Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 229
Filter
Add more filters

Publication year range
1.
Immunity ; 55(12): 2271-2284.e7, 2022 12 13.
Article in English | MEDLINE | ID: mdl-36384135

ABSTRACT

The NLRP3 inflammasome plays a central role in antimicrobial defense as well as in the context of sterile inflammatory conditions. NLRP3 activity is governed by two independent signals: the first signal primes NLRP3, rendering it responsive to the second signal, which then triggers inflammasome formation. Our understanding of how NLRP3 priming contributes to inflammasome activation remains limited. Here, we show that IKKß, a kinase activated during priming, induces recruitment of NLRP3 to phosphatidylinositol-4-phosphate (PI4P), a phospholipid enriched on the trans-Golgi network. NEK7, a mitotic spindle kinase that had previously been thought to be indispensable for NLRP3 activation, was redundant for inflammasome formation when IKKß recruited NLRP3 to PI4P. Studying iPSC-derived human macrophages revealed that the IKKß-mediated NEK7-independent pathway constitutes the predominant NLRP3 priming mechanism in human myeloid cells. Our results suggest that PI4P binding represents a primed state into which NLRP3 is brought by IKKß activity.


Subject(s)
Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Humans , I-kappa B Kinase , Inflammasomes/metabolism , Mice, Inbred C57BL , NIMA-Related Kinases/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Protein Serine-Threonine Kinases/metabolism , trans-Golgi Network/metabolism
2.
Cell ; 152(3): 584-98, 2013 Jan 31.
Article in English | MEDLINE | ID: mdl-23374351

ABSTRACT

Eukaryotic cells have a layer of heterochromatin at the nuclear periphery. To investigate mechanisms regulating chromatin distribution, we analyzed heterochromatin organization in different tissues and species, including mice with mutations in the lamin B receptor (Lbr) and lamin A (Lmna) genes that encode nuclear envelope (NE) proteins. We identified LBR- and lamin-A/C-dependent mechanisms tethering heterochromatin to the NE. The two tethers are sequentially used during cellular differentiation and development: first the LBR- and then the lamin-A/C-dependent tether. The absence of both LBR and lamin A/C leads to loss of peripheral heterochromatin and an inverted architecture with heterochromatin localizing to the nuclear interior. Myoblast transcriptome analyses indicated that selective disruption of the LBR- or lamin-A-dependent heterochromatin tethers have opposite effects on muscle gene expression, either increasing or decreasing, respectively. These results show how changes in NE composition contribute to regulating heterochromatin positioning, gene expression, and cellular differentiation during development.


Subject(s)
Heterochromatin/metabolism , Lamin Type A/metabolism , Muscle Development , Myoblasts/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Gene Expression Profiling , Mice , Myoblasts/cytology , Nuclear Envelope/metabolism , Lamin B Receptor
3.
Cell ; 152(5): 1146-59, 2013 Feb 28.
Article in English | MEDLINE | ID: mdl-23434322

ABSTRACT

Tet proteins oxidize 5-methylcytosine (mC) to generate 5-hydroxymethyl (hmC), 5-formyl (fC), and 5-carboxylcytosine (caC). The exact function of these oxidative cytosine bases remains elusive. We applied quantitative mass-spectrometry-based proteomics to identify readers for mC and hmC in mouse embryonic stem cells (mESC), neuronal progenitor cells (NPC), and adult mouse brain tissue. Readers for these modifications are only partially overlapping, and some readers, such as Rfx proteins, display strong specificity. Interactions are dynamic during differentiation, as for example evidenced by the mESC-specific binding of Klf4 to mC and the NPC-specific binding of Uhrf2 to hmC, suggesting specific biological roles for mC and hmC. Oxidized derivatives of mC recruit distinct transcription regulators as well as a large number of DNA repair proteins in mouse ES cells, implicating the DNA damage response as a major player in active DNA demethylation.


