Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Front Immunol ; 12: 667922, 2021.
Article in English | MEDLINE | ID: mdl-34194431

ABSTRACT

Group 2 innate lymphoid cells (ILC2s) are emerging as important cellular regulators of homeostatic and disease-associated immune processes. The cytokine interleukin-33 (IL-33) promotes ILC2-dependent inflammation and immunity, with IL-33 having been shown to activate NF-κB in a wide variety of cell types. However, it is currently unclear which NF-κB members play an important role in IL-33-dependent ILC2 biology. Here, we identify the NF-κB family member c-Rel as a critical component of the IL-33-dependent activation of ILC2s. Although c-Rel is dispensable for ILC2 development, it is critical for ILC2 function in the lung, with c-Rel-deficient (c-Rel-/- ) mice present a significantly reduced response to papain- and IL-33-induced lung inflammation. We also show that the absence of c-Rel reduces the IL-33-dependent expansion of ILC2 precursors and lower levels of IL-5 and IL-13 cytokine production by mature ILC2s in the lung. Together, these results identify the IL-33-c-Rel axis as a central control point of ILC2 activation and function.


Subject(s)
Immunity, Innate/drug effects , Interleukin-33/pharmacology , Lung/drug effects , Lymphocyte Activation/drug effects , Lymphocytes/drug effects , Pneumonia/metabolism , Proto-Oncogene Proteins c-rel/metabolism , Animals , Bone Marrow/drug effects , Bone Marrow/immunology , Bone Marrow/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Interleukin-13/metabolism , Interleukin-5/metabolism , Lung/immunology , Lung/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B p50 Subunit/genetics , NF-kappa B p50 Subunit/metabolism , Papain , Pneumonia/chemically induced , Pneumonia/genetics , Pneumonia/immunology , Proto-Oncogene Proteins c-rel/genetics
3.
J Autoimmun ; 70: 52-62, 2016 06.
Article in English | MEDLINE | ID: mdl-27068879

ABSTRACT

The properties of CD4(+) regulatory T cell (Treg) subsets are dictated by distinct patterns of gene expression determined by FOXP3 and different combinations of various transcription factors. Here we show the NF-κB transcription factor RelA is constitutively active in naïve and effector Tregs. The conditional inactivation of Rela in murine FOXP3(+) cells induces a rapid onset, multi-focal autoimmune disease that depends on RelA being expressed in conventional T cells. In addition to promoting Treg lineage stability, RelA determines the size of the effector Treg population, a function influenced by the presence or absence of RelA in conventional T cells. These findings showing that RelA controls Treg stability and promotes the competitive fitness of effector Tregs highlight the importance of RelA activity in peripheral Treg induced tolerance.


Subject(s)
Immune Tolerance , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Transcription Factor RelA/metabolism , Animals , Antibodies/blood , Antibodies/immunology , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Autoimmune Diseases/pathology , Autoimmunity , Biomarkers , Cluster Analysis , Cytokines/blood , Cytokines/metabolism , Disease Models, Animal , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Profiling , Immune Tolerance/genetics , Immunomodulation , Immunophenotyping , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Lymphocyte Count , Male , Mice , Mice, Transgenic , Phenotype , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Transcription Factor RelA/genetics
4.
PLoS One ; 9(11): e111524, 2014.
Article in English | MEDLINE | ID: mdl-25375135

ABSTRACT

Alterations in the immune cell profile and the induction of inflammation within adipose tissue are a hallmark of obesity in mice and humans. Dual-specificity phosphatase 2 (DUSP2) is widely expressed within the immune system and plays a key role promoting immune and inflammatory responses dependent on mitogen-activated protein kinase (MAPK) activity. We hypothesised that the absence of DUSP2 would protect mice against obesity-associated inflammation and insulin resistance. Accordingly, male and female littermate mice that are either wild-type (wt) or homozygous for a germ-line null mutation of the dusp2 gene (dusp2-/-) were fed either a standard chow diet (SCD) or high fat diet (HFD) for 12 weeks prior to metabolic phenotyping. Compared with mice fed the SCD, all mice consuming the HFD became obese, developed glucose intolerance and insulin resistance, and displayed increased macrophage recruitment and markers of inflammation in epididymal white adipose tissue. The absence of DUSP2, however, had no effect on the development of obesity or adipose tissue inflammation. Whole body insulin sensitivity in male mice was unaffected by an absence of DUSP2 in response to either the SCD or HFD; however, HFD-induced insulin resistance was slightly, but significantly, reduced in female dusp2-/- mice. In conclusion, DUSP2 plays no role in regulating obesity-associated inflammation and only a minor role in controlling insulin sensitivity following HFD in female, but not male, mice. These data indicate that rather than DUSP2 being a pan regulator of MAPK dependent immune cell mediated inflammation, it appears to differentially regulate inflammatory responses that have a MAPK component.


