Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters










Publication year range
1.
Immunol Cell Biol ; 94(1): 66-78, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26084385

ABSTRACT

Defects in apoptosis can cause autoimmune disease. Loss-of-function mutations in the 'death receptor' FAS impair the deletion of autoreactive lymphocytes in the periphery, leading to progressive lymphadenopathy and systemic lupus erythematosus-like autoimmune disease in mice (Fas(lpr/lpr) (mice homozygous for the lymphoproliferation inducing spontaneous mutation)) and humans. The REL/nuclear factor-κB (NF-κB) transcription factors regulate a broad range of immune effector functions and are also implicated in various autoimmune diseases. We generated compound mutant mice to investigate the individual functions of the NF-κB family members NF-κB1, NF-κB2 and c-REL in the various autoimmune pathologies of Fas(lpr/lpr) mutant mice. We show that loss of each of these transcription factors resulted in amelioration of many classical features of autoimmune disease, including hypergammaglobulinaemia, anti-nuclear autoantibodies and autoantibodies against tissue-specific antigens. Remarkably, only c-REL deficiency substantially reduced immune complex-mediated glomerulonephritis and extended the lifespan of Fas(lpr/lpr) mice. Interestingly, compared with the Fas(lpr/lpr) animals, Fas(lpr/lpr)nfkb2(-/-) mice presented with a dramatic acceleration and augmentation of lymphadenopathy that was accompanied by severe lung pathology due to extensive lymphocytic infiltration. The Fas(lpr/lpr)nfkb1(-/-) mice exhibited the combined pathologies caused by defects in FAS-mediated apoptosis and premature ageing due to loss of NF-κB1. These findings demonstrate that different NF-κB family members exert distinct roles in the development of the diverse autoimmune and lymphoproliferative pathologies that arise in Fas(lpr/lpr) mice, and suggest that pharmacological targeting of c-REL should be considered as a strategy for therapeutic intervention in autoimmune diseases.


Subject(s)
Lupus Erythematosus, Systemic/complications , Lymphatic Diseases/complications , NF-kappa B p50 Subunit/deficiency , NF-kappa B p52 Subunit/deficiency , Proto-Oncogene Proteins c-rel/metabolism , fas Receptor/metabolism , Animals , Autoantibodies/blood , Chemokines/blood , Chemokines/metabolism , Dermatitis/blood , Dermatitis/complications , Dermatitis/immunology , Forkhead Transcription Factors/metabolism , Genotype , Hypergammaglobulinemia/blood , Hypergammaglobulinemia/complications , Immune Tolerance/immunology , Leukocytes/pathology , Longevity , Lupus Erythematosus, Systemic/blood , Lymphatic Diseases/blood , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , NF-kappa B p50 Subunit/metabolism , NF-kappa B p52 Subunit/metabolism , Organ Specificity , Splenomegaly/blood , Transcription Factors/metabolism , AIRE Protein
2.
Cell Death Differ ; 22(5): 767-78, 2015 May.
Article in English | MEDLINE | ID: mdl-25361085

ABSTRACT

FASL/FAS signaling imposes a critical barrier against autoimmune disease and lymphadenopathy. Mutant mice unable to produce membrane-bound FASL (FasL(Δm/Δm)), a prerequisite for FAS-induced apoptosis, develop lymphadenopathy and systemic autoimmune disease with immune complex-mediated glomerulonephritis. Prior to disease onset, FasL(Δm/Δm) mice contain abnormally high numbers of leukocytes displaying activated and elevated NF-κB-regulated cytokine levels, indicating that NF-κB-dependent inflammation may be a key pathological driver in this multifaceted autoimmune disease. We tested this hypothesis by genetically impairing canonical or non-canonical NF-κB signaling in FasL(Δm/Δm) mice by deleting the c-Rel or NF-κB2 genes, respectively. Although the loss of NF-κB2 reduced the levels of inflammatory cytokines and autoantibodies, the impact on animal survival was minor due to substantially accelerated and exacerbated lymphoproliferative disease. In contrast, a marked increase in lifespan resulting from the loss of c-REL coincided with a striking reduction in classical parameters of autoimmune pathology, including the levels of cytokines and antinuclear autoantibodies. Notably, the decrease in regulatory T-cell numbers associated with loss of c-REL did not exacerbate autoimmunity in FasL(Δm/Δm)c-rel(-/-) mice. These findings indicate that selective inhibition of c-REL may be an attractive strategy for the treatment of autoimmune pathologies driven by defects in FASL/FAS signaling that would be expected to circumvent many of the complications caused by pan-NF-κB inhibition.


