Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Nat Immunol ; 25(3): 512-524, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38356059

ABSTRACT

Interleukin-23 (IL-23) is a proinflammatory cytokine mainly produced by myeloid cells that promotes tumor growth in various preclinical cancer models and correlates with adverse outcomes. However, as to how IL-23 fuels tumor growth is unclear. Here, we found tumor-associated macrophages to be the main source of IL-23 in mouse and human tumor microenvironments. Among IL-23-sensing cells, we identified a subset of tumor-infiltrating regulatory T (Treg) cells that display a highly suppressive phenotype across mouse and human tumors. The use of three preclinical models of solid cancer in combination with genetic ablation of Il23r in Treg cells revealed that they are responsible for the tumor-promoting effect of IL-23. Mechanistically, we found that IL-23 sensing represents a crucial signal driving the maintenance and stabilization of effector Treg cells involving the transcription factor Foxp3. Our data support that targeting the IL-23/IL-23R axis in cancer may represent a means of eliciting antitumor immunity.


Subject(s)
Interleukin-23 , Neoplasms , Animals , Humans , Mice , Cytokines , Interleukin-23/genetics , Neoplasms/genetics , T-Lymphocytes , Tumor Microenvironment
2.
Nat Immunol ; 11(11): 1057-62, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20935646

ABSTRACT

After being activated by antigen, helper T lymphocytes switch from a resting state to clonal expansion. This switch requires inactivation of the transcription factor Foxo1, a suppressor of proliferation expressed in resting helper T lymphocytes. In the early antigen-dependent phase of expansion, Foxo1 is inactivated by antigen receptor-mediated post-translational modifications. Here we show that in the late phase of expansion, Foxo1 was no longer post-translationally regulated but was inhibited post-transcriptionally by the interleukin 2 (IL-2)-induced microRNA miR-182. Specific inhibition of miR-182 in helper T lymphocytes limited their population expansion in vitro and in vivo. Our results demonstrate a central role for miR-182 in the physiological regulation of IL-2-driven helper T cell-mediated immune responses and open new therapeutic possibilities.


Subject(s)
Interleukin-2/immunology , MicroRNAs/immunology , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Arthritis/immunology , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
3.
J Autoimmun ; 89: 41-52, 2018 05.
Article in English | MEDLINE | ID: mdl-29183643

ABSTRACT

In T lymphocytes, expression of miR-148a is induced by T-bet and Twist1, and is specific for pro-inflammatory Th1 cells. In these cells, miR-148a inhibits the expression of the pro-apoptotic protein Bim and promotes their survival. Here we use sequence-specific cholesterol-modified oligonucleotides against miR-148a (antagomir-148a) for the selective elimination of pro-inflammatory Th1 cells in vivo. In the murine model of transfer colitis, antagomir-148a treatment reduced the number of pro-inflammatory Th1 cells in the colon of colitic mice by 50% and inhibited miR-148a expression by 71% in the remaining Th1 cells. Expression of Bim protein in colonic Th1 cells was increased. Antagomir-148a-mediated reduction of Th1 cells resulted in a significant amelioration of colitis. The effect of antagomir-148a was selective for chronic inflammation. Antigen-specific memory Th cells that were generated by an acute immune reaction to nitrophenylacetyl-coupled chicken gamma globulin (NP-CGG) were not affected by treatment with antagomir-148a, both during the effector and the memory phase. In addition, antibody titers to NP-CGG were not altered. Thus, antagomir-148a might qualify as an effective drug to selectively deplete pro-inflammatory Th1 cells of chronic inflammation without affecting the protective immunological memory.


