Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
1.
Nucleic Acids Res ; 2024 May 27.
Article in English | MEDLINE | ID: mdl-38801077

ABSTRACT

Acute myeloid leukemia (AML) is a hematological malignancy characterized by abnormal proliferation and accumulation of immature myeloid cells in the bone marrow. Inflammation plays a crucial role in AML progression, but excessive activation of cell-intrinsic inflammatory pathways can also trigger cell death. IRF2BP2 is a chromatin regulator implicated in AML pathogenesis, although its precise role in this disease is not fully understood. In this study, we demonstrate that IRF2BP2 interacts with the AP-1 heterodimer ATF7/JDP2, which is involved in activating inflammatory pathways in AML cells. We show that IRF2BP2 is recruited by the ATF7/JDP2 dimer to chromatin and counteracts its gene-activating function. Loss of IRF2BP2 leads to overactivation of inflammatory pathways, resulting in strongly reduced proliferation. Our research indicates that a precise equilibrium between activating and repressive transcriptional mechanisms creates a pro-oncogenic inflammatory environment in AML cells. The ATF7/JDP2-IRF2BP2 regulatory axis is likely a key regulator of this process and may, therefore, represent a promising therapeutic vulnerability for AML. Thus, our study provides new insights into the molecular mechanisms underlying AML pathogenesis and identifies a potential therapeutic target for AML treatment.

2.
Nucleic Acids Res ; 51(2): 574-594, 2023 01 25.
Article in English | MEDLINE | ID: mdl-36537216

ABSTRACT

The lysine acetyltransferase KAT6A (MOZ, MYST3) belongs to the MYST family of chromatin regulators, facilitating histone acetylation. Dysregulation of KAT6A has been implicated in developmental syndromes and the onset of acute myeloid leukemia (AML). Previous work suggests that KAT6A is recruited to its genomic targets by a combinatorial function of histone binding PHD fingers, transcription factors and chromatin binding interaction partners. Here, we demonstrate that a winged helix (WH) domain at the very N-terminus of KAT6A specifically interacts with unmethylated CpG motifs. This DNA binding function leads to the association of KAT6A with unmethylated CpG islands (CGIs) genome-wide. Mutation of the essential amino acids for DNA binding completely abrogates the enrichment of KAT6A at CGIs. In contrast, deletion of a second WH domain or the histone tail binding PHD fingers only subtly influences the binding of KAT6A to CGIs. Overexpression of a KAT6A WH1 mutant has a dominant negative effect on H3K9 histone acetylation, which is comparable to the effects upon overexpression of a KAT6A HAT domain mutant. Taken together, our work revealed a previously unrecognized chromatin recruitment mechanism of KAT6A, offering a new perspective on the role of KAT6A in gene regulation and human diseases.


Subject(s)
Chromatin , Histone Acetyltransferases , Histones , Humans , Chromatin/genetics , CpG Islands/genetics , DNA , Histone Acetyltransferases/metabolism , Histones/metabolism , Acetylation
3.
Proc Natl Acad Sci U S A ; 119(36): e2120680119, 2022 09 06.
Article in English | MEDLINE | ID: mdl-35998224

ABSTRACT

The systemic immune response to viral infection is shaped by master transcription factors, such as NF-κB, STAT1, or PU.1. Although long noncoding RNAs (lncRNAs) have been suggested as important regulators of transcription factor activity, their contributions to the systemic immunopathologies observed during SARS-CoV-2 infection have remained unknown. Here, we employed a targeted single-cell RNA sequencing approach to reveal lncRNAs differentially expressed in blood leukocytes during severe COVID-19. Our results uncover the lncRNA PIRAT (PU.1-induced regulator of alarmin transcription) as a major PU.1 feedback-regulator in monocytes, governing the production of the alarmins S100A8/A9, key drivers of COVID-19 pathogenesis. Knockout and transgene expression, combined with chromatin-occupancy profiling, characterized PIRAT as a nuclear decoy RNA, keeping PU.1 from binding to alarmin promoters and promoting its binding to pseudogenes in naïve monocytes. NF-κB-dependent PIRAT down-regulation during COVID-19 consequently releases a transcriptional brake, fueling alarmin production. Alarmin expression is additionally enhanced by the up-regulation of the lncRNA LUCAT1, which promotes NF-κB-dependent gene expression at the expense of targets of the JAK-STAT pathway. Our results suggest a major role of nuclear noncoding RNA networks in systemic antiviral responses to SARS-CoV-2 in humans.


