Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 119
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Immunity ; 47(4): 710-722.e6, 2017 10 17.
Article in English | MEDLINE | ID: mdl-29045902

ABSTRACT

Gastro-intestinal helminth infections trigger the release of interleukin-33 (IL-33), which induces type-2 helper T cells (Th2 cells) at the site of infection to produce IL-13, thereby contributing to host resistance in a T cell receptor (TCR)-independent manner. Here, we show that, as a prerequisite for IL-33-induced IL-13 secretion, Th2 cells required the expression of the epidermal growth factor receptor (EGFR) and of its ligand, amphiregulin, for the formation of a signaling complex between T1/ST2 (the IL-33R) and EGFR. This shared signaling complex allowed IL-33 to induce the EGFR-mediated activation of the MAP-kinase signaling pathway and consequently the expression of IL-13. Lack of EGFR expression on T cells abrogated IL-13 expression in infected tissues and impaired host resistance. EGFR expression on Th2 cells was TCR-signaling dependent, and therefore, our data reveal a mechanism by which antigen presentation controls the innate effector function of Th2 cells at the site of inflammation.


Subject(s)
ErbB Receptors/immunology , Interleukin-13/immunology , Interleukin-33/immunology , Receptors, Antigen, T-Cell/immunology , Th2 Cells/immunology , Amphiregulin/immunology , Amphiregulin/metabolism , Animals , Cell Line , Cells, Cultured , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Expression/genetics , Gene Expression/immunology , Gene Expression Profiling/methods , HEK293 Cells , Humans , Interleukin-13/genetics , Interleukin-13/metabolism , Interleukin-33/genetics , Interleukin-33/metabolism , MAP Kinase Signaling System/immunology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nematospiroides dubius/immunology , Nematospiroides dubius/physiology , Nocardia/immunology , Nocardia/physiology , Nocardia Infections/immunology , Nocardia Infections/metabolism , Nocardia Infections/microbiology , Receptors, Antigen, T-Cell/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Strongylida Infections/immunology , Strongylida Infections/metabolism , Strongylida Infections/parasitology , Th2 Cells/metabolism
2.
Rheumatology (Oxford) ; 62(8): 2887-2897, 2023 08 01.
Article in English | MEDLINE | ID: mdl-36625523

ABSTRACT

OBJECTIVES: How the local inflammatory environment regulates epigenetic changes in the context of inflammatory arthritis remains unclear. Here we assessed the transcriptional and active enhancer profile of monocytes derived from the inflamed joints of JIA patients, a model well-suited for studying inflammatory arthritis. METHODS: RNA sequencing and H3K27me3 chromatin immunoprecipitation sequencing (ChIP-seq) were used to analyse the transcriptional and epigenetic profile, respectively, of JIA synovial fluid-derived monocytes. RESULTS: Synovial-derived monocytes display an activated phenotype, which is regulated on the epigenetic level. IFN signalling-associated genes are increased and epigenetically altered in synovial monocytes, indicating a driving role for IFN in establishing the local inflammatory phenotype. Treatment of synovial monocytes with the Janus-associated kinase (JAK) inhibitor ruxolitinib, which inhibits IFN signalling, transformed the activated enhancer landscape and reduced disease-associated gene expression, thereby inhibiting the inflammatory phenotype. CONCLUSION: This study provides novel insights into epigenetic regulation of inflammatory arthritis patient-derived monocytes and highlights the therapeutic potential of epigenetic modulation for the treatment of inflammatory rheumatic diseases.


Subject(s)
Arthritis , Monocytes , Humans , Monocytes/metabolism , Epigenesis, Genetic , Arthritis/metabolism , Synovial Fluid/metabolism , Phenotype
3.
Immunity ; 38(2): 275-84, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-23333074

ABSTRACT

Epidermal growth factor receptor (EGFR) is known to be critically involved in tissue development and homeostasis as well as in the pathogenesis of cancer. Here we showed that Foxp3(+) regulatory T (Treg) cells express EGFR under inflammatory conditions. Stimulation with the EGF-like growth factor Amphiregulin (AREG) markedly enhanced Treg cell function in vitro, and in a colitis and tumor vaccination model we showed that AREG was critical for efficient Treg cell function in vivo. In addition, mast cell-derived AREG fully restored optimal Treg cell function. These findings reveal EGFR as a component in the regulation of local immune responses and establish a link between mast cells and Treg cells. Targeting of this immune regulatory mechanism may contribute to the therapeutic successes of EGFR-targeting treatments in cancer patients.