Subject(s)
Antagomirs/genetics , Colitis/immunology , Colon/immunology , Inflammation/immunology , MicroRNAs/genetics , Th1 Cells/physiology , Animals , Cell Differentiation , Cells, Cultured , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism
4.
Eur J Immunol ; 45(4): 1206-15, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25678371

ABSTRACT

B cells undergo affinity maturation and class switch recombination of their immunoglobulin receptors during a germinal center (GC) reaction, before they differentiate into long-lived antibody-secreting plasma cells (PCs). Transcription factors such as Bach2 and Mitf are essential during this process, as they delay premature differentiation of GC B cells by repressing Blimp-1 and IRF4, two transcription factors required for terminal PC differentiation. Therefore, Bach2 and Mitf expression must be attenuated in activated B cells to allow terminal PC differentiation, but the precise mechanism remains enigmatic. Here, we provide evidence that miR-148a, a small noncoding microRNA, fosters PC differentiation and survival. Next-generation sequencing revealed that miR-148a is the most abundant microRNA in primary human and murine PCs, and its expression is upregulated in activated murine B cells and coincides with Blimp-1 synthesis. miR-148a targets Bach2, Mitf and proapoptotic factors such as PTEN and Bim. When prematurely expressed, miR-148a promotes the differentiation and survival of plasmablasts and reduces frequencies of IgG1(+) cells in primary B-cell cultures. In summary, we propose that miR-148a is a new player in the regulatory network controlling terminal PC differentiation and could, therefore, be a therapeutic target for interfering with PC differentiation and survival.


Subject(s)
Basic-Leucine Zipper Transcription Factors/biosynthesis , Cell Differentiation/genetics , MicroRNAs/physiology , Microphthalmia-Associated Transcription Factor/biosynthesis , Plasma Cells/cytology , Animals , Apoptosis Regulatory Proteins/biosynthesis , B-Lymphocytes/immunology , Base Sequence , Bcl-2-Like Protein 11 , Cell Differentiation/immunology , Cell Survival , Gene Knockdown Techniques , Germinal Center/cytology , HEK293 Cells , Humans , Immunoglobulin Class Switching/genetics , Immunoglobulin Class Switching/immunology , Interferon Regulatory Factors/biosynthesis , Lymphocyte Activation/genetics , Membrane Proteins/biosynthesis , Mice , MicroRNAs/genetics , PTEN Phosphohydrolase/biosynthesis , Positive Regulatory Domain I-Binding Factor 1 , Proto-Oncogene Proteins/biosynthesis , Repressor Proteins/biosynthesis , Sequence Analysis, DNA
5.
Eur J Immunol ; 45(4): 1192-205, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25486906

ABSTRACT

Repeatedly activated T helper 1 (Th1) cells present during chronic inflammation can efficiently adapt to the inflammatory milieu, for example, by expressing the transcription factor Twist1, which limits the immunopathology caused by Th1 cells. Here, we show that in repeatedly activated murine Th1 cells, Twist1 and T-bet induce expression of microRNA-148a (miR-148a). miR-148a regulates expression of the proapoptotic gene Bim, resulting in a decreased Bim/Bcl2 ratio. Inhibition of miR-148a by antagomirs in repeatedly activated Th1 cells increases the expression of Bim, leading to enhanced apoptosis. Knockdown of Bim expression by siRNA in miR-148a antagomir-treated cells restores viability of the Th1 cells, demonstrating that miR-148a controls survival by regulating Bim expression. Thus, Twist1 and T-bet not only control the differentiation and function of Th1 cells, but also their persistence in chronic inflammation.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Apoptosis/genetics , Gene Expression Regulation , Membrane Proteins/genetics , MicroRNAs/physiology , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/genetics , T-Box Domain Proteins/physiology , Th1 Cells/immunology , Twist-Related Protein 1/metabolism , Animals , Arthritis, Rheumatoid/immunology , Bcl-2-Like Protein 11 , Cell Survival/immunology , Cells, Cultured , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Nuclear Proteins/genetics , RNA Interference , RNA, Small Interfering , T-Box Domain Proteins/genetics , Twist-Related Protein 1/genetics
6.
J Biol Chem ; 289(39): 26752-26761, 2014 Sep 26.
Article in English | MEDLINE | ID: mdl-25037220