Subject(s)
Autoimmune Diseases/metabolism , Fas Ligand Protein/metabolism , Mutation , NF-kappa B p52 Subunit/metabolism , Proto-Oncogene Proteins c-rel/metabolism , fas Receptor/metabolism , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , Autoimmune Diseases/prevention & control , Fas Ligand Protein/genetics , Mice , Mice, Knockout , NF-kappa B p52 Subunit/genetics , Proto-Oncogene Proteins c-rel/genetics , Signal Transduction/genetics , fas Receptor/genetics
3.
Blood Cancer J ; 3: e105, 2013 Mar 22.
Article in English | MEDLINE | ID: mdl-23524590

ABSTRACT

The Ras/Raf/MEK/extracellular signal regulated kinase (ERK) (Ras/mitogen-activated protein kinases (MAPK)) signal transduction pathway is a crucial mediator of many fundamental biological processes, including cellular proliferation, survival, angiogenesis and migration. Aberrant signalling through the Ras/MAPK cascade is common in a wide array of malignancies, including multiple myeloma (MM), making it an appealing candidate for the development of novel targeted therapies. In this review, we explore our current understanding of the Ras/MAPK pathway and its role in MM. Additionally, we summarise the current status of small molecule inhibitors of MEK under clinical evaluation, and discuss future approaches required to optimise their use.

5.
Oncogene ; 25(51): 6781-99, 2006 Oct 30.
Article in English | MEDLINE | ID: mdl-17072328

ABSTRACT

The nuclear factor-kappaB (NF-kappaB) signalling pathway serves a crucial role in regulating the transcriptional responses of physiological processes that include cell division, cell survival, differentiation, immunity and inflammation. Here we outline studies using mouse models in which the core components of the NF-kappaB pathway, namely the IkappaB kinase subunits (IKKalpha, IKKbeta and NEMO), the IkappaB proteins (IkappaBalpha, IkappaBbeta, IkappaBvarepsilon and Bcl-3) and the five NF-kappaB transcription factors (NF-kappaB1, NF-kappaB2, c-Rel, RelA and RelB), have been genetically manipulated using transgenic and knockout technology.


Subject(s)
Models, Animal , NF-kappa B/metabolism , Signal Transduction , Animals , Female , Male , Mice , Mice, Knockout , Mice, Transgenic
7.
Immunity ; 15(4): 545-55, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11672537

ABSTRACT

PU.1(+/-)Spi-B(-/-) mice exhibit reduced numbers of immature and mature B lymphocytes, which exhibit severe defects in response to BCR-mediated stimulation and poor survival. We found that expression of c-rel, a member of the Rel/NF-kappa B family, is dramatically reduced in PU.1(+/-)Spi-B(-/-) splenic B cells. Analysis of the murine c-rel promoter identified three PU.1/Spi-B binding sites critical for c-rel promoter activity. Furthermore, reintroduction of Rel protein restored wild-type B cell numbers to mice reconstituted with PU.1(+/-)Spi-B(-/-) bone marrow. These findings are the first to demonstrate that a member of the Rel/NF-kappa B family is directly regulated by Ets proteins and dissect the molecular basis for the function of two Ets factors, PU.1 and Spi-B, in promoting B lymphocyte survival.


