Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Cell ; 172(1-2): 162-175.e14, 2018 01 11.
Article in English | MEDLINE | ID: mdl-29328911

ABSTRACT

Long-term epigenetic reprogramming of innate immune cells in response to microbes, also termed "trained immunity," causes prolonged altered cellular functionality to protect from secondary infections. Here, we investigated whether sterile triggers of inflammation induce trained immunity and thereby influence innate immune responses. Western diet (WD) feeding of Ldlr-/- mice induced systemic inflammation, which was undetectable in serum soon after mice were shifted back to a chow diet (CD). In contrast, myeloid cell responses toward innate stimuli remained broadly augmented. WD-induced transcriptomic and epigenomic reprogramming of myeloid progenitor cells led to increased proliferation and enhanced innate immune responses. Quantitative trait locus (QTL) analysis in human monocytes trained with oxidized low-density lipoprotein (oxLDL) and stimulated with lipopolysaccharide (LPS) suggested inflammasome-mediated trained immunity. Consistently, Nlrp3-/-/Ldlr-/- mice lacked WD-induced systemic inflammation, myeloid progenitor proliferation, and reprogramming. Hence, NLRP3 mediates trained immunity following WD and could thereby mediate the potentially deleterious effects of trained immunity in inflammatory diseases.


Subject(s)
Cellular Reprogramming , Diet, Western , Epigenesis, Genetic , Immunity, Innate , Immunologic Memory , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Adult , Aged , Animals , Cells, Cultured , Female , Humans , Lipoproteins, LDL/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , Myeloid Cells/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Quantitative Trait Loci , Receptors, LDL/genetics
2.
Immunity ; 50(5): 1232-1248.e14, 2019 05 21.
Article in English | MEDLINE | ID: mdl-31027998

ABSTRACT

Regulatory T cells (Treg cells) are important for preventing autoimmunity and maintaining tissue homeostasis, but whether Treg cells can adopt tissue- or immune-context-specific suppressive mechanisms is unclear. Here, we found that the enzyme hydroxyprostaglandin dehydrogenase (HPGD), which catabolizes prostaglandin E2 (PGE2) into the metabolite 15-keto PGE2, was highly expressed in Treg cells, particularly those in visceral adipose tissue (VAT). Nuclear receptor peroxisome proliferator-activated receptor-γ (PPARγ)-induced HPGD expression in VAT Treg cells, and consequential Treg-cell-mediated generation of 15-keto PGE2 suppressed conventional T cell activation and proliferation. Conditional deletion of Hpgd in mouse Treg cells resulted in the accumulation of functionally impaired Treg cells specifically in VAT, causing local inflammation and systemic insulin resistance. Consistent with this mechanism, humans with type 2 diabetes showed decreased HPGD expression in Treg cells. These data indicate that HPGD-mediated suppression is a tissue- and context-dependent suppressive mechanism used by Treg cells to maintain adipose tissue homeostasis.


Subject(s)
Dinoprostone/analogs & derivatives , Dinoprostone/metabolism , Hydroxyprostaglandin Dehydrogenases/metabolism , Intra-Abdominal Fat/immunology , T-Lymphocytes, Regulatory/enzymology , T-Lymphocytes, Regulatory/immunology , 3T3 Cells , Animals , Cell Line , Diabetes Mellitus, Type 2/metabolism , HEK293 Cells , Homeostasis/immunology , Humans , Hydroxyprostaglandin Dehydrogenases/genetics , Insulin Resistance/genetics , Intra-Abdominal Fat/cytology , Jurkat Cells , Lymphocyte Activation/immunology , Male , Mice , Mice, Knockout , STAT5 Transcription Factor/metabolism
3.
Immunity ; 47(6): 1051-1066.e12, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29262348

