Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Nature ; 623(7988): 803-813, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37938781

ABSTRACT

Patients with autoimmune polyendocrinopathy syndrome type 1 (APS-1) caused by autosomal recessive AIRE deficiency produce autoantibodies that neutralize type I interferons (IFNs)1,2, conferring a predisposition to life-threatening COVID-19 pneumonia3. Here we report that patients with autosomal recessive NIK or RELB deficiency, or a specific type of autosomal-dominant NF-κB2 deficiency, also have neutralizing autoantibodies against type I IFNs and are at higher risk of getting life-threatening COVID-19 pneumonia. In patients with autosomal-dominant NF-κB2 deficiency, these autoantibodies are found only in individuals who are heterozygous for variants associated with both transcription (p52 activity) loss of function (LOF) due to impaired p100 processing to generate p52, and regulatory (IκBδ activity) gain of function (GOF) due to the accumulation of unprocessed p100, therefore increasing the inhibitory activity of IκBδ (hereafter, p52LOF/IκBδGOF). By contrast, neutralizing autoantibodies against type I IFNs are not found in individuals who are heterozygous for NFKB2 variants causing haploinsufficiency of p100 and p52 (hereafter, p52LOF/IκBδLOF) or gain-of-function of p52 (hereafter, p52GOF/IκBδLOF). In contrast to patients with APS-1, patients with disorders of NIK, RELB or NF-κB2 have very few tissue-specific autoantibodies. However, their thymuses have an abnormal structure, with few AIRE-expressing medullary thymic epithelial cells. Human inborn errors of the alternative NF-κB pathway impair the development of AIRE-expressing medullary thymic epithelial cells, thereby underlying the production of autoantibodies against type I IFNs and predisposition to viral diseases.


Subject(s)
Autoantibodies , Genetic Predisposition to Disease , Interferon Type I , NF-kappa B , Humans , Autoantibodies/immunology , COVID-19/genetics , COVID-19/immunology , Gain of Function Mutation , Heterozygote , I-kappa B Proteins/deficiency , I-kappa B Proteins/genetics , Interferon Type I/antagonists & inhibitors , Interferon Type I/immunology , Loss of Function Mutation , NF-kappa B/deficiency , NF-kappa B/genetics , NF-kappa B p52 Subunit/deficiency , NF-kappa B p52 Subunit/genetics , Pneumonia, Viral/genetics , Pneumonia, Viral/immunology , Thymus Gland/abnormalities , Thymus Gland/immunology , Thymus Gland/pathology , Thyroid Epithelial Cells/metabolism , Thyroid Epithelial Cells/pathology , AIRE Protein , NF-kappaB-Inducing Kinase
2.
Blood Cancer J ; 10(3): 38, 2020 03 13.
Article in English | MEDLINE | ID: mdl-32170099

ABSTRACT

Aberrant NF-κB activation is a hallmark of most B-cell malignancies. Recurrent inactivating somatic mutations in the NFKBIE gene, which encodes IκBε, an inhibitor of NF-κB-inducible activity, are reported in several B-cell malignancies with highest frequencies in chronic lymphocytic leukemia and primary mediastinal B-cell lymphoma, and account for a fraction of NF-κB pathway activation. The impact of NFKBIE deficiency on B-cell development and function remains, however, largely unknown. Here, we show that Nfkbie-deficient mice exhibit an amplification of marginal zone B cells and an expansion of B1 B-cell subsets. In germinal center (GC)-dependent immune response, Nfkbie deficiency triggers expansion of GC B-cells through increasing cell proliferation in a B-cell autonomous manner. We also show that Nfkbie deficiency results in hyperproliferation of a B1 B-cell subset and leads to increased NF-κB activation in these cells upon Toll-like receptor stimulation. Nfkbie deficiency cooperates with mutant MYD88 signaling and enhances B-cell proliferation in vitro. In aged mice, Nfkbie absence drives the development of an oligoclonal indolent B-cell lymphoproliferative disorders, resembling monoclonal B-cell lymphocytosis. Collectively, these findings shed light on an essential role of IκBε in finely tuning B-cell development and function.


Subject(s)
I-kappa B Proteins/deficiency , Leukemia, Lymphocytic, Chronic, B-Cell/etiology , Proto-Oncogene Proteins/deficiency , Animals , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Mice
3.
J Immunol ; 204(8): 2033-2042, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32144162

ABSTRACT

IκBζ (encoded by the Nfkbiz) is a member of the nuclear IκB family, which is involved in the expression of secondary response genes based on signals from TLR or IL-1R. ST2L, an IL-33R, is a member of the IL-1R family and abundantly expressed in tissue-resident immune cells, such as mast cells and innate lymphoid cells; however, its downstream signaling pathway remains unelucidated. In this study, we examined the role of IκBζ in ST2L-mediated cytokine and chemokine production in mast cells. Murine bone marrow cells were differentiated ex vivo into bone marrow-derived mast cells (BMMCs). The treatment of BMMCs with IL-33 transiently induced robust IκBζ expression. Of the 40 cytokines and chemokines examined using a cytokine and chemokine array, the concentrations of IL-6, IL-13, CCL2, CCL3, and TNF-α in the supernatant were augmented by IL-33. The deletion of IκBζ in BMMCs resulted in a significant reduction of the production of these mediators and the expression of their mRNA. NF-κB p50 but not p65 translocated to the nucleus by IL-33 and was not affected by the deletion of IκBζ. However, induction of IκBζ and the resultant cytokine and chemokine productions were significantly inhibited by pretreatment with an NF-κB inhibitor. The deletion of IκBζ did not affect the phosphorylation of ERK, p38 MAPK, or JNK by IL-33, and the treatment with inhibitors of these mitogen-activated kinases failed to abolish the expression of Nfkbiz Our findings suggest that IκBζ augments IL-33-dependent cytokine and chemokine production in BMMCs through the action of NF-κB.