Subject(s)
ErbB Receptors/immunology , Glycoproteins/immunology , Intercellular Signaling Peptides and Proteins/immunology , T-Lymphocytes, Regulatory/immunology , Amphiregulin , Animals , Antibodies, Neutralizing/pharmacology , Cell Communication/immunology , Colitis/chemically induced , Colitis/immunology , Colitis/metabolism , Colitis/pathology , EGF Family of Proteins , ErbB Receptors/genetics , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Gene Expression Regulation/drug effects , Glycoproteins/antagonists & inhibitors , Glycoproteins/genetics , Glycoproteins/pharmacology , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/pharmacology , Lymphocyte Activation/drug effects , Mast Cells/drug effects , Mast Cells/immunology , Mast Cells/metabolism , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Membrane Proteins/administration & dosage , Membrane Proteins/immunology , Mice , Mice, Transgenic , Peptide Fragments/administration & dosage , Peptide Fragments/immunology , Signal Transduction/drug effects , Signal Transduction/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism
4.
Immunity ; 39(2): 259-71, 2013 Aug 22.
Article in English | MEDLINE | ID: mdl-23973222

ABSTRACT

Stable Foxp3 expression is required for the development of functional regulatory T (Treg) cells. Here, we demonstrate that the expression of the transcription factor Foxp3 can be regulated through the polyubiquitination of multiple lysine residues, resulting in proteasome-mediated degradation. Expression of the deubiquitinase (DUB) USP7 was found to be upregulated and active in Treg cells, being associated with Foxp3 in the nucleus. Ectopic expression of USP7 decreased Foxp3 polyubiquitination and increased Foxp3 expression. Conversely, either treatment with DUB inhibitor or USP7 knockdown decreased endogenous Foxp3 protein expression and decreased Treg-cell-mediated suppression in vitro. Furthermore, in a murine adoptive-transfer-induced colitis model, either inhibition of DUB activity or USP7 knockdown in Treg cells abrogated their ability to resolve inflammation in vivo. Our data reveal a molecular mechanism in which rapid temporal control of Foxp3 expression in Treg cells can be regulated by USP7, thereby modulating Treg cell numbers and function.


Subject(s)
Colitis/immunology , Forkhead Transcription Factors/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Ubiquitin Thiolesterase/metabolism , Adoptive Transfer , Animals , Cell Differentiation/immunology , Cell Line , DNA-Binding Proteins/genetics , Disease Models, Animal , Endopeptidases/metabolism , HEK293 Cells , Homeodomain Proteins/genetics , Humans , Immediate-Early Proteins/antagonists & inhibitors , Immediate-Early Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , RNA Interference , RNA, Small Interfering , Ubiquitin Thiolesterase/genetics , Ubiquitin-Specific Peptidase 7 , Ubiquitination
5.
Immunity ; 39(2): 298-310, 2013 Aug 22.
Article in English | MEDLINE | ID: mdl-23954131

ABSTRACT

Foxp3 is crucial for both the development and function of regulatory T (Treg) cells; however, the posttranslational mechanisms regulating Foxp3 transcriptional output remain poorly defined. Here, we demonstrate that T cell factor 1 (TCF1) and Foxp3 associates in Treg cells and that active Wnt signaling disrupts Foxp3 transcriptional activity. A global chromatin immunoprecipitation sequencing comparison in Treg cells revealed considerable overlap between Foxp3 and Wnt target genes. The activation of Wnt signaling reduced Treg-mediated suppression both in vitro and in vivo, whereas disruption of Wnt signaling in Treg cells enhanced their suppressive capacity. The activation of effector T cells increased Wnt3a production, and Wnt3a levels were found to be greatly increased in mononuclear cells isolated from synovial fluid versus peripheral blood of arthritis patients. We propose a model in which Wnt produced under inflammatory conditions represses Treg cell function, allowing a productive immune response, but, if uncontrolled, could lead to the development of autoimmunity.


Subject(s)
Arthritis/immunology , Colitis/immunology , Forkhead Transcription Factors/metabolism , Hepatocyte Nuclear Factor 1-alpha/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Cell Proliferation , Cells, Cultured , Forkhead Transcription Factors/genetics , HEK293 Cells , Hepatocyte Nuclear Factor 1-alpha/genetics , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Synovial Fluid/cytology , T-Lymphocytes, Regulatory/metabolism , Wnt Signaling Pathway , Wnt3A Protein/metabolism , beta Catenin/genetics , beta Catenin/metabolism
6.
J Immunol ; 201(8): 2193-2200, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30301837

ABSTRACT

T cell factor, the effector transcription factor of the WNT signaling pathway, was so named because of the primary observation that it is indispensable for T cell development in the thymus. Since this discovery, the role of this signaling pathway has been extensively studied in T cell development, hematopoiesis, and stem cells; however, its functional role in mature T cells has remained relatively underinvestigated. Over the last few years, various studies have demonstrated that T cell factor can directly influence T cell function and the differentiation of Th1, Th2, Th17, regulatory T cell, follicular helper CD4+ T cell subsets, and CD8+ memory T cells. In this paper, we discuss the molecular mechanisms underlying these observations and place them in the general context of immune responses. Furthermore, we explore the implications and limitations of these findings for WNT manipulation as a therapeutic approach for treating immune-related diseases.


