Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Sci Adv ; 10(10): eadl1122, 2024 Mar 08.
Article in English | MEDLINE | ID: mdl-38446892

ABSTRACT

Coxsackievirus B (CVB) infection of pancreatic ß cells is associated with ß cell autoimmunity and type 1 diabetes. We investigated how CVB affects human ß cells and anti-CVB T cell responses. ß cells were efficiently infected by CVB in vitro, down-regulated human leukocyte antigen (HLA) class I, and presented few, selected HLA-bound viral peptides. Circulating CD8+ T cells from CVB-seropositive individuals recognized a fraction of these peptides; only another subfraction was targeted by effector/memory T cells that expressed exhaustion marker PD-1. T cells recognizing a CVB epitope cross-reacted with ß cell antigen GAD. Infected ß cells, which formed filopodia to propagate infection, were more efficiently killed by CVB than by CVB-reactive T cells. Our in vitro and ex vivo data highlight limited CD8+ T cell responses to CVB, supporting the rationale for CVB vaccination trials for type 1 diabetes prevention. CD8+ T cells recognizing structural and nonstructural CVB epitopes provide biomarkers to differentially follow response to infection and vaccination.


Subject(s)
Coxsackievirus Infections , Diabetes Mellitus, Type 1 , Insulin-Secreting Cells , Humans , CD8-Positive T-Lymphocytes , Antibodies , Epitopes , Peptides , Antiviral Agents
2.
bioRxiv ; 2023 Aug 21.
Article in English | MEDLINE | ID: mdl-37662376

ABSTRACT

Coxsackievirus B (CVB) infection of pancreatic ß cells is associated with ß-cell autoimmunity. We investigated how CVB impacts human ß cells and anti-CVB T-cell responses. ß cells were efficiently infected by CVB in vitro, downregulated HLA Class I and presented few, selected HLA-bound viral peptides. Circulating CD8+ T cells from CVB-seropositive individuals recognized only a fraction of these peptides, and only another sub-fraction was targeted by effector/memory T cells that expressed the exhaustion marker PD-1. T cells recognizing a CVB epitope cross-reacted with the ß-cell antigen GAD. Infected ß cells, which formed filopodia to propagate infection, were more efficiently killed by CVB than by CVB-reactive T cells. Thus, our in-vitro and ex-vivo data highlight limited T-cell responses to CVB, supporting the rationale for CVB vaccination trials for type 1 diabetes prevention. CD8+ T cells recognizing structural and non-structural CVB epitopes provide biomarkers to differentially follow response to infection and vaccination.

3.
Front Endocrinol (Lausanne) ; 13: 861985, 2022.
Article in English | MEDLINE | ID: mdl-35498413

ABSTRACT

Although type 1 diabetes (T1D) is primarily a disease of the pancreatic beta-cells, understanding of the disease-associated alterations in the whole pancreas could be important for the improved treatment or the prevention of the disease. We have characterized the whole-pancreas gene expression of patients with recently diagnosed T1D from the Diabetes Virus Detection (DiViD) study and non-diabetic controls. Furthermore, another parallel dataset of the whole pancreas and an additional dataset from the laser-captured pancreatic islets of the DiViD patients and non-diabetic organ donors were analyzed together with the original dataset to confirm the results and to get further insights into the potential disease-associated differences between the exocrine and the endocrine pancreas. First, higher expression of the core acinar cell genes, encoding for digestive enzymes, was detected in the whole pancreas of the DiViD patients when compared to non-diabetic controls. Second, In the pancreatic islets, upregulation of immune and inflammation related genes was observed in the DiViD patients when compared to non-diabetic controls, in line with earlier publications, while an opposite trend was observed for several immune and inflammation related genes at the whole pancreas tissue level. Third, strong downregulation of the regenerating gene family (REG) genes, linked to pancreatic islet growth and regeneration, was observed in the exocrine acinar cell dominated whole-pancreas data of the DiViD patients when compared with the non-diabetic controls. Fourth, analysis of unique features in the transcriptomes of each DiViD patient compared with the other DiViD patients, revealed elevated expression of central antiviral immune response genes in the whole-pancreas samples, but not in the pancreatic islets, of one DiViD patient. This difference in the extent of antiviral gene expression suggests different statuses of infection in the pancreas at the time of sampling between the DiViD patients, who were all enterovirus VP1+ in the islets by immunohistochemistry based on earlier studies. The observed features, indicating differences in the function, status and interplay between the exocrine and the endocrine pancreas of recent onset T1D patients, highlight the importance of studying both compartments for better understanding of the molecular mechanisms of T1D.


