Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Sci Signal ; 16(791): eabo4094, 2023 06 27.
Article in English | MEDLINE | ID: mdl-37368952

ABSTRACT

The inhibitor of κB kinase (IKK) complex regulates the activation of the nuclear factor κB (NF-κB) family of transcription factors. In addition, IKK represses extrinsic cell death pathways dependent on receptor-interacting serine/threonine-protein kinase 1 (RIPK1) by directly phosphorylating this kinase. Here, we showed that peripheral naïve T cells in mice required the continued expression of IKK1 and IKK2 for their survival; however, the loss of these cells was only partially prevented when extrinsic cell death pathways were blocked by either deleting Casp8 (which encodes the apoptosis-inducing caspase 8) or inhibiting the kinase activity of RIPK1. Inducible deletion of Rela (which encodes the NF-κB p65 subunit) in mature CD4+ T cells also resulted in loss of naïve CD4+ T cells and in reduced abundance of the interleukin-7 receptor (IL-7R) encoded by the NF-κB target Il7r, revealing an additional reliance upon NF-κB for the long-term survival of mature T cells. Together, these data indicate that the IKK-dependent survival of naïve CD4+ T cells depends on both repression of extrinsic cell death pathways and activation of an NF-κB-dependent survival program.


Subject(s)
I-kappa B Kinase , NF-kappa B , Animals , Mice , Apoptosis/genetics , Cell Survival/genetics , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , T-Lymphocytes/metabolism
2.
Front Immunol ; 13: 1067164, 2022.
Article in English | MEDLINE | ID: mdl-36532075

ABSTRACT

The Inhibitor of Kappa B Kinase (IKK) complex is a critical regulator of NF-κB activation. More recently, IKK has also been shown to repress RIPK1 dependent extrinsic cell death pathways by directly phosphorylating RIPK1 at serine 25. In T cells, IKK expression is essential for normal development in the thymus, by promoting survival of thymocytes independently of NF-κB activation. RIPK1 undergoes extensive phosphorylation following TNF stimulation in T cells, though which targets are required to repress RIPK1 has not been defined. Here, we show that TNF induced phosphorylation of RIPK1 at S25 is IKK dependent. We test the relevance of this phosphorylation event in T cells using mice with a RIPK1S25D phosphomimetic point mutation to endogenous RIPK1. We find that this mutation protects T cells from TNF induced cell death when IKK activity is inhibited in vitro, and can rescues development of IKK deficient thymocytes in vivo to a degree comparable with kinase dead RIPK1D138N. Together, these data show that phosphorylation of RIPK1S25 by IKK represents a key regulatory event promoting survival of T cells by IKK.


Subject(s)
NF-kappa B , Serine , Mice , Animals , Phosphorylation , NF-kappa B/metabolism , Serine/metabolism , Apoptosis , Tumor Necrosis Factor-alpha/metabolism , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Cell Death , Thymocytes/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
3.
Development ; 147(19)2020 10 07.
Article in English | MEDLINE | ID: mdl-32907850

ABSTRACT

Pre-T-cell receptor (TCR) signal transduction is required for developing thymocytes to differentiate from CD4-CD8- double-negative (DN) cell to CD4+CD8+ double-positive (DP) cell. Notch signalling is required for T-cell fate specification and must be maintained throughout ß-selection, but inappropriate Notch activation in DN4 and DP cells is oncogenic. Here, we show that pre-TCR signalling leads to increased expression of the transcriptional repressor Bcl6 and that Bcl6 is required for differentiation to DP. Conditional deletion of Bcl6 from thymocytes reduced pre-TCR-induced differentiation to DP cells, disrupted expansion and enrichment of intracellular TCRß+ cells within the DN population and increased DN4 cell death. Deletion also increased Notch1 activation and Notch-mediated transcription in the DP population. Thus, Bcl6 is required in thymocyte development for efficient differentiation from DN3 to DP and to attenuate Notch1 activation in DP cells. Given the importance of inappropriate NOTCH1 signalling in T-cell acute lymphoblastic leukaemia (T-ALL), and the involvement of BCL6 in other types of leukaemia, this study is important to our understanding of T-ALL.