Subject(s)
Cytokines/biosynthesis , I-kappa B Proteins/metabolism , Interleukin-33/immunology , Mast Cells/metabolism , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/immunology , Animals , Cytokines/immunology , I-kappa B Proteins/deficiency , Mast Cells/immunology , Mice , Mice, Knockout , Mice, Transgenic , NF-kappa B/metabolism
4.
Immunol Cell Biol ; 97(5): 485-497, 2019 05.
Article in English | MEDLINE | ID: mdl-30597621

ABSTRACT

Impaired classical NF-κB pathway signaling causes reduced antibody responses to T-independent (TI) antigens. We investigated the potential reasons for defective TI responses in mice lacking the atypical inhibitory kappa B (IκB) protein of the NF-κB pathway, IκBNS. Analyses of the plasma cell compartment in vitro and in vivo after challenge with lipopolysaccharide (LPS) showed significant decreases in the frequencies of plasma cells in the absence of IκBNS. In vitro activation of B cells via the B cell receptor or via Toll-like receptor 4 revealed that early activation events were unaffected in IκBNS-deficient B cells, while proliferation was reduced compared to in similarly stimulated wildtype (wt) B cells. IκBNS-deficient B cells also displayed impaired upregulation of the transmembrane activator and calcium modulator cyclophilin ligand interactor (TACI), which is essential for TI responses, and decreased sensitivity to TACI ligands upon stimulation. Furthermore, IκBNS-deficient B cells, in contrast to wt B cells, displayed altered expression of IRF4, Blimp-1 and Pax5 upon LPS-induced differentiation, indicating impaired transcriptional regulation of plasma cell generation.


Subject(s)
Cell Differentiation , Gene Expression Regulation/immunology , I-kappa B Proteins/deficiency , Plasma Cells/immunology , Transmembrane Activator and CAML Interactor Protein/immunology , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , I-kappa B Proteins/immunology , Mice , Mice, Knockout , Plasma Cells/cytology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Transmembrane Activator and CAML Interactor Protein/genetics
5.
Immunology ; 147(4): 453-63, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26749055

ABSTRACT

Although a major function of B cells is to mediate humoral immunity by producing antigen-specific antibodies, a specific subset of B cells is important for immune suppression, which is mainly mediated by the secretion of the anti-inflammatory cytokine interleukin-10 (IL-10). However, the mechanism by which IL-10 is induced in B cells has not been fully elucidated. Here, we report that IκBNS , an inducible nuclear IκB protein, is important for Toll-like receptor (TLR)-mediated IL-10 production in B cells. Studies using IκB(NS) knockout mice revealed that the number of IL-10-producing B cells is reduced in IκB(NS)(-/-) spleens and that the TLR-mediated induction of cytoplasmic IL-10-positive cells and IL-10 secretion in B cells are impaired in the absence of IκB(NS). The impairment of IL-10 production by a lack of IκB(NS) was not observed in TLR-triggered macrophages or T-cell-receptor-stimulated CD4(+) CD25(+) T cells. In addition, IκB(NS)-deficient B cells showed reduced expression of Prdm1 and Irf4 and failed to generate IL-10(+) CD138(+) plasmablasts. These results suggest that IκB(NS) is selectively required for IL-10 production in B cells responding to TLR signals, so defining an additional role for IκB(NS) in the control of the B-cell-mediated immune responses.


Subject(s)
B-Lymphocytes/immunology , B-Lymphocytes/metabolism , I-kappa B Proteins/metabolism , Interleukin-10/biosynthesis , Toll-Like Receptors/metabolism , Animals , B-Lymphocytes/cytology , Cell Differentiation/genetics , I-kappa B Proteins/deficiency , I-kappa B Proteins/genetics , Interleukin-10/genetics , Lipopolysaccharides/immunology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Mice , Mice, Knockout , NF-kappa B/metabolism , Plasma Cells/cytology , Plasma Cells/immunology , Plasma Cells/metabolism , Promoter Regions, Genetic , Protein Binding , Spleen , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
6.
Cytokine ; 78: 20-1, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26615569

ABSTRACT

Psoriasis is an immune-mediated chronic inflammatory skin disease with a complex etiology. The proinflammatory cytokine IL-17A is known to play key role in the pathogenesis of psoriasis, and recently anti-IL-17A antibodies have been approved for psoriasis treatment. Here, we discuss our recent findings demonstrating that IκBζ, a transcriptional co-activator, plays a crucial role in the development of psoriasis by mediating IL-17A-driven effects. These findings have significant implications as they uncover a novel crucial regulatory mechanism involved in psoriasis development, and identify IκBζ as a possible future target in the treatment of psoriasis and other IL-17A-driven diseases.