Subject(s)
5-Methylcytosine/analysis , Cytosine/analogs & derivatives , DNA Methylation , 5-Methylcytosine/metabolism , Animals , Brain/cytology , Brain/metabolism , Cytosine/analysis , Cytosine/metabolism , DNA Glycosylases/metabolism , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/metabolism , Kruppel-Like Factor 4 , Mass Spectrometry , Mice , Oxidation-Reduction , Proto-Oncogene Proteins/metabolism , Regulatory Factor X Transcription Factors , Stem Cells/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism
4.
Nat Immunol ; 15(6): 538-45, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24777530

ABSTRACT

Double-stranded DNA (dsDNA) in the cytoplasm triggers the production of interleukin 1ß (IL-1ß) as an antiviral host response, and deregulation of the pathways involved can promote inflammatory disease. Here we report a direct cytosolic interaction between the DNA-damage sensor Rad50 and the innate immune system adaptor CARD9. Transfection of dendritic cells with dsDNA or infection of dendritic cells with a DNA virus induced the formation of dsDNA-Rad50-CARD9 signaling complexes for activation of the transcription factor NF-κB and the generation of pro-IL-1ß. Primary cells conditionally deficient in Rad50 or lacking CARD9 consequently exhibited defective DNA-induced production of IL-1ß, and Card9(-/-) mice had impaired inflammatory responses after infection with a DNA virus in vivo. Our results define a cytosolic DNA-recognition pathway for inflammation and a physical and functional connection between a conserved DNA-damage sensor and the innate immune response to pathogens.


Subject(s)
CARD Signaling Adaptor Proteins/immunology , DNA Repair Enzymes/immunology , DNA, Viral/immunology , DNA-Binding Proteins/immunology , Interleukin-1beta/biosynthesis , Vaccinia virus/immunology , Acid Anhydride Hydrolases , Adaptor Proteins, Signal Transducing/immunology , Animals , B-Cell CLL-Lymphoma 10 Protein , CARD Signaling Adaptor Proteins/genetics , Cell Line , Cytosol/immunology , DNA Repair Enzymes/genetics , DNA-Binding Proteins/genetics , Dendritic Cells/immunology , Enzyme Activation , Humans , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Knockout , NF-kappa B/immunology , Signal Transduction , Toll-Like Receptor 4/biosynthesis , Toll-Like Receptor 9/biosynthesis , Vaccinia virus/genetics
5.
Immunity ; 47(6): 1067-1082.e12, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29246441

ABSTRACT

Roquin proteins preclude spontaneous T cell activation and aberrant differentiation of T follicular helper (Tfh) or T helper 17 (Th17) cells. Here we showed that deletion of Roquin-encoding alleles specifically in regulatory T (Treg) cells also caused the activation of conventional T cells. Roquin-deficient Treg cells downregulated CD25, acquired a follicular Treg (Tfr) cell phenotype, and suppressed germinal center reactions but could not protect from colitis. Roquin inhibited the PI3K-mTOR signaling pathway by upregulation of Pten through interfering with miR-17∼92 binding to an overlapping cis-element in the Pten 3' UTR, and downregulated the Foxo1-specific E3 ubiquitin ligase Itch. Loss of Roquin enhanced Akt-mTOR signaling and protein synthesis, whereas inhibition of PI3K or mTOR in Roquin-deficient T cells corrected enhanced Tfh and Th17 or reduced iTreg cell differentiation. Thereby, Roquin-mediated control of PI3K-mTOR signaling prevents autoimmunity by restraining activation and differentiation of conventional T cells and specialization of Treg cells.


Subject(s)
Colitis/immunology , Phosphatidylinositol 3-Kinases/immunology , Repressor Proteins/immunology , TOR Serine-Threonine Kinases/immunology , Ubiquitin-Protein Ligases/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Cell Differentiation , Colitis/genetics , Colitis/pathology , Disease Models, Animal , Female , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/immunology , Gene Expression Regulation , Germinal Center/immunology , Germinal Center/pathology , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/genetics , MicroRNAs/immunology , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/immunology , Phosphatidylinositol 3-Kinases/genetics , Primary Cell Culture , Repressor Proteins/deficiency , Repressor Proteins/genetics , Signal Transduction , Spleen/immunology , Spleen/pathology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , TOR Serine-Threonine Kinases/genetics , Th17 Cells/immunology , Th17 Cells/pathology , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics
7.
Nature ; 587(7832): 145-151, 2020 11.
Article in English | MEDLINE | ID: mdl-32908311