Subject(s)
Receptor, Notch1/metabolism , Thymocytes/cytology , Thymocytes/metabolism , Animals , Blotting, Western , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/physiology , Flow Cytometry , Genotype , Mice , Receptor, Notch1/genetics , Signal Transduction/genetics , Signal Transduction/physiology
4.
Immunity ; 50(2): 348-361.e4, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30737145

ABSTRACT

NF-κB (nuclear factor κB) signaling is considered critical for single positive (SP) thymocyte development because loss of upstream activators of NF-κB, such as the IKK complex, arrests their development. We found that the compound ablation of RelA, cRel, and p50, required for canonical NF-κB transcription, had no impact upon thymocyte development. While IKK-deficient thymocytes were acutely sensitive to tumor necrosis factor (TNF)-induced cell death, Rel-deficient cells remained resistant, calling into question the importance of NF-κB as the IKK target required for thymocyte survival. Instead, we found that IKK controlled thymocyte survival by repressing cell-death-inducing activity of the serine/threonine kinase RIPK1. We observed that RIPK1 expression was induced during development of SP thymocytes and that IKK was required to prevent RIPK1-kinase-dependent death of SPs in vivo. Finally, we showed that IKK was required to protect Rel-deficient thymocytes from RIPK1-dependent cell death, underscoring the NF-κB-independent function of IKK during thymic development.


Subject(s)
I-kappa B Kinase/metabolism , NF-kappa B/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Thymocytes/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , Gene Expression Regulation/drug effects , I-kappa B Kinase/genetics , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Thymocytes/cytology , Thymocytes/drug effects , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/pharmacology
5.
Eur J Immunol ; 49(1): 66-78, 2019 01.
Article in English | MEDLINE | ID: mdl-30365177

ABSTRACT

The interferon-inducible transmembrane (Ifitm/Fragilis) genes encode homologous proteins that are induced by IFNs. Here, we show that IFITM proteins regulate murine CD4+ Th cell differentiation. Ifitm2 and Ifitm3 are expressed in wild-type (WT) CD4+ T cells. On activation, Ifitm3 was downregulated and Ifitm2 was upregulated. Resting Ifitm-family-deficient CD4+ T cells had higher expression of Th1-associated genes than WT and purified naive Ifitm-family-deficient CD4+ T cells differentiated more efficiently to Th1, whereas Th2 differentiation was inhibited. Ifitm-family-deficient mice, but not Ifitm3-deficient mice, were less susceptible than WT to induction of allergic airways disease, with a weaker Th2 response and less severe disease and lower Il4 but higher Ifng expression and IL-27 secretion. Thus, the Ifitm family is important in adaptive immunity, influencing Th1/Th2 polarization, and Th2 immunopathology.


Subject(s)
Hypersensitivity/immunology , Inflammation/immunology , Membrane Proteins/metabolism , Respiratory System/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Animals , Cell Differentiation/genetics , Cells, Cultured , Interferon-gamma/metabolism , Interleukin-27/metabolism , Interleukin-4/metabolism , Lymphocyte Activation/genetics , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Th1-Th2 Balance/genetics
6.
Oncotarget ; 8(15): 24163-24176, 2017 Apr 11.
Article in English | MEDLINE | ID: mdl-28445929

ABSTRACT

Kif7 is a ciliary kinesin motor protein that regulates mammalian Hedgehog pathway activation through influencing structure of the primary cilium. Here we show that Kif7 is required for normal T-cell development, despite the fact that T-cells lack primary cilia. Analysis of Kif7-deficient thymus showed that Kif7-deficiency increases the early CD44+CD25+CD4-CD8- thymocyte progenitor population but reduces differentiation to CD4+CD8+ double positive (DP) cell. At the transition from DP to mature T-cell, Kif7-deficiency selectively delayed maturation to the CD8 lineage. Expression of CD5, which correlates with TCR signal strength, was reduced on DP and mature CD4 and CD8 cells, as a result of thymocyte-intrinsic Kif7-deficiency, and Kif7-deficient T-cells from radiation chimeras activated less efficiently when stimulated with anti-CD3 and anti-CD28 in vitro. Kif7-deficient thymocytes showed higher expression of the Hedgehog target gene Ptch1 than WT, but were less sensitive to treatment with recombinant Shh, and Kif7-deficient T-cell development was refractory to neutralisation of endogenous Hh proteins, indicating that Kif7-deficient thymocytes were unable to interpret changes in the Hedgehog signal. In addition, Kif7-deficiency reduced cell-surface MHCII expression on thymic epithelial cells.