Subject(s)
COVID-19 , Gene Expression Regulation , Monocytes , RNA, Long Noncoding , SARS-CoV-2 , Alarmins/genetics , COVID-19/genetics , COVID-19/immunology , Humans , Janus Kinases/genetics , Monocytes/immunology , NF-kappa B/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA-Seq , SARS-CoV-2/immunology , STAT Transcription Factors/genetics , Signal Transduction/genetics , Single-Cell Analysis
4.
Gut ; 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38834297

ABSTRACT

OBJECTIVE: Highly malignant pancreatic ductal adenocarcinoma (PDAC) is characterised by an abundant immunosuppressive and fibrotic tumour microenvironment (TME). Future therapeutic attempts will therefore demand the targeting of tumours and stromal compartments in order to be effective. Here we investigate whether dual specificity and tyrosine phosphorylation-regulated kinase 1B (DYRK1B) fulfil these criteria and represent a promising anticancer target in PDAC. DESIGN: We used transplantation and autochthonous mouse models of PDAC with either genetic Dyrk1b loss or pharmacological DYRK1B inhibition, respectively. Mechanistic interactions between tumour cells and macrophages were studied in direct or indirect co-culture experiments. Histological analyses used tissue microarrays from patients with PDAC. Additional methodological approaches included bulk mRNA sequencing (transcriptomics) and proteomics (secretomics). RESULTS: We found that DYRK1B is mainly expressed by pancreatic epithelial cancer cells and modulates the influx and activity of TME-associated macrophages through effects on the cancer cells themselves as well as through the tumour secretome. Mechanistically, genetic ablation or pharmacological inhibition of DYRK1B strongly attracts tumoricidal macrophages and, in addition, downregulates the phagocytosis checkpoint and 'don't eat me' signal CD24 on cancer cells, resulting in enhanced tumour cell phagocytosis. Consequently, tumour cells lacking DYRK1B hardly expand in transplantation experiments, despite their rapid growth in culture. Furthermore, combining a small-molecule DYRK1B-directed therapy with mammalian target of rapamycin inhibition and conventional chemotherapy stalls the growth of established tumours and results in a significant extension of life span in a highly aggressive autochthonous model of PDAC. CONCLUSION: In light of DYRK inhibitors currently entering clinical phase testing, our data thus provide a novel and clinically translatable approach targeting both the cancer cell compartment and its microenvironment.

5.
EMBO J ; 39(1): e101533, 2020 01 02.
Article in English | MEDLINE | ID: mdl-31701553

ABSTRACT

How cytokine-driven changes in chromatin topology are converted into gene regulatory circuits during inflammation still remains unclear. Here, we show that interleukin (IL)-1α induces acute and widespread changes in chromatin accessibility via the TAK1 kinase and NF-κB at regions that are highly enriched for inflammatory disease-relevant SNPs. Two enhancers in the extended chemokine locus on human chromosome 4 regulate the IL-1α-inducible IL8 and CXCL1-3 genes. Both enhancers engage in dynamic spatial interactions with gene promoters in an IL-1α/TAK1-inducible manner. Microdeletions of p65-binding sites in either of the two enhancers impair NF-κB recruitment, suppress activation and biallelic transcription of the IL8/CXCL2 genes, and reshuffle higher-order chromatin interactions as judged by i4C interactome profiles. Notably, these findings support a dominant role of the IL8 "master" enhancer in the regulation of sustained IL-1α signaling, as well as for IL-8 and IL-6 secretion. CRISPR-guided transactivation of the IL8 locus or cross-TAD regulation by TNFα-responsive enhancers in a different model locus supports the existence of complex enhancer hierarchies in response to cytokine stimulation that prime and orchestrate proinflammatory chromatin responses downstream of NF-κB.


Subject(s)
Chromatin/metabolism , Enhancer Elements, Genetic/genetics , Gene Expression Regulation/drug effects , Inflammation Mediators/metabolism , Interleukin-1alpha/pharmacology , MAP Kinase Kinase Kinases/metabolism , NF-kappa B/metabolism , Binding Sites , Cells, Cultured , Chemokines/metabolism , Chromatin/chemistry , Chromatin/genetics , HeLa Cells , Humans , MAP Kinase Kinase Kinases/genetics , NF-kappa B/genetics , Signal Transduction , Tumor Necrosis Factor-alpha/pharmacology
6.
EMBO Rep ; 23(12): e54685, 2022 12 06.
Article in English | MEDLINE | ID: mdl-36215678

ABSTRACT

Increased lactate levels in the tissue microenvironment are a well-known feature of chronic inflammation. However, the role of lactate in regulating T cell function remains controversial. Here, we demonstrate that extracellular lactate predominantly induces deregulation of the Th17-specific gene expression program by modulating the metabolic and epigenetic status of Th17 cells. Following lactate treatment, Th17 cells significantly reduced their IL-17A production and upregulated Foxp3 expression through ROS-driven IL-2 secretion. Moreover, we observed increased levels of genome-wide histone H3K18 lactylation, a recently described marker for active chromatin in macrophages, in lactate-treated Th17 cells. In addition, we show that high lactate concentrations suppress Th17 pathogenicity during intestinal inflammation in mice. These results indicate that lactate is capable of reprogramming pro-inflammatory T cell phenotypes into regulatory T cells.