ABSTRACT

Human in vitro generated monocyte-derived dendritic cells (moDCs) and macrophages are used clinically, e.g., to induce immunity against cancer. However, their physiological counterparts, ontogeny, transcriptional regulation, and heterogeneity remains largely unknown, hampering their clinical use. High-dimensional techniques were used to elucidate transcriptional, phenotypic, and functional differences between human in vivo and in vitro generated mononuclear phagocytes to facilitate their full potential in the clinic. We demonstrate that monocytes differentiated by macrophage colony-stimulating factor (M-CSF) or granulocyte macrophage colony-stimulating factor (GM-CSF) resembled in vivo inflammatory macrophages, while moDCs resembled in vivo inflammatory DCs. Moreover, differentiated monocytes presented with profound transcriptomic, phenotypic, and functional differences. Monocytes integrated GM-CSF and IL-4 stimulation combinatorically and temporally, resulting in a mode- and time-dependent differentiation relying on NCOR2. Finally, moDCs are phenotypically heterogeneous and therefore necessitate the use of high-dimensional phenotyping to open new possibilities for better clinical tailoring of these cellular therapies.


Subject(s)
Dendritic Cells/immunology , Interleukin-4/immunology , Macrophages/immunology , Monocytes/immunology , Nuclear Receptor Co-Repressor 2/immunology , Signal Transduction/immunology , Cell Differentiation , Cell Lineage , Dendritic Cells/cytology , Dendritic Cells/drug effects , Gene Expression Profiling , Gene Expression Regulation , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Immunophenotyping , Interleukin-4/genetics , Interleukin-4/pharmacology , Macrophage Activation , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/cytology , Macrophages/drug effects , Monocytes/cytology , Monocytes/drug effects , Nuclear Receptor Co-Repressor 2/genetics , Primary Cell Culture , Time Factors , Transcription, Genetic
4.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Article in English | MEDLINE | ID: mdl-33372158

ABSTRACT

Macrophages are the principal immune cells of the epididymis and testis, but their origins, heterogeneity, development, and maintenance are not well understood. Here, we describe distinct populations of epididymal and testicular macrophages that display an organ-specific cellular identity. Combining in vivo fate-mapping, chimeric and parabiotic mouse models with in-depth cellular analyses, we found that CD64hiMHCIIlo and CD64loMHCIIhi macrophage populations of epididymis and testis arise sequentially from yolk sac erythro-myeloid progenitors, embryonic hematopoiesis, and nascent neonatal monocytes. While monocytes were the major developmental source of both epididymal and testicular macrophages, both populations self-maintain in the steady-state independent of bone marrow hematopoietic precursors. However, after radiation-induced macrophage ablation or during infection, bone marrow-derived circulating monocytes are recruited to the epididymis and testis, giving rise to inflammatory macrophages that promote tissue damage. These results define the layered ontogeny, maintenance and inflammatory response of macrophage populations in the male reproductive organs.


Subject(s)
Infertility, Male/immunology , Macrophages/immunology , Macrophages/metabolism , Animals , Cell Differentiation , Cell Lineage , Epididymis/immunology , Epididymis/metabolism , Infertility, Male/metabolism , Infertility, Male/physiopathology , Male , Mice , Mice, Inbred C57BL , Monocytes/immunology , Testis/immunology , Testis/metabolism
5.
J Immunol ; 195(9): 4446-55, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26416280

ABSTRACT

Cytokines and IFNs downstream of innate immune pathways are critical for mounting an appropriate immune response to microbial infection. However, the expression of these inflammatory mediators is tightly regulated, as uncontrolled production can result in tissue damage and lead to chronic inflammatory conditions and autoimmune diseases. Activating transcription factor 3 (ATF3) is an important transcriptional modulator that limits the inflammatory response by controlling the expression of a number of cytokines and chemokines. However, its role in modulating IFN responses remains poorly defined. In this study, we demonstrate that ATF3 expression in macrophages is necessary for governing basal IFN-ß expression, as well as the magnitude of IFN-ß cytokine production following activation of innate immune receptors. We found that ATF3 acted as a transcriptional repressor and regulated IFN-ß via direct binding to a previously unidentified specific regulatory site distal to the Ifnb1 promoter. Additionally, we observed that ATF3 itself is a type I IFN-inducible gene, and that ATF3 further modulates the expression of a subset of inflammatory genes downstream of IFN signaling, suggesting it constitutes a key component of an IFN negative feedback loop. Consistent with this, macrophages deficient in Atf3 showed enhanced viral clearance in lymphocytic choriomeningitis virus and vesicular stomatitis virus infection models. Our study therefore demonstrates an important role for ATF3 in modulating IFN responses in macrophages by controlling basal and inducible levels of IFNß, as well as the expression of genes downstream of IFN signaling.