Subject(s)
Diabetes Mellitus, Type 1 , Pancreas, Exocrine , Antiviral Agents , Diabetes Mellitus, Type 1/metabolism , Humans , Inflammation/metabolism , Pancreas/metabolism , Transcriptome
4.
Curr Diab Rep ; 19(9): 82, 2019 08 10.
Article in English | MEDLINE | ID: mdl-31401790

ABSTRACT

PURPOSE OF REVIEW: We provide an overview of the current knowledge regarding the natural history of human type 1 diabetes (T1D) and the documented associations between virus infections (in particular the enteroviruses) and disease development. We review studies that examine whether T1D-specific risk alleles in genes involved in the function of the immune system can alter susceptibility to virus infections or affect the magnitude of the host antiviral response. We also highlight where the major gaps in our knowledge exist and consider possible implications that new insights gained from the discussed gene-environment interaction studies may bring. RECENT FINDINGS: A commonality between several of the studied T1D risk variants studied is their role in modulating the host immune response to viral infection. Generally, little support exists indicating that the risk variants increase susceptibility to infection and moreover, they usually appear to predispose the immune system towards a hyper-reactive state, decrease the risk of infection, and/or favor the establishment of viral persistence. In conclusion, although the current number of studies is limited, this type of research can provide important insights into the mechanisms that are central to disease pathogenesis and further describe how genetic and environmental factors jointly influence the risk of T1D development. The latter may provide genetic markers that could be used for patient stratification and for the selection of method(s) for disease prevention.


Subject(s)
Diabetes Mellitus, Type 1/etiology , Enterovirus Infections/genetics , Gene-Environment Interaction , Alleles , Autoimmunity , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/virology , Enterovirus/genetics , Enterovirus/immunology , Enterovirus Infections/immunology , Genetic Predisposition to Disease , Humans , Insulin-Secreting Cells/immunology , Phenotype , Polymorphism, Genetic
5.
Diabetes ; 68(10): 2024-2034, 2019 10.
Article in English | MEDLINE | ID: mdl-31311800

ABSTRACT

The appearance of type 1 diabetes (T1D)-associated autoantibodies is the first and only measurable parameter to predict progression toward T1D in genetically susceptible individuals. However, autoantibodies indicate an active autoimmune reaction, wherein the immune tolerance is already broken. Therefore, there is a clear and urgent need for new biomarkers that predict the onset of the autoimmune reaction preceding autoantibody positivity or reflect progressive ß-cell destruction. Here we report the mRNA sequencing-based analysis of 306 samples including fractionated samples of CD4+ and CD8+ T cells as well as CD4-CD8- cell fractions and unfractionated peripheral blood mononuclear cell samples longitudinally collected from seven children who developed ß-cell autoimmunity (case subjects) at a young age and matched control subjects. We identified transcripts, including interleukin 32 (IL32), that were upregulated before T1D-associated autoantibodies appeared. Single-cell RNA sequencing studies revealed that high IL32 in case samples was contributed mainly by activated T cells and NK cells. Further, we showed that IL32 expression can be induced by a virus and cytokines in pancreatic islets and ß-cells, respectively. The results provide a basis for early detection of aberrations in the immune system function before T1D and suggest a potential role for IL32 in the pathogenesis of T1D.


Subject(s)
Autoantibodies , Autoimmunity/physiology , Diabetes Mellitus, Type 1/diagnosis , Insulin-Secreting Cells/immunology , Biomarkers/blood , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line , Child, Preschool , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/immunology , Disease Progression , Early Diagnosis , Female , Humans , Infant , Male
6.
Cell Rep ; 19(9): 1888-1901, 2017 05 30.
Article in English | MEDLINE | ID: mdl-28564606

ABSTRACT

The development of therapeutic strategies to combat immune-associated diseases requires the molecular mechanisms of human Th17 cell differentiation to be fully identified and understood. To investigate transcriptional control of Th17 cell differentiation, we used primary human CD4+ T cells in small interfering RNA (siRNA)-mediated gene silencing and chromatin immunoprecipitation followed by massive parallel sequencing (ChIP-seq) to identify both the early direct and indirect targets of STAT3. The integrated dataset presented in this study confirms that STAT3 is critical for transcriptional regulation of early human Th17 cell differentiation. Additionally, we found that a number of SNPs from loci associated with immune-mediated disorders were located at sites where STAT3 binds to induce Th17 cell specification. Importantly, introduction of such SNPs alters STAT3 binding in DNA affinity precipitation assays. Overall, our study provides important insights for modulating Th17-mediated pathogenic immune responses in humans.