Subject(s)
Lactic Acid , Th17 Cells , Animals , Mice , Epigenomics
7.
Nucleic Acids Res ; 50(14): 7925-7937, 2022 08 12.
Article in English | MEDLINE | ID: mdl-35848919

ABSTRACT

Signal transduction pathways often involve transcription factors that promote activation of defined target gene sets. The transcription factor RBPJ is the central player in Notch signaling and either forms an activator complex with the Notch intracellular domain (NICD) or a repressor complex with corepressors like KYOT2/FHL1. The balance between these two antagonizing RBPJ-complexes depends on the activation state of the Notch receptor regulated by cell-to-cell interaction, ligand binding and proteolytic cleavage events. Here, we depleted RBPJ in mature T-cells lacking active Notch signaling and performed RNA-Seq, ChIP-Seq and ATAC-seq analyses. RBPJ depletion leads to upregulation of many Notch target genes. Ectopic expression of NICD1 activates several Notch target genes and enhances RBPJ occupancy. Based on gene expression changes and RBPJ occupancy we define four different clusters, either RBPJ- and/or Notch-regulated genes. Importantly, we identify early (Hes1 and Hey1) and late Notch-responsive genes (IL2ra). Similarly, to RBPJ depletion, interfering with transcriptional repression by squelching with cofactor KYOT2/FHL1, leads to upregulation of Notch target genes. Taken together, RBPJ is not only an essential part of the Notch co-activator complex but also functions as a repressor in a Notch-independent manner.


Subject(s)
Immunoglobulin J Recombination Signal Sequence-Binding Protein , Receptors, Notch , T-Lymphocytes , Gene Expression Regulation , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction , T-Lymphocytes/metabolism
8.
PLoS Genet ; 17(2): e1009318, 2021 02.
Article in English | MEDLINE | ID: mdl-33600407

ABSTRACT

The generation of lineage-specific gene expression programmes that alter proliferation capacity, metabolic profile and cell type-specific functions during differentiation from multipotent stem cells to specialised cell types is crucial for development. During differentiation gene expression programmes are dynamically modulated by a complex interplay between sequence-specific transcription factors, associated cofactors and epigenetic regulators. Here, we study U-shaped (Ush), a multi-zinc finger protein that maintains the multipotency of stem cell-like hemocyte progenitors during Drosophila hematopoiesis. Using genomewide approaches we reveal that Ush binds to promoters and enhancers and that it controls the expression of three gene classes that encode proteins relevant to stem cell-like functions and differentiation: cell cycle regulators, key metabolic enzymes and proteins conferring specific functions of differentiated hemocytes. We employ complementary biochemical approaches to characterise the molecular mechanisms of Ush-mediated gene regulation. We uncover distinct Ush isoforms one of which binds the Nucleosome Remodeling and Deacetylation (NuRD) complex using an evolutionary conserved peptide motif. Remarkably, the Ush/NuRD complex specifically contributes to the repression of lineage-specific genes but does not impact the expression of cell cycle regulators or metabolic genes. This reveals a mechanism that enables specific and concerted modulation of functionally related portions of a wider gene expression programme. Finally, we use genetic assays to demonstrate that Ush and NuRD regulate enhancer activity during hemocyte differentiation in vivo and that both cooperate to suppress the differentiation of lamellocytes, a highly specialised blood cell type. Our findings reveal that Ush coordinates proliferation, metabolism and cell type-specific activities by isoform-specific cooperation with an epigenetic regulator.


Subject(s)
Cell Cycle/genetics , Drosophila Proteins/metabolism , Fatty Acids/metabolism , Gene Expression Regulation, Developmental/genetics , Hematopoiesis/genetics , Hemocytes/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Transcription Factors/metabolism , Amino Acid Motifs , Animals , Cell Line , Cell Proliferation/genetics , Cell Survival/genetics , Chromatin Immunoprecipitation Sequencing , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Enhancer Elements, Genetic , Gene Ontology , Promoter Regions, Genetic , Protein Isoforms , RNA Interference , RNA-Seq , Transcription Factors/genetics
9.
EMBO J ; 38(20): e102096, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31483066