Subject(s)
T-Lymphocyte Subsets/immunology , T-Lymphocytes/immunology , Wnt Signaling Pathway/immunology , Animals , Cell Differentiation , Cytokines/metabolism , Humans , Immunologic Memory , Lymphocyte Activation , TCF Transcription Factors/metabolism
7.
Nucleic Acids Res ; 46(18): 9578-9590, 2018 10 12.
Article in English | MEDLINE | ID: mdl-30137431

ABSTRACT

Expression of the transcription factor SOX4 is often elevated in human cancers, where it generally correlates with tumor-progression and poor-disease outcome. Reduction of SOX4 expression results in both diminished tumor-incidence and metastasis. In breast cancer, TGF-ß-mediated induction of SOX4 has been shown to contribute to epithelial-to-mesenchymal transition (EMT), which controls pro-metastatic events. Here, we identify SMAD3 as a novel, functionally relevant SOX4 interaction partner. Genome-wide analysis showed that SOX4 and SMAD3 co-occupy a large number of genomic loci in a cell-type specific manner. Moreover, SOX4 expression was required for TGF-ß-mediated induction of a subset of SMAD3/SOX4-co-bound genes regulating migration and extracellular matrix-associated processes, and correlating with poor-prognosis. These findings identify SOX4 as an important SMAD3 co-factor controlling transcription of pro-metastatic genes and context-dependent shaping of the cellular response to TGF-ß. Targeted disruption of the interaction between these factors may have the potential to disrupt pro-oncogenic TGF-ß signaling, thereby impairing tumorigenesis.


Subject(s)
Breast Neoplasms/genetics , SOXC Transcription Factors/genetics , Smad3 Protein/genetics , Transforming Growth Factor beta/genetics , Breast Neoplasms/pathology , Carcinogenesis/genetics , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Prognosis , Signal Transduction , Transcription, Genetic
8.
Gastroenterology ; 155(1): 130-143.e15, 2018 07.
Article in English | MEDLINE | ID: mdl-29604290

ABSTRACT

BACKGROUND & AIMS: Congenital diarrheal disorders are rare inherited intestinal disorders characterized by intractable, sometimes life-threatening, diarrhea and nutrient malabsorption; some have been associated with mutations in diacylglycerol-acyltransferase 1 (DGAT1), which catalyzes formation of triacylglycerol from diacylglycerol and acyl-CoA. We investigated the mechanisms by which DGAT1 deficiency contributes to intestinal failure using patient-derived organoids. METHODS: We collected blood samples from 10 patients, from 6 unrelated pedigrees, who presented with early-onset severe diarrhea and/or vomiting, hypoalbuminemia, and/or (fatal) protein-losing enteropathy with intestinal failure; we performed next-generation sequencing analysis of DNA from 8 patients. Organoids were generated from duodenal biopsies from 3 patients and 3 healthy individuals (controls). Caco-2 cells and patient-derived dermal fibroblasts were transfected or transduced with vectors that express full-length or mutant forms of DGAT1 or full-length DGAT2. We performed CRISPR/Cas9-guided disruption of DGAT1 in control intestinal organoids. Cells and organoids were analyzed by immunoblot, immunofluorescence, flow cytometry, chromatography, quantitative real-time polymerase chain reaction, and for the activity of caspases 3 and 7. RESULTS: In the 10 patients, we identified 5 bi-allelic loss-of-function mutations in DGAT1. In patient-derived fibroblasts and organoids, the mutations reduced expression of DGAT1 protein and altered triacylglycerol metabolism, resulting in decreased lipid droplet formation after oleic acid addition. Expression of full-length DGAT2 in patient-derived fibroblasts restored formation of lipid droplets. Organoids derived from patients with DGAT1 mutations were more susceptible to lipid-induced cell death than control organoids. CONCLUSIONS: We identified a large cohort of patients with congenital diarrheal disorders with mutations in DGAT1 that reduced expression of its product; dermal fibroblasts and intestinal organoids derived from these patients had altered lipid metabolism and were susceptible to lipid-induced cell death. Expression of full-length wildtype DGAT1 or DGAT2 restored normal lipid metabolism in these cells. These findings indicate the importance of DGAT1 in fat metabolism and lipotoxicity in the intestinal epithelium. A fat-free diet might serve as the first line of therapy for patients with reduced DGAT1 expression. It is important to identify genetic variants associated with congenital diarrheal disorders for proper diagnosis and selection of treatment strategies.