Subject(s)
Cell Differentiation/genetics , Genome-Wide Association Study , STAT3 Transcription Factor/metabolism , Th17 Cells/cytology , Transcription, Genetic , Autoimmune Diseases/genetics , Base Sequence , Binding Sites , Cell Differentiation/drug effects , Cytokines/pharmacology , Gene Expression Regulation/drug effects , Gene Regulatory Networks/drug effects , Humans , Kinetics , Polymorphism, Single Nucleotide/genetics , Protein Binding/drug effects , Th17 Cells/drug effects , Transcription, Genetic/drug effects
7.
Oncotarget ; 7(12): 13416-28, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-26967054

ABSTRACT

Uncontrolled Th17 cell activity is associated with cancer and autoimmune and inflammatory diseases. To validate the potential relevance of mouse models of targeting the Th17 pathway in human diseases we used RNA sequencing to compare the expression of coding and non-coding transcripts during the priming of Th17 cell differentiation in both human and mouse. In addition to already known targets, several transcripts not previously linked to Th17 cell polarization were found in both species. Moreover, a considerable number of human-specific long non-coding RNAs were identified that responded to cytokines stimulating Th17 cell differentiation. We integrated our transcriptomics data with known disease-associated polymorphisms and show that conserved regulation pinpoints genes that are relevant to Th17 cell-mediated human diseases and that can be modelled in mouse. Substantial differences observed in non-coding transcriptomes between the two species as well as increased overlap between Th17 cell-specific gene expression and disease-associated polymorphisms underline the need of parallel analysis of human and mouse models. Comprehensive analysis of genes regulated during Th17 cell priming and their classification to conserved and non-conserved between human and mouse facilitates translational research, pointing out which candidate targets identified in human are worth studying by using in vivo mouse models.


Subject(s)
Biomarkers/metabolism , Polymorphism, Single Nucleotide , Th17 Cells/immunology , Th17 Cells/metabolism , Transcriptome , Animals , Cells, Cultured , Humans , Infant, Newborn , Mice , Mice, Inbred C57BL , Sequence Analysis, RNA
8.
J Hepatol ; 61(1): 132-8, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24613361

ABSTRACT

BACKGROUND & AIMS: Recent evidence suggests that in animals gut microbiota composition (GMC) affects the onset and progression of hepatic fat accumulation. The aim of this study was to investigate in humans whether subjects with high hepatic fat content (HHFC) differ in their GMC from those with low hepatic fat content (LHFC), and whether these differences are associated with body composition, biomarkers and abdominal adipose tissue inflammation. METHODS: Hepatic fat content (HFC) was measured using proton magnetic resonance spectroscopy ((1)H MRS). Fecal GMC was profiled by 16S rRNA fluorescence in situ hybridization and flow cytometry. Adipose tissue gene expression was analyzed using Affymetrix microarrays and quantitative PCR. RESULTS: The HHFC group had unfavorable GMC described by lower amount of Faecalibacterium prausnitzii (FPrau) (p<0.05) and relatively higher Enterobacteria than the LHFC group. Metabolically dysbiotic GMC associated with HOMA-IR and triglycerides (p<0.05 for both). Several inflammation-related adipose tissue genes were differentially expressed and correlated with HFC (p<0.05). In addition, the expression of certain genes correlated with GMC dysbiosis, i.e., low FPrau-to-Bacteroides ratio. CONCLUSIONS: HHFC subjects differ unfavorably in their GMC from LHFC subjects. Adipose tissue inflammation may be an important link between GMC, metabolic disturbances, and hepatic fat accumulation.