ABSTRACT

Engineered p53 mutant mice are valuable tools for delineating p53 functions in tumor suppression and cancer therapy. Here, we have introduced the R178E mutation into the Trp53 gene of mice to specifically ablate the cooperative nature of p53 DNA binding. Trp53R178E mice show no detectable target gene regulation and, at first sight, are largely indistinguishable from Trp53-/- mice. Surprisingly, stabilization of p53R178E in Mdm2-/- mice nevertheless triggers extensive apoptosis, indicative of residual wild-type activities. Although this apoptotic activity suffices to trigger lethality of Trp53R178E ;Mdm2-/- embryos, it proves insufficient for suppression of spontaneous and oncogene-driven tumorigenesis. Trp53R178E mice develop tumors indistinguishably from Trp53-/- mice and tumors retain and even stabilize the p53R178E protein, further attesting to the lack of significant tumor suppressor activity. However, Trp53R178E tumors exhibit remarkably better chemotherapy responses than Trp53-/- ones, resulting in enhanced eradication of p53-mutated tumor cells. Together, this provides genetic proof-of-principle evidence that a p53 mutant can be highly tumorigenic and yet retain apoptotic activity which provides a survival benefit in the context of cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Leukemia, Myeloid, Acute/prevention & control , Lymphoma/prevention & control , Mutation , Proto-Oncogene Proteins c-mdm2/physiology , Tumor Suppressor Protein p53/physiology , Animals , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Cycle , Disease Models, Animal , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Lymphoma/genetics , Lymphoma/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Tumor Cells, Cultured
10.
Mol Cancer ; 21(1): 191, 2022 10 03.
Article in English | MEDLINE | ID: mdl-36192757

ABSTRACT

BACKGROUND: In vivo gene editing of somatic cells with CRISPR nucleases has facilitated the generation of autochthonous mouse tumors, which are initiated by genetic alterations relevant to the human disease and progress along a natural timeline as in patients. However, the long and variable, orthotopic tumor growth in inner organs requires sophisticated, time-consuming and resource-intensive imaging for longitudinal disease monitoring and impedes the use of autochthonous tumor models for preclinical studies. METHODS: To facilitate a more widespread use, we have generated a reporter mouse that expresses a Cre-inducible luciferase from Gaussia princeps (GLuc), which is secreted by cells in an energy-consuming process and can be measured quantitatively in the blood as a marker for the viable tumor load. In addition, we have developed a flexible, complementary toolkit to rapidly assemble recombinant adenoviruses (AVs) for delivering Cre recombinase together with CRISPR nucleases targeting cancer driver genes. RESULTS: We demonstrate that intratracheal infection of GLuc reporter mice with CRISPR-AVs efficiently induces lung tumors driven by mutations in the targeted cancer genes and simultaneously activates the GLuc transgene, resulting in GLuc secretion into the blood by the growing tumor. GLuc blood levels are easily and robustly quantified in small-volume blood samples with inexpensive equipment, enable tumor detection already several months before the humane study endpoint and precisely mirror the kinetics of tumor development specified by the inducing gene combination. CONCLUSIONS: Our study establishes blood-based GLuc monitoring as an inexpensive, rapid, high-throughput and animal-friendly method to longitudinally monitor autochthonous tumor growth in preclinical studies.


Subject(s)
Copepoda , Lung Neoplasms , Animals , Clustered Regularly Interspaced Short Palindromic Repeats , Copepoda/genetics , Copepoda/metabolism , Gene Editing , Genes, Reporter , Humans , Luciferases/genetics , Luciferases/metabolism , Lung Neoplasms/genetics , Mice
11.
Nucleic Acids Res ; 48(7): 3496-3512, 2020 04 17.
Article in English | MEDLINE | ID: mdl-32107550

ABSTRACT

Aberrant Notch signaling plays a pivotal role in T-cell acute lymphoblastic leukemia (T-ALL) and chronic lymphocytic leukemia (CLL). Amplitude and duration of the Notch response is controlled by ubiquitin-dependent proteasomal degradation of the Notch1 intracellular domain (NICD1), a hallmark of the leukemogenic process. Here, we show that HDAC3 controls NICD1 acetylation levels directly affecting NICD1 protein stability. Either genetic loss-of-function of HDAC3 or nanomolar concentrations of HDAC inhibitor apicidin lead to downregulation of Notch target genes accompanied by a local reduction of histone acetylation. Importantly, an HDAC3-insensitive NICD1 mutant is more stable but biologically less active. Collectively, these data show a new HDAC3- and acetylation-dependent mechanism that may be exploited to treat Notch1-dependent leukemias.


Subject(s)
Histone Deacetylases/metabolism , Leukemia/metabolism , Receptor, Notch1/metabolism , Signal Transduction , Animals , Cell Line , Cell Line, Tumor , Histone Deacetylase Inhibitors/pharmacology , Humans , Leukemia/enzymology , Lysine/metabolism , Mice , Mutation , Peptides, Cyclic/pharmacology , Protein Stability , Receptor, Notch1/chemistry , Receptor, Notch1/genetics
12.
Proc Natl Acad Sci U S A ; 116(44): 22288-22293, 2019 10 29.
Article in English | MEDLINE | ID: mdl-31611375

