-17 is known to induce local recruitment of neutrophils, but the direct impact of IL-17 on the lung epithelium is poorly understood. In this study, we describe a novel mouse model of spontaneous IL-17-driven lung inflammation that exhibits many similarities to asthma in humans. We have found that STAT3 hyperactivity in T lymphocytes causes an expansion of Th17 cells, which home preferentially to the lungs. IL-17 secretion then leads to neutrophil infiltration and lung epithelial changes, in turn leading to a chronic inflammatory state with increased mucus production and decreased lung function. We used this model to investigate the effects of IL-17 activity on airway epithelium and identified CXCL5 and MIP-2 as important factors in neutrophil recruitment. The neutralization of IL-17 greatly reduces pulmonary neutrophilia, underscoring a key role for IL-17 in promoting chronic airway inflammation. These findings emphasize the role of IL-17 in mediating neutrophil-driven pulmonary inflammation and highlight a new mouse model that may be used for the development of novel therapies targeting Th17 cells in asthma and other chronic pulmonary diseases.


Subject(s)
Asthma/immunology , Immune System Diseases/immunology , Interleukin-17/immunology , Leukocyte Disorders/immunology , Neutrophils/immunology , Respiratory Mucosa/immunology , Animals , Asthma/metabolism , Cell Separation , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Interleukin-17/metabolism , Mice , Mice, Inbred C57BL , Pneumonia/immunology , Pneumonia/metabolism , Real-Time Polymerase Chain Reaction , Respiratory Mucosa/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Transfection
12.
Cell ; 148(4): 739-51, 2012 Feb 17.
Article in English | MEDLINE | ID: mdl-22341446

ABSTRACT

B cells infected by Epstein-Barr virus (EBV), a transforming virus endemic in humans, are rapidly cleared by the immune system, but some cells harboring the virus persist for life. Under conditions of immunosuppression, EBV can spread from these cells and cause life-threatening pathologies. We have generated mice expressing the transforming EBV latent membrane protein 1 (LMP1), mimicking a constitutively active CD40 coreceptor, specifically in B cells. Like human EBV-infected cells, LMP1+ B cells were efficiently eliminated by T cells, and breaking immune surveillance resulted in rapid, fatal lymphoproliferation and lymphomagenesis. The lymphoma cells expressed ligands for a natural killer (NK) cell receptor, NKG2D, and could be targeted by an NKG2D-Fc fusion protein. These experiments indicate a central role for LMP1 in the surveillance and transformation of EBV-infected B cells in vivo, establish a preclinical model for B cell lymphomagenesis in immunosuppressed patients, and validate a new therapeutic approach.


Subject(s)
Disease Models, Animal , Herpesvirus 4, Human , Immunologic Surveillance , Lymphoma/immunology , Lymphoma/therapy , Viral Matrix Proteins/metabolism , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Humans , Immunotherapy , Lymphoma/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , NK Cell Lectin-Like Receptor Subfamily K/immunology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Viral Matrix Proteins/genetics
13.
Curr Top Microbiol Immunol ; 349: 97-114, 2011.
Article in English | MEDLINE | ID: mdl-21267708

ABSTRACT

NF-κB transcription factors are critical regulators of many biological processes such as innate and adaptive immune responses, inflammation, cell proliferation and programmed cell death. This versatility necessitates a highly complex and tightly coordinated control of the signaling pathways leading to their activation. Here, we review the role of proteolysis in the regulation of NF-κB activity, more specifically the contribution of the well-known ubiquitin-proteasome system and the involvement of proteolytic activity of caspases and calpains.