ABSTRACT

Nuclear compartments have diverse roles in regulating gene expression, yet the molecular forces and components that drive compartment formation remain largely unclear1. The long non-coding RNA Xist establishes an intra-chromosomal compartment by localizing at a high concentration in a territory spatially close to its transcription locus2 and binding diverse proteins3-5 to achieve X-chromosome inactivation (XCI)6,7. The XCI process therefore serves as a paradigm for understanding how RNA-mediated recruitment of various proteins induces a functional compartment. The properties of the inactive X (Xi)-compartment are known to change over time, because after initial Xist spreading and transcriptional shutoff a state is reached in which gene silencing remains stable even if Xist is turned off8. Here we show that the Xist RNA-binding proteins PTBP19, MATR310, TDP-4311 and CELF112 assemble on the multivalent E-repeat element of Xist7 and, via self-aggregation and heterotypic protein-protein interactions, form a condensate1 in the Xi. This condensate is required for gene silencing and for the anchoring of Xist to the Xi territory, and can be sustained in the absence of Xist. Notably, these E-repeat-binding proteins become essential coincident with transition to the Xist-independent XCI phase8, indicating that the condensate seeded by the E-repeat underlies the developmental switch from Xist-dependence to Xist-independence. Taken together, our data show that Xist forms the Xi compartment by seeding a heteromeric condensate that consists of ubiquitous RNA-binding proteins, revealing an unanticipated mechanism for heritable gene silencing.


Subject(s)
Gene Silencing , RNA, Long Noncoding/genetics , RNA-Binding Proteins/metabolism , Animals , CELF1 Protein/metabolism , Cell Line , DNA-Binding Proteins/metabolism , Female , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Humans , In Situ Hybridization, Fluorescence , Male , Mice , Nuclear Matrix-Associated Proteins/metabolism , Polypyrimidine Tract-Binding Protein/metabolism , X Chromosome Inactivation/genetics
8.
EMBO J ; 40(11): e102277, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33876849

ABSTRACT

The ongoing outbreak of severe acute respiratory syndrome (SARS) coronavirus 2 (SARS-CoV-2) demonstrates the continuous threat of emerging coronaviruses (CoVs) to public health. SARS-CoV-2 and SARS-CoV share an otherwise non-conserved part of non-structural protein 3 (Nsp3), therefore named as "SARS-unique domain" (SUD). We previously found a yeast-2-hybrid screen interaction of the SARS-CoV SUD with human poly(A)-binding protein (PABP)-interacting protein 1 (Paip1), a stimulator of protein translation. Here, we validate SARS-CoV SUD:Paip1 interaction by size-exclusion chromatography, split-yellow fluorescent protein, and co-immunoprecipitation assays, and confirm such interaction also between the corresponding domain of SARS-CoV-2 and Paip1. The three-dimensional structure of the N-terminal domain of SARS-CoV SUD ("macrodomain II", Mac2) in complex with the middle domain of Paip1, determined by X-ray crystallography and small-angle X-ray scattering, provides insights into the structural determinants of the complex formation. In cellulo, SUD enhances synthesis of viral but not host proteins via binding to Paip1 in pBAC-SARS-CoV replicon-transfected cells. We propose a possible mechanism for stimulation of viral translation by the SUD of SARS-CoV and SARS-CoV-2.