Subject(s)
Diacylglycerol O-Acyltransferase/genetics , Duodenum/metabolism , Fibroblasts/metabolism , Hypoalbuminemia/genetics , Lipid Metabolism Disorders/genetics , Organoids/metabolism , Protein-Losing Enteropathies/genetics , Caco-2 Cells , Case-Control Studies , Caspase 3/metabolism , Caspase 7/metabolism , Child , Child, Preschool , Consanguinity , Dermis/cytology , Diacylglycerol O-Acyltransferase/deficiency , Female , High-Throughput Nucleotide Sequencing , Humans , Male , Netherlands , Phorbols , Real-Time Polymerase Chain Reaction , Sequence Analysis, DNA , Turkey
9.
Pediatr Blood Cancer ; 66(8): e27785, 2019 08.
Article in English | MEDLINE | ID: mdl-31044544

ABSTRACT

BACKGROUND: Acute myeloid leukemia (AML) is a heterogeneous disease regarding morphology, immunophenotyping, genetic abnormalities, and clinical behavior. The overall survival rate of pediatric AML is 60% to 70%, and has not significantly improved over the past two decades. Children with Down syndrome (DS) are at risk of developing acute megakaryoblastic leukemia (AMKL), which can be preceded by a transient myeloproliferative disorder during the neonatal period. Intensification of current treatment protocols is not feasible due to already high treatment-related morbidity and mortality. Instead, more targeted therapies with less severe side effects are highly needed. PROCEDURE: To identify potential novel therapeutic targets for myeloid disorders in children, including DS-AMKL and non-DS-AML, we performed an unbiased compound screen of 80 small molecules targeting epigenetic regulators in three pediatric AML cell lines that are representative for different subtypes of pediatric AML. Three candidate compounds were validated and further evaluated in normal myeloid precursor cells during neutrophil differentiation and in (pre-)leukemic pediatric patient cells. RESULTS: Candidate drugs LMK235, NSC3852, and bromosporine were effective in all tested pediatric AML cell lines with antiproliferative, proapoptotic, and differentiation effects. Out of these three compounds, the pan-histone deacetylase inhibitor NSC3852 specifically induced growth arrest and apoptosis in pediatric AML cells, without disrupting normal neutrophil differentiation. CONCLUSION: NSC3852 is a potential candidate drug for further preclinical testing in pediatric AML and DS-AMKL.


Subject(s)
Drug Screening Assays, Antitumor/methods , Epigenesis, Genetic , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/chemistry , Hydroxyquinolines/pharmacology , Leukemia, Myeloid, Acute/pathology , Nitroso Compounds/pharmacology , Apoptosis , Cell Proliferation , Child , Down Syndrome/drug therapy , Down Syndrome/genetics , Down Syndrome/pathology , High-Throughput Screening Assays , Histone Deacetylases/genetics , Humans , Leukemia, Megakaryoblastic, Acute/drug therapy , Leukemia, Megakaryoblastic, Acute/genetics , Leukemia, Megakaryoblastic, Acute/pathology , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemoid Reaction/drug therapy , Leukemoid Reaction/genetics , Leukemoid Reaction/pathology , Prognosis , Tumor Cells, Cultured
10.
Nature ; 504(7480): 451-5, 2013 Dec 19.
Article in English | MEDLINE | ID: mdl-24226773

ABSTRACT

Intestinal microbes provide multicellular hosts with nutrients and confer resistance to infection. The delicate balance between pro- and anti-inflammatory mechanisms, essential for gut immune homeostasis, is affected by the composition of the commensal microbial community. Regulatory T cells (Treg cells) expressing transcription factor Foxp3 have a key role in limiting inflammatory responses in the intestine. Although specific members of the commensal microbial community have been found to potentiate the generation of anti-inflammatory Treg or pro-inflammatory T helper 17 (TH17) cells, the molecular cues driving this process remain elusive. Considering the vital metabolic function afforded by commensal microorganisms, we reasoned that their metabolic by-products are sensed by cells of the immune system and affect the balance between pro- and anti-inflammatory cells. We tested this hypothesis by exploring the effect of microbial metabolites on the generation of anti-inflammatory Treg cells. We found that in mice a short-chain fatty acid (SCFA), butyrate, produced by commensal microorganisms during starch fermentation, facilitated extrathymic generation of Treg cells. A boost in Treg-cell numbers after provision of butyrate was due to potentiation of extrathymic differentiation of Treg cells, as the observed phenomenon was dependent on intronic enhancer CNS1 (conserved non-coding sequence 1), essential for extrathymic but dispensable for thymic Treg-cell differentiation. In addition to butyrate, de novo Treg-cell generation in the periphery was potentiated by propionate, another SCFA of microbial origin capable of histone deacetylase (HDAC) inhibition, but not acetate, which lacks this HDAC-inhibitory activity. Our results suggest that bacterial metabolites mediate communication between the commensal microbiota and the immune system, affecting the balance between pro- and anti-inflammatory mechanisms.