Subject(s)
B-Lymphocytes/immunology , DNA-Binding Proteins/physiology , Proto-Oncogene Proteins c-rel/genetics , Proto-Oncogene Proteins/physiology , Trans-Activators/physiology , Transcription Factors/physiology , Animals , Binding Sites , Bone Marrow Cells/immunology , Bone Marrow Transplantation , Cell Line , Cell Survival , Cells, Cultured , DNA-Binding Proteins/genetics , Down-Regulation , Gene Expression Regulation , Mice , Mice, Knockout , Promoter Regions, Genetic , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-rel/biosynthesis , RNA, Messenger/biosynthesis , Spleen/immunology , Trans-Activators/genetics , Transcription Factors/genetics , Transfection
8.
J Exp Med ; 194(8): 1021-32, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11602633

ABSTRACT

Interleukin 12 (IL-12) is a 70-kD proinflammatory cytokine produced by antigen presenting cells that is essential for the induction of T helper type 1 development. It comprises 35-kD (p35) and 40-kD (p40) polypeptides encoded by separate genes that are induced by a range of stimuli that include lipopolysaccharide (LPS), DNA, and CD40 ligand. To date, the regulation of IL-12 expression at the transcriptional level has mainly been examined in macrophages and restricted almost exclusively to the p40 gene. Here we show that in CD8(+) dendritic cells, major producers of IL-12 p70, the Rel/nuclear factor (NF)-kappaB signaling pathway is necessary for the induction of IL-12 in response to microbial stimuli. In contrast to macrophages which require c-Rel for p40 transcription, in CD8(+) dendritic cells, the induced expression of p35 rather than p40 by inactivated Staphylococcus aureus, DNA, or LPS is c-Rel dependent and regulated directly by c-Rel complexes binding to the p35 promoter. This data establishes the IL-12 p35 gene as a new target of c-Rel and shows that the regulation of IL-12 p70 expression at the transcriptional level by Rel/NF-kappaB is controlled through both the p35 and p40 genes in a cell type-specific fashion.


Subject(s)
CD8 Antigens , Dendritic Cells/immunology , Gene Expression Regulation , Interleukin-12/genetics , Proto-Oncogene Proteins c-rel/metabolism , Transcription, Genetic , Animals , Biomarkers , Dendritic Cells/cytology , Female , Interleukin-12/biosynthesis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Promoter Regions, Genetic , Proto-Oncogene Proteins c-rel/genetics , Proto-Oncogene Proteins c-rel/physiology
9.
J Immunol ; 166(8): 4949-57, 2001 Apr 15.
Article in English | MEDLINE | ID: mdl-11290773

ABSTRACT

The Fas death receptor plays a key role in the killing of target cells by NK cells and CTLs and in activation-induced cell death of mature T lymphocytes. These cytotoxic pathways are dependent on induction of Fas expression by cytokines such as TNF-alpha and IFN-gamma or by signals generated after TCR engagement. Although much of our knowledge of the Fas death pathway has been generated from murine studies, little is known about regulatory mechanisms important for murine Fas expression. To this end, we have molecularly cloned a region of the murine Fas promoter that is responsible for mediating TNF-alpha and PMA/PHA-induced expression. We demonstrate here that induction of Fas expression by both stimuli is critically dependent on two sites that associate with RelA-containing NF-kappaB complexes. To determine whether RelA and/or other NF-kappaB subunits are also important for regulating Fas expression in primary T cells, we used CD4 T cells from RelA(-/-), c-Rel(-/-), and p50(-/-) mice. Although proliferative responses were significantly impaired, expression of Fas and activation-induced cell death was unaffected in T cells obtained from these different mice. Importantly, we show that unlike fibroblasts, which consist primarily of RelA-containing NF-kappaB complexes, T cells have high levels of both RelA and c-Rel complexes, suggesting that Fas expression in T cells may be dependent on redundant functions of these NF-kappaB subunits.