Subject(s)
Activating Transcription Factor 3/genetics , Interferon-beta/genetics , Macrophages/metabolism , Transcriptome/genetics , Activating Transcription Factor 3/metabolism , Animals , Cells, Cultured , Dendritic Cells/drug effects , Dendritic Cells/metabolism , HEK293 Cells , Humans , Immunoblotting , Interferon-beta/metabolism , Interferon-beta/pharmacology , Macrophages/drug effects , Mice, Inbred C57BL , Mice, Knockout , Monocytes/drug effects , Monocytes/metabolism , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic/genetics , Protein Binding , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcriptome/drug effects
6.
Front Immunol ; 14: 1107397, 2023.
Article in English | MEDLINE | ID: mdl-37559728

ABSTRACT

CD4+ T cells play a central role in the adaptive immune response through their capacity to activate, support and control other immune cells. Although these cells have become the focus of intense research, a comprehensive understanding of the underlying regulatory networks that orchestrate CD4+ T cell function and activation is still incomplete. Here, we analyzed a large transcriptomic dataset consisting of 48 different human CD4+ T cell conditions. By performing reverse network engineering, we identified six common denominators of CD4+ T cell functionality (CREB1, E2F3, AHR, STAT1, NFAT5 and NFATC3). Moreover, we also analyzed condition-specific genes which led us to the identification of the transcription factor MEOX1 in Treg cells. Expression of MEOX1 was comparable to FOXP3 in Treg cells and can be upregulated by IL-2. Epigenetic analyses revealed a permissive epigenetic landscape for MEOX1 solely in Treg cells. Knockdown of MEOX1 in Treg cells revealed a profound impact on downstream gene expression programs and Treg cell suppressive capacity. These findings in the context of CD4+ T cells contribute to a better understanding of the transcriptional networks and biological mechanisms controlling CD4+ T cell functionality, which opens new avenues for future therapeutic strategies.


Subject(s)
Gene Expression Regulation , T-Lymphocytes, Regulatory , Humans , Gene Regulatory Networks , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Transcription Factors/metabolism , Homeodomain Proteins/genetics
7.
Sci Rep ; 13(1): 5506, 2023 04 04.
Article in English | MEDLINE | ID: mdl-37016052

ABSTRACT

Epigenetic features such as DNA accessibility dictate transcriptional regulation in a cell type- and cell state- specific manner, and mapping this in health vs. disease in clinically relevant material is opening the door to new mechanistic insights and new targets for therapy. Assay for Transposase Accessible Chromatin Sequencing (ATAC-seq) allows chromatin accessibility profiling from low cell input, making it tractable on rare cell populations, such as regulatory T (Treg) cells. However, little is known about the compatibility of the assay with cryopreserved rare cell populations. Here we demonstrate the robustness of an ATAC-seq protocol comparing primary Treg cells recovered from fresh or cryopreserved PBMC samples, in the steady state and in response to stimulation. We extend this method to explore the feasibility of conducting simultaneous quantitation of chromatin accessibility and transcriptome from a single aliquot of 50,000 cryopreserved Treg cells. Profiling of chromatin accessibility and gene expression in parallel within the same pool of cells controls for cellular heterogeneity and is particularly beneficial when constrained by limited input material. Overall, we observed a high correlation of accessibility patterns and transcription factor dynamics between fresh and cryopreserved samples. Furthermore, highly similar transcriptomic profiles were obtained from whole cells and from the supernatants recovered from ATAC-seq reactions. We highlight the feasibility of applying these techniques to profile the epigenomic landscape of cells recovered from cryopreservation biorepositories.