Subject(s)
I-kappa B Proteins/genetics , I-kappa B Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Psoriasis/immunology , Psoriasis/physiopathology , Adaptor Proteins, Signal Transducing , Animals , Animals, Genetically Modified , Cytokines/metabolism , Disease Models, Animal , Humans , I-kappa B Proteins/deficiency , Inflammation , Interleukin-17/metabolism , Keratinocytes/metabolism , Mice , Nuclear Proteins/deficiency , Signal Transduction
7.
J Immunol ; 194(6): 2888-98, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25694610

ABSTRACT

IL-17-producing Th17 cells mediate immune responses against a variety of fungal and bacterial infections. Signaling via NF-κB has been linked to the development and maintenance of Th17 cells. We analyzed the role of the unusual inhibitor of NF-κB, IκBNS, in the proliferation and effector cytokine production of murine Th17 cells. Our study demonstrates that nuclear IκBNS is crucial for murine Th17 cell generation. IκBNS is highly expressed in Th17 cells; in the absence of IκBNS, the frequencies of IL-17A-producing cells are drastically reduced. This was measured in vitro under Th17-polarizing conditions and confirmed in two colitis models. Mechanistically, murine IκBNS (-/-) Th17 cells were less proliferative and expressed markedly reduced levels of IL-2, IL-10, MIP-1α, and GM-CSF. Citrobacter rodentium was used as a Th17-inducing infection model, in which IκBNS (-/-) mice displayed an increased bacterial burden and diminished tissue damage. These results demonstrate the important function of Th17 cells in pathogen clearance, as well as in inflammation-associated pathology. We identified IκBNS to be crucial for the generation and function of murine Th17 cells upon inflammation and infection. Our findings may have implications for the therapy of autoimmune conditions, such as inflammatory bowel disease, and for the treatment of gut-tropic infections.


Subject(s)
Cell Differentiation/immunology , Citrobacter rodentium/immunology , Colitis/immunology , Enterobacteriaceae Infections/immunology , I-kappa B Proteins/immunology , Th17 Cells/immunology , Animals , Blotting, Western , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Cell Proliferation/genetics , Citrobacter rodentium/physiology , Colitis/genetics , Colitis/metabolism , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Enterobacteriaceae Infections/genetics , Enterobacteriaceae Infections/microbiology , Flow Cytometry , Gene Expression/immunology , Host-Pathogen Interactions/immunology , I-kappa B Proteins/deficiency , I-kappa B Proteins/genetics , Mice, 129 Strain , Mice, Knockout , NF-kappa B/immunology , NF-kappa B/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Th17 Cells/metabolism
8.
Proc Natl Acad Sci U S A ; 111(39): E4119-26, 2014 Sep 30.
Article in English | MEDLINE | ID: mdl-25228759

ABSTRACT

B-1 cells mediate early protection against infection by responding to T cell-independent (TI) antigens found on the surface of various pathogens. Mice with impaired expression of the atypical IκB protein IκBNS have markedly reduced frequencies of B-1 cells. We used a mouse strain with dysfunctional IκBNS derived from an N-ethyl-N-nitrosourea (ENU) screen, named bumble, to investigate the point in the development of B-1 cells where IκBNS is required. The presence of wild-type (wt) peritoneal cells in mixed wt/bumble chimeras did not rescue the development of bumble B-1 cells, but wt peritoneal cells transferred to bumble mice restored natural IgM levels and response to TI antigens. The bumble and wt mice displayed similar levels of fetal liver B-1 progenitors and splenic neonatal transitional B (TrB) cells, both of which were previously shown to give rise to B-1 cells. Interestingly, we found that a subset of wt neonatal TrB cells expressed common B-1a markers (TrB-1a) and that this cell population was absent in the bumble neonatal spleen. Sorted TrB-1a (CD93(+)IgM(+)CD5(+)) cells exclusively generated B-1a cells when adoptively transferred, whereas sorted CD93(+)IgM(+)CD5(-) cells gave rise to B-2 cells and, to a lesser extent, B-1b and B-1a cells. This study identifies a phenotypically distinct splenic population of TrB-1a cells and establishes that the development of B-1a cells is blocked before this stage in the absence of IκBNS.


Subject(s)
B-Lymphocyte Subsets/immunology , I-kappa B Proteins/deficiency , Proteins/immunology , Adoptive Transfer , Animals , Animals, Newborn , Antigens, T-Independent/administration & dosage , B-Lymphocyte Subsets/cytology , B-Lymphocyte Subsets/metabolism , Cell Differentiation/immunology , I-kappa B Proteins/genetics , I-kappa B Proteins/immunology , Immunoglobulin M/metabolism , Intracellular Signaling Peptides and Proteins , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Proteins/genetics
9.
Immunity ; 37(6): 998-1008, 2012 Dec 14.
Article in English | MEDLINE | ID: mdl-23200824

ABSTRACT

Forkhead box P3 positive (Foxp3(+)) regulatory T (Treg) cells suppress immune responses and regulate peripheral tolerance. Here we show that the atypical inhibitor of NFκB (IκB) IκB(NS) drives Foxp3 expression via association with the promoter and the conserved noncoding sequence 3 (CNS3) of the Foxp3 locus. Consequently, IκB(NS) deficiency leads to a substantial reduction of Foxp3(+) Treg cells in vivo and impaired Foxp3 induction upon transforming growth factor-ß (TGF-ß) treatment in vitro. Moreover, fewer Foxp3(+) Treg cells developed from IκB(NS)-deficient CD25(-)CD4(+) T cells adoptively transferred into immunodeficient recipients. Importantly, IκB(NS) was required for the transition of immature GITR(+)CD25(+)Foxp3(-) thymic Treg cell precursors into Foxp3(+) cells. In contrast to mice lacking c-Rel or Carma1, IκB(NS)-deficient mice do not show reduced Treg precursor cells. Our results demonstrate that IκB(NS) critically regulates Treg cell development in the thymus and during gut inflammation, indicating that strategies targeting IκB(NS) could modulate the Treg cell compartment.