Subject(s)
Adipose Tissue/pathology , Liver/pathology , Microbiota , Adipose Tissue/metabolism , Adult , Body Composition , Cross-Sectional Studies , Digestive System/microbiology , Female , Gene Expression , Humans , Inflammation/pathology , Insulin Resistance , Male , Middle Aged , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/microbiology , Non-alcoholic Fatty Liver Disease/pathology , Triglycerides/blood
9.
Semin Immunol ; 25(4): 282-90, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24246225

ABSTRACT

Molecular mechanisms guiding naïve T helper cell differentiation into functionally specified effector cells are intensively studied. The rapidly growing knowledge is mainly achieved by using mouse cells or disease models. Comparatively exiguous data is gathered from human primary cells although they provide the "ultimate model" for immunology in man, have been exploited in many original studies paving the way for the field, and can be analyzed more easily than ever with the help of modern technology and methods. As usage of mouse models is unavoidable in translational research, parallel human and mouse studies should be performed to assure the relevancy of the hypothesis created during the basic research. In this review, we give an overview on the status of the studies conducted with human primary cells aiming at elucidating the mechanisms instructing the priming of T helper cell subtypes. The special emphasis is given to the recent high-throughput studies. In addition, by comparing the human and mouse studies we intend to point out the regulatory mechanisms and questions which are lacking examination with human primary cells.


Subject(s)
Cell Differentiation/genetics , Gene Expression Regulation , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/metabolism , Cell Differentiation/immunology , Epigenesis, Genetic , Humans , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Helper-Inducer/immunology , Transcription, Genetic
10.
PLoS One ; 8(7): e68414, 2013.
Article in English | MEDLINE | ID: mdl-23874618

ABSTRACT

The potential impact of nanoparticles on the environment and on human health has attracted considerable interest worldwide. The amount of transcriptomics data, in which tissues and cell lines are exposed to nanoparticles, increases year by year. In addition to the importance of the original findings, this data can have value in broader context when combined with other previously acquired and published results. In order to facilitate the efficient usage of the data, we have developed the NanoMiner web resource (http://nanominer.cs.tut.fi/), which contains 404 human transcriptome samples exposed to various types of nanoparticles. All the samples in NanoMiner have been annotated, preprocessed and normalized using standard methods that ensure the quality of the data analyses and enable the users to utilize the database systematically across the different experimental setups and platforms. With NanoMiner it is possible to 1) search and plot the expression profiles of one or several genes of interest, 2) cluster the samples within the datasets, 3) find differentially expressed genes in various nanoparticle studies, 4) detect the nanoparticles causing differential expression of selected genes, 5) analyze enriched Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways and Gene Ontology (GO) terms for the detected genes and 6) search the expression values and differential expressions of the genes belonging to a specific KEGG pathway or Gene Ontology. In sum, NanoMiner database is a valuable collection of microarray data which can be also used as a data repository for future analyses.


Subject(s)
Gene Expression Profiling , Nanoparticles/chemistry , Nanotechnology , Software , Statistics as Topic , Transcriptome/genetics , Cluster Analysis , Gene Expression Regulation , Humans , Molecular Sequence Annotation , Particulate Matter
11.
PLoS One ; 8(7): e68415, 2013.
Article in English | MEDLINE | ID: mdl-23894303

ABSTRACT

A comprehensive in vitro assessment of two commercial metal oxide nanoparticles, TiO2 and ZnO, was performed using human monocyte-derived macrophages (HMDM), monocyte-derived dendritic cells (MDDC), and Jurkat T cell leukemia-derived cell line. TiO2 nanoparticles were found to be non-toxic whereas ZnO nanoparticles caused dose-dependent cell death. Subsequently, global gene expression profiling was performed to identify transcriptional response underlying the cytotoxicity caused by ZnO nanoparticles. Analysis was done with doses 1 µg/ml and 10 µg/ml after 6 and 24 h of exposure. Interestingly, 2703 genes were significantly differentially expressed in HMDM upon exposure to 10 µg/ml ZnO nanoparticles, while in MDDCs only 12 genes were affected. In Jurkat cells, 980 genes were differentially expressed. It is noteworthy that only the gene expression of metallothioneins was upregulated in all the three cell types and a notable proportion of the genes were regulated in a cell type-specific manner. Gene ontology analysis revealed that the top biological processes disturbed in HMDM and Jurkat cells were regulating cell death and growth. In addition, genes controlling immune system development were affected. Using a panel of modified ZnO nanoparticles, we obtained an additional support that the cellular response to ZnO nanoparticles is largely dependent on particle dissolution and show that the ligand used to modify ZnO nanoparticles modulates Zn(2+) leaching. Overall, the study provides an extensive resource of transcriptional markers for mediating ZnO nanoparticle-induced toxicity for further mechanistic studies, and demonstrates the value of assessing nanoparticle responses through a combined transcriptomics and bioinformatics approach.