Subject(s)
Butyrates/metabolism , Cell Differentiation , Intestinal Mucosa/metabolism , Intestines/microbiology , Symbiosis , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism , Acetylation , Animals , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Enhancer Elements, Genetic/genetics , Fermentation , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Histone Deacetylases/metabolism , Inflammation Mediators/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestines/cytology , Intestines/immunology , Introns/genetics , Lymphocyte Count , Male , Mice , Mice, Inbred C57BL , Starch/metabolism , T-Lymphocytes, Regulatory/immunology
11.
J Biol Chem ; 292(19): 7904-7920, 2017 05 12.
Article in English | MEDLINE | ID: mdl-28302725

ABSTRACT

Infantile-onset inflammatory bowel disease (IO IBD) is an invalidating illness with an onset before 2 years of age and has a complex pathophysiology in which genetic factors are important. Homozygosity mapping and whole-exome sequencing in an IO IBD patient and subsequent sequencing of the candidate gene in 12 additional IO IBD patients revealed two patients with two mutated ankyrin repeat and zinc-finger domain-containing 1 (ANKZF1) alleles (homozygous ANKZF1 R585Q mutation and compound heterozygous ANKZF1 E152K and V32_Q87del mutations, respectively) and two patients with one mutated ANKZF1 allele. Although the function of ANKZF1 in mammals had not been previously evaluated, we show that ANKZF1 has an indispensable role in the mitochondrial response to cellular stress. ANKZF1 is located diffusely in the cytoplasm and translocates to the mitochondria upon cellular stress. ANKZF1 depletion reduces mitochondrial integrity and mitochondrial respiration under conditions of cellular stress. The ANKZF1 mutations identified in IO IBD patients with two mutated ANKZF1 alleles result in dysfunctional ANKZF1, as shown by an increased level of apoptosis in patients' lymphocytes, a decrease in mitochondrial respiration in patient fibroblasts with a homozygous ANKZF1 R585Q mutation, and an inability of ANKZF1 R585Q and E152K to rescue the phenotype of yeast deficient in Vms1, the yeast homologue of ANKZF1. These data indicate that loss-of-function mutations in ANKZF1 result in deregulation of mitochondrial integrity, and this may play a pathogenic role in the development of IO IBD.


Subject(s)
Ankyrin Repeat/genetics , Carrier Proteins/genetics , Inflammatory Bowel Diseases/genetics , Zinc Fingers , Age of Onset , Alleles , Apoptosis , Carrier Proteins/metabolism , Cell Line, Tumor , Child, Preschool , Exome , Female , Fibroblasts/metabolism , Genome, Human , HEK293 Cells , Homozygote , Humans , Infant , Inflammation , Inflammatory Bowel Diseases/metabolism , Lymphocytes/cytology , Male , Mitochondria/metabolism , Mutation , Phenotype , RNA, Small Interfering/metabolism , Sequence Analysis, DNA , Zinc/chemistry
12.
Blood ; 125(11): 1782-92, 2015 Mar 12.
Article in English | MEDLINE | ID: mdl-25568349

ABSTRACT

C/EBPε, a member of the CCAAT/enhancer binding protein (C/EBP) family of transcription factors, is exclusively expressed in myeloid cells and regulates transition from the promyelocytic stage to the myelocytic stage of neutrophil development, being indispensable for secondary and tertiary granule formation. Knowledge concerning the functional role of C/EBPε posttranslational modifications is limited to studies concerning phosphorylation and sumoylation. In the current study, using ectopic expression and ex vivo differentiation of CD34(+) hematopoietic progenitor cells, we demonstrate that C/EBPε is acetylated, which was confirmed by mass spectrometry analysis, identifying 4 acetylated lysines in 3 distinct functional domains. Regulation of C/EBPε acetylation levels by the p300 acetyltransferase and the sirtuin 1 deacetylase controls transcriptional activity, which can at least in part be explained by modulation of DNA binding. During neutrophil development, acetylation of lysines 121 and 198 were found to be crucial for terminal neutrophil differentiation and the expression of neutrophil-specific granule proteins, including lactoferrin and collagenase. Taken together, our data illustrate a critical role for acetylation in the functional regulation of C/EBPε activity during terminal neutrophil development.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , Neutrophils/cytology , Neutrophils/metabolism , Acetylation , Animals , CCAAT-Enhancer-Binding Proteins/chemistry , CCAAT-Enhancer-Binding Proteins/genetics , COS Cells , Cell Differentiation , Cell Line, Tumor , Chlorocebus aethiops , Collagenases/metabolism , HL-60 Cells , Humans , Lactoferrin/metabolism , Lysine/chemistry , Myelopoiesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sirtuin 1/metabolism , Transcription, Genetic , p300-CBP Transcription Factors/metabolism
13.
Stem Cells ; 34(6): 1651-63, 2016 06.
Article in English | MEDLINE | ID: mdl-26930546