Subject(s)
Apoptosis/immunology , Lymphocyte Activation , NF-kappa B/physiology , Receptors, Antigen, T-Cell/physiology , Tumor Necrosis Factor-alpha/physiology , fas Receptor/biosynthesis , 3T3 Cells , Animals , Apoptosis/drug effects , Apoptosis/genetics , Base Sequence , Binding Sites/genetics , Binding Sites/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Cloning, Molecular , Down-Regulation/genetics , Down-Regulation/immunology , Fas Ligand Protein , Fibroblasts/metabolism , Humans , Jurkat Cells , Ligands , Lymphocyte Activation/drug effects , Lymphocyte Activation/genetics , Membrane Glycoproteins/biosynthesis , Mice , Mice, Inbred C57BL , Molecular Sequence Data , NF-kappa B/deficiency , NF-kappa B/genetics , NF-kappa B/metabolism , NF-kappa B p50 Subunit , Phytohemagglutinins/pharmacology , Promoter Regions, Genetic/immunology , Proto-Oncogene Proteins c-rel/deficiency , Proto-Oncogene Proteins c-rel/genetics , Tetradecanoylphorbol Acetate/pharmacology , Transcription Factor RelA , fas Receptor/genetics , fas Receptor/metabolism
11.
EMBO J ; 19(23): 6351-60, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-11101508

ABSTRACT

Rel and RelA, individually dispensable for lymphopoiesis, serve unique functions in activated B and T cells. Here their combined roles in lymphocyte development were examined in chimeric mice repopulated with c-rel(-/-) rela(-/-) fetal liver hemopoietic stem cells. Mice engrafted with double-mutant cells lacked mature IgM(lo)IgD(hi) B cells, and numbers of peripheral CD4(+) and CD8(+) T cells were markedly reduced. The absence of mature B cells was associated with impaired survival that coincided with reduced expression of bcl-2 and A1. bcl-2 transgene expression not only prevented apoptosis and increased peripheral B-cell numbers, but also induced further maturation to an IgM(lo)IgD(hi) phenotype. In contrast, the survival of double-mutant T cells was normal and the bcl-2 transgene could not rectify the peripheral T-cell deficit. These findings indicate that Rel and RelA serve essential, albeit redundant, functions during the later antigen-independent stages of B- and T-cell maturation, with these transcription factors promoting the survival of peripheral B cells in part by upregulating Bcl-2.


Subject(s)
NF-kappa B/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-rel/metabolism , Up-Regulation , Animals , Apoptosis , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Survival , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fluorescent Antibody Technique , Hematopoietic Stem Cells , Immunoglobulin D/metabolism , Liver/embryology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mutation , NF-kappa B/genetics , Phenotype , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-rel/genetics , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/metabolism , Time Factors , Transcription Factor RelA , Transcription Factors/metabolism , Transgenes
12.
Immunol Rev ; 176: 134-40, 2000 Aug.
Article in English | MEDLINE | ID: mdl-11043773

ABSTRACT

The Rel/nuclear factor (NF)-kappaB family of transcription factors have been implicated in the regulation of a wide variety of genes, in particular those encoding proteins crucial to the function of the immune system. Through the use of mutant mice that lack one or more of these proteins, we have begun to examine the individual and combined roles of Rel, RelA and NF-kappaB1 in B-cell development and function. Here we outline and discuss how these transcription factors operate as differentiation stage-specific regulators of B-cell development, survival, division and immunoglobulin expression, emphasizing those Rel/NF-kappaB-regulated genes that mediate these functions.


Subject(s)
B-Lymphocytes/immunology , B-Lymphocytes/metabolism , NF-kappa B/metabolism , Proto-Oncogene Proteins c-rel/metabolism , Animals , B-Lymphocytes/cytology , Cell Differentiation , Cell Division , Gene Expression Regulation , Genes, rel , Humans , Immunoglobulin Isotypes/metabolism , Lymphocyte Activation , Mice
13.
J Clin Invest ; 105(12): 1799-806, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10862795