Subject(s)
NF-kappa B/physiology , Animals , Calpain/physiology , Caspases/physiology , Humans , I-kappa B Kinase/metabolism , Proteasome Endopeptidase Complex/physiology , Signal Transduction
14.
Nat Immunol ; 10(6): 647-54, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19412180

ABSTRACT

By genetically ablating IkappaB kinase (IKK)-mediated activation of the transcription factor NF-kappaB in the B cell lineage and by analyzing a mouse mutant in which immunoglobulin lambda-chain-positive B cells are generated in the absence of rearrangements in the locus encoding immunoglobulin kappa-chain, we define here two distinct, consecutive phases of early B cell development that differ in their dependence on IKK-mediated NF-kappaB signaling. During the first phase, in which NF-kappaB signaling is dispensable, predominantly kappa-chain-positive B cells are generated, which undergo efficient receptor editing. In the second phase, predominantly lambda-chain-positive B cells are generated whose development is ontogenetically timed to occur after rearrangements of the locus encoding kappa-chain. This second phase of development is dependent on NF-kappaB signals, which can be substituted by transgenic expression of the prosurvival factor Bcl-2.


Subject(s)
B-Lymphocytes/cytology , Immunoglobulin kappa-Chains/metabolism , Immunoglobulin lambda-Chains/metabolism , NF-kappa B/metabolism , Animals , B-Lymphocytes/metabolism , Cell Differentiation , I-kappa B Kinase/genetics , Immunoglobulin kappa-Chains/genetics , Immunoglobulin lambda-Chains/genetics , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , NF-kappa B/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2 , Signal Transduction
15.
Proc Natl Acad Sci U S A ; 105(31): 10883-8, 2008 Aug 05.
Article in English | MEDLINE | ID: mdl-18663224

ABSTRACT

BAFF-R-dependent activation of the alternative NF-kappaB pathway plays an essential role in mature B cell survival. Mutations leading to overexpression of NIK and deletion of the TRAF3 gene are implicated in human multiple myeloma. We show that overexpression of NIK in mouse B lymphocytes amplifies alternative NF-kappaB activation and peripheral B cell numbers in a BAFF-R-dependent manner, whereas uncoupling NIK from TRAF3-mediated control causes maximal p100 processing and dramatic hyperplasia of BAFF-R-independent B cells. NIK controls alternative NF-kappaB signaling by increasing the protein levels of its negative regulator TRAF3 in a dose-dependent fashion. This mechanism keeps NIK protein levels below detection even when they cause B cell hyperplasia, so that contributions of NIK to B cell pathologies can easily be overlooked.


Subject(s)
B-Cell Activating Factor/metabolism , B-Lymphocytes/metabolism , Gene Expression Regulation/immunology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/immunology , Animals , B-Cell Activation Factor Receptor/metabolism , B-Lymphocytes/immunology , Blotting, Northern , Blotting, Western , Cell Proliferation , Cell Survival/immunology , Flow Cytometry , Immunohistochemistry , Mice , NF-kappa B/metabolism , Protein Structure, Tertiary , TNF Receptor-Associated Factor 3/metabolism , NF-kappaB-Inducing Kinase
17.
Immunity ; 24(6): 729-739, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16782029

ABSTRACT

The maintenance of mature B cells hinges on signals emitted from the BAFF-R cell-surface receptor, but the nature of these signals is incompletely understood. Inhibition of canonical NF-kappaB transcription factor activity through ablation of the essential scaffold protein NEMO arrests B cell development at the same stage as BAFF-R deficiency. Correspondingly, activation of this pathway by constitutively active IkappaB Kinase2 renders B cell survival independent of BAFF-R:BAFF interactions and prevents proapoptotic PKCdelta nuclear translocation. In addition, canonical NF-kappaB activity mediates differentiation and proper localization of follicular and marginal zone B cells in the absence of BAFF-R, but not CD19. By replacing BAFF-R signals, constitutive canonical NF-kappaB signaling, a hallmark of various B cell lymphomas, causes accumulation of resting B cells and promotes their proliferation and survival upon activation, but does not per se induce lymphomagenesis. Therefore, canonical NF-kappaB activity can substitute for BAFF-R signals in B cell development and pathogenesis.