Subject(s)
Forkhead Transcription Factors/metabolism , I-kappa B Proteins/metabolism , Proteins/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Adoptive Transfer , Animals , Apoptosis , Cell Differentiation/immunology , Forkhead Transcription Factors/genetics , Gene Expression Regulation , I-kappa B Proteins/deficiency , I-kappa B Proteins/genetics , Immunomodulation , Intracellular Signaling Peptides and Proteins , Lymphocyte Activation/immunology , Male , Mice , Mice, Transgenic , NF-kappa B p50 Subunit/metabolism , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins c-rel/metabolism , T-Lymphocytes, Regulatory/drug effects , Transforming Growth Factor beta/pharmacology
10.
Mol Neurodegener ; 7: 47, 2012 Sep 19.
Article in English | MEDLINE | ID: mdl-22992283

ABSTRACT

BACKGROUND: The transcription factor NFκB is an important mediator of cell survival and inflammation in the immune system. In the central nervous system (CNS), NFκB signaling has been implicated in regulating neuronal survival following acute pathologic damage such as traumatic brain injury (TBI) and stroke. NFκB is normally bound by the principal inhibitory protein, IκBα, and sequestered in the cytoplasm. Activation of NFκB requires the degradation of IκBα, thereby freeing NFκB to translocate to the nucleus and activate the target genes. Mice deficient in IκBα display deregulated and sustained NFκB activation and early postnatal lethality, highlighting a critical role of IκBα in NFκB regulation. RESULTS: We investigated the role of IκBα in regulating NFκB activity in the brain and the effects of the NFκB/IκBα pathway in mediating neuroinflammation under both physiological and brain injury conditions. We report that astrocytes, but not neurons, exhibit prominent NFκB activity, and that basal NFκB activity in astrocytes is elevated in the absence of IκBα. By generating mice with brain-specific deletion of IκBα, we show that IκBα deficiency does not compromise normal brain development. However, basal neuroinflammation detected by GFAP and Iba1 immunoreactivity is elevated. This leads to impaired inflammatory responses following TBI and worsened brain damage including higher blood brain barrier permeability, increased injury volumes and enlarged ventricle volumes. CONCLUSIONS: We conclude that, in the CNS, astrocyte is the primary cell type subject to NFκB regulation. We further demonstrate that IκBα plays an important role in regulating NFκB activity in the brain and a robust NFκB/IκBα-mediated neuroinflammatory response immediately following TBI is beneficial.


Subject(s)
Astrocytes/metabolism , Brain Injuries/metabolism , Brain/metabolism , I-kappa B Proteins/deficiency , Recovery of Function/physiology , Animals , Blood-Brain Barrier/physiology , Brain/growth & development , Brain/pathology , Brain Injuries/pathology , Glial Fibrillary Acidic Protein , Inflammation/metabolism , Mice , NF-kappa B/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Neurons/pathology , Phosphorylation , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/metabolism
11.
J Clin Invest ; 122(10): 3563-78, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23006329

ABSTRACT

The strength and duration of NF-κB signaling are tightly controlled by multiple negative feedback mechanisms. However, in cancer cells, these feedback loops are overridden through unclear mechanisms to sustain oncogenic activation of NF-κB signaling. Previously, we demonstrated that overexpression of miR-30e* directly represses IκBα expression and leads to hyperactivation of NF-κB. Here, we report that miR-182 was overexpressed in a different set of gliomas with relatively lower miR-30e* expression and that miR-182 directly suppressed cylindromatosis (CYLD), an NF-κB negative regulator. This suppression of CYLD promoted ubiquitin conjugation of NF-κB signaling pathway components and induction of an aggressive phenotype of glioma cells both in vitro and in vivo. Furthermore, we found that TGF-ß induced miR-182 expression, leading to prolonged NF-κB activation. Importantly, the results of these experiments were consistent with an identified significant correlation between miR-182 levels with TGF-ß hyperactivation and activated NF-κB in a cohort of human glioma specimens. These findings uncover a plausible mechanism for sustained NF-κB activation in malignant gliomas and may suggest a new target for clinical intervention in human cancer.


Subject(s)
Brain Neoplasms/metabolism , Glioma/metabolism , MicroRNAs/physiology , NF-kappa B/metabolism , Neoplasm Proteins/metabolism , RNA, Neoplasm/physiology , Transforming Growth Factor beta/physiology , Animals , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Cell Line, Tumor/transplantation , Deubiquitinating Enzyme CYLD , Genes, Reporter , Glioma/genetics , Glioma/pathology , Humans , I-kappa B Kinase/metabolism , I-kappa B Proteins/deficiency , Mice , Mice, Nude , MicroRNAs/antagonists & inhibitors , MicroRNAs/biosynthesis , MicroRNAs/genetics , NF-KappaB Inhibitor alpha , Neoplasm Invasiveness , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neovascularization, Pathologic/physiopathology , Phosphorylation , Protein Processing, Post-Translational , RNA/biosynthesis , RNA/genetics , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Signal Transduction/drug effects , Smad Proteins/biosynthesis , Smad Proteins/genetics , Transcription, Genetic , Transforming Growth Factor beta/pharmacology , Tumor Suppressor Proteins/biosynthesis , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/physiology , Ubiquitination/drug effects
12.
J Neuroinflammation ; 9: 15, 2012 Jan 20.
Article in English | MEDLINE | ID: mdl-22260436