ABSTRACT

Rheumatoid arthritis (RA) is a complex disease, with contributions from systemic autoimmunity and local inflammation. Persistent synovial joint inflammation and invasive synovial pannus tissue lead to joint destruction. RA is characterized by the production of inflammatory mediators, many of which are regulated by the Rel/NF-kappaB transcription factors. Although an attractive target for therapeutic intervention in inflammatory diseases, Rel/NF-kappaB is involved in normal physiology, thus global inhibition could be harmful. An alternate approach is to identify and target the Rel/NF-kappaB subunits critical for components of disease. To assess this, mice with null mutations in c-rel or nfkb1 were used to examine directly the roles of c-Rel and p50 in models of acute and chronic inflammatory arthritis. We found c-Rel-deficient mice were resistant to collagen-induced arthritis but had a normal response in an acute, destructive arthritis model (methylated BSA/IL-1 induced arthritis) suggesting c-Rel is required for systemic but not local joint disease. In contrast, p50-deficient mice were refractory to induction of both the chronic and acute arthritis models, showing this subunit is essential for local joint inflammation and destruction. Our data suggest Rel/NF-kappaB subunits play distinct roles in the pathogenesis of inflammatory arthritis and may provide a rationale for more specific therapeutic blockade of Rel/NF-kappaB in RA.


Subject(s)
Arthritis, Experimental/genetics , Genes, rel , NF-kappa B/metabolism , Proto-Oncogene Proteins c-rel/metabolism , Acute Disease , Animals , Arthritis, Experimental/pathology , Arthritis, Experimental/physiopathology , Chronic Disease , Collagen/immunology , Crosses, Genetic , Inflammation , Joints/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/deficiency , NF-kappa B/genetics , NF-kappa B p50 Subunit , Proto-Oncogene Proteins c-rel/deficiency , Transcription Factors/metabolism
14.
J Exp Med ; 191(8): 1281-92, 2000 Apr 17.
Article in English | MEDLINE | ID: mdl-10770796

ABSTRACT

In lymphocytes, the Rel transcription factor is essential in establishing a pattern of gene expression that promotes cell proliferation, survival, and differentiation. Here we show that mitogen-induced expression of interferon (IFN) regulatory factor 4 (IRF-4), a lymphoid-specific member of the IFN family of transcription factors, is Rel dependent. Consistent with IRF-4 functioning as a repressor of IFN-induced gene expression, the absence of IRF-4 expression in c-rel(-/-) B cells coincided with a greater sensitivity of these cells to the antiproliferative activity of IFNs. In turn, enforced expression of an IRF-4 transgene restored IFN modulated c-rel(-/-) B cell proliferation to that of wild-type cells. This cross-regulation between two different signaling pathways represents a novel mechanism that Rel/nuclear factor kappaB can repress the transcription of IFN-regulated genes in a cell type-specific manner.


Subject(s)
B-Lymphocytes/metabolism , DNA-Binding Proteins/genetics , NF-kappa B/metabolism , Proto-Oncogene Proteins c-rel/metabolism , Transcription Factors/genetics , Animals , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Base Sequence , DNA Primers/genetics , Gene Expression Regulation/drug effects , Humans , Interferons/metabolism , Interferons/pharmacology , Jurkat Cells , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogens/pharmacology , NF-kappa B/genetics , Promoter Regions, Genetic , Proto-Oncogene Proteins c-rel/genetics
15.
Int J Biochem Cell Biol ; 31(10): 1209-19, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10582348

ABSTRACT

In mammals, Rel/NF-kappa B proteins are a small family of transcription factors which serve as pivotal regulators of immune, inflammatory and acute phase responses. Pathways leading to the activation of Rel/NF-kappa B have recently been dissected in some detail and shown to converge on a unique high molecular weight cytoplasmic complex that includes several kinases and regulatory molecules. Moreover, gene targeting experiments have identified novel roles for Rel/NF-kappa B proteins in the development and maturation of hemopoietic precursors as well as in the function of mature cells in the immune system. These include regulating the cell cycle, controlling cell survival and providing a link between the innate and adaptive immune systems. Since the dysregulation of Rel/NF-kappa B function is associated with various pathologies including inflammatory and neoplastic disease, new insights into the role of Rel/NF-kappa B in human disease may provide a basis for therapeutic strategies in the treatment of chronic inflammatory diseases and certain malignancies.