ABSTRACT

Autophagy is a highly regulated catabolic process that involves sequestration and lysosomal degradation of cytosolic components such as damaged organelles and misfolded proteins. While autophagy can be considered to be a general cellular housekeeping process, it has become clear that it may also play cell type-dependent functional roles. In this study, we analyzed the functional importance of autophagy in human hematopoietic stem/progenitor cells (HSPCs), and how this is regulated during differentiation. Western blot-based analysis of LC3-II and p62 levels, as well as flow cytometry-based autophagic vesicle quantification, demonstrated that umbilical cord blood-derived CD34(+) /CD38(-) immature hematopoietic progenitors show a higher autophagic flux than CD34(+) /CD38(+) progenitors and more differentiated myeloid and erythroid cells. This high autophagic flux was critical for maintaining stem and progenitor function since knockdown of autophagy genes ATG5 or ATG7 resulted in reduced HSPC frequencies in vitro as well as in vivo. The reduction in HSPCs was not due to impaired differentiation, but at least in part due to reduced cell cycle progression and increased apoptosis. This is accompanied by increased expression of p53, proapoptotic genes BAX and PUMA, and the cell cycle inhibitor p21, as well as increased levels of cleaved caspase-3 and reactive oxygen species. Taken together, our data demonstrate that autophagy is an important regulatory mechanism for human HSCs and their progeny, reducing cellular stress and promoting survival. Stem Cells 2016;34:1651-1663.


Subject(s)
Antigens, CD34/metabolism , Autophagy-Related Protein 5/metabolism , Autophagy-Related Protein 7/metabolism , Autophagy , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Animals , Apoptosis , Cell Count , Cell Cycle , Cell Differentiation , Fetal Blood/cytology , Gene Knockdown Techniques , Humans , Intracellular Space/metabolism , Mice , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/metabolism , Reactive Oxygen Species/metabolism
14.
Trends Immunol ; 35(8): 368-78, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25047417

ABSTRACT

Forkhead box (FOX)P3 is a requisite transcription factor for the development and maintenance of immunosuppressive function of regulatory T (Treg) cells, and therefore for immune homeostasis. Post-translational modifications (PTMs) can transiently alter the functionality of transcription factors, and recent evidence reveals that FOXP3 can be regulated by various PTMs including acetylation, ubiquitination, and phosphorylation. Here, we review the current understanding of how these modifications control FOXP3, including regulation of DNA binding, transactivation potential, and proteasomal degradation. We place these findings in the context of the biology of Treg cells, and discuss both limitations in translating biochemical findings into in vivo functions and the opportunities presented by a better understanding of the molecular mechanisms that can transiently control FOXP3 activity in response to environmental cues.


Subject(s)
Forkhead Transcription Factors/metabolism , Proteasome Endopeptidase Complex/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Cellular Microenvironment , Homeostasis , Humans , Immune Tolerance , Protein Processing, Post-Translational , Transcriptional Activation
15.
Pediatr Res ; 81(1-1): 127-135, 2017 01.
Article in English | MEDLINE | ID: mdl-27632779

ABSTRACT

BACKGROUND: Hypoxic-ischemic (HI) encephalopathy causes mortality and severe morbidity in neonates. Treatments with a therapeutic window >6 h are currently not available. Here, we explored whether delayed transplantation of allogenic neural stem cells (NSCs) at 10 d after HI could be a tool to repair HI brain injury and improve behavioral impairments. METHODS: HI was induced in 9-d-old mice. Animals received NSCs or vehicle intracranially in the hippocampus at 10 d post-HI. Sensorimotor performance was assessed by cylinder rearing test. Lesion size, synaptic integrity, and fate of injected NSCs were determined by immuno-stainings. Neuroinflammation was studied by immuno-stainings of brain sections, primary glial cultures, and TNFα ELISA. RESULTS: NSC transplantation at 10 d post-insult induced long-term improvement of motor performance and synaptic integrity, and reduced lesion size compared to vehicle-treatment. HI-induced neuroinflammation was reduced after NSC treatment, at least partially by factors secreted by NSCs. Injected NSCs migrated toward and localized at the damaged hippocampus. Transplanted NSCs differentiated toward the neuronal lineage and formed a niche with endogenous precursors. CONCLUSION: Our study provides evidence of the efficacy of NSC transplantation late after HI as a tool to reduce neonatal HI brain injury through regeneration of the lesion.