ABSTRACT

The NF-κB/REL-family of transcription factors plays a central role in coordinating the expression of a wide variety of genes controlling immune responses including autoimmunity of the central nervous system (CNS). The inactive form of NF-κB consists of a heterodimer which is complexed with its inhibitor, IκB. Conditional knockout-mice for IκBα in myeloid cells (lysMCreIκBα(fl/fl)) have been generated and are characterized by a constitutive activation of NF-κB proteins allowing the study of this transcription factor in myelin-oligodendrocyte-glycoprotein induced experimental autoimmune encephalomyelitis (MOG-EAE), a well established experimental model for autoimmune demyelination of the CNS.In comparison to controls, lysMCreIκBα(fl/fl) mice developed a more severe clinical course of EAE. Upon histological analysis on day 15 p.i., there was an over two fold increased infiltration of T-cells and macrophages/microglia. In addition, lysMCreIκBα(fl/fl) mice displayed an increased expression of the NF-κB dependent factor inducible nitric oxide synthase in inflamed lesions. These changes in the CNS are associated with increased numbers of CD11b positive splenocytes and a higher expression of Ly6c on monocytes in the periphery. Well in accordance with these changes in the myeloid cell compartment, there was an increased production of the monocyte cytokines interleukin(IL)-12 p70, IL-6 and IL-1beta in splenocytes. In contrast, production of the T-cell associated cytokines interferon gamma (IFN-gamma) and IL-17 was not influenced.In summary, myeloid cell derived NF-κB plays a crucial role in autoimmune inflammation of the CNS and drives a pathogenic role of monocytes and macrophages independently from T-cells.


Subject(s)
Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , I-kappa B Proteins/metabolism , Macrophages/metabolism , Monocytes/metabolism , Animals , CD11b Antigen/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Enzyme-Linked Immunosorbent Assay/methods , Flow Cytometry , Glycoproteins/adverse effects , I-kappa B Proteins/deficiency , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin-Oligodendrocyte Glycoprotein , Neutrophil Infiltration/drug effects , Neutrophil Infiltration/immunology , Nitric Oxide Synthase Type II/metabolism , Peptide Fragments/adverse effects , Spleen/cytology
13.
Clin Microbiol Rev ; 24(3): 490-7, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21734245

ABSTRACT

Autosomal recessive IRAK-4 and MyD88 deficiencies predispose affected patients to recurrent invasive pyogenic bacterial infection. Both defects result in the selective impairment of cellular responses to Toll-like receptors (TLRs) other than TLR3 and of cellular responses to most interleukin-1 receptors (IL-1Rs), including IL-1R, IL-18R, and IL-33R. Hypomorphic mutations in the X-linked NEMO gene and hypermorphic mutations in the autosomal IKBA gene cause X-linked recessive and autosomal dominant anhidrotic ectodermal dysplasia with immunodeficiency (EDA-ID) syndromes. Both of these defects impair NF-κB-mediated cellular responses to multiple receptors, including TLRs, IL-1Rs, and tumor necrosis factor receptors (TNF-Rs). They therefore confer a much broader predisposition to infections than that for IRAK-4 and MyD88 deficiencies. These disorders were initially thought to be rare but have now been diagnosed in over 170 patients worldwide. We review here the infectious diseases affecting patients with inborn errors of NF-κB-dependent TLR and IL-1R immunity.


Subject(s)
Bacterial Infections/metabolism , I-kappa B Kinase/deficiency , I-kappa B Proteins/deficiency , Interleukin-1 Receptor-Associated Kinases/deficiency , Myeloid Differentiation Factor 88/deficiency , Bacterial Infections/genetics , Humans , I-kappa B Kinase/genetics , I-kappa B Proteins/genetics , Interleukin-1 Receptor-Associated Kinases/genetics , Mutation , Myeloid Differentiation Factor 88/genetics , NF-KappaB Inhibitor alpha
14.
Med Sci (Paris) ; 27(6-7): 619-25, 2011.
Article in French | MEDLINE | ID: mdl-21718646

ABSTRACT

The IKKε kinase, an atypical member of the IKK family of kinases, was recently identified as an oncogene overexpressed in over 30% of breast cancers. Besides its role in the regulation of the NF-κB transcription factor, which is well recognized for its implication in the development of breast cancers, IKKε was shown to phosphorylate numerous targets. Analysis of the phosphorylation of some of these substrates in the context of breast cancer highlighted new oncogenic signaling pathways that constitute potential targets for new therapies. Interestingly, IKKε is involved in the development of resistance to Tamoxifène. Thus, IKKε is a promising therapeutic target for newly developed breast cancer treatment.