Subject(s)
B-Lymphocytes/immunology , I-kappa B Kinase/physiology , Lymphoma, B-Cell/immunology , Membrane Proteins/metabolism , NF-kappa B/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Active Transport, Cell Nucleus , Animals , Antigens, CD19/genetics , Antigens, CD19/metabolism , B-Cell Activation Factor Receptor , Cell Nucleus/enzymology , Cell Proliferation , Hyperplasia , I-kappa B Kinase/genetics , Lymphocyte Activation , Lymphoma, B-Cell/genetics , Membrane Proteins/genetics , Mice , Mice, Mutant Strains , Protein Kinase C-delta/analysis , Protein Kinase C-delta/metabolism , Receptors, Tumor Necrosis Factor/genetics , Spleen/pathology
18.
Biochem Biophys Res Commun ; 308(4): 744-9, 2003 Sep 05.
Article in English | MEDLINE | ID: mdl-12927781

ABSTRACT

P100, which is encoded by NF-kappa B2, inhibits Rel dimers. It can also be processed into p52, one of the DNA binding sub-units of NF-kappa B/Rel factors. Several p100 C-terminal truncations that result from gene rearrangements are associated with lymphomagenesis. Here, we characterized a new p100 mutant that we termed p100HB. It originates from a point-mutation that generates a premature stop-codon, and thus the protein lacks the last 125 amino acids. We have detected p100HB in several human tumor cell lines. The truncated protein is mainly unprocessed, and although it still binds Rel dimers, it has reduced inhibitory potency compared to p100 and translocates into the nucleus. Thus, p100HB may be associated with deregulated NF-kappa B/Rel functions.


Subject(s)
Mutation , NF-kappa B/chemistry , NF-kappa B/genetics , Active Transport, Cell Nucleus , Adaptor Proteins, Signal Transducing , Base Sequence , Blotting, Western , Carrier Proteins/chemistry , Codon , Dimerization , Exons , Gene Expression Regulation , Humans , I-kappa B Proteins/metabolism , Jurkat Cells , Lymphocytes/metabolism , Microscopy, Confocal , Molecular Sequence Data , NF-kappa B p52 Subunit , Plasmids/metabolism , Point Mutation , Precipitin Tests , Protein Structure, Tertiary , Protein Transport , RNA, Messenger/metabolism , Time Factors , Transfection , Tumor Cells, Cultured
19.
J Biol Chem ; 278(26): 23278-84, 2003 Jun 27.
Article in English | MEDLINE | ID: mdl-12709443

ABSTRACT

Tumor necrosis factor-alpha (TNF-alpha) and lymphotoxin-beta receptor (LTbetaR) signaling both play important roles in inflammatory and immune responses through activation of NF-kappaB. Using various deficient mouse embryonic fibroblast cells, we have compared the signaling pathways leading to NF-kappaB induction in response to TNF-alpha and LTbetaR activation. We demonstrate that LTbetaR ligation induces not only RelA/p50 dimers but also RelB/p50 dimers, whereas TNF-alpha induces only RelA/p50 dimers. LTbetaR-induced binding of RelB/p50 requires processing of p100 that is mediated by IKKalpha but is independent of IKKbeta, NEMO/IKKgamma, and RelA. Moreover, we show that RelB, p50, and p100 can associate in the same complex and that TNF-alpha but not LTbeta signaling increases the association of p100 with RelB/p50 dimers in the nucleus, leading to the specific inhibition of RelB DNA binding. These results suggest that the alternative NF-kappaB pathway based on p100 processing may account not only for the activation of RelB/p52 dimers but also for that of RelB/p50 dimers and that p100 regulates the binding activity of RelB/p50 dimers via at least two distinct mechanisms depending on the signaling pathway involved.


Subject(s)
NF-kappa B/metabolism , Nuclear Proteins/physiology , Proto-Oncogene Proteins/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/physiology , Active Transport, Cell Nucleus , Animals , DNA/metabolism , Dimerization , Endonucleases , Fibroblasts/metabolism , Gene Expression Regulation , I-kappa B Kinase , I-kappa B Proteins/metabolism , Lymphotoxin beta Receptor , Mice , NF-kappa B p50 Subunit , Nuclear Proteins/metabolism , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/antagonists & inhibitors , Receptors, Tumor Necrosis Factor/physiology , Transcription Factor RelB , Transcription Factors/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...