Subject(s)
Chromatin , T-Lymphocytes, Regulatory , Humans , Chromatin/genetics , Leukocytes, Mononuclear , High-Throughput Nucleotide Sequencing/methods , Transcriptome
8.
Nat Commun ; 12(1): 681, 2021 01 29.
Article in English | MEDLINE | ID: mdl-33514719

ABSTRACT

Endothelial cells play a critical role in the adaptation of tissues to injury. Tissue ischemia induced by infarction leads to profound changes in endothelial cell functions and can induce transition to a mesenchymal state. Here we explore the kinetics and individual cellular responses of endothelial cells after myocardial infarction by using single cell RNA sequencing. This study demonstrates a time dependent switch in endothelial cell proliferation and inflammation associated with transient changes in metabolic gene signatures. Trajectory analysis reveals that the majority of endothelial cells 3 to 7 days after myocardial infarction acquire a transient state, characterized by mesenchymal gene expression, which returns to baseline 14 days after injury. Lineage tracing, using the Cdh5-CreERT2;mT/mG mice followed by single cell RNA sequencing, confirms the transient mesenchymal transition and reveals additional hypoxic and inflammatory signatures of endothelial cells during early and late states after injury. These data suggest that endothelial cells undergo a transient mes-enchymal activation concomitant with a metabolic adaptation within the first days after myocardial infarction but do not acquire a long-term mesenchymal fate. This mesenchymal activation may facilitate endothelial cell migration and clonal expansion to regenerate the vascular network.


Subject(s)
Endothelium/pathology , Epithelial-Mesenchymal Transition/genetics , Myocardial Infarction/pathology , Myocardium/pathology , Animals , Cell Movement/genetics , Cell Plasticity/genetics , Cell Proliferation/genetics , Cells, Cultured , Disease Models, Animal , Endothelial Cells/pathology , Endothelium/cytology , Genes, Reporter/genetics , Human Umbilical Vein Endothelial Cells , Humans , Luminescent Proteins/genetics , Male , Mice , Mice, Transgenic , Myocardium/cytology , RNA-Seq , Single-Cell Analysis
9.
Nat Commun ; 11(1): 4267, 2020 08 26.
Article in English | MEDLINE | ID: mdl-32848148

ABSTRACT

While footprinting analysis of ATAC-seq data can theoretically enable investigation of transcription factor (TF) binding, the lack of a computational tool able to conduct different levels of footprinting analysis has so-far hindered the widespread application of this method. Here we present TOBIAS, a comprehensive, accurate, and fast footprinting framework enabling genome-wide investigation of TF binding dynamics for hundreds of TFs simultaneously. We validate TOBIAS using paired ATAC-seq and ChIP-seq data, and find that TOBIAS outperforms existing methods for bias correction and footprinting. As a proof-of-concept, we illustrate how TOBIAS can unveil complex TF dynamics during zygotic genome activation in both humans and mice, and propose how zygotic Dux activates cascades of TFs, binds to repeat elements and induces expression of novel genetic elements.


Subject(s)
Chromatin Immunoprecipitation Sequencing/methods , Transcription Factors/metabolism , Transcriptional Activation , Zygote/metabolism , Animals , Binding Sites/genetics , Embryonic Development/genetics , Epigenesis, Genetic , Female , Genome, Human , Homeodomain Proteins/metabolism , Humans , Kinetics , Mice , Promoter Regions, Genetic , Proof of Concept Study , Protein Binding/genetics , Species Specificity
10.
Nat Neurosci ; 23(3): 351-362, 2020 03.
Article in English | MEDLINE | ID: mdl-32042176