Subject(s)
Dual Specificity Phosphatase 2/genetics , Glucose Intolerance/genetics , Inflammation/genetics , Insulin Resistance/genetics , Obesity/genetics , Adipose Tissue/metabolism , Animals , Blood Glucose/metabolism , Diet, High-Fat , Dual Specificity Phosphatase 2/metabolism , Female , Glucose Intolerance/metabolism , Glucose Tolerance Test , Inflammation/etiology , Inflammation/metabolism , Insulin/blood , Male , Mice , Mutation , Obesity/complications , Obesity/metabolism , Sex Factors
5.
Nat Immunol ; 15(1): 15-25, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24352326

ABSTRACT

The NF-κB signal transduction pathway is best known as a major regulator of innate and adaptive immune responses, yet there is a growing appreciation of its importance in immune cell development, particularly of T lineage cells. In this Review, we discuss how the temporal regulation of NF-κB controls the stepwise differentiation and antigen-dependent selection of conventional and specialized subsets of T cells in response to T cell receptor and costimulatory, cytokine and growth factor signals.


Subject(s)
NF-kappa B/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Thymocytes/immunology , Cell Lineage/immunology , Humans , Models, Immunological , NF-kappa B/metabolism , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Thymocytes/cytology , Thymocytes/metabolism
6.
Transcription ; 3(5): 245-9, 2012.
Article in English | MEDLINE | ID: mdl-22885978

ABSTRACT

In the thymus, sequential antigen and cytokine receptor signals direct the stepwise differentiation of multi-potential CD4+CD8+ thymic precursors into Foxp3+ CD4 regulatory T cells (Tregs). In this Point of View article we discuss our recent findings about how the c-Rel transcription factor orchestrates this developmental process.


Subject(s)
Proto-Oncogene Proteins c-rel/metabolism , T-Lymphocytes, Regulatory/cytology , Animals , Cell Differentiation , Forkhead Transcription Factors/metabolism , Mice , Mice, Transgenic , T-Lymphocytes, Regulatory/metabolism , Thymus Gland/cytology , Thymus Gland/metabolism
7.
Immunol Rev ; 246(1): 272-85, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22435561

ABSTRACT

Although the diverse functions served by the nuclear factor-κB (NF-κB) pathway in virtually all cell types are typically employed to deal with stress responses, NF-κB transcription factors also play key roles in the development of hemopoietic cells. This review focuses on how NF-κB transcription factors control various aspects of thymic T-cell and myeloid cell differentiation that include its roles in hemopoietic precursors, conventional αß T cells, CD4(+) regulatory T cells, natural killer T cells, γδ T cells, macrophages, and dendritic cells.


Subject(s)
Hematopoiesis/physiology , NF-kappa B/chemistry , NF-kappa B/metabolism , Protein Subunits/metabolism , Animals , Cell Lineage , Humans , Myeloid Cells/metabolism , T-Lymphocytes/metabolism , Thymocytes/metabolism
8.
EMBO J ; 31(3): 692-706, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22124325

ABSTRACT

The role of specific members of the NF-κB family of transcription factors in CD8 T-cell selection and development is largely unknown. Here, we show that mice lacking NF-κB1 develop a unique population of conventional CD8 single-positive (SP) thymocytes with memory T cell-like properties that populate peripheral immune organs. Development of this memory-like population is not due to PLZF(+) thymocytes and instead coincides with changes in CD8 T-cell selection. These include a reduction in the efficiency of negative selection and a dependence on MHC class Ia or Ib expressed by haematopoietic cells. These findings indicate that NF-κB1 regulates multiple events in the thymus that collectively inhibit the excess development of CD8(+) thymocytes with memory cell characteristics.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/physiology , NF-kappa B/physiology , Thymus Gland/cytology , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Immunophenotyping , Interleukin-4/biosynthesis , NF-kappa B/genetics , Signal Transduction
9.
J Exp Med ; 206(13): 3001-14, 2009 Dec 21.
Article in English | MEDLINE | ID: mdl-19995950