ABSTRACT

Th2 memory lymphocytes have imprinted their Il4 genes epigenetically for expression in dependence of T cell receptor restimulation. However, in a given restimulation, not all Th cells with a memory for IL-4 expression express IL-4. Here, we show that in reactivated Th2 cells, the transcription factors NFATc2, NF-kB p65, c-Maf, p300, Brg1, STAT6, and GATA-3 assemble at the Il4 promoter in Th2 cells expressing IL-4 but not in Th2 cells not expressing it. NFATc2 is critical for assembly of this transcription factor complex. Because NFATc2 translocation into the nucleus occurs in an all-or-none fashion, dependent on complete dephosphorylation by calcineurin, NFATc2 controls the frequencies of cells reexpressing Il4, translates analog differences in T cell receptor stimulation into a digital decision for Il4 reexpression, and instructs all reexpressing cells to express the same amount of IL-4. This analog-to-digital conversion may be critical for the immune system to respond to low concentrations of antigens.


Subject(s)
Cell Nucleus/metabolism , Gene Expression Regulation/physiology , Interleukin-4/biosynthesis , NFATC Transcription Factors/metabolism , Response Elements/physiology , Th2 Cells/metabolism , Active Transport, Cell Nucleus/physiology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Nucleus/genetics , Cell Nucleus/immunology , DNA Helicases/genetics , DNA Helicases/immunology , DNA Helicases/metabolism , E1A-Associated p300 Protein/genetics , E1A-Associated p300 Protein/immunology , E1A-Associated p300 Protein/metabolism , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/immunology , GATA3 Transcription Factor/metabolism , Interleukin-4/genetics , Interleukin-4/immunology , Mice , Mice, Inbred BALB C , Mice, Transgenic , NFATC Transcription Factors/genetics , NFATC Transcription Factors/immunology , Nuclear Proteins/genetics , Nuclear Proteins/immunology , Nuclear Proteins/metabolism , Phosphorylation/physiology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , STAT6 Transcription Factor/genetics , STAT6 Transcription Factor/immunology , STAT6 Transcription Factor/metabolism , Th2 Cells/cytology , Th2 Cells/immunology , Transcription Factor RelA/genetics , Transcription Factor RelA/immunology , Transcription Factor RelA/metabolism , Transcription Factors/genetics , Transcription Factors/immunology , Transcription Factors/metabolism
7.
J Immunol ; 183(11): 7362-70, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19917688

ABSTRACT

Phosphorylation of CARMA1 is a crucial event initiating the assembly of IkappaB kinase and JNK signaling complexes downstream of activated Ag receptors. We previously mapped three protein kinase C (PKC) target sites in murine CARMA1 in vitro, and demonstrated that mutation of two of these serines (S564 and S657) resulted in reduced NF-kappaB activation, whereas mutation of the third serine (S649) had no clear effect. In this study, we report that when low concentrations of Ag receptor activators are used, loss of S649 (by mutation to alanine) promotes enhanced IkappaB kinase and JNK activation in both B and T cell lines. Reconstitution of CARMA1(-/-) DT40 B cells with CARMA1 S649A leads to increased cell death and reduced cell growth in comparison to wild-type CARMA1, likely a result of enhanced JNK activation. To directly determine whether S649 is modified in vivo, we generated phospho-specific Abs recognizing phospho-S649, and phospho-S657 as a positive control. Although phospho-S657 peaked and declined rapidly after Ag receptor stimulation, phospho-S649 occurred later and was maintained for a significantly longer period poststimulation in both B and T cells. Interestingly, phospho-S657 was completely abolished in PKCbeta-deficient B cells, whereas delayed phosphorylation at S649 was partially intact and depended, in part, upon novel PKC activity. Thus, distinct PKC-mediated CARMA1 phosphorylation events exert opposing effects on the activation status of CARMA1. We propose that early phosphorylation events at S657 and S564 promote the initial assembly of the CARMA1 signalosome, whereas later phosphorylation at S649 triggers CARMA1 down-regulation.