Subject(s)
Dendritic Cells/drug effects , Engineering , Gene Expression Profiling , Macrophages/drug effects , Nanoparticles , Titanium/pharmacology , Zinc Oxide/pharmacology , Adult , Computational Biology , Dendritic Cells/cytology , Dendritic Cells/metabolism , Humans , Jurkat Cells , Macrophages/cytology , Macrophages/metabolism , Monocytes/cytology , Nanotechnology , Titanium/chemistry , Transcription, Genetic/drug effects , Zinc Oxide/chemistry
12.
BMC Genomics ; 13: 572, 2012 Oct 30.
Article in English | MEDLINE | ID: mdl-23110343

ABSTRACT

BACKGROUND: A proper balance between different T helper (Th) cell subsets is necessary for normal functioning of the adaptive immune system. Revealing key genes and pathways driving the differentiation to distinct Th cell lineages provides important insight into underlying molecular mechanisms and new opportunities for modulating the immune response. Previous computational methods to quantify and visualize kinetic differential expression data of three or more lineages to identify reciprocally regulated genes have relied on clustering approaches and regression methods which have time as a factor, but have lacked methods which explicitly model temporal behavior. RESULTS: We studied transcriptional dynamics of human umbilical cord blood T helper cells cultured in absence and presence of cytokines promoting Th1 or Th2 differentiation. To identify genes that exhibit distinct lineage commitment dynamics and are specific for initiating differentiation to different Th cell subsets, we developed a novel computational methodology (LIGAP) allowing integrative analysis and visualization of multiple lineages over whole time-course profiles. Applying LIGAP to time-course data from multiple Th cell lineages, we identified and experimentally validated several differentially regulated Th cell subset specific genes as well as reciprocally regulated genes. Combining differentially regulated transcriptional profiles with transcription factor binding site and pathway information, we identified previously known and new putative transcriptional mechanisms involved in Th cell subset differentiation. All differentially regulated genes among the lineages together with an implementation of LIGAP are provided as an open-source resource. CONCLUSIONS: The LIGAP method is widely applicable to quantify differential time-course dynamics of many types of datasets and generalizes to any number of conditions. It summarizes all the time-course measurements together with the associated uncertainty for visualization and manual assessment purposes. Here we identified novel human Th subset specific transcripts as well as regulatory mechanisms important for the initiation of the Th cell subset differentiation.


Subject(s)
Cell Differentiation/genetics , Cell Lineage/genetics , Fetal Blood/metabolism , Gene Expression Regulation , Th1 Cells/metabolism , Th2 Cells/metabolism , Transcriptome , Adaptive Immunity/genetics , Binding Sites , Cell Differentiation/immunology , Cell Lineage/drug effects , Fetal Blood/cytology , Fetal Blood/drug effects , Gene Expression Profiling , Gene Expression Regulation/drug effects , Humans , Interleukin-12/immunology , Interleukin-12/pharmacology , Interleukin-2/immunology , Interleukin-2/pharmacology , Lymphocyte Activation , Primary Cell Culture , Protein Binding , Signal Transduction/drug effects , Systems Biology , Th1 Cells/cytology , Th1 Cells/drug effects , Th2 Cells/cytology , Th2 Cells/drug effects , Transcription Factors/genetics , Transcription Factors/immunology
13.
Blood ; 119(23): e151-60, 2012 Jun 07.
Article in English | MEDLINE | ID: mdl-22544700

ABSTRACT

Th17 cells play an essential role in the pathogenesis of autoimmune and inflammatory diseases. Most of our current understanding on Th17 cell differentiation relies on studies carried out in mice, whereas the molecular mechanisms controlling human Th17 cell differentiation are less well defined. In this study, we identified gene expression changes characterizing early stages of human Th17 cell differentiation through genome-wide gene expression profiling. CD4(+) cells isolated from umbilical cord blood were used to determine detailed kinetics of gene expression after initiation of Th17 differentiation with IL1ß, IL6, and TGFß. The differential expression of selected candidate genes was further validated at protein level and analyzed for specificity in initiation of Th17 compared with initiation of other Th subsets, namely Th1, Th2, and iTreg. This first genome-wide profiling of transcriptomics during the induction of human Th17 differentiation provides a starting point for defining gene regulatory networks and identifying new candidates regulating Th17 differentiation in humans.