Subject(s)
Coronavirus Papain-Like Proteases/metabolism , Gene Expression Regulation, Viral , Peptide Initiation Factors/metabolism , RNA-Binding Proteins/metabolism , RNA-Dependent RNA Polymerase/metabolism , SARS-CoV-2/physiology , Severe acute respiratory syndrome-related coronavirus/physiology , Viral Nonstructural Proteins/metabolism , Amino Acid Sequence , Bacterial Proteins , Chromatography, Gel , Coronavirus Papain-Like Proteases/chemistry , Crystallography, X-Ray , Genes, Reporter , HEK293 Cells , Humans , Immunoprecipitation , Luminescent Proteins , Models, Molecular , Peptide Initiation Factors/chemistry , Protein Binding , Protein Biosynthesis , Protein Conformation , Protein Domains , Protein Interaction Mapping , RNA, Viral/genetics , RNA-Binding Proteins/chemistry , RNA-Dependent RNA Polymerase/chemistry , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Ribosome Subunits/metabolism , Severe acute respiratory syndrome-related coronavirus/genetics , SARS-CoV-2/genetics , Scattering, Small Angle , Sequence Alignment , Sequence Homology, Amino Acid , Viral Nonstructural Proteins/chemistry , X-Ray Diffraction
9.
Blood ; 141(9): 1023-1035, 2023 03 02.
Article in English | MEDLINE | ID: mdl-35981498

ABSTRACT

Fms-like tyrosine kinase 3 (FLT3) is often overexpressed or constitutively activated by internal tandem duplication (ITD) and tyrosine kinase domain (TKD) mutations in acute myeloid leukemia (AML). Despite the use of receptor tyrosine kinase inhibitors (TKI) in FLT3-ITD-positive AML, the prognosis of patients is still poor, and further improvement of therapy is required. Targeting FLT3 independent of mutations by antibody-drug conjugates (ADCs) is a promising strategy for AML therapy. Here, we report the development and preclinical characterization of a novel FLT3-targeting ADC, 20D9-ADC, which was generated by applying the innovative P5 conjugation technology. In vitro, 20D9-ADC mediated potent cytotoxicity to Ba/F3 cells expressing transgenic FLT3 or FLT3-ITD, to AML cell lines, and to FLT3-ITD-positive patient-derived xenograft AML cells. In vivo, 20D9-ADC treatment led to a significant tumor reduction and even durable complete remission in AML xenograft models. Furthermore, 20D9-ADC demonstrated no severe hematotoxicity in in vitro colony formation assays using concentrations that were cytotoxic in AML cell line treatment. The combination of 20D9-ADC with the TKI midostaurin showed strong synergy in vitro and in vivo, leading to reduction of aggressive AML cells below the detection limit. Our data indicate that targeting FLT3 with an advanced new-generation ADC is a promising and potent antileukemic strategy, especially when combined with FLT3-TKI in FLT3-ITD-positive AML.


Subject(s)
Antineoplastic Agents , Leukemia, Myeloid, Acute , Humans , fms-Like Tyrosine Kinase 3/genetics , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mutation
10.
Nature ; 572(7771): E22, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31375785

ABSTRACT

An Amendment to this paper has been published and can be accessed via a link at the top of the paper.

11.
Nature ; 570(7761): 395-399, 2019 06.
Article in English | MEDLINE | ID: mdl-31168090

ABSTRACT

The nucleus of mammalian cells displays a distinct spatial segregation of active euchromatic and inactive heterochromatic regions of the genome1,2. In conventional nuclei, microscopy shows that euchromatin is localized in the nuclear interior and heterochromatin at the nuclear periphery1,2. Genome-wide chromosome conformation capture (Hi-C) analyses show this segregation as a plaid pattern of contact enrichment within euchromatin and heterochromatin compartments3, and depletion between them. Many mechanisms for the formation of compartments have been proposed, such as attraction of heterochromatin to the nuclear lamina2,4, preferential attraction of similar chromatin to each other1,4-12, higher levels of chromatin mobility in active chromatin13-15 and transcription-related clustering of euchromatin16,17. However, these hypotheses have remained inconclusive, owing to the difficulty of disentangling intra-chromatin and chromatin-lamina interactions in conventional nuclei18. The marked reorganization of interphase chromosomes in the inverted nuclei of rods in nocturnal mammals19,20 provides an opportunity to elucidate the mechanisms that underlie spatial compartmentalization. Here we combine Hi-C analysis of inverted rod nuclei with microscopy and polymer simulations. We find that attractions between heterochromatic regions are crucial for establishing both compartmentalization and the concentric shells of pericentromeric heterochromatin, facultative heterochromatin and euchromatin in the inverted nucleus. When interactions between heterochromatin and the lamina are added, the same model recreates the conventional nuclear organization. In addition, our models allow us to rule out mechanisms of compartmentalization that involve strong euchromatin interactions. Together, our experiments and modelling suggest that attractions between heterochromatic regions are essential for the phase separation of the active and inactive genome in inverted and conventional nuclei, whereas interactions of the chromatin with the lamina are necessary to build the conventional architecture from these segregated phases.