ABSTRACT

During thymopoiesis, a unique program of gene expression promotes the development of CD4 regulatory T (T reg) cells. Although Foxp3 maintains a pattern of gene expression necessary for T reg cell function, other transcription factors are emerging as important determinants of T reg cell development. We show that the NF-kappaB transcription factor c-Rel is highly expressed in thymic T reg cells and that in c-rel(-/-) mice, thymic T reg cell numbers are markedly reduced as a result of a T cell-intrinsic defect that is manifest during thymocyte development. Although c-Rel is not essential for TGF-beta conversion of peripheral CD4(+)CD25(-) T cells into CD4(+)Foxp3(+) cells, it is required for optimal homeostatic expansion of peripheral T reg cells. Despite a lower number of peripheral T reg cells in c-rel(-/-) mice, the residual peripheral c-rel(-/-) T reg cells express normal levels of Foxp3, display a pattern of cell surface markers and gene expression similar to those of wild-type T reg cells, and effectively suppress effector T cell function in culture and in vivo. Collectively, our results indicate that c-Rel is important for both the thymic development and peripheral homeostatic proliferation of T reg cells.


Subject(s)
Forkhead Transcription Factors/physiology , Lymphopoiesis , Proto-Oncogene Proteins c-rel/physiology , T-Lymphocytes, Regulatory/physiology , Animals , Animals, Newborn , Cell Survival , Colitis/prevention & control , Genes, bcl-2 , Lymphocyte Activation , Lymphocyte Count , Mice , Mice, Inbred C57BL , Thymus Gland/cytology
10.
Immunol Cell Biol ; 85(6): 471-5, 2007.
Article in English | MEDLINE | ID: mdl-17637697

ABSTRACT

Following encounters with microbes, cellular activation programs that involve the control of proliferation and survival are initiated in follicular B cells either via the B-cell receptor in a specific antigen-defined manner, or through Toll-like receptors (TLRs) that recognize specific microbial products. This review summarizes and discusses recent findings that shed light on how the nuclear factor kappaB pathway controls and coordinates B-cell division and survival following TLR4 engagement.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/immunology , Lymphocyte Activation/immunology , Signal Transduction/immunology , Toll-Like Receptors/immunology , Animals , B-Lymphocytes/enzymology , Cell Survival , Humans , NF-kappa B
11.
Nat Immunol ; 7(3): 274-83, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16474395

ABSTRACT

Mitogen-activated protein kinases facilitate many cellular processes and are essential for immune cell function. Their activity is controlled by kinases and dual-specificity phosphatases. A comprehensive microarray analysis of human leukocytes identified DUSP2 (encoding the phosphatase PAC-1) as one of the most highly induced transcripts in activated immune cells. We generated Dusp2(-/-) mice and found considerably reduced inflammatory responses in the 'K/BxN' model of rheumatoid arthritis. PAC-1 deficiency led to increased activity of Jun kinase (Jnk) but unexpected impairment of the activity of extracellular signal-regulated kinase (Erk) and the kinase p38, reduced activity of the transcription factor Elk1 and a complex of mobilized transcription factor NFAT and the AP-1 transcription factor and decreased effector immune cell function. Thus, PAC-1 is a key positive regulator of inflammatory cell signaling and effector functions, mediated through Jnk and Erk mitogen-activated protein kinase crosstalk.


Subject(s)
Inflammation/immunology , Leukocytes/immunology , Protein Tyrosine Phosphatases/immunology , Protein Tyrosine Phosphatases/metabolism , Animals , Arthritis, Experimental/immunology , Dual Specificity Phosphatase 2 , Gene Expression , Gene Expression Profiling , Humans , Leukocytes/metabolism , MAP Kinase Kinase 4/immunology , MAP Kinase Kinase 4/metabolism , Mice , Mitogen-Activated Protein Kinases/immunology , Mitogen-Activated Protein Kinases/metabolism , Polymerase Chain Reaction , Protein Phosphatase 2 , Protein Tyrosine Phosphatases/deficiency , Receptor Cross-Talk/immunology
12.
Blood ; 106(10): 3457-64, 2005 Nov 15.
Article in English | MEDLINE | ID: mdl-16037393