ABSTRACT

Cancer development is driven by activated oncogenes and loss of tumor suppressors. While oncogene inhibitors have entered routine clinical practice, tumor suppressor reactivation therapy remains to be established. For the most frequently inactivated tumor suppressor p53, genetic mouse models have demonstrated regression of p53-null tumors upon p53 reactivation. While this was shown in tumor models driven by p53 loss as the initiating lesion, many human tumors initially develop in the presence of wild-type p53, acquire aberrations in the p53 pathway to bypass p53-mediated tumor suppression, and inactivate p53 itself only at later stages during metastatic progression or therapy. To explore the efficacy of p53 reactivation in this scenario, we used a reversibly switchable p53 (p53ERTAM) mouse allele to generate Eµ-Myc-driven lymphomas in the presence of active p53 and, after full lymphoma establishment, switched off p53 to model late-stage p53 inactivation. Although these lymphomas had evolved in the presence of active p53, later loss and subsequent p53 reactivation surprisingly activated p53 target genes triggering massive apoptosis, tumor regression, and long-term cure of the majority of animals. Mechanistically, the reactivation response was dependent on Cdkn2a/p19Arf, which is commonly silenced in p53 wild-type lymphomas, but became reexpressed upon late-stage p53 inactivation. Likewise, human p53 wild-type tumor cells with CRISPR-engineered switchable p53ERTAM alleles responded to p53 reactivation when CDKN2A/p14ARF function was restored or mimicked with Mdm2 inhibitors. Together, these experiments provide genetic proof of concept that tumors can respond, in an ARF-dependent manner, to p53 reactivation even if p53 inactivation has occurred late during tumor evolution.


Subject(s)
Carcinogenesis/genetics , Genetic Therapy/methods , Lymphoma/genetics , Tumor Suppressor Protein p53/genetics , 3T3 Cells , Animals , Apoptosis , Cyclin-Dependent Kinase Inhibitor p16/metabolism , HCT116 Cells , Humans , Lymphoma/therapy , Mice , Tumor Suppressor Protein p53/metabolism
13.
J Biol Chem ; 295(33): 11388-11407, 2020 08 14.
Article in English | MEDLINE | ID: mdl-32303635

ABSTRACT

Cleavage of influenza virus hemagglutinin (HA) by host proteases is essential for virus infectivity. HA of most influenza A and B (IAV/IBV) viruses is cleaved at a monobasic motif by trypsin-like proteases. Previous studies have reported that transmembrane serine protease 2 (TMPRSS2) is essential for activation of H7N9 and H1N1pdm IAV in mice but that H3N2 IAV and IBV activation is independent of TMPRSS2 and carried out by as-yet-undetermined protease(s). Here, to identify additional H3 IAV- and IBV-activating proteases, we used RNA-Seq to investigate the protease repertoire of murine lower airway tissues, primary type II alveolar epithelial cells (AECIIs), and the mouse lung cell line MLE-15. Among 13 candidates identified, TMPRSS4, TMPRSS13, hepsin, and prostasin activated H3 and IBV HA in vitro IBV activation and replication was reduced in AECIIs from Tmprss2/Tmprss4-deficient mice compared with WT or Tmprss2-deficient mice, indicating that murine TMPRSS4 is involved in IBV activation. Multicycle replication of H3N2 IAV and IBV in AECIIs of Tmprss2/Tmprss4-deficient mice varied in sensitivity to protease inhibitors, indicating that different, but overlapping, sets of murine proteases facilitate H3 and IBV HA cleavages. Interestingly, human hepsin and prostasin orthologs did not activate H3, but they did activate IBV HA in vitro Our results indicate that TMPRSS4 is an IBV-activating protease in murine AECIIs and suggest that TMPRSS13, hepsin, and prostasin cleave H3 and IBV HA in mice. They further show that hepsin and prostasin orthologs might contribute to the differences observed in TMPRSS2-independent activation of H3 in murine and human airways.


Subject(s)
Influenza A Virus, H3N2 Subtype/physiology , Influenza B virus/physiology , Influenza, Human/enzymology , Orthomyxoviridae Infections/enzymology , Peptide Hydrolases/metabolism , Virus Activation , Animals , Cell Line , Dogs , Enzyme Activation/drug effects , Gene Expression Profiling , HEK293 Cells , Host-Pathogen Interactions/drug effects , Humans , Influenza A Virus, H3N2 Subtype/drug effects , Influenza B virus/drug effects , Influenza, Human/drug therapy , Influenza, Human/genetics , Influenza, Human/virology , Lung/enzymology , Lung/metabolism , Lung/virology , Madin Darby Canine Kidney Cells , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/virology , Peptide Hydrolases/genetics , Protease Inhibitors/pharmacology , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Virus Activation/drug effects
14.
RNA Biol ; 18(5): 604-618, 2021 05.
Article in English | MEDLINE | ID: mdl-33622174