Subject(s)
Gene Expression Profiling , Th17 Cells/cytology , Th17 Cells/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation , Cells, Cultured , Fetal Blood/cytology , Gene Expression Regulation , Humans , Interleukin-17/analysis , Interleukin-17/genetics , Interleukin-17/immunology , Interleukin-1beta/immunology , Interleukin-6/immunology , Th17 Cells/metabolism , Transforming Growth Factor beta/immunology
14.
Immunity ; 32(6): 852-62, 2010 Jun 25.
Article in English | MEDLINE | ID: mdl-20620947

ABSTRACT

Dissecting the molecular mechanisms by which T helper (Th) cells differentiate to effector Th2 cells is important for understanding the pathogenesis of immune-mediated diseases, such as asthma and allergy. Because the STAT6 transcription factor is an upstream mediator required for interleukin-4 (IL-4)-induced Th2 cell differentiation, its targets include genes important for this process. Using primary human CD4(+) T cells, and by blocking STAT6 with RNAi, we identified a number of direct and indirect targets of STAT6 with ChIP sequencing. The integration of these data sets with detailed kinetics of IL-4-driven transcriptional changes showed that STAT6 was predominantly needed for the activation of transcription leading to the Th2 cell phenotype. This integrated genome-wide data on IL-4- and STAT6-mediated transcription provide a unique resource for studies on Th cell differentiation and, in particular, for designing interventions of human Th2 cell responses.


Subject(s)
Cell Differentiation/immunology , Gene Expression Regulation/immunology , Interleukin-4/immunology , STAT6 Transcription Factor/immunology , Th2 Cells/cytology , Gene Expression , Gene Expression Profiling , Genome-Wide Association Study , Humans , Interleukin-4/genetics , Oligonucleotide Array Sequence Analysis , STAT6 Transcription Factor/genetics , Th2 Cells/immunology , Transcription, Genetic
15.
Blood ; 116(9): 1443-53, 2010 Sep 02.
Article in English | MEDLINE | ID: mdl-20522714

ABSTRACT

Special AT-rich binding protein 1 (SATB1) is a global chromatin organizer and a transcription factor regulated by interleukin-4 (IL-4) during the early T helper 2 (Th2) cell differentiation. Here we show that SATB1 controls multiple IL-4 target genes involved in human Th cell polarization or function. Among the genes regulated by SATB1 is that encoding the cytokine IL-5, which is predominantly produced by Th2 cells and plays a key role in the development of eosinophilia in asthma. We demonstrate that, during the early Th2 cell differentiation, IL-5 expression is repressed through direct binding of SATB1 to the IL-5 promoter. Furthermore, SATB1 knockdown-induced up-regulation of IL-5 is partly counteracted by down-regulating GATA3 expression using RNAi in polarizing Th2 cells. Our results suggest that a competitive mechanism involving SATB1 and GATA3 regulates IL-5 transcription, and provide new mechanistic insights into the stringent regulation of IL-5 expression during human Th2 cell differentiation.


Subject(s)
Cell Differentiation , Cell Proliferation , Interleukin-5/genetics , Matrix Attachment Region Binding Proteins/metabolism , Th2 Cells/cytology , Biomarkers/metabolism , Blotting, Western , CD4-Positive T-Lymphocytes/metabolism , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , Gene Expression Profiling , Humans , Infant, Newborn , Interleukin-5/metabolism , Luciferases/metabolism , Matrix Attachment Region Binding Proteins/antagonists & inhibitors , Matrix Attachment Region Binding Proteins/genetics , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Th2 Cells/metabolism , Transcription, Genetic , Transcriptional Activation , Transfection
16.
BMC Genomics ; 10: 618, 2009 Dec 18.
Article in English | MEDLINE | ID: mdl-20017957

ABSTRACT

BACKGROUND: Chromatin immunoprecipitation coupled with massively parallel sequencing (ChIP-seq) is increasingly being applied to study transcriptional regulation on a genome-wide scale. While numerous algorithms have recently been proposed for analysing the large ChIP-seq datasets, their relative merits and potential limitations remain unclear in practical applications. RESULTS: The present study compares the state-of-the-art algorithms for detecting transcription factor binding sites in four diverse ChIP-seq datasets under a variety of practical research settings. First, we demonstrate how the biological conclusions may change dramatically when the different algorithms are applied. The reproducibility across biological replicates is then investigated as an internal validation of the detections. Finally, the predicted binding sites with each method are compared to high-scoring binding motifs as well as binding regions confirmed in independent qPCR experiments. CONCLUSIONS: In general, our results indicate that the optimal choice of the computational approach depends heavily on the dataset under analysis. In addition to revealing valuable information to the users of this technology about the characteristics of the binding site detection approaches, the systematic evaluation framework provides also a useful reference to the developers of improved algorithms for ChIP-seq data.