ABSTRACT

Reticuloendotheliosis viral oncogene homolog/nuclear factor of kappa light polypeptide gene enhancer in B cells 1 (Rel/NF-kappaB) activation is a ubiquitous outcome of engaging Toll-like receptors (TLRs), yet the cell-type-specific functions of this pathway in response to particular microbial signals remain poorly defined. Here we show that NF-kappaB1 and C-Rel, Rel/NF-kappaB proteins induced in conventional dendritic cells (cDCs) and plasmacytoid dendritic cells (pDCs) by cytosine-phosphate-guanosine (CpG) DNA, a TLR-9 ligand, serve markedly different functions in these DC subsets. With the exception of impaired interleukin-12 (IL-12) production, cultured Nfkb1(-/-)C-Rel(-/-) cDCs responded relatively normally to CpG DNA. In contrast, CpG-treated Nfkb1(-/-)C-Rel(-/-) pDCs, which were still able to produce type I interferon and regulated on activation normal T-cell expressed and secreted (RANTES), but not IL-6 or IL-12, failed to acquire an activated dendritic phenotype and underwent apoptosis. Although the TLR-9-mediated death of Nfkb1(-/-)C-Rel(-/-) pDCs, which coincided with a failure to up-regulate the prosurvival proteins B-cell lymphoma apoptosis regulator xL (Bcl-x(L)) and A1, was blocked by Bcl-2 transgene expression, this inhibition of apoptosis still failed to rescue the differentiation defects. This indicated that these NF-kappaB transcription factors independently regulate TLR-9-mediated pDC morphogenesis and survival. Collectively, these findings establish that NF-kappaB1 and c-Rel, while largely dispensable for TLR-9-induced cDC activation, are critical for regulating differentiation and survival programs during pDC activation.


Subject(s)
Cell Differentiation/immunology , Dendritic Cells/immunology , NF-kappa B p50 Subunit/immunology , Plasma Cells/immunology , Proto-Oncogene Proteins c-rel/immunology , Signal Transduction/immunology , Toll-Like Receptor 9/immunology , Animals , Cell Death/drug effects , Cell Death/immunology , Cell Survival/drug effects , Cell Survival/immunology , Cells, Cultured , CpG Islands/immunology , Cytokines/biosynthesis , Cytokines/immunology , Dendritic Cells/cytology , Genes, bcl-2/genetics , Genes, bcl-2/immunology , Mice , Mice, Knockout , NF-kappa B p50 Subunit/genetics , Oligodeoxyribonucleotides/immunology , Oligodeoxyribonucleotides/pharmacology , Plasma Cells/cytology , Proto-Oncogene Proteins c-rel/genetics , Reticuloendotheliosis virus/immunology , Signal Transduction/drug effects , bcl-X Protein/genetics , bcl-X Protein/immunology
13.
Mol Cell Biol ; 24(13): 5733-45, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15199130

ABSTRACT

Determining the roles of Rel/NF-kappaB transcription factors in mouse skin development with loss-of-function mutants has been limited by redundancy among these proteins and by embryonic lethality associated with the absence of RelA. Using mice lacking RelA and c-rel, which survive throughout embryogenesis on a tumor necrosis factor alpha (TNF-alpha)-deficient background (rela(-/-) c-rel(-/-) tnfalpha(-/-)), we show that c-rel and RelA are required for normal epidermal development. Although mutant fetuses fail to form tylotrich hair and have a thinner epidermis, mutant keratinocyte progenitors undergo terminal differentiation to form an outer cornified layer. Mutant basal keratinocytes are abnormally small, exhibit a delay in G(1) progression, and fail to form keratinocyte colonies in culture. In contrast to the reduced proliferation of mutant keratinocytes during embryogenesis, skin grafting experiments revealed that the mutant epidermis develops a TNF-alpha-dependent hyperproliferative condition. Collectively, our findings indicate that RelA and c-rel control the development of the epidermis and associated appendages during embryogenesis and regulate epidermal homeostasis in a postnatal environment through the suppression of innate immune-mediated inflammation.


Subject(s)
Homeostasis , NF-kappa B/physiology , Proto-Oncogene Proteins c-rel/physiology , Skin/growth & development , Age Factors , Animals , Embryo, Mammalian , Hair Follicle/growth & development , Inflammation/embryology , Inflammation/etiology , Mice , Mice, Knockout , Mutation , NF-kappa B/genetics , Phenotype , Proto-Oncogene Proteins c-rel/genetics , Signal Transduction , Skin/chemistry , Skin/embryology , Transcription Factor RelA , Transcription Factors/physiology , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/physiology
14.
Mol Cell ; 10(6): 1283-94, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12504005

ABSTRACT

Rel/NF-kappaB transcription factors regulate the division and survival of B lymphocytes. Here we show that B cells lacking NF-kappaB1 and c-Rel fail to increase in size upon mitogenic stimulation due to a reduction in induced c-myc expression. Mitogen-induced B cell growth, although not markedly impaired by FRAP/mTOR or MEK inhibitors, required phosphatidylinositol 3-kinase (PI3K) activity. Inhibition of PI3K-dependent growth coincided with a block in the nuclear import of NF-kappaB1/c-Rel dimers and a failure to upregulate c-myc. In addition, PI3K was shown to be necessary for a transcription-independent increase in c-Myc protein levels that accompanies mitogenic activation. Collectively, these findings establish a role for Rel/NF-kappaB signaling in the mitogen-induced growth of mammalian cells, which in B lymphocytes requires a PI3K/c-myc-dependent pathway.