Subject(s)
Cell Compartmentation , Cell Nucleus/metabolism , Heterochromatin/metabolism , Animals , Cell Compartmentation/genetics , Cell Nucleus/genetics , Euchromatin/genetics , Euchromatin/metabolism , Heterochromatin/genetics , Mice , Models, Biological , Nuclear Lamina/genetics , Nuclear Lamina/metabolism , Time Factors
12.
Mol Cell ; 65(4): 631-643.e4, 2017 Feb 16.
Article in English | MEDLINE | ID: mdl-28212749

ABSTRACT

Single-cell RNA sequencing (scRNA-seq) offers new possibilities to address biological and medical questions. However, systematic comparisons of the performance of diverse scRNA-seq protocols are lacking. We generated data from 583 mouse embryonic stem cells to evaluate six prominent scRNA-seq methods: CEL-seq2, Drop-seq, MARS-seq, SCRB-seq, Smart-seq, and Smart-seq2. While Smart-seq2 detected the most genes per cell and across cells, CEL-seq2, Drop-seq, MARS-seq, and SCRB-seq quantified mRNA levels with less amplification noise due to the use of unique molecular identifiers (UMIs). Power simulations at different sequencing depths showed that Drop-seq is more cost-efficient for transcriptome quantification of large numbers of cells, while MARS-seq, SCRB-seq, and Smart-seq2 are more efficient when analyzing fewer cells. Our quantitative comparison offers the basis for an informed choice among six prominent scRNA-seq methods, and it provides a framework for benchmarking further improvements of scRNA-seq protocols.


Subject(s)
Embryonic Stem Cells/chemistry , High-Throughput Nucleotide Sequencing , RNA/genetics , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Animals , Base Sequence , Cell Line , Computer Simulation , Cost-Benefit Analysis , High-Throughput Nucleotide Sequencing/economics , Mice , Models, Economic , RNA/isolation & purification , Sequence Analysis, RNA/economics , Single-Cell Analysis/economics
13.
Mol Cell ; 67(4): 550-565.e5, 2017 Aug 17.
Article in English | MEDLINE | ID: mdl-28803780

ABSTRACT

DNA methylation is an essential epigenetic mark in mammals that has to be re-established after each round of DNA replication. The protein UHRF1 is essential for this process; it has been proposed that the protein targets newly replicated DNA by cooperatively binding hemi-methylated DNA and H3K9me2/3, but this model leaves a number of questions unanswered. Here, we present evidence for a direct recruitment of UHRF1 by the replication machinery via DNA ligase 1 (LIG1). A histone H3K9-like mimic within LIG1 is methylated by G9a and GLP and, compared with H3K9me2/3, more avidly binds UHRF1. Interaction with methylated LIG1 promotes the recruitment of UHRF1 to DNA replication sites and is required for DNA methylation maintenance. These results further elucidate the function of UHRF1, identify a non-histone target of G9a and GLP, and provide an example of a histone mimic that coordinates DNA replication and DNA methylation maintenance.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , DNA Ligase ATP/metabolism , DNA Methylation , DNA Replication , DNA/biosynthesis , Epigenesis, Genetic , Histocompatibility Antigens/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Protein Processing, Post-Translational , Animals , CCAAT-Enhancer-Binding Proteins/chemistry , CCAAT-Enhancer-Binding Proteins/genetics , DNA/genetics , DNA Ligase ATP/chemistry , DNA Ligase ATP/genetics , Embryonic Stem Cells/enzymology , HEK293 Cells , HeLa Cells , Histocompatibility Antigens/chemistry , Histocompatibility Antigens/genetics , Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/genetics , Histones/metabolism , Humans , Lysine , Methylation , Mice , Models, Molecular , Molecular Mimicry , Mutation , Protein Binding , Protein Conformation , Structure-Activity Relationship , Transfection , Tudor Domain , Ubiquitin-Protein Ligases
14.
Nucleic Acids Res ; 51(6): 2671-2690, 2023 04 11.
Article in English | MEDLINE | ID: mdl-36806742