Subject(s)
Hypoxia-Ischemia, Brain/therapy , Neural Stem Cells/transplantation , Animals , Animals, Newborn , Behavior, Animal , Cell Movement , Disease Models, Animal , Female , Hippocampus/pathology , Hypoxia-Ischemia, Brain/pathology , Hypoxia-Ischemia, Brain/physiopathology , Inflammation/pathology , Inflammation/therapy , Male , Mice , Mice, Inbred C57BL , Psychomotor Performance , Time Factors , Transplantation, Homologous
16.
J Immunol ; 195(7): 3180-9, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26324768

ABSTRACT

Regulatory T cell (Treg) activity is modulated by a cooperative complex between the transcription factor NFAT and FOXP3, a lineage specification factor for Tregs. FOXP3/NFAT interaction is required to repress expression of IL-2, upregulate expression of the Treg markers CTLA4 and CD25, and confer suppressor function to Tregs. However, FOXP3 is expressed transiently in conventional CD4(+) T cells upon TCR stimulation and may lead to T cell hyporesponsiveness. We found that a short synthetic peptide able to inhibit FOXP3/NFAT interaction impaired suppressor activity of conventional Tregs in vitro. Specific inhibition of FOXP3/NFAT interaction with this inhibitory peptide revealed that FOXP3 downregulates NFAT-driven promoter activity of CD40L and IL-17. Inhibition of FOXP3/NFAT interaction upregulated CD40L expression on effector T cells and enhanced T cell proliferation and IL-2, IFN-γ, IL-6, or IL-17 production in response to TCR stimulation. The inhibitory peptide impaired effector T cell conversion into induced Tregs in the presence of TGF-ß. Moreover, in vivo peptide administration showed antitumor efficacy in mice bearing Hepa129 or TC1 tumor cells when combined with sorafenib or with an antitumor vaccine, respectively. Our results suggest that inhibition of NFAT/FOXP3 interaction might improve antitumor immunotherapies.


Subject(s)
CD40 Ligand/biosynthesis , Forkhead Transcription Factors/metabolism , Interleukin-17/biosynthesis , NFATC Transcription Factors/metabolism , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antineoplastic Agents/pharmacology , CD40 Ligand/genetics , CTLA-4 Antigen/biosynthesis , Cell Proliferation/genetics , Female , Forkhead Transcription Factors/antagonists & inhibitors , Humans , Immunotherapy , Interferon-gamma/biosynthesis , Interleukin-17/genetics , Interleukin-2/biosynthesis , Interleukin-2 Receptor alpha Subunit/biosynthesis , Interleukin-6/biosynthesis , Jurkat Cells , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , NFATC Transcription Factors/antagonists & inhibitors , Neoplasms/therapy , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Ovalbumin/immunology , Peptide Fragments/pharmacology , Phenylurea Compounds/pharmacology , Promoter Regions, Genetic/genetics , Sorafenib , Transforming Growth Factor beta/metabolism
17.
Blood ; 124(23): 3431-40, 2014 Nov 27.
Article in English | MEDLINE | ID: mdl-25267198

ABSTRACT

The forkhead transcription factor FOXP1 is involved in B-cell development and function and is generally regarded as an oncogene in activated B-cell-like subtype of diffuse large B-cell lymphoma (DLBCL) and mucosa-associated lymphoid tissue lymphoma, lymphomas relying on constitutive nuclear factor κB (NF-κB) activity for survival. However, the mechanism underlying its putative oncogenic activity has not been established. By gene expression microarray, upon overexpression or silencing of FOXP1 in primary human B cells and DLBCL cell lines, combined with chromatin immunoprecipitation followed by next-generation sequencing, we established that FOXP1 directly represses a set of 7 proapoptotic genes. Low expression of these genes, encoding the BH3-only proteins BIK and Harakiri, the p53-regulatory proteins TP63, RASSF6, and TP53INP1, and AIM2 and EAF2, is associated with poor survival in DLBCL patients. In line with these findings, we demonstrated that FOXP1 promotes the expansion of primary mature human B cells by inhibiting caspase-dependent apoptosis, without affecting B-cell proliferation. Furthermore, FOXP1 is dependent upon, and cooperates with, NF-κB signaling to promote B-cell expansion and survival. Taken together, our data indicate that, through direct repression of proapoptotic genes, (aberrant) expression of FOXP1 complements (constitutive) NF-κB activity to promote B-cell survival and can thereby contribute to B-cell homeostasis and lymphomagenesis.


Subject(s)
Apoptosis Regulatory Proteins/genetics , B-Lymphocytes/physiology , Forkhead Transcription Factors/physiology , Gene Expression Regulation, Leukemic , NF-kappa B/physiology , Repressor Proteins/physiology , Transcription, Genetic , Apoptosis Regulatory Proteins/metabolism , Cell Survival/genetics , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Down-Regulation , Gene Expression Profiling , Humans , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Microarray Analysis , Up-Regulation
18.
J Biol Chem ; 289(8): 5000-12, 2014 Feb 21.
Article in English | MEDLINE | ID: mdl-24356959