Subject(s)
B-Lymphocytes/cytology , Genes, myc/genetics , Genes, rel , NF-kappa B/metabolism , Oncogene Proteins v-rel/physiology , Phosphatidylinositol 3-Kinases/metabolism , Transcription, Genetic , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/physiology , Cell Division/genetics , Homeostasis , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/deficiency , NF-kappa B/genetics , Oncogene Proteins v-rel/deficiency , Oncogene Proteins v-rel/genetics
15.
Mol Cell Biol ; 22(15): 5563-74, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12101248

ABSTRACT

Mounting evidence implicates deregulated Rel/NF-kappaB signaling as a common feature of lymphoid malignancies. Despite the fact that they promote the survival and proliferation of normal lymphocytes, the underlying mechanisms by which various Rel/NF-kappaB proteins with different transcriptional regulatory capacities might facilitate transformation remain to be established. Here we show that the proliferation and tumorigenicity of Abelson murine leukemia virus (A-MuLV)-transformed pre-B cells are enhanced in the absence of NF-kappaB1 and that this coincides with elevated levels of cyclin D1. Support for a link between cyclin D1 expression and v-Abl transformation came from the finding that proliferation of transformed pre-B cells was reduced in the absence of cyclin D1, while enforced cyclin D1 expression increased the proliferation and tumorigenicity of wild-type transformants. A reduction in endogenous cyclin D1 levels that coincided with NF-kappaB1 transgene reversal of enhanced nfkb1(-/-) pre-B-cell transformation, coupled with NF-kappaB1 inhibition of v-Abl-induced kappaB-dependent murine cyclin D1 transcription, lends support to a model in which v-Abl-induced cyclin D1 transcription in transformed pre-B cells is controlled by Rel/NF-kappaB dimers with different activities.


Subject(s)
Cell Transformation, Neoplastic , Cyclin D1/metabolism , Gene Expression Regulation, Neoplastic , Genes, abl , Leukemia, Experimental/etiology , Lymphoid Tissue/metabolism , NF-kappa B/physiology , Abelson murine leukemia virus/pathogenicity , Animals , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , B-Lymphocytes/virology , Bone Marrow Cells/pathology , Bone Marrow Cells/virology , Bone Marrow Transplantation , Dimerization , G1 Phase , Genes, abl/genetics , Leukemia, Experimental/pathology , Lymphoid Tissue/pathology , Lymphoid Tissue/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/deficiency , Protein Subunits , Proto-Oncogene Proteins c-rel/deficiency , Proto-Oncogene Proteins c-rel/genetics , Proto-Oncogene Proteins c-rel/metabolism , Stem Cells/metabolism , Stem Cells/pathology , Stem Cells/virology , Survival Rate , Thymoma/etiology , Thymoma/pathology
16.
Proc Natl Acad Sci U S A ; 99(7): 4514-9, 2002 Apr 02.
Article in English | MEDLINE | ID: mdl-11930006

ABSTRACT

Transcription factors NF-kappaB1 and c-Rel, individually dispensable during embryogenesis, serve similar, yet distinct, roles in the function of mature hemopoietic cells. Redundancy among Rel/NF-kappaB family members prompted an examination of the combined roles of c-Rel and NF-kappaB1 by using mice that lack both proteins. Embryonic development and the maturation of hemopoietic progenitors were unaffected in nfkb1(-/-)c-rel(-/-) mice. Peripheral T cell populations developed normally, but follicular, marginal zone, and CD5(+) peritoneal B cell populations all were reduced. In culture, a failure of mitogen-stimulated nfkb1(-/-)c-rel(-/-) B cells to proliferate was caused by a cell cycle defect in early G(1) that prevented growth. In vivo, defects in humoral immunity and splenic architecture seen in nfkb1(-/-) and c-rel(-/-) mice were exacerbated in the double mutant mice. These findings demonstrate that in the B lineage overlapping roles for NF-kappaB1 and c-Rel appear to be restricted to regulating the activation and function of mature cells.


Subject(s)
B-Lymphocytes/physiology , Cell Lineage , Lymphocyte Activation , NF-kappa B/physiology , Proto-Oncogene Proteins c-rel/physiology , Animals , Antibody Formation , Cell Division/drug effects , Hematopoiesis , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...