Subject(s)
Hematopoiesis/physiology , NF-kappa B/physiology , Proto-Oncogene Proteins c-rel/physiology , Proto-Oncogene Proteins/physiology , Transcription Factors/physiology , Animals , Cell Cycle , Cell Survival , Humans , Inflammation/physiopathology , NF-kappa B/immunology , NF-kappa B p50 Subunit , Neoplasms/physiopathology , Proto-Oncogene Proteins/immunology , Proto-Oncogene Proteins c-rel/immunology , Signal Transduction , Transcription Factor RelA , Transcription Factor RelB , Transcription Factors/immunology
16.
Oncogene ; 18(49): 6888-95, 1999 Nov 22.
Article in English | MEDLINE | ID: mdl-10602464

ABSTRACT

Rel/NF-kappaB transcription factors have been implicated in regulating a wide variety of genes important in cellular processes that include cell division, cell survival, differentiation and immunity. Here genetic models in which various Rel/NF-kappaB and IkappaB proteins have either been over-expressed or deleted in mice will be reviewed. Although expressed fairly ubiquitously, homozygous disruption of individual Rel/NF-kappaB genes generally affects the development of proper immune cell function. One exception is rela, which is essential for embryonic liver development. The disruption of genes encoding the individual subunits of the IkappaB kinase, namely IKKalpha and IKKbeta, has demonstrated that IKKbeta transmits the response to most common NF-kappaB inducing agents, whereas IKKalpha has an unexpected role in keratinocyte differentiation. Future studies will no doubt focus on the effect of multiple gene disruptions of members of this signaling pathway, on tissue-specific disruptions of these genes, and on the use of these mice as models for human diseases.


Subject(s)
NF-kappa B/physiology , Proto-Oncogene Proteins/physiology , Transcription Factors/physiology , Animals , B-Cell Lymphoma 3 Protein , I-kappa B Kinase , Mice , Mice, Knockout , Mice, Transgenic , Mutation , Protein Serine-Threonine Kinases/physiology , Transcription Factor RelA , Transcription Factor RelB
17.
Proc Natl Acad Sci U S A ; 96(21): 11848-53, 1999 Oct 12.
Article in English | MEDLINE | ID: mdl-10518539

ABSTRACT

Individual Rel/NF-kappaB transcription factors, although dispensable for the development and maturation of most hemopoietic cells, are critical regulators of normal immune function. Redundancy among these proteins prompted us to examine the role of Rel and RelA in hemopoiesis by using mice that lack both subunits. Because of the death of double-mutant fetuses at day 13.5 of gestation (E13.5), E12 fetal liver hemopoietic progenitors were used for in vitro cultures and for repopulating stem cell studies in lethally irradiated normal recipient mice. Most striking, Rel/RelA-deficient hemopoietic precursors failed to promote the survival of myeloablated mice. This phenotype was associated with several defects including a reduction of spleen colony-forming unit progenitors, impaired erythropoiesis, and a deregulated expansion of granulocytes. In vitro progenitor assays also revealed that Rel or RelA serves an antiapoptotic role during monocyte differentiation. Despite the combined loss of Rel and RelA leading to these hemopoietic defects, c-rel(-/-)rela(-/-) stem cells contributed to the development of all lineages in mice engrafted with double-mutant fetal liver cells and normal bone marrow cells, albeit in a reduced fashion compared with controls. Collectively, these data indicate the loss of Rel and RelA does not appear to affect pluripotent stem cells; rather, Rel and RelA serve redundant functions in regulating differentiation and survival of committed progenitors in multiple hemopoietic lineages.