ABSTRACT

Monocyte-derived and tissue-resident macrophages are ontogenetically distinct components of the innate immune system. Assessment of their respective functions in pathology is complicated by changes to the macrophage phenotype during inflammation. Here we find that Cxcr4-CreER enables permanent genetic labeling of hematopoietic stem cells (HSCs) and distinguishes HSC-derived monocytes from microglia and other tissue-resident macrophages. By combining Cxcr4-CreER-mediated lineage tracing with Cxcr4 inhibition or conditional Cxcr4 ablation in photothrombotic stroke, we find that Cxcr4 promotes initial monocyte infiltration and subsequent territorial restriction of monocyte-derived macrophages to infarct tissue. After transient focal ischemia, Cxcr4 deficiency reduces monocyte infiltration and blunts the expression of pattern recognition and defense response genes in monocyte-derived macrophages. This is associated with an altered microglial response and deteriorated outcomes. Thus, Cxcr4 is essential for an innate-immune-system-mediated defense response after cerebral ischemia. We further propose Cxcr4-CreER as a universal tool to study functions of HSC-derived cells.


Subject(s)
Brain Ischemia/immunology , Hematopoietic Stem Cells/immunology , Microglia/immunology , Monocytes/immunology , Receptors, CXCR4/metabolism , Stroke/immunology , Animals , Brain Ischemia/pathology , Cell Lineage , Cerebral Infarction/immunology , Cerebral Infarction/pathology , Hematopoietic Stem Cells/pathology , Immunity, Innate/genetics , Ischemic Attack, Transient/immunology , Ischemic Attack, Transient/pathology , Mice, Inbred C57BL , Mice, Knockout , Microglia/pathology , Monocytes/pathology , Receptors, CXCR4/genetics , Receptors, CXCR4/immunology , Stroke/pathology , Thrombosis/pathology , Treatment Outcome
11.
iScience ; 23(1): 100780, 2020 Jan 24.
Article in English | MEDLINE | ID: mdl-31918046

ABSTRACT

Acute myeloid leukemia (AML) is a severe, mostly fatal hematopoietic malignancy. We were interested in whether transcriptomic-based machine learning could predict AML status without requiring expert input. Using 12,029 samples from 105 different studies, we present a large-scale study of machine learning-based prediction of AML in which we address key questions relating to the combination of machine learning and transcriptomics and their practical use. We find data-driven, high-dimensional approaches-in which multivariate signatures are learned directly from genome-wide data with no prior knowledge-to be accurate and robust. Importantly, these approaches are highly scalable with low marginal cost, essentially matching human expert annotation in a near-automated workflow. Our results support the notion that transcriptomics combined with machine learning could be used as part of an integrated -omics approach wherein risk prediction, differential diagnosis, and subclassification of AML are achieved by genomics while diagnosis could be assisted by transcriptomic-based machine learning.

12.
Cell Host Microbe ; 28(2): 322-334.e5, 2020 08 12.
Article in English | MEDLINE | ID: mdl-32544459

ABSTRACT

Induction of trained immunity by Bacille-Calmette-Guérin (BCG) vaccination mediates beneficial heterologous effects, but the mechanisms underlying its persistence and magnitude remain elusive. In this study, we show that BCG vaccination in healthy human volunteers induces a persistent transcriptional program connected to myeloid cell development and function within the hematopoietic stem and progenitor cell (HSPC) compartment in the bone marrow. We identify hepatic nuclear factor (HNF) family members 1a and b as crucial regulators of this transcriptional shift. These findings are corroborated by higher granulocyte numbers in BCG-vaccinated infants, HNF1 SNP variants that correlate with trained immunity, and elevated serum concentrations of the HNF1 target alpha-1 antitrypsin. Additionally, transcriptomic HSPC remodeling was epigenetically conveyed to peripheral CD14+ monocytes, displaying an activated transcriptional signature three months after BCG vaccination. Taken together, transcriptomic, epigenomic, and functional reprogramming of HSPCs and peripheral monocytes is a hallmark of BCG-induced trained immunity in humans.