Subject(s)
B-Lymphocytes/enzymology , CARD Signaling Adaptor Proteins/metabolism , Guanylate Cyclase/metabolism , Protein Kinase C/metabolism , Signal Transduction/immunology , T-Lymphocytes/enzymology , Animals , B-Lymphocytes/immunology , Blotting, Western , CARD Signaling Adaptor Proteins/immunology , Cell Line , Chickens , Down-Regulation , Enzyme Activation , Guanylate Cyclase/immunology , Humans , I-kappa B Kinase/immunology , I-kappa B Kinase/metabolism , Immunoprecipitation , MAP Kinase Kinase 4/immunology , MAP Kinase Kinase 4/metabolism , Mice , Phosphorylation , Protein Kinase C/immunology , Serine/metabolism , T-Lymphocytes/immunology
8.
Sci Immunol ; 6(65): eabf3111, 2021 Nov 19.
Article in English | MEDLINE | ID: mdl-34797691

ABSTRACT

Medullary thymic epithelial cells (mTECs) are key antigen-presenting cells mediating T cell tolerance to prevent harmful autoimmunity. mTECs both negatively select self-reactive T cells and promote the development of thymic regulatory T cells (tTregs) that mediate peripheral tolerance. The relative importance of these two mechanisms of thymic education to prevent autoimmunity is unclear. We generated a mouse model to specifically target the development and function of mTECs by conditional ablation of the NF-κB­inducing kinase (NIK) in the TEC compartment. In contrast to germline-deficient NIK−/− mice, Foxn1CreNIKfl/fl mice rapidly developed fatal T cell­dependent multiorgan autoimmunity shortly after birth. Thymic transplantation and adoptive transfer experiments demonstrated that autoimmunity arises specifically from the emergence of dysfunctional tTregs. Thus, Treg function, rather than negative selection, enforces the protection of peripheral tissues from autoimmune attack.


Subject(s)
Autoimmunity , Epithelial Cells/immunology , Forkhead Transcription Factors/immunology , Protein Serine-Threonine Kinases/immunology , T-Lymphocytes, Regulatory/immunology , Thymus Gland/immunology , Animals , Humans , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Knockout , Protein Serine-Threonine Kinases/deficiency , Thymus Gland/cytology , NF-kappaB-Inducing Kinase
9.
Cell Rep ; 31(13): 107826, 2020 06 30.
Article in English | MEDLINE | ID: mdl-32610127

ABSTRACT

Regulatory T cells (Tregs) represent a major population in the control of immune homeostasis and autoimmunity. Here we show that Fos-like 2 (Fosl2), a TCR-induced AP1 transcription factor, represses Treg development and controls autoimmunity. Mice overexpressing Fosl2 (Fosl2tg) indeed show a systemic inflammatory phenotype, with immune infiltrates in multiple organs. This phenotype is absent in Fosl2tg × Rag2-/- mice lacking T and B cells, and Fosl2 induces T cell-intrinsic reduction of Treg development that is responsible for the inflammatory phenotype. Fosl2tg T cells can transfer inflammation, which is suppressed by the co-delivery of Tregs, while Fosl2 deficiency in T cells reduces the severity of autoimmunity in the EAE model. We find that Fosl2 could affect expression of FoxP3 and other Treg development genes. Our data highlight the importance of AP1 transcription factors, in particular Fosl2, during T cell development to determine Treg differentiation and control autoimmunity.


Subject(s)
Autoimmunity , Fos-Related Antigen-2/metabolism , Inflammation/immunology , Inflammation/pathology , T-Lymphocytes, Regulatory/immunology , Transcription Factor AP-1/metabolism , Animals , Bone Marrow/pathology , DNA-Binding Proteins/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Forkhead Transcription Factors/metabolism , Gene Deletion , Mice, Inbred C57BL , Phenotype , Receptors, Antigen, T-Cell/metabolism
10.
Nat Commun ; 11(1): 2570, 2020 05 22.
Article in English | MEDLINE | ID: mdl-32444631

ABSTRACT

At present, it is not clear how memory B lymphocytes are maintained over time, and whether only as circulating cells or also residing in particular tissues. Here we describe distinct populations of isotype-switched memory B lymphocytes (Bsm) of murine spleen and bone marrow, identified according to individual transcriptional signature and B cell receptor repertoire. A population of marginal zone-like cells is located exclusively in the spleen, while a population of quiescent Bsm is found only in the bone marrow. Three further resident populations, present in spleen and bone marrow, represent transitional and follicular B cells and B1 cells, respectively. A population representing 10-20% of spleen and bone marrow memory B cells is the only one qualifying as circulating. In the bone marrow, all cells individually dock onto VCAM1+ stromal cells and, reminiscent of resident memory T and plasma cells, are void of activation, proliferation and mobility.