Subject(s)
Breast Neoplasms/metabolism , I-kappa B Proteins/physiology , Neoplasm Proteins/physiology , Proto-Oncogene Proteins/physiology , Animals , Antineoplastic Agents, Hormonal/pharmacology , Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/etiology , Breast Neoplasms/genetics , Cell Transformation, Neoplastic , Deubiquitinating Enzyme CYLD , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , I-kappa B Proteins/antagonists & inhibitors , I-kappa B Proteins/chemistry , I-kappa B Proteins/deficiency , Mice , Models, Biological , Neoplasm Proteins/antagonists & inhibitors , Oncogenes , Phosphorylation , Protein Processing, Post-Translational , Protein Structure, Tertiary , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/deficiency , Signal Transduction , Tamoxifen/pharmacology , Tamoxifen/therapeutic use , Transcription Factors/physiology , Tumor Suppressor Proteins/metabolism
15.
J Immunol ; 186(4): 2412-21, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21228348

ABSTRACT

Bcl-3 is an atypical member of the IκB family that has the potential to positively or negatively modulate nuclear NF-κB activity in a context-dependent manner. Bcl-3's biologic impact is complex and includes roles in tumorigenesis and diverse immune responses, including innate immunity. Bcl-3 may mediate LPS tolerance, suppressing cytokine production, but it also seems to contribute to defense against select systemic bacterial challenges. However, the potential role of Bcl-3 in organ-specific host defense against bacteria has not been addressed. In this study, we investigated the relevance of Bcl-3 in a lung challenge with the Gram-negative pathogen Klebsiella pneumoniae. In contrast to wild-type mice, Bcl-3-deficient mice exhibited significantly increased susceptibility toward K. pneumoniae pneumonia. The mutant mice showed increased lung damage marked by neutrophilic alveolar consolidation, and they failed to clear bacteria in lungs, which correlated with increased bacteremic dissemination. Loss of Bcl-3 incurred a dramatic cytokine imbalance in the lungs, which was characterized by higher levels of IL-10 and a near total absence of IFN-γ. Moreover, Bcl-3-deficient mice displayed increased lung production of the neutrophil-attracting chemokines CXCL-1 and CXCL-2. Alveolar macrophages and neutrophils are important to antibacterial lung defense. In vitro stimulation of Bcl-3-deficient alveolar macrophages with LPS or heat-killed K. pneumoniae recapitulated the increase in IL-10 production, and Bcl-3-deficient neutrophils were impaired in intracellular bacterial killing. These findings suggest that Bcl-3 is critically involved in lung defense against Gram-negative bacteria, modulating functions of several cells to facilitate efficient clearance of bacteria.


Subject(s)
I-kappa B Proteins/physiology , Klebsiella Infections/immunology , Klebsiella pneumoniae/immunology , Pneumonia, Bacterial/immunology , Proto-Oncogene Proteins/physiology , Transcription Factors/physiology , Animals , B-Cell Lymphoma 3 Protein , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Bronchoalveolar Lavage Fluid/microbiology , Genetic Predisposition to Disease , I-kappa B Proteins/deficiency , I-kappa B Proteins/genetics , Klebsiella Infections/pathology , Klebsiella Infections/prevention & control , Macrophages, Alveolar/immunology , Macrophages, Alveolar/microbiology , Macrophages, Alveolar/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Multigene Family/immunology , Neutrophils/immunology , Neutrophils/microbiology , Neutrophils/pathology , Pneumonia, Bacterial/pathology , Pneumonia, Bacterial/prevention & control , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Transcription Factors/deficiency , Transcription Factors/genetics
16.
Eye Contact Lens ; 36(5): 269-81, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20703156

ABSTRACT

This review addresses three subjects: the innate immunity of the ocular surface epithelium, innate immunity and ocular surface inflammation, and Stevens-Johnson syndrome (SJS) and abnormality of innate immunity. In innate immunity of the ocular surface epithelium, ocular surface epithelial cells respond selectively to microbial components and induce limited inflammation, whereas immune-competent cells such as macrophages can recognize various microbial components through Toll-like receptors (TLRs) and induce inflammation to exclude the microbes. The difference between macrophages and ocular surface epithelial cells may be caused by the dissimilarity in the degree of coexistence with commensal bacteria. The unique innate immune response of ocular surface epithelium might contribute to coexistence with commensal bacteria. In innate immunity and ocular surface inflammation, we speculate that an abnormality in the proper innate immunity of the ocular surface may result in ocular surface inflammation. Our investigation shows that TLR3 positively regulates the late-phase reaction of experimental allergic conjunctivitis, which causes reduced eosinophilic conjunctival inflammation in TLR3KO (knockout) mice and pronounced eosinophilic conjunctival inflammation in TLR3Tg mice. We also demonstrate that human ocular surface epithelial cells can be induced to express many transcripts, including antiviral innate immune response-related genes and allergy-related genes, through polyI:C stimulation. Furthermore, we show that IkappaBzeta KO mice exhibit severe, spontaneous ocular surface inflammation accompanied by the eventual loss of almost all goblet cells and spontaneous perioral inflammation. IkappaBzeta is induced by diverse pathogen-associated molecular patterns and regulates nuclear factor-kappaB activity, possibly to prevent excessive inflammation in the presence of bacterial components. The spontaneous ocular surface inflammation observed in IkappaBzeta KO mice suggested that dysfunction/abnormality of innate immunity can play a role in ocular surface inflammation. In SJS and abnormality of innate immunity, we considered the possibility that there may be an association between SJS and a disordered innate immune response. In gene expression analysis of CD14 cells, we found that IL4R gene expression was different in patients with SJS/toxic epidermal necrolysis (TEN) and controls on lipopolysaccharide stimulation, being downregulated in patients with SJS/TEN and slightly upregulated in the controls. The expression of IkappaBzeta- and interleukin (IL)-1alpha-specific mRNA in patients with SJS/TEN was lower than in normal controls after 1-hour culture. Although SJS/TEN can be induced by drugs, not all individuals treated with these drugs developed SJS/TEN. Because the incidence of SJS/TEN is very low, we suspected a genetic predisposition and performed single-nucleotide polymorphism (SNP) association analysis using candidate genes associated with innate immunity, apoptosis, or allergy. We found that TLR3 SNP rs.3775296 and IL4R SNP rs.1801275 (Gln551Arg) were strongly associated (P<0.0005) with SJS/TEN with ocular surface complications, FasL rs.3830150 SNP was mildly associated (P<0.005), and IL13 rs.20541 (Arg110Gln) and IkappaBzeta SNP rs.595788G/A exhibited a weak association (P<0.05). Genetic and environmental factors may play a role in an integrated cause of SJS, and there is the possibility of an association between SJS and a disordered innate immunity.