Subject(s)
BCG Vaccine/immunology , Granulocytes/cytology , Hematopoiesis/immunology , Hematopoietic Stem Cells/cytology , Monocytes/cytology , Bone Marrow/immunology , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Cytokines/metabolism , Female , Healthy Volunteers , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 1-beta/genetics , Humans , Lipopolysaccharide Receptors/metabolism , Male , Monocytes/immunology , Mycobacterium bovis/immunology , Transcription, Genetic/genetics , Transcriptome/genetics , Vaccination , Young Adult , alpha 1-Antitrypsin/blood
13.
Nucleic Acids Res ; 35(Database issue): D834-40, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17202173

ABSTRACT

Genome-oriented plant research delivers rapidly increasing amount of plant genome data. Comprehensive and structured information resources are required to structure and communicate genome and associated analytical data for model organisms as well as for crops. The increase in available plant genomic data enables powerful comparative analysis and integrative approaches. PlantsDB aims to provide data and information resources for individual plant species and in addition to build a platform for integrative and comparative plant genome research. PlantsDB is constituted from genome databases for Arabidopsis, Medicago, Lotus, rice, maize and tomato. Complementary data resources for cis elements, repetive elements and extensive cross-species comparisons are implemented. The PlantsDB portal can be reached at http://mips.gsf.de/projects/plants.


Subject(s)
Databases, Nucleic Acid , Genome, Plant , Arabidopsis/genetics , Base Sequence , Conserved Sequence , DNA, Plant/chemistry , Fabaceae/genetics , Genomics , Internet , Solanum lycopersicum/genetics , Plant Proteins/chemistry , Poaceae/genetics , Promoter Regions, Genetic , Repetitive Sequences, Nucleic Acid , Sequence Homology, Amino Acid , Software , Systems Integration , User-Computer Interface
14.
Cell Rep ; 29(5): 1221-1235.e5, 2019 10 29.
Article in English | MEDLINE | ID: mdl-31665635

ABSTRACT

Tumor-associated macrophages (TAMs) are frequently the most abundant immune cells in cancers and are associated with poor survival. Here, we generated TAM molecular signatures from K14cre;Cdh1flox/flox;Trp53flox/flox (KEP) and MMTV-NeuT (NeuT) transgenic mice that resemble human invasive lobular carcinoma (ILC) and HER2+ tumors, respectively. Determination of TAM-specific signatures requires comparison with healthy mammary tissue macrophages to avoid overestimation of gene expression differences. TAMs from the two models feature a distinct transcriptomic profile, suggesting that the cancer subtype dictates their phenotype. The KEP-derived signature reliably correlates with poor overall survival in ILC but not in triple-negative breast cancer patients, indicating that translation of murine TAM signatures to patients is cancer subtype dependent. Collectively, we show that a transgenic mouse tumor model can yield a TAM signature relevant for human breast cancer outcome prognosis and provide a generalizable strategy for determining and applying immune cell signatures provided the murine model reflects the human disease.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Gene Expression Profiling , Macrophages/metabolism , Mammary Neoplasms, Animal/pathology , Transcription, Genetic , Animals , Carcinogenesis/genetics , Carcinogenesis/pathology , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic , Humans , Mammary Neoplasms, Animal/genetics , Mice, Inbred BALB C , Mice, Transgenic , Phenotype , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Survival Analysis , Transcriptome/genetics , Treatment Outcome
15.
Arch Microbiol ; 190(3): 281-99, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18592220

ABSTRACT

HaloLex is a software system for the central management, integration, curation, and web-based visualization of genomic and other -omics data for any given microorganism. The system has been employed for the manual curation of three haloarchaeal genomes, namely Halobacterium salinarum (strain R1), Natronomonas pharaonis, and Haloquadratum walsbyi. HaloLex, in particular, enables the integrated analysis of genome-wide proteomic results with the underlying genomic data. This has proven indispensable to generate reliable gene predictions for GC-rich genomes, which, due to their characteristically low abundance of stop codons, are known to be hard targets for standard gene finders, especially concerning start codon assignment. The proteomic identification of more than 600 N-terminal peptides has greatly increased the reliability of the start codon assignment for Halobacterium salinarum. Application of homology-based methods to the published genome of Haloarcula marismortui allowed to detect 47 previously unidentified genes (a problem that is particularly serious for short protein sequences) and to correct more than 300 start codon misassignments.