ABSTRACT

The establishment of cellular identity is driven by transcriptional and epigenetic regulators of the chromatin proteome - the chromatome. Comprehensive analyses of the chromatome composition and dynamics can therefore greatly improve our understanding of gene regulatory mechanisms. Here, we developed an accurate mass spectrometry (MS)-based proteomic method called Chromatin Aggregation Capture (ChAC) followed by Data-Independent Acquisition (DIA) and analyzed chromatome reorganizations during major phases of pluripotency. This enabled us to generate a comprehensive atlas of proteomes, chromatomes, and chromatin affinities for the ground, formative and primed pluripotency states, and to pinpoint the specific binding and rearrangement of regulatory components. These comprehensive datasets combined with extensive analyses identified phase-specific factors like QSER1 and JADE1/2/3 and provide a detailed foundation for an in-depth understanding of mechanisms that govern the phased progression of pluripotency. The technical advances reported here can be readily applied to other models in development and disease.


Subject(s)
Chromatin , Embryonic Stem Cells , Pluripotent Stem Cells , Proteomics , Chromatin/genetics , Mass Spectrometry/methods , Proteome/genetics , Proteome/metabolism , Proteomics/methods , Humans , Animals , Mice , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism
15.
Blood ; 139(5): 690-703, 2022 02 03.
Article in English | MEDLINE | ID: mdl-34657154

ABSTRACT

The cellular mechanisms required to ensure homeostasis of the hematopoietic niche and the ability of this niche to support hematopoiesis upon stress remain elusive. We here identify Wnt5a in Osterix+ mesenchymal progenitor and stem cells (MSPCs) as a critical factor for niche-dependent hematopoiesis. Mice lacking Wnt5a in MSPCs suffer from stress-related bone marrow (BM) failure and increased mortality. Niche cells devoid of Wnt5a show defective actin stress fiber orientation due to an elevated activity of the small GTPase CDC42. This results in incorrect positioning of autophagosomes and lysosomes, thus reducing autophagy and increasing oxidative stress. In MSPCs from patients from BM failure states which share features of peripheral cytopenia and hypocellular BM, we find similar defects in actin stress fiber orientation, reduced and incorrect colocalization of autophagosomes and lysosomes, and CDC42 activation. Strikingly, a short pharmacological intervention to attenuate elevated CDC42 activation in vivo in mice prevents defective actin-anchored autophagy in MSPCs, salvages hematopoiesis and protects against lethal cytopenia upon stress. In summary, our study identifies Wnt5a as a restriction factor for niche homeostasis by affecting CDC42-regulated actin stress-fiber orientation and autophagy upon stress. Our data further imply a critical role for autophagy in MSPCs for adequate support of hematopoiesis by the niche upon stress and in human diseases characterized by peripheral cytopenias and hypocellular BM.


Subject(s)
Autophagy , Bone Marrow Failure Disorders/metabolism , Hematopoiesis , Mesenchymal Stem Cells/cytology , Animals , Cells, Cultured , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Mesenchymal Stem Cells/metabolism , Mice , Oxidative Stress , Wnt-5a Protein/metabolism
16.
Nucleic Acids Res ; 50(21): e125, 2022 11 28.
Article in English | MEDLINE | ID: mdl-36189882