ABSTRACT

Most hereditary periodic fever syndromes are mediated by deregulated IL-1ß secretion. The generation of mature IL-1ß requires two signals: one that induces synthesis of inflammasome components and substrates and a second that activates inflammasomes. The mechanisms that mediate autoinflammation in mevalonate kinase deficiency, a periodic fever disease characterized by a block in isoprenoid biosynthesis, are poorly understood. In studying the effects of isoprenoid shortage on IL-1 ß generation, we identified a new inflammasome activation signal that originates from defects in autophagy. We find that hypersecretion of IL-1ß and IL-18 requires reactive oxygen species and is associated with an oxidized redox status of monocytes but not lymphocytes. IL-1ß hypersecretion by monocytes involves decreased mitochondrial stability, release of mitochondrial content into the cytosol and attenuated autophagosomal degradation. Defective autophagy, as established by ATG7 knockdown, results in prolonged cytosolic retention of damaged mitochondria and increased IL-1ß secretion. Finally, activation of autophagy in healthy but not mevalonate kinase deficiency patient cells reduces IL-1ß secretion. Together, these results indicate that defective autophagy can prime monocytes for mitochondria-mediated NLRP3 inflammasome activation, thereby contributing to hypersecretion of IL-1ß in mevalonate kinase deficiency.


Subject(s)
Disease Susceptibility/metabolism , Disease Susceptibility/pathology , Interleukin-1beta/metabolism , Mitochondria/metabolism , Monocytes/metabolism , Monocytes/pathology , Adolescent , Autophagy , Cell Line , Child , Child, Preschool , Cytosol/metabolism , DNA, Mitochondrial/metabolism , Humans , Inflammasomes/metabolism , Membrane Potential, Mitochondrial , Mevalonate Kinase Deficiency/metabolism , Mevalonate Kinase Deficiency/pathology , Models, Biological , Oxidation-Reduction , Terpenes/metabolism
19.
Biochem J ; 460(1): 25-34, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24762137

ABSTRACT

Growth factors inactivate the FOXO (forkhead box O) transcription factors through PI3K (phosphoinositide 3-kinase) and PKB (protein kinase B). By comparing microarray data from multiple model systems, we identified HBP1 (high-mobility group-box protein 1) as a novel downstream target of this pathway. HBP1 mRNA was down-regulated by PDGF (platelet-derived growth factor), FGF (fibroblast growth factor), PI3K and PKB, whereas it was up-regulated by FOXO factors. This observation was confirmed in human and murine fibroblasts as well as in cell lines derived from leukaemia, breast adenocarcinoma and colon carcinoma. Bioinformatics analysis led to the identification of a conserved consensus FOXO-binding site in the HBP1 promoter. By luciferase activity assay and ChIP, we demonstrated that FOXO bound to this site and regulated the HBP1 promoter activity in a PI3K-dependent manner. Silencing of HBP1 by shRNA increased the proliferation of human fibroblasts in response to growth factors, suggesting that HBP1 limits cell growth. Finally, by analysing a transcriptomics dataset from The Cancer Genome Atlas, we observed that HBP1 expression was lower in breast tumours that had lost FOXO expression. In conclusion, HBP1 is a novel target of the PI3K/FOXO pathway and controls cell proliferation in response to growth factors.


Subject(s)
Down-Regulation/genetics , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic , High Mobility Group Proteins/antagonists & inhibitors , High Mobility Group Proteins/genetics , Intercellular Signaling Peptides and Proteins/genetics , Phosphatidylinositol 3-Kinase/genetics , Proto-Oncogene Proteins c-akt/genetics , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Animals , CHO Cells , Cells, Cultured , Conserved Sequence , Cricetinae , Cricetulus , Fibroblasts/drug effects , Fibroblasts/metabolism , Forkhead Box Protein O1 , Forkhead Transcription Factors/biosynthesis , HEK293 Cells , High Mobility Group Proteins/biosynthesis , Humans , MCF-7 Cells , Male , Mice , NIH 3T3 Cells , Phosphatidylinositol 3-Kinase/biosynthesis , Promoter Regions, Genetic , Protein Binding/genetics , Proto-Oncogene Proteins c-akt/biosynthesis , Repressor Proteins/biosynthesis , Signal Transduction/genetics
20.
Nat Rev Immunol ; 4(11): 889-99, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15516968

ABSTRACT

It is more than a decade since the discovery of the first forkhead-box (FOX) transcription factor in the fruit fly Drosophila melanogaster. In the intervening time, there has been an explosion in the identification and characterization of members of this family of proteins. Importantly, in the past few years, it has become clear that members of the FOX family have crucial roles in various aspects of immune regulation, from lymphocyte survival to thymic development. This review focuses on FOXP3, FOXN1, FOXJ1 and members of the FOXO subfamily and their function in the immune system.


Subject(s)
DNA-Binding Proteins/immunology , Immune System/physiology , Trans-Activators/immunology , Transcription Factors/immunology , Animals , Forkhead Transcription Factors , Humans , Immune System/immunology , Lymphocyte Activation/immunology , Mice , T-Lymphocytes, Helper-Inducer/immunology , Thymus Gland/embryology , Thymus Gland/immunology
SELECTION OF CITATIONS
SEARCH DETAIL