Subject(s)
Genome, Archaeal , Halobacteriaceae/genetics , Software , Amino Acid Sequence , Archaeal Proteins/genetics , Codon, Initiator/genetics , Computational Biology/methods , Genes, Archaeal , Genomics , Information Management , Molecular Sequence Data , Open Reading Frames , Proteomics , Sequence Alignment , Sequence Homology, Amino Acid
16.
Cell Rep ; 22(4): 967-978, 2018 01 23.
Article in English | MEDLINE | ID: mdl-29386138

ABSTRACT

Maintenance of metabolic homeostasis requires adaption of gene regulation to the cellular energy state via transcriptional regulators. Here, we identify a role of ceramide synthase (CerS) Schlank, a multiple transmembrane protein containing a catalytic lag1p motif and a homeodomain, which is poorly studied in CerSs, as a transcriptional regulator. ChIP experiments show that it binds promoter regions of lipases lipase3 and magro via its homeodomain. Mutation of nuclear localization site 2 (NLS2) within the homeodomain leads to loss of DNA binding and deregulated gene expression, and NLS2 mutants can no longer adjust the transcriptional response to changing lipid levels. This mechanism is conserved in mammalian CerS2 and emphasizes the importance of the CerS protein rather than ceramide synthesis. This study demonstrates a double role of CerS Schlank as an enzyme and a transcriptional regulator, sensing lipid levels and transducing the information to the level of gene expression.


Subject(s)
Ceramides/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Gene Expression Regulation/genetics , Sphingosine N-Acyltransferase/genetics , Animals
17.
BMC Bioinformatics ; 8: 320, 2007 Aug 30.
Article in English | MEDLINE | ID: mdl-17760972

ABSTRACT

BACKGROUND: Apollo, a genome annotation viewer and editor, has become a widely used genome annotation and visualization tool for distributed genome annotation projects. When using Apollo for annotation, database updates are carried out by uploading intermediate annotation files into the respective database. This non-direct database upload is laborious and evokes problems of data synchronicity. RESULTS: To overcome these limitations we extended the Apollo data adapter with a generic, configurable web service client that is able to retrieve annotation data in a GAME-XML-formatted string and pass it on to Apollo's internal input routine. CONCLUSION: This Apollo web service adapter, Apollo2Go, simplifies the data exchange in distributed projects and aims to render the annotation process more comfortable. The Apollo2Go software is freely available from ftp://ftpmips.gsf.de/plants/apollo_webservice.


Subject(s)
Chromosome Mapping/methods , Databases, Genetic , Documentation/methods , Information Storage and Retrieval/methods , Internet , Software , User-Computer Interface , Database Management Systems
18.
Oncoimmunology ; 6(4): e1295203, 2017.
Article in English | MEDLINE | ID: mdl-28507805

ABSTRACT

Myeloid-derived suppressor cells (MDSC) are critical in regulating immune responses by suppressing antigen presenting cells (APC) and T cells. We previously observed that incubation of peripheral blood monocytes with interleukin (IL)-10 during their differentiation to monocyte-derived dendritic cells (moDCs) results in the generation of an APC population with a CD14+HLA-DRlowphenotype (IL-10-APC) with reduced stimulatory capacity similar to human MDSC. Co-incubation experiments now revealed that the addition of IL-10-APC to moDC caused a reduction of DC-induced T-cell proliferation, of the expression of maturation markers, and of secreted cytokines and chemokines such as TNF-α, IL-6, MIP-1α and Rantes. Addition of IL-10-APC increased the immunosuppressive molecule osteoactivin and its corresponding receptor syndecan-4 on moDC. Moreover, CD14+HLA-DRlow MDSC isolated from healthy donors expressed high levels of osteoactivin, which was even further upregulated by the auxiliary addition of IL-10. Using transcriptome analysis, we identified a set of molecules and pathways mediating these effects. In addition, we found that IL-10-APC as well as human isolated MDSC expressed higher levels of programmed death (PD)-1, PD-ligand-1 (PD-L1), glucocorticoid-induced-tumor-necrosis-factor-receptor-related-protein (GITR) and GITR-ligand. Inhibition of osteoactivin, syndecan-4, PD-1 or PD-L1 on MDSC by using blocking antibodies restored the stimulatory capacity of DC in co-incubation experiments. Activation of MDSC with Dectin-1 ligand curdlan reduced the expression of osteoactivin and PD-L1. Our results demonstrate that osteoactivin/syndecan-4 and PD-/PD-L1 are key molecules that are profoundly involved in the inhibitory effects of MDSC on DC function and might be promising tools for clinical application.