Subject(s)
Hematopoietic Stem Cells/physiology , NF-kappa B/genetics , NF-kappa B/physiology , Proto-Oncogene Proteins c-rel/genetics , Proto-Oncogene Proteins c-rel/physiology , Animals , Cell Differentiation , Cells, Cultured , Erythropoiesis , Flow Cytometry , Gene Expression Regulation, Developmental , Liver/embryology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Monocytes/metabolism , Stem Cells , Time Factors , Up-Regulation
18.
Int Immunol ; 11(9): 1431-9, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10464164

ABSTRACT

Rel, a haemopoietic cell-restricted member of the NF-kappaB/Rel family of transcription factors, has recently been shown to be important in the function of B and T lymphocytes. In an attempt to understand the role of this protein in the immune response, we examined the ability of Rel(-/-) mice to counter an influenza virus infection. Normal levels of virus-specific cytotoxic T cells induced in Rel(-/-) mice were able to clear virus from the lungs, albeit with somewhat delayed kinetics compared to normal mice. Rel(-/-) mice did, however, display a markedly reduced T cell proliferative response to the virus, and exhibited impaired local and systemic influenza virus-specific antibody responses. This defect was sufficient to result in an inability of vaccinated mice, but not of previously infected mice, to acquire antibody-dependent protective immunity to reinfection with the same virus. These findings establish that during the response to influenza virus, Rel function allows optimal development of humoral immunity, a role that apparently cannot be fulfilled by other NF-kappaB/Rel proteins.


Subject(s)
Antibodies, Viral/blood , Orthomyxoviridae Infections/immunology , Proto-Oncogene Proteins c-rel/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antibody Specificity , Disease Models, Animal , Immunity, Cellular , Lung/immunology , Lung/virology , Mice , Mice, Knockout/virology , Orthomyxoviridae/immunology , Orthomyxoviridae Infections/virology , T-Lymphocytes, Cytotoxic/metabolism , Vaccination
19.
Eur J Immunol ; 29(3): 928-35, 1999 03.
Article in English | MEDLINE | ID: mdl-10092097

ABSTRACT

The family of Rel/NF-kappaB transcription factors is a crucial regulator of various cellular responses. Using Rel-deficient (c-rel-/-) mice crossed with T cell receptor (TCR)-transgenic mice we show that Rel is neither required for positive selection of major histocompatibility complex (MHC)-restricted T cells nor for deletion of thymocytes bearing autoreactive antigen receptors. Our studies also demonstrate that Rel is dispensable for T lymphocyte apoptosis. Rel is, however, essential for antigen-induced activation of mature T cells and its absence exacerbates the anergic state. These results indicate that thymocytes and mature T cells differ in their requirement for Rel in mediating TCR-induced responses.


Subject(s)
Apoptosis/immunology , Proto-Oncogene Proteins/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocytes/immunology , Thymus Gland/immunology , Transcription Factors/immunology , Animals , CD3 Complex/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation , Cell Nucleus/metabolism , Clonal Anergy , Female , Isoantibodies/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-rel , Receptors, Antigen, T-Cell, alpha-beta/genetics , T-Lymphocytes/cytology , Thymus Gland/cytology , Transcription Factors/genetics
20.
Genes Dev ; 13(4): 400-11, 1999 Feb 15.
Article in English | MEDLINE | ID: mdl-10049356

ABSTRACT

In response to different extracellular signals, Rel/NF-kappaB transcription factors are critical regulators of apoptosis in a variety of cell types. Here we show that in normal B and T cells, expression of the Bcl-2 prosurvival homolog, A1, is rapidly induced in a Rel-dependent manner by mitogens. In B-cell lines derived from c-rel-/- mice, which like primary cells lacking Rel undergo apoptosis in response to antigen receptor ligation, constitutive expression of an A1 transgene inhibits this pathway to cell death. These findings are the first to show that Rel/NF-kappaB regulates physiologically the expression of a Bcl-2-like protein that is critical for the control of cell survival during lymphocyte activation.


Subject(s)
Apoptosis/genetics , B-Lymphocytes/metabolism , Proteins/genetics , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins/metabolism , Receptors, Antigen, B-Cell/metabolism , Transcription, Genetic/genetics , Animals , Base Sequence , DNA-Binding Proteins/genetics , Gene Expression Regulation/genetics , Lymphocyte Activation/drug effects , Mice , Mice, Knockout , Minor Histocompatibility Antigens , Mitogens/pharmacology , Molecular Sequence Data , NF-kappa B/genetics , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-rel , RNA, Messenger/metabolism , Sequence Analysis, DNA , T-Lymphocytes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...