Subject(s)
Conjunctivitis/immunology , Immunity, Innate , Keratitis/immunology , Animals , Conjunctivitis/etiology , Conjunctivitis/metabolism , Conjunctivitis/pathology , Fas Ligand Protein/genetics , Gene Expression , Goblet Cells/pathology , Humans , I-kappa B Proteins/deficiency , I-kappa B Proteins/metabolism , Immunity, Innate/genetics , Interleukin-1alpha/genetics , Keratitis/genetics , Keratitis/metabolism , Keratitis/pathology , Polymorphism, Single Nucleotide , Protein Isoforms/deficiency , Protein Isoforms/metabolism , RNA, Messenger/metabolism , Receptors, Interleukin-4/genetics , Receptors, Interleukin-4/metabolism , Stevens-Johnson Syndrome/genetics , Stevens-Johnson Syndrome/immunology , Stevens-Johnson Syndrome/metabolism , Toll-Like Receptor 3/genetics , Toll-Like Receptors/metabolism
17.
Mol Cell ; 39(4): 570-82, 2010 Aug 27.
Article in English | MEDLINE | ID: mdl-20797629

ABSTRACT

Proinflammatory cytokines activate NF-kappaB using the IkappaB kinase (IKK) complex that phosphorylates inhibitory proteins (IkappaBs) at N-terminal sites resulting in their ubiquitination and degradation in the cytoplasm. Although ultraviolet (UV) irradiation does not lead to IKK activity, it activates NF-kappaB by an unknown mechanism through IkappaBalpha degradation without N-terminal phosphorylation. Here, we describe an adaptor function of nuclear IKKbeta in UV-induced IkappaBalpha degradation. UV irradiation induces the nuclear translocation of IkappaBalpha and association with IKKbeta, which constitutively interacts with beta-TrCP through heterogeneous ribonucleoprotein-U (hnRNP-U) leading to IkappaBalpha ubiquitination and degradation. Furthermore, casein kinase 2 (CK2) and p38 associate with IKKbeta and promote IkappaBalpha degradation by phosphorylation at C-terminal sites. Thus, nuclear IKKbeta acts as an adaptor protein for IkappaBalpha degradation in UV-induced NF-kappaB activation. NF-kappaB activated by the nuclear IKKbeta adaptor protein suppresses anti-apoptotic gene expression and promotes UV-induced cell death.


Subject(s)
Cell Nucleus/radiation effects , I-kappa B Kinase/metabolism , I-kappa B Proteins/metabolism , NF-kappa B/metabolism , Protein Processing, Post-Translational/radiation effects , Ultraviolet Rays , Active Transport, Cell Nucleus , Animals , Apoptosis/radiation effects , Casein Kinase II/metabolism , Cell Line , Cell Nucleus/metabolism , Cell Nucleus/pathology , Fibroblasts/metabolism , Fibroblasts/radiation effects , Heterogeneous-Nuclear Ribonucleoprotein U/metabolism , Humans , I-kappa B Kinase/deficiency , I-kappa B Kinase/genetics , I-kappa B Proteins/deficiency , I-kappa B Proteins/genetics , Mice , Mice, Knockout , Mutation , NF-KappaB Inhibitor alpha , NF-kappa B/genetics , Phosphorylation , Time Factors , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Transfection , Ubiquitination , beta-Transducin Repeat-Containing Proteins/genetics , beta-Transducin Repeat-Containing Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
18.
Exp Cell Res ; 316(19): 3317-27, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20466000

ABSTRACT

BACKGROUND AND AIMS: B-cell lymphoma/leukemia (BCL)-10 and reactive oxygen species mediate two pathways of NF-κB (RelA) activation by lipopolysaccharide (LPS) in human colonic epithelial cells. The pathway for LPS activation of RelB by the non-canonical pathway (RelB) in non-myeloid cells was not yet reported, but important for understanding the range of potential microbial LPS-induced effects in inflammatory bowel disease. METHODS: Experiments were performed in human colonic epithelial cells and in mouse embryonic fibroblasts deficient in components of the IkappaB kinase (IKK) signalosome, in order to detect mediators of the non-canonical pathway of NF-κB activation, including nuclear RelB and p52 and phospho- and total NF-κB inducing kinase (NIK). BCL10 was silenced by siRNA and effects of mutations of specific phosphorylation sites of BCL10 (Ser138Gly and Ser218Gly) were determined. RESULTS: By the non-canonical pathway, LPS exposure increased nuclear RelB and p52, and phospho-NIK, with no change in total NIK. Phosphorylation of BCL10 serine 138 was required for NIK phosphorylation, since mutation of this residue eliminated the increases in phospho-NIK and nuclear RelB and p52. Mutations of either serine 138 or serine 218 reduced RelA, p50, and phospho-IκBα of the canonical pathway. Effects of LPS stimulation and BCL10 silencing on NIK phosphorylation were demonstrated in confocal images. CONCLUSIONS: LPS induces activation of both canonical and non-canonical pathways of NF-κB in human colonic epithelial cells, and the non-canonical pathway requires phosphorylations of BCL10 (serine 138) and NIK. These findings demonstrate the important role of BCL10 in mediating LPS-induced inflammation in human colonic epithelial cells and may open new avenues for therapeutic interventions.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Lipopolysaccharides/pharmacology , NF-kappa B/metabolism , Phosphoserine/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Aged , Animals , B-Cell CLL-Lymphoma 10 Protein , Colon/cytology , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Fibroblasts/metabolism , Gene Silencing , Humans , I-kappa B Proteins/deficiency , I-kappa B Proteins/metabolism , Interleukin-8/metabolism , Male , Mice , Mutation/genetics , NF-KappaB Inhibitor alpha , Phosphorylation/drug effects , Structure-Activity Relationship , NF-kappaB-Inducing Kinase
19.
J Neurochem ; 110(2): 765-78, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19522780