Subject(s)
B-Lymphocytes/immunology , Bone Marrow Cells/immunology , Immunoglobulin Class Switching , Immunologic Memory , Spleen/immunology , Adjuvants, Immunologic/pharmacology , Animals , Animals, Wild/immunology , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , Bone Marrow Cells/cytology , Cell Cycle , Cell Proliferation/genetics , Gene Expression Regulation/immunology , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Antigen, B-Cell/genetics , Receptors, Antigen, B-Cell/immunology , Spleen/cytology , Stromal Cells/cytology , Vascular Cell Adhesion Molecule-1/metabolism
11.
Nat Protoc ; 14(7): 1946-1969, 2019 07.
Article in English | MEDLINE | ID: mdl-31160786

ABSTRACT

The interrogation of single cells is revolutionizing biology, especially our understanding of the immune system. Flow cytometry is still one of the most versatile and high-throughput approaches for single-cell analysis, and its capability has been recently extended to detect up to 28 colors, thus approaching the utility of cytometry by time of flight (CyTOF). However, flow cytometry suffers from autofluorescence and spreading error (SE) generated by errors in the measurement of photons mainly at red and far-red wavelengths, which limit barcoding and the detection of dim markers. Consequently, development of 28-color fluorescent antibody panels for flow cytometry is laborious and time consuming. Here, we describe the steps that are required to successfully achieve 28-color measurement capability. To do this, we provide a reference map of the fluorescence spreading errors in the 28-color space to simplify panel design and predict the success of fluorescent antibody combinations. Finally, we provide detailed instructions for the computational analysis of such complex data by existing, popular algorithms (PhenoGraph and FlowSOM). We exemplify our approach by designing a high-dimensional panel to characterize the immune system, but we anticipate that our approach can be used to design any high-dimensional flow cytometry panel of choice. The full protocol takes a few days to complete, depending on the time spent on panel design and data analysis.


Subject(s)
Antibodies, Monoclonal , Flow Cytometry/methods , Fluorescent Antibody Technique/methods , Single-Cell Analysis/methods , Algorithms , Biomarkers , Buffers , Coloring Agents , Computational Biology/methods , Forkhead Transcription Factors/immunology , Humans , Signaling Lymphocytic Activation Molecule Family/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology
12.
Front Immunol ; 9: 2813, 2018.
Article in English | MEDLINE | ID: mdl-30574141

ABSTRACT

Proinflammatory type 1 T helper (Th1) cells are enriched in inflamed tissues and contribute to the maintenance of chronic inflammation in rheumatic diseases. Here we show that the microRNA- (miR-) 31 is upregulated in murine Th1 cells with a history of repeated reactivation and in memory Th cells isolated from the synovial fluid of patients with rheumatic joint disease. Knock-down of miR-31 resulted in the upregulation of genes associated with cytoskeletal rearrangement and motility and induced the expression of target genes involved in T cell activation, chemokine receptor- and integrin-signaling. Accordingly, inhibition of miR-31 resulted in increased migratory activity of repeatedly activated Th1 cells. The transcription factors T-bet and FOXO1 act as positive and negative regulators of T cell receptor (TCR)-mediated miR-31 expression, respectively. Taken together, our data show that a gene regulatory network involving miR-31, T-bet, and FOXO1 controls the migratory behavior of proinflammatory Th1 cells.