Subject(s)
Cell Differentiation/genetics , Epithelial Cells/metabolism , Kinesins/genetics , Major Histocompatibility Complex/genetics , Thymocytes/cytology , Thymocytes/metabolism , Thymus Gland/physiology , Animals , Biomarkers , Gene Expression , Genotype , Hedgehog Proteins/metabolism , Major Histocompatibility Complex/immunology , Mice , Mice, Knockout , Phenotype , Signal Transduction , Thymocytes/immunology
7.
J Dev Biol ; 4(3): 22, 2016 Jul 16.
Article in English | MEDLINE | ID: mdl-27504268

ABSTRACT

T cells develop in the thymus, which provides an essential environment for T cell fate specification, and for the differentiation of multipotent progenitor cells into major histocompatibility complex (MHC)-restricted, non-autoreactive T cells. Here we review the role of the Hedgehog signalling pathway in T cell development, thymic epithelial cell (TEC) development, and thymocyte-TEC cross-talk in the embryonic mouse thymus during the last week of gestation.

8.
Oncotarget ; 6(30): 28646-60, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26415229

ABSTRACT

Developing thymocytes require pre-TCR signalling to differentiate from CD4-CD8- double negative to CD4+CD8+ double positive cell. Here we followed the transcriptional response to pre-TCR signalling in a synchronised population of differentiating double negative thymocytes. This time series analysis revealed a complex transcriptional response, in which thousands of genes were up and down-regulated before changes in cell surface phenotype were detected. Genome-wide measurement of RNA degradation of individual genes showed great heterogeneity in the rate of degradation between different genes. We therefore used time course expression and degradation data and a genome wide transcriptional modelling (GWTM) strategy to model the transcriptional response of genes up-regulated on pre-TCR signal transduction. This analysis revealed five major temporally distinct transcriptional activities that up regulate transcription through time, whereas down-regulation of expression occurred in three waves. Our model thus placed known regulators in a temporal perspective, and in addition identified novel candidate regulators of thymocyte differentiation.


Subject(s)
Cell Differentiation , Models, Genetic , Protein Precursors/genetics , Receptors, Antigen, T-Cell/genetics , Thymocytes/metabolism , Transcription, Genetic , Animals , Cells, Cultured , Cluster Analysis , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental , Genetic Markers , Genome-Wide Association Study , Genotype , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , Homeodomain Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Phenotype , Protein Precursors/immunology , Protein Precursors/metabolism , RNA/genetics , RNA/metabolism , RNA Stability , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , Thymocytes/immunology , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism
9.
J Cell Sci ; 128(11): 2085-95, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25908851

ABSTRACT

Different tissues contain diverse and dynamic cellular niches, providing distinct signals to tissue-resident or migratory infiltrating immune cells. Hedgehog (Hh) proteins are secreted inter-cellular signalling molecules, which are essential during development and are important in cancer, post-natal tissue homeostasis and repair. Hh signalling mediated by the Hh-responsive transcription factor Gli2 also has multiple roles in T-lymphocyte development and differentiation. Here, we investigate the function of Gli2 in T-cell signalling and activation. Gene transcription driven by the Gli2 transcriptional activator isoform (Gli2A) attenuated T-cell activation and proliferation following T-cell receptor (TCR) stimulation. Expression of Gli2A in T-cells altered gene expression profiles, impaired the TCR-induced Ca(2+) flux and nuclear expression of NFAT2, suppressed upregulation of molecules essential for activation, and attenuated signalling pathways upstream of the AP-1 and NFκB complexes, leading to reduced activation of these important transcription factors. Inhibition of physiological Hh-dependent transcription increased NFκB activity upon TCR ligation. These data are important for understanding the molecular mechanisms of immunomodulation, particularly in tissues where Hh proteins or other Gli-activating ligands such as TGFß are upregulated, including during inflammation, tissue damage and repair, and in tumour microenvironments.


Subject(s)
Kruppel-Like Transcription Factors/genetics , NF-kappa B/genetics , Receptors, Antigen, T-Cell/genetics , Signal Transduction/genetics , Transcription Factor AP-1/genetics , Transcriptional Activation/genetics , Animals , Cell Differentiation/genetics , Cell Proliferation/genetics , Gene Expression Regulation/genetics , Hedgehog Proteins/genetics , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , NFATC Transcription Factors/genetics , T-Lymphocytes/metabolism , Transcriptome/genetics , Transforming Growth Factor beta/genetics , Up-Regulation/genetics , Zinc Finger Protein Gli2
10.
J Am Coll Cardiol ; 58(7): 752-61, 2011 Aug 09.
Article in English | MEDLINE | ID: mdl-21816313