ABSTRACT

A persisting obstacle in human immunology is that blood-derived leukocytes are notoriously difficult to manipulate at the RNA level. Therefore, our knowledge about immune-regulatory RNA-networks is largely based on tumour cell-line and rodent knockout models, which do not fully mimic human leukocyte biology. Here, we exploit straightforward cell penetrating peptide (CPP) chemistry to enable efficient loss-of-function phenotyping of regulatory RNAs in primary human blood-derived cells. The classical CPP octaarginine (R8) enabled antisense peptide-nucleic-acid (PNA) oligomer delivery into nearly 100% of human blood-derived macrophages without apparent cytotoxicity even up to micromolar concentrations. In a proof-of-principle experiment, we successfully de-repressed the global microRNA-155 regulome in primary human macrophages using a PNA-R8 oligomer, which phenocopies a CRISPR-Cas9 induced gene knockout. Interestingly, although it is often believed that fairly high concentrations (µM) are needed to achieve antisense activity, our PNA-R8 was effective at 200 nM. RNA-seq characterized microRNA-155 as a broad-acting riboregulator, feedback restraining a late myeloid differentiation-induced pro-inflammatory network, comprising MyD88-signalling and ubiquitin-proteasome components. Our results highlight the important role of the microRNA machinery in fine-control of blood-derived human phagocyte immunity and open the door for further studies on regulatory RNAs in difficult-to-transfect primary human immune cells.


Subject(s)
Inflammation/genetics , MicroRNAs/physiology , Oligonucleotides, Antisense/pharmacology , Phagocytes/drug effects , Cells, Cultured , Gene Knockdown Techniques , Humans , Inflammation/metabolism , MicroRNAs/genetics , Myeloid Cells/drug effects , Myeloid Cells/metabolism , Myeloid Cells/physiology , Phagocytes/immunology , Phagocytes/metabolism , Primary Cell Culture , RNA Interference/drug effects , Transcriptome/drug effects , U937 Cells
15.
J Immunol ; 202(4): 1099-1111, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30651342

ABSTRACT

RNA editing by adenosine deaminases acting on dsRNA (ADAR) has become of increasing medical relevance, particularly because aberrant ADAR1 activity has been associated with autoimmunity and malignancies. However, the role of ADAR1 in dendritic cells (DC), representing critical professional APCs, is unknown. We have established conditional murine CD11c Cre-mediated ADAR1 gene ablation, which did not induce general apoptosis in CD11c+ cells but instead manifests in cell type-specific effects in DC subpopulations. Bone marrow-derived DC subset analysis revealed an incapacity to differentiate CD103 DC+ in both bulk bone marrow and purified pre-DC lineage progenitor assays. ADAR1 deficiency further resulted in a preferential systemic loss of CD8+/CD103+ DCs, revealing critical dependency on ADAR1, whereas other DC subpopulations were moderately affected or unaffected. Additionally, alveolar macrophages were depleted and dysfunctional, resembling pulmonary alveolar proteinosis. These results reveal an unrecognized role of ADAR1 in DC subset homeostasis and unveils the cell type-specific effects of RNA editing.


Subject(s)
Adenosine Deaminase/metabolism , Dendritic Cells/immunology , Homeostasis/immunology , Macrophages, Alveolar/immunology , Animals , Cell Proliferation , Dendritic Cells/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA Editing , T-Lymphocytes/cytology , T-Lymphocytes/immunology
16.
Nucleic Acids Res ; 47(22): 11649-11666, 2019 12 16.
Article in English | MEDLINE | ID: mdl-31701127

ABSTRACT

CoREST has been identified as a subunit of several protein complexes that generate transcriptionally repressive chromatin structures during development. However, a comprehensive analysis of the CoREST interactome has not been carried out. We use proteomic approaches to define the interactomes of two dCoREST isoforms, dCoREST-L and dCoREST-M, in Drosophila. We identify three distinct histone deacetylase complexes built around a common dCoREST/dRPD3 core: A dLSD1/dCoREST complex, the LINT complex and a dG9a/dCoREST complex. The latter two complexes can incorporate both dCoREST isoforms. By contrast, the dLSD1/dCoREST complex exclusively assembles with the dCoREST-L isoform. Genome-wide studies show that the three dCoREST complexes associate with chromatin predominantly at promoters. Transcriptome analyses in S2 cells and testes reveal that different cell lineages utilize distinct dCoREST complexes to maintain cell-type-specific gene expression programmes: In macrophage-like S2 cells, LINT represses germ line-related genes whereas other dCoREST complexes are largely dispensable. By contrast, in testes, the dLSD1/dCoREST complex prevents transcription of germ line-inappropriate genes and is essential for spermatogenesis and fertility, whereas depletion of other dCoREST complexes has no effect. Our study uncovers three distinct dCoREST complexes that function in a lineage-restricted fashion to repress specific sets of genes thereby maintaining cell-type-specific gene expression programmes.