Subject(s)
Cell Movement/immunology , MicroRNAs/immunology , Th1 Cells/immunology , Animals , Cell Movement/genetics , Female , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/immunology , Humans , Inflammation/genetics , Inflammation/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , MicroRNAs/genetics , T-Box Domain Proteins/genetics , T-Box Domain Proteins/immunology
13.
Sci Transl Med ; 10(469)2018 11 28.
Article in English | MEDLINE | ID: mdl-30487251

ABSTRACT

Allogeneic hematopoietic cell transplantation (allo-HCT) not only is an effective treatment for several hematologic malignancies but can also result in potentially life-threatening graft-versus-host disease (GvHD). GvHD is caused by T cells within the allograft attacking nonmalignant host tissues; however, these same T cells mediate the therapeutic graft-versus-leukemia (GvL) response. Thus, there is an urgent need to understand how to mechanistically uncouple GvL from GvHD. Using preclinical models of full and partial MHC-mismatched HCT, we here show that the granulocyte-macrophage colony-stimulating factor (GM-CSF) produced by allogeneic T cells distinguishes between the two processes. GM-CSF drives GvHD pathology by licensing donor-derived phagocytes to produce inflammatory mediators such as interleukin-1ß and reactive oxygen species. In contrast, GM-CSF did not affect allogeneic T cells or their capacity to eliminate leukemic cells, retaining undiminished GvL responses. Last, tissue biopsies and peripheral blood mononuclear cells from patients with grade IV GvHD showed an elevation of GM-CSF-producing T cells, suggesting that GM-CSF neutralization has translational potential in allo-HCT.


Subject(s)
Graft vs Host Disease/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Immunity/drug effects , Leukemia/immunology , Myeloid Cells/metabolism , Animals , Graft vs Host Disease/pathology , Hematopoietic Stem Cell Transplantation , Histocompatibility Antigens/metabolism , Humans , Interferon-gamma/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Myeloid Cells/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Transplantation, Homologous
14.
Transplantation ; 100(4): 898-907, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26444957

ABSTRACT

BACKGROUND: MicroRNAs (miRNAs, miR) hold important roles in the posttranscriptional regulation of gene expression. Their function has been correlated with kidney disease, and they might represent a new class of biomarkers for frequent evaluation of renal graft status. We analyzed their potential in identifying severe T cell-mediated vascular rejection (TCMVR) (Banff 4-II/III) in kidney transplanted patients. METHODS: Microarray experiments and semiquantitative real-time reverse transcription polymerase chain reaction were performed with total RNA isolated from blood cells of kidney graft recipients. Initial microarray analysis revealed 23 differentially expressed miRNAs distinguishing patients with TCMVR from patients with stable grafts. From these, we validated and further determined the expression of 6 differentially expressed miRNAs and 2 control miRNAs in 161 samples from patients with T cell-mediated rejection (Banff 3-Borderline, Banff 4-I/II/III), Banff-2 antibody-mediated rejection, Banff-5 interstitial fibrosis/tubular atrophy, in samples from stable patients and in samples from patients with urinary tract infection using real-time reverse transcription polymerase chain reaction. RESULTS: Expression levels of all 6 candidate miRNAs were significantly downregulated in blood of TCMVR patients compared to the other groups and displayed high sensitivities and specificities for diagnosing TCMVR. The combination of 5 miRNAs, identified by an unbiased multivariate logistic regression followed by cross-validation, enhanced the sensitivity and specificity for the diagnosis of TCMVR after renal transplantation. CONCLUSIONS: The combined measurement of miRNA-15B, miRNA-16, miRNA-103A, miRNA-106A, and miRNA-107 may help to better identify TCMVR after renal transplantation in a precise and clinically applicable way.