Subject(s)
Algorithms , Chromatin Immunoprecipitation/methods , Computational Biology/methods , Sequence Analysis, DNA/methods , Transcription Factors/metabolism , Binding Sites , Software
17.
Proteomics ; 9(4): 1087-98, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19180534

ABSTRACT

Interleukin 4 (IL-4) has an indispensable role in the differentiation of naive T helper (Th) cells toward the Th2 phenotype and induction of B cells to produce the IgE class of Igs. By regulating these two cell types, IL-4 has a pre-eminent role in regulation of allergic inflammation. IL-4-mediated regulation of T and B cell functions is largely transmitted through signal transducer and activator of transcription 6 (Stat6). In this study, we have used metabolic labeling and 2-D electrophoresis to detect differences in the proteomes of IL-4 stimulated spleen mononuclear cells of Stat6-/- and wild type mice and MS/MS for protein identification. With this methodology, we identified 49 unique proteins from 21 protein spots to be differentially expressed. Interestingly, in Stat6-/- CD4(+) cells the expression of isoform 2 of core binding factor b (CBFb2) was enhanced. CBFb is a non-DNA binding cofactor for the Runx family of transcription factors, which have been implicated in regulation of Th cell differentiation. We also found cellular nucleic acid protein (CNBP) to be downregulated in Stat6-/- cells. None of the proteins identified in this study have previously been reported to be regulated via Stat6. The results highlight the importance of exploiting proteomics tools to complement the studies on Stat6 target genes identified through transcriptional profiling.


Subject(s)
Gene Expression Regulation/physiology , Interleukin-4/metabolism , Lymphocytes/metabolism , STAT6 Transcription Factor/metabolism , Animals , Core Binding Factor beta Subunit/genetics , Core Binding Factor beta Subunit/metabolism , Electrophoresis, Gel, Two-Dimensional , Lymphocytes/ultrastructure , Mice , Mice, Inbred BALB C , Mice, Knockout , Microscopy, Fluorescence , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , STAT6 Transcription Factor/genetics , Spleen/cytology , Tandem Mass Spectrometry
18.
Mol Cell Proteomics ; 8(1): 32-44, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18701445

ABSTRACT

T helper (Th) cells differentiate into functionally distinct effector cell subsets of which Th1 and Th2 cells are best characterized. Besides T cell receptor signaling, IL-12-induced STAT4 and T-bet- and IL-4-induced STAT6 and GATA3 signaling pathways are the major players regulating the Th1 and Th2 differentiation process, respectively. However, there are likely to be other yet unknown factors or pathways involved. In this study we used quantitative proteomics exploiting cleavable ICAT labeling and LC-MS/MS to identify IL-4-regulated proteins from the microsomal fractions of CD4(+) cells extracted from umbilical cord blood. We were able to identify 557 proteins of which 304 were also quantified. This study resulted in the identification of the down-regulation of small GTPases GIMAP1 and GIMAP4 by IL-4 during Th2 differentiation. We also showed that both GIMAP1 and GIMAP4 genes are up-regulated by IL-12 and other Th1 differentiation-inducing cytokines in cells induced to differentiate toward Th1 lineage and down-regulated by IL-4 in cells induced to Th2. Our results indicate that the GIMAP (GTPase of the immunity-associated protein) family of proteins is differentially regulated during Th cell differentiation.