Subject(s)
Chromatin/metabolism , Co-Repressor Proteins/metabolism , Drosophila Proteins/metabolism , Gene Expression Regulation/genetics , Histone Deacetylases/metabolism , Oxidoreductases, N-Demethylating/metabolism , Animals , Cell Line , Drosophila melanogaster/embryology , Epigenesis, Genetic/genetics , Gene Expression Profiling , Histone Deacetylases/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Protein Isoforms/genetics , Transcription Factors/metabolism , Transcriptome/genetics
17.
PLoS Genet ; 14(1): e1007193, 2018 01.
Article in English | MEDLINE | ID: mdl-29381691

ABSTRACT

Diverse Polycomb repressive complexes 1 (PRC1) play essential roles in gene regulation, differentiation and development. Six major groups of PRC1 complexes that differ in their subunit composition have been identified in mammals. How the different PRC1 complexes are recruited to specific genomic sites is poorly understood. The Polycomb Ring finger protein PCGF6, the transcription factors MGA and E2F6, and the histone-binding protein L3MBTL2 are specific components of the non-canonical PRC1.6 complex. In this study, we have investigated their role in genomic targeting of PRC1.6. ChIP-seq analysis revealed colocalization of MGA, L3MBTL2, E2F6 and PCGF6 genome-wide. Ablation of MGA in a human cell line by CRISPR/Cas resulted in complete loss of PRC1.6 binding. Rescue experiments revealed that MGA recruits PRC1.6 to specific loci both by DNA binding-dependent and by DNA binding-independent mechanisms. Depletion of L3MBTL2 and E2F6 but not of PCGF6 resulted in differential, locus-specific loss of PRC1.6 binding illustrating that different subunits mediate PRC1.6 loading to distinct sets of promoters. Mga, L3mbtl2 and Pcgf6 colocalize also in mouse embryonic stem cells, where PRC1.6 has been linked to repression of germ cell-related genes. Our findings unveil strikingly different genomic recruitment mechanisms of the non-canonical PRC1.6 complex, which specify its cell type- and context-specific regulatory functions.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , DNA/metabolism , E2F6 Transcription Factor/physiology , Nuclear Proteins/physiology , Polycomb Repressive Complex 1/metabolism , Transcription Factors/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cells, Cultured , E2F6 Transcription Factor/genetics , Gene Expression Regulation , Gene Knockdown Techniques , HEK293 Cells , Human Embryonic Stem Cells/physiology , Humans , Mice , Mouse Embryonic Stem Cells/physiology , Nuclear Proteins/genetics , Protein Binding/genetics , Transcription Factors/genetics
18.
Int J Mol Sci ; 22(4)2021 Feb 07.
Article in English | MEDLINE | ID: mdl-33562308

ABSTRACT

CTRP-3 (C1q/TNF-related protein-3) is an adipokine with endocrine and immunological function. The impact of adipocyte CTRP-3 production on systemic CTRP-3 concentrations and on adipocyte biology is unknown. A murine model of adipocyte CTRP-3 knockout (KO) was established (via the Cre/loxP system). Serum adipokine levels were quantified by ELISA and adipose tissue (AT) gene expression by real-time PCR. Preadipocytes were isolated from AT and differentiated into adipocytes. Comparative transcriptome analysis was applied in adipocytes and liver tissue. Body weight and AT mass were reduced in CTRP-3 KO mice together with decreased serum leptin. In primary cells from visceral AT of KO mice, expression of adiponectin, progranulin, and resistin was induced, while peroxisome proliferator activated receptor γ (PPARγ) was decreased. M1/M2 macrophage polarization markers were shifted to a more anti-inflammatory phenotype. CTRP-3 expression in AT did not contribute to serum concentrations. AT and liver morphology remained unaffected by CTRP-3 KO. Myelin transcription factor 1-like (Myt1l) was identified as a highly upregulated gene. In conclusion, adipocyte CTRP-3 has a role in adipogenesis and AT weight gain whereas adipocyte differentiation is not impaired by CTRP-3 deficiency. Since no effects on circulating CTRP-3 levels were observed, the impact of adipocyte CTRP-3 KO is limited to adipose tissue. Modified AT gene expression indicates a rather anti-inflammatory phenotype.