Subject(s)
Graft Rejection/blood , Graft Rejection/genetics , Immunity, Cellular/genetics , Kidney Transplantation/adverse effects , MicroRNAs/blood , MicroRNAs/genetics , T-Lymphocytes/immunology , Area Under Curve , Cluster Analysis , Down-Regulation , Gene Expression Profiling/methods , Genetic Markers , Graft Rejection/diagnosis , Graft Rejection/immunology , Humans , Logistic Models , Multivariate Analysis , Oligonucleotide Array Sequence Analysis , Predictive Value of Tests , ROC Curve , Real-Time Polymerase Chain Reaction , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Risk Factors , Treatment Outcome
15.
J Immunol Methods ; 426: 128-33, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26187895

ABSTRACT

Depending on their physiological expression level, microRNAs (miRNA) address different target genes, thus have different biological functions. To identify these, the physiological expression has to be blocked. Here, we describe the use of inhibitory cholesterol-modified oligonucleotides (Antagomirs) to inhibit miRNAs selectively in primary human and murine T and B lymphocytes. Due to their lipophilic cholesterol tag Antagomirs enter primary lymphocytes efficiently and quantitatively. We show here that at concentrations of 0.125 to 1µM, Antagomirs selectively inhibit expression of their target miRNA up to 99.5% without affecting cell viability.


Subject(s)
B-Lymphocytes/immunology , Gene Knockdown Techniques , MicroRNAs/antagonists & inhibitors , Oligonucleotides/genetics , Th1 Cells/immunology , Animals , Biological Transport , Cells, Cultured , Humans , Mice , Mice, Inbred C57BL , MicroRNAs/biosynthesis , MicroRNAs/genetics , Spleen/cytology , Spleen/immunology
16.
J Exp Med ; 212(2): 217-33, 2015 Feb 09.
Article in English | MEDLINE | ID: mdl-25646266

ABSTRACT

The co-stimulators ICOS (inducible T cell co-stimulator) and CD28 are both important for T follicular helper (TFH) cells, yet their individual contributions are unclear. Here, we show that each molecule plays an exclusive role at different stages of TFH cell development. While CD28 regulated early expression of the master transcription factor Bcl-6, ICOS co-stimulation was essential to maintain the phenotype by regulating the novel TFH transcription factor Klf2 via Foxo1. Klf2 directly binds to Cxcr5, Ccr7, Psgl-1, and S1pr1, and low levels of Klf2 were essential to maintain this typical TFH homing receptor pattern. Blocking ICOS resulted in relocation of fully developed TFH cells back to the T cell zone and reversion of their phenotype to non-TFH effector cells, which ultimately resulted in breakdown of the germinal center response. Our study describes for the first time the exclusive role of ICOS and its downstream signaling in the maintenance of TFH cells by controlling their anatomical localization in the B cell follicle.


Subject(s)
Gene Expression Regulation , Inducible T-Cell Co-Stimulator Protein/metabolism , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Phenotype , T-Lymphocyte Subsets , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Animals , CD28 Antigens/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Down-Regulation , Female , Forkhead Box Protein O1 , Forkhead Transcription Factors/metabolism , Gene Expression , Germinal Center/immunology , Germinal Center/metabolism , Humans , Inducible T-Cell Co-Stimulator Ligand/metabolism , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , Palatine Tonsil/immunology , Palatine Tonsil/metabolism , Protein Binding , Receptors, CCR7/metabolism , Receptors, CXCR5/metabolism , Signal Transduction , T-Lymphocytes, Helper-Inducer/cytology
17.
Nat Commun ; 6: 8576, 2015 Oct 13.
Article in English | MEDLINE | ID: mdl-26815406

ABSTRACT

Regulatory T-cells induced via IL-2 and TGFß in vitro (iTreg) suppress immune cells and are potential therapeutics during autoimmunity. However, several reports described their re-differentiation into pathogenic cells in vivo and loss of their key functional transcription factor (TF) FOXP3 after T-cell antigen receptor (TCR)-signalling in vitro. Here, we show that TCR-activation antagonizes two necessary TFs for foxp3 gene transcription, which are themselves regulated by phosphorylation. Although the tyrosine phosphatase PTPN2 is induced to restrain IL-2-mediated phosphorylation of the TF STAT5, expression of the TF FOXO1 is downregulated and miR-182, a suppressor of FOXO1 expression, is upregulated. TGFß counteracts the FOXP3-depleting TCR-signal by reassuring FOXO1 expression and by re-licensing STAT5 phosphorylation. Overexpressed phosphorylation-independent active versions of FOXO1 and STAT5 or knockdown of PTPN2 restores FOXP3 expression despite TCR-signal and absence of TGFß. This study suggests novel targets for stabilisation and less dangerous application of iTreg during devastating inflammation.