ABSTRACT

We previously showed that Nuclear Factor kappaB (NF-kappaB) inactivation in astrocytes leads to improved functional recovery following spinal cord injury (SCI). This correlated with reduced expression of pro-inflammatory mediators and chondroitin sulfate proteoglycans, and increased white matter preservation. Hence we hypothesized that inactivation of astrocytic NF-kappaB would create a more permissive environment for axonal sprouting and regeneration. We induced both contusive and complete transection SCI in GFAP-Inhibitor of kappaB-dominant negative (GFAP-IkappaBalpha-dn) and wild-type (WT) mice and performed retrograde [fluorogold (FG)] and anterograde [biotinylated dextran amine (BDA)] tracing 8 weeks after injury. Following contusive SCI, more FG-labeled cells were found in motor cortex, reticular formation, and raphe nuclei of transgenic mice. Spared and sprouting BDA-positive corticospinal axons were found caudal to the lesion in GFAP-IkappaBalpha-dn mice. Higher numbers of FG-labeled neurons were detected immediately rostral to the lesion in GFAP-IkappaBalpha-dn mice, accompanied by increased expression of synaptic and axonal growth-associated molecules. After transection, however, no FG-labeled neurons or BDA-filled axons were found rostral and caudal to the lesion, respectively, in either genotype. These data demonstrated that inhibiting astroglial NF-kappaB resulted in a growth-supporting terrain promoting sparing and sprouting, rather than regeneration, of supraspinal and propriospinal circuitries essential for locomotion, hence contributing to the improved functional recovery observed after SCI in GFAP-IkappaBalpha-dn mice.


Subject(s)
Astrocytes/pathology , Astrocytes/physiology , Axons/pathology , Axons/physiology , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , Spinal Cord Injuries/genetics , Spinal Cord Injuries/metabolism , Animals , Astrocytes/cytology , Astrocytes/metabolism , Female , Genetic Therapy , I-kappa B Proteins/deficiency , I-kappa B Proteins/genetics , I-kappa B Proteins/physiology , Mice , Mice, Transgenic , NF-KappaB Inhibitor alpha , Nerve Regeneration/genetics , Nerve Regeneration/physiology , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology , Spinal Cord Injuries/therapy
20.
J Immunol ; 182(5): 2628-40, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19234157

ABSTRACT

In the CNS, the transcription factor NF-kappaB is a key regulator of inflammation and secondary injury processes. Following trauma or disease, the expression of NF-kappaB-dependent genes is activated, leading to both protective and detrimental effects. In this study, we show that transgenic inactivation of astroglial NF-kappaB (glial fibrillary acidic protein-IkappaB alpha-dominant-negative mice) resulted in reduced disease severity and improved functional recovery following experimental autoimmune encephalomyelitis. At the chronic stage of the disease, transgenic mice exhibited an overall higher presence of leukocytes in spinal cord and brain, and a markedly higher percentage of CD8(+)CD122(+) T regulatory cells compared with wild type, which correlated with the timing of clinical recovery. We also observed that expression of proinflammatory genes in both spinal cord and cerebellum was delayed and reduced, whereas the loss of neuronal-specific molecules essential for synaptic transmission was limited compared with wild-type mice. Furthermore, death of retinal ganglion cells in affected retinas was almost abolished, suggesting the activation of neuroprotective mechanisms. Our data indicate that inhibiting NF-kappaB in astrocytes results in neuroprotective effects following experimental autoimmune encephalomyelitis, directly implicating astrocytes in the pathophysiology of this disease.


Subject(s)
Astrocytes/immunology , Astrocytes/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Inflammation Mediators/antagonists & inhibitors , NF-kappa B/antagonists & inhibitors , Animals , Central Nervous System/immunology , Central Nervous System/metabolism , Central Nervous System/pathology , Chronic Disease , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Female , Gene Expression Profiling , Glial Fibrillary Acidic Protein/deficiency , Glial Fibrillary Acidic Protein/genetics , I-kappa B Proteins/deficiency , I-kappa B Proteins/genetics , Inflammation Mediators/physiology , Male , Mice , Mice, Transgenic , NF-KappaB Inhibitor alpha , NF-kappa B/physiology , Oligonucleotide Array Sequence Analysis , Severity of Illness Index
SELECTION OF CITATIONS
SEARCH DETAIL
...