Subject(s)
Adipocytes/cytology , Adipogenesis , Adipokines/metabolism , Adipose Tissue, White/cytology , Gene Expression Regulation , Adipocytes/metabolism , Adipokines/genetics , Adipokines/physiology , Adipose Tissue, White/metabolism , Animals , Mice , Mice, Inbred C57BL , Mice, Transgenic , Transcriptome
19.
Int J Mol Sci ; 22(2)2021 Jan 12.
Article in English | MEDLINE | ID: mdl-33445503

ABSTRACT

Histone variants differ in amino acid sequence, expression timing and genomic localization sites from canonical histones and convey unique functions to eukaryotic cells. Their tightly controlled spatial and temporal deposition into specific chromatin regions is accomplished by dedicated chaperone and/or remodeling complexes. While quantitatively identifying the chaperone complexes of many human H2A variants by using mass spectrometry, we also found additional members of the known H2A.Z chaperone complexes p400/TIP60/NuA4 and SRCAP. We discovered JAZF1, a nuclear/nucleolar protein, as a member of a p400 sub-complex containing MBTD1 but excluding ANP32E. Depletion of JAZF1 results in transcriptome changes that affect, among other pathways, ribosome biogenesis. To identify the underlying molecular mechanism contributing to JAZF1's function in gene regulation, we performed genome-wide ChIP-seq analyses. Interestingly, depletion of JAZF1 leads to reduced H2A.Z acetylation levels at > 1000 regulatory sites without affecting H2A.Z nucleosome positioning. Since JAZF1 associates with the histone acetyltransferase TIP60, whose depletion causes a correlated H2A.Z deacetylation of several JAZF1-targeted enhancer regions, we speculate that JAZF1 acts as chromatin modulator by recruiting TIP60's enzymatic activity. Altogether, this study uncovers JAZF1 as a member of a TIP60-containing p400 chaperone complex orchestrating H2A.Z acetylation at regulatory regions controlling the expression of genes, many of which are involved in ribosome biogenesis.


Subject(s)
Co-Repressor Proteins/metabolism , DNA-Binding Proteins/metabolism , Histones/metabolism , Regulatory Sequences, Nucleic Acid , Acetylation , Cell Line , Chromatin Assembly and Disassembly , Computational Biology/methods , DNA Helicases/metabolism , Enhancer Elements, Genetic , Gene Expression Regulation , Genomics/methods , Humans , Introns , Lysine Acetyltransferase 5/metabolism , Molecular Chaperones/metabolism , Multiprotein Complexes , Protein Binding , Ribosomes , Transcription Factors/metabolism
20.
Eur Respir J ; 56(5)2020 Nov.
Article in English | MEDLINE | ID: mdl-32616594

ABSTRACT

While severe coronavirus infections, including Middle East respiratory syndrome coronavirus (MERS-CoV), cause lung injury with high mortality rates, protective treatment strategies are not approved for clinical use.We elucidated the molecular mechanisms by which the cyclophilin inhibitors cyclosporin A (CsA) and alisporivir (ALV) restrict MERS-CoV to validate their suitability as readily available therapy in MERS-CoV infection.Calu-3 cells and primary human alveolar epithelial cells (hAECs) were infected with MERS-CoV and treated with CsA or ALV or inhibitors targeting cyclophilin inhibitor-regulated molecules including calcineurin, nuclear factor of activated T-cells (NFATs) or mitogen-activated protein kinases. Novel CsA-induced pathways were identified by RNA sequencing and manipulated by gene knockdown or neutralising antibodies. Viral replication was quantified by quantitative real-time PCR and 50% tissue culture infective dose. Data were validated in a murine MERS-CoV infection model.Both CsA and ALV reduced MERS-CoV titres and viral RNA replication in Calu-3 cells and hAECs, improving epithelial integrity. While neither calcineurin nor NFAT inhibition reduced MERS-CoV propagation, blockade of c-Jun N-terminal kinase diminished infectious viral particle release but not RNA accumulation. Importantly, CsA induced interferon regulatory factor 1 (IRF1), a pronounced type III interferon (IFNλ) response and expression of antiviral genes. Downregulation of IRF1 or IFNλ increased MERS-CoV propagation in the presence of CsA. Importantly, oral application of CsA reduced MERS-CoV replication in vivo, correlating with elevated lung IFNλ levels and improved outcome.We provide evidence that cyclophilin inhibitors efficiently decrease MERS-CoV replication in vitro and in vivo via upregulation of inflammatory antiviral cell responses, in particular IFNλ. CsA might therefore represent a promising candidate for treating MERS-CoV infection.


Subject(s)
Coronavirus Infections/prevention & control , Cyclophilins/antagonists & inhibitors , Cyclosporine/pharmacology , Interferons/metabolism , Middle East Respiratory Syndrome Coronavirus/drug effects , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/virology , Animals , Calcineurin Inhibitors/pharmacology , Cell Culture Techniques , Coronavirus Infections/metabolism , Disease Models, Animal , Humans , Interferon Regulatory Factor-1/drug effects , Interferon Regulatory Factor-1/metabolism , Interferons/drug effects , Mice , Middle East Respiratory Syndrome Coronavirus/physiology , Virus Replication/drug effects , Interferon Lambda
SELECTION OF CITATIONS
SEARCH DETAIL