Subject(s)
Forkhead Transcription Factors/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 2/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes, Regulatory/metabolism , Animals , Blotting, Western , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Female , Flow Cytometry , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Male , Mice , Protein Tyrosine Phosphatase, Non-Receptor Type 2/genetics , Receptors, Antigen, T-Cell/genetics
18.
Sci Transl Med ; 6(241): 241ra80, 2014 Jun 18.
Article in English | MEDLINE | ID: mdl-24944195

ABSTRACT

Although T helper 17 (TH17) cells have been acknowledged as crucial mediators of autoimmune tissue damage, the effector cytokines responsible for their pathogenicity still remain poorly defined, particularly in humans. In mouse models of autoimmunity, the pathogenicity of TH17 cells has recently been associated with their production of granulocyte-macrophage colony-stimulating factor (GM-CSF). We analyzed the regulation of GM-CSF expression by human TH cell subsets. Surprisingly, the induction of GM-CSF expression by human TH cells is constrained by the interleukin-23 (IL-23)/ROR-γt/TH17 cell axis but promoted by the IL-12/T-bet/TH1 cell axis. IL-2-mediated signal transducer and activator of transcription 5 (STAT5) signaling induced GM-CSF expression in naïve and memory TH cells, whereas STAT3 signaling blocked it. The opposite effect was observed for IL-17 expression. Ex vivo, GM-CSF(+) TH cells that coexpress interferon-γ and T-bet could be distinguished by differential chemokine receptor expression from a previously uncharacterized subset of GM-CSF-only-producing TH cells that did not express TH1, TH2, and TH17 signature cytokines or master transcription factors. Our findings demonstrate distinct and counterregulatory pathways for the generation of IL-17- and GM-CSF-producing cells and also suggest a pathogenic role for GM-CSF(+) T cells in the inflamed brain of multiple sclerosis (MS) patients. This provides not only a scientific rationale for depleting T cell-derived GM-CSF in MS patients but also multiple new molecular checkpoints for therapeutic GM-CSF suppression, which, unlike in mice, do not associate with the TH17 but instead with the TH1 axis.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Interleukin-17/metabolism , T-Lymphocytes, Helper-Inducer/metabolism , Humans , STAT3 Transcription Factor/metabolism , Signal Transduction
19.
Ann N Y Acad Sci ; 1247: 46-55, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22236474

ABSTRACT

The Forkhead box O (FoxO) family of transcription factors is important for the maintenance of immunological homeostasis and tolerance by controlling the development and function of B and T lymphocytes. Because dysregulation in FoxO activity can result in chronic inflammation and autoimmunity, the transcriptional activity of FoxO proteins is tightly controlled and generally dependent on complex posttranslational modifications that lead either to their nuclear entry and subsequent activation or, alternatively, to their nuclear export. The phosphatidylinositol 3-kinase (PI3K)-protein kinase B (PKB/Akt) axis represents the major pathway phosphorylating and thereby inactivating FoxO proteins. However, recent results have revealed an additional posttranscriptional mechanism of FoxO inactivation by microRNAs. The discovery of this molecular pathway may provide a new therapeutic avenue for the modulation of FoxO activity in immune-mediated diseases using either microRNA targeting antagomirs or synthetic microRNA mimics, a topic that is addressed in this review.


Subject(s)
Forkhead Transcription Factors/genetics , Lymphocytes/metabolism , MicroRNAs/metabolism , Signal Transduction , Adaptive Immunity/genetics , Animals , Forkhead Transcription Factors/metabolism , Humans , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA Processing, Post-Transcriptional
SELECTION OF CITATIONS
SEARCH DETAIL