Subject(s)
Cell Differentiation , GTP-Binding Proteins/genetics , Membrane Proteins/genetics , Proteomics , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/metabolism , Alternative Splicing/drug effects , Cell Differentiation/drug effects , Down-Regulation/drug effects , Fetal Blood/cytology , GTP-Binding Proteins/chemistry , GTP-Binding Proteins/metabolism , Humans , Interferon-alpha/pharmacology , Interleukin-18/pharmacology , Interleukin-4/pharmacology , Mass Spectrometry , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Microsomes/drug effects , Microsomes/metabolism , Proteome/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , STAT1 Transcription Factor/metabolism , STAT6 Transcription Factor/metabolism , Signal Transduction/drug effects , T-Lymphocytes, Helper-Inducer/drug effects , Up-Regulation/drug effects
19.
J Immunol ; 178(6): 3648-60, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17339462

ABSTRACT

Th cell subtypes, Th1 and Th2, are involved in the pathogenesis or progression of many immune-mediated diseases, such as type 1 diabetes and asthma, respectively. Defining the molecular networks and factors that direct Th1 and Th2 cell differentiation will help to understand the pathogenic mechanisms causing these diseases. Some of the key factors regulating this differentiation have been identified, however, they alone do not explain the process in detail. To identify novel factors directing the early differentiation, we have studied the transcriptomes of human Th1 and Th2 cells after 2, 6, and 48 h of polarization at the genome scale. Based on our current and previous studies, 288 genes or expressed sequence tags, representing approximately 1-1.5% of the human genome, are regulated in the process during the first 2 days. These transcriptional profiles revealed genes coding for components of certain pathways, such as RAS oncogene family and G protein-coupled receptor signaling, to be differentially regulated during the early Th1 and Th2 cell differentiation. Importantly, numerous novel genes with unknown functions were identified. By using short-hairpin RNA knockdown, we show that a subset of these genes is regulated by IL-4 through STAT6 signaling. Furthermore, we demonstrate that one of the IL-4 regulated genes, NDFIP2, promotes IFN-gamma production by the polarized human Th1 lymphocytes. Among the novel genes identified, there may be many factors that play a crucial role in the regulation of the differentiation process together with the previously known factors and are potential targets for developing therapeutics to modulate Th1 and Th2 responses.


Subject(s)
Cell Differentiation/physiology , Gene Expression Regulation/physiology , Genome, Human/physiology , Th1 Cells/physiology , Th2 Cells/physiology , Transcription, Genetic/physiology , Cells, Cultured , Gene Expression Profiling , Humans , Interleukin-4/immunology , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , STAT6 Transcription Factor/immunology , Signal Transduction/immunology , Time Factors
20.
Clin Cancer Res ; 12(16): 4812-21, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16914566

ABSTRACT

PURPOSE: Increased production of Th2 cytokines characterizes Sezary syndrome, the leukemic form of cutaneous T-cell lymphomas (CTCL). To identify the molecular background and to study whether shared by the most common CTCL subtype, mycosis fungoides, we analyzed the gene expression profiles in both subtypes. EXPERIMENTAL DESIGN: Freshly isolated cells from 30 samples, representing skin, blood, and enriched CD4(+) cell populations of mycosis fungoides and Sezary syndrome, were analyzed with Affymetrix (Santa Clara, CA) oligonucleotide microarrays, quantitative PCR, or immunohistochemistry. The gene expression profiles were combined with findings of comparative genomic hybridization of the same samples to identify chromosomal changes affecting the aberrant gene expression. RESULTS: We identified a set of Th1-specific genes [e.g., TBX21 (T-bet), NKG7, and SCYA5 (RANTES)] to be down-regulated in Sezary syndrome as well as in a proportion of mycosis fungoides samples. In both Sezary syndrome and mycosis fungoides blood samples, the S100P and LIR9 gene expression was up-regulated. In lesional skin, IL7R and CD52 were up-regulated. Integration of comparative genomic hybridization and transcriptomic data identified chromosome arms 1q, 3p, 3q, 4q, 12q, 16p, and 16q as likely targets for new CTCL-associated gene aberrations. CONCLUSIONS: Our findings revealed several new genes involved in CTCL pathogenesis and potential therapeutic targets. Down-regulation of a set of genes involved in Th1 polarization, including the major Th1-polarizing factor, TBX21, was for the first time associated with CTCL. In addition, a plausible explanation for the proliferative response of CTCL cells to locally produced interleukin-7 was revealed.


Subject(s)
Mycosis Fungoides/genetics , Mycosis Fungoides/immunology , Sezary Syndrome/genetics , Sezary Syndrome/immunology , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Th1 Cells/physiology , Cytotoxicity, Immunologic/genetics , Cytotoxicity, Immunologic/immunology , Down-Regulation , Gene Dosage , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry/methods , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction/methods , Th1 Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...