19.
Cell Res ; 26(2): 151-70, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26729620

ABSTRACT

Differentiation of inflammatory macrophages from monocytes is characterized by an orderly integration of epigenetic and transcriptional regulatory mechanisms guided by lineage-determining transcription factors such as PU.1. Further activation of macrophages leads to a stimulus- or microenvironment-specific signal integration with subsequent transcriptional control established by the action of tissue- or signal-associated transcription factors. Here, we assess four histone modifications during human macrophage activation and integrate this information with the gene expression data from 28 different macrophage activation conditions in combination with GM-CSF. Bioinformatically, for inflammatory macrophages we define a unique network of transcriptional and epigenetic regulators (TRs), which was characterized by accessible promoters independent of the activation signal. In contrast to the general accessibility of promoters of TRs, mRNA expression of central TRs belonging to the TR network displayed stimulus-specific expression patterns, indicating a second level of transcriptional regulation beyond epigenetic chromatin changes. In contrast, stringent integration of epigenetic and transcriptional regulation was observed in networks of TRs established from somatic tissues and tissue macrophages. In these networks, clusters of TRs with permissive histone marks were associated with high gene expression whereas clusters with repressive chromatin marks were associated with absent gene expression. Collectively, these results support that macrophage activation during inflammation in contrast to lineage determination is mainly regulated transcriptionally by a pre-defined TR network.


Subject(s)
Chromatin/genetics , Gene Regulatory Networks/genetics , Inflammation/genetics , Macrophages/metabolism , Animals , Epigenesis, Genetic/genetics , Gene Expression/genetics , Gene Expression Regulation/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Mice , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics
20.
Sci Rep ; 5: 11126, 2015 Jun 09.
Article in English | MEDLINE | ID: mdl-26057209

ABSTRACT

Regulatory networks for differentiation and pluripotency in embryonic stem (ES) cells have long been suggested to be mutually exclusive. However, with the identification of many new components of these networks ranging from epigenetic, transcriptional, and translational to even post-translational mechanisms, the cellular states of pluripotency and early differentiation might not be strictly bi-modal, but differentiating stem cells appear to go through phases of simultaneous expression of stemness and differentiation genes. Translational regulators such as RNA binding proteins (RBPs) and micro RNAs (miRNAs) might be prime candidates for guiding a cell from pluripotency to differentiation. Using Trim71, one of two members of the Tripartite motif (Trim) protein family with RNA binding activity expressed in murine ES cells, we demonstrate that Trim71 is not involved in regulatory networks of pluripotency but regulates neural differentiation. Loss of Trim71 in mES cells leaves stemness and self-maintenance of these cells intact, but many genes required for neural development are up-regulated at the same time. Concordantly, Trim71(-/-) mES show increased neural marker expression following treatment with retinoic acid. Our findings strongly suggest that Trim71 keeps priming steps of differentiation in check, which do not pre-require a loss of the pluripotency network in ES cells.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Neural Plate/cytology , Transcription Factors/physiology , 3' Untranslated Regions , Animals , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental/physiology , Mice , Transcription Factors/genetics , Transcription, Genetic/physiology
SELECTION OF CITATIONS
SEARCH DETAIL