ABSTRACT

OBJECTIVES: In this study we investigate: 1) the role of multidrug resistance protein-4 (MRP4), an organic anion unidirectional transporter, in modulating aspirin action on human platelet cyclooxygenase (COX)-1; and 2) whether the impairment of aspirin-COX-1 interaction, found in coronary artery bypass grafting (CABG) patients, could be dependent on MRP4-mediated transport. BACKGROUND: Platelets of CABG patients present a reduced sensitivity to aspirin despite in vivo and in vitro drug treatment. Aspirin is an organic anion and could be a substrate for MRP4. METHODS: Intracellular aspirin concentration and drug COX-1 activity, measured by thrombin-induced thromboxane B2 (TxB2) production, were evaluated in platelets obtained from healthy volunteers (HV) and hematopoietic-progenitor cell cultures reducing or not reducing MRP4-mediated transport. Platelet MRP4 expression was evaluated, in platelets from HV and CABG patients, by dot-blot or by immunogold-electromicrographs or immunofluorescence-microscopy analysis. RESULTS: Inhibition of MRP4-mediated transport by dipyridamole or Mk-571 increases aspirin entrapment and its in vitro effect on COX-1 activity (142.7 ± 34.6 pg/10(8) cells vs. 343.7 ± 169.3 pg/108 cells TxB2-production). Platelets derived from megakaryocytes transfected with MRP4 small interfering ribonucleic acid have a higher aspirin entrapment and drug COX-1 activity. Platelets from CABG patients showed a high expression of MRP4 whose in vitro inhibition enhanced aspirin effect on COX-1 (349 ± 141 pg/108 cells vs. 1,670 ± 646 pg/108 cells TxB2-production). CONCLUSIONS: Aspirin is a substrate for MRP4 and can be extruded from platelet through its transportation. Aspirin effect on COX-1 is little-related to MRP4-mediated aspirin transport in HV, but in CABG patients with MRP4 over-expression, its pharmacological inhibition enhances aspirin action in an efficient way.


Subject(s)
Aspirin/pharmacokinetics , Blood Platelets/metabolism , Coronary Artery Bypass , Fibrinolytic Agents/pharmacokinetics , Multidrug Resistance-Associated Proteins/pharmacology , Platelet Aggregation Inhibitors/pharmacokinetics , Adult , Aspirin/pharmacology , Biological Transport/drug effects , Blood Platelets/drug effects , Cells, Cultured , Cyclic AMP/pharmacology , Cyclooxygenase 1/metabolism , Dinoprostone/metabolism , Drug Interactions , Drug Resistance , Female , Fibrinolytic Agents/pharmacology , Humans , Male , Middle Aged , Multidrug Resistance-Associated Proteins/metabolism , Platelet Aggregation , Platelet Aggregation Inhibitors/pharmacology , Propionates/pharmacology , Prostaglandin-Endoperoxide Synthases/metabolism , Quinolines/pharmacology , RNA, Small Interfering/metabolism , Salicylic Acid/pharmacokinetics , Thromboxane B2/metabolism
11.
J Immunol ; 186(11): 6199-206, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21508258

ABSTRACT

Notch3 overexpression has been previously shown to positively regulate the generation and function of naturally occurring regulatory T cells and the expression of Foxp3, in cooperation with the pTα/pre-TCR pathway. In this study, we show that Notch3 triggers the trans activation of Foxp3 promoter depending on the T cell developmental stage. Moreover, we discovered a novel CSL/NF-κB overlapping binding site within the Foxp3 promoter, and we demonstrate that the activation of NF-κB, mainly represented by p65-dependent canonical pathway, plays a positive role in Notch3-dependent regulation of Foxp3 transcription. Accordingly, the deletion of protein kinase C, which mediates canonical NF-κB activation, markedly reduces regulatory T cell number and per cell Foxp3 expression in transgenic mice with a constitutive activation of Notch3 signaling. Collectively, our data indicate that the cooperation among Notch3, protein kinase C, and p65/NF-κB subunit modulates Foxp3 expression, adding new insights in the understanding of the molecular mechanisms involved in regulatory T cell homeostasis and function.


Subject(s)
Forkhead Transcription Factors/metabolism , NF-kappa B/metabolism , Receptors, Notch/metabolism , Signal Transduction , Animals , Cell Line , Cells, Cultured , Flow Cytometry , Forkhead Transcription Factors/genetics , Immunoblotting , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Promoter Regions, Genetic/genetics , Protein Binding , Protein Kinase C/genetics , Protein Kinase C/metabolism , Protein Kinase C-theta , Receptor, Notch3 , Receptors, Notch/genetics , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes, Regulatory/metabolism , Thymus Gland/cytology , Thymus Gland/metabolism , Time Factors , Transcription Factor RelA/metabolism , Transcription, Genetic , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...