ABSTRACT

The reversible attachment of ubiquitin governs the interaction, activity and degradation of proteins whereby the type and target of this conjugation determine the biological response. The investigation of this complex and multi-faceted protein ubiquitination mostly relies on painstaking biochemical analyses. Here, we employ recombinant binding domains to probe the ubiquitination of proteins in living cells. We immobilize GFP-fused proteins of interest at a distinct cellular structure and detect their ubiquitination state with red fluorescent ubiquitin binders. With this ubiquitin fluorescent three-hybrid (ubiF3H) assay we identified HP1ß as a novel ubiquitination target of UHRF1. The use of linkage specific ubiquitin binding domains enabled the discrimination of K48 and K63 linked protein ubiquitination. To enhance signal-to-noise ratio, we implemented fluorescence complementation (ubiF3Hc) with split YFP. Using in addition a cell cycle marker we could show that HP1ß is mostly ubiquitinated by UHRF1 during S phase and deubiquitinated by the protease USP7. With this complementation assay we could also directly detect the ubiquitination of the tumor suppressor p53 and monitor its inhibition by the anti-cancer drug Nutlin-3. Altogether, we demonstrate the utility of the ubiF3H assay to probe the ubiquitination of specific proteins and to screen for ligases, proteases and small molecules controlling this posttranslational modification.


Subject(s)
Protein Processing, Post-Translational , Ubiquitin-Protein Ligases , Ubiquitination , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism
17.
Nucleic Acids Res ; 50(15): 8491-8511, 2022 08 26.
Article in English | MEDLINE | ID: mdl-35904814

ABSTRACT

DNA methylation (5-methylcytosine (5mC)) is critical for genome stability and transcriptional regulation in mammals. The discovery that ten-eleven translocation (TET) proteins catalyze the oxidation of 5mC to 5-hydroxymethylcytosine (5hmC), 5-formylcytosine (5fC), and 5-carboxylcytosine (5caC) revolutionized our perspective on the complexity and regulation of DNA modifications. However, to what extent the regulatory functions of TET1 can be attributed to its catalytic activity remains unclear. Here, we use genome engineering and quantitative multi-omics approaches to dissect the precise catalytic vs. non-catalytic functions of TET1 in murine embryonic stem cells (mESCs). Our study identifies TET1 as an essential interaction hub for multiple chromatin modifying complexes and a global regulator of histone modifications. Strikingly, we find that the majority of transcriptional regulation depends on non-catalytic functions of TET1. In particular, we show that TET1 is critical for the establishment of H3K9me3 and H4K20me3 at endogenous retroviral elements (ERVs) and their silencing that is independent of its canonical role in DNA demethylation. Furthermore, we provide evidence that this repression of ERVs depends on the interaction between TET1 and SIN3A. In summary, we demonstrate that the non-catalytic functions of TET1 are critical for regulation of gene expression and the silencing of endogenous retroviruses in mESCs.


Subject(s)
DNA-Binding Proteins/metabolism , Endogenous Retroviruses , Proto-Oncogene Proteins/metabolism , 5-Methylcytosine/metabolism , Animals , Cytosine/metabolism , DNA Demethylation , DNA Methylation , DNA-Binding Proteins/genetics , Endogenous Retroviruses/genetics , Endogenous Retroviruses/metabolism , Gene Expression , Mammals/genetics , Mice , Proto-Oncogene Proteins/genetics
18.
Nucleic Acids Res ; 50(21): 12527-12542, 2022 11 28.
Article in English | MEDLINE | ID: mdl-36420895

ABSTRACT

Ubiquitin-like with PHD and RING finger domain-containing protein 1 (UHRF1)-dependent DNA methylation is essential for maintaining cell fate during cell proliferation. Developmental pluripotency-associated 3 (DPPA3) is an intrinsically disordered protein that specifically interacts with UHRF1 and promotes passive DNA demethylation by inhibiting UHRF1 chromatin localization. However, the molecular basis of how DPPA3 interacts with and inhibits UHRF1 remains unclear. We aimed to determine the structure of the mouse UHRF1 plant homeodomain (PHD) complexed with DPPA3 using nuclear magnetic resonance. Induced α-helices in DPPA3 upon binding of UHRF1 PHD contribute to stable complex formation with multifaceted interactions, unlike canonical ligand proteins of the PHD domain. Mutations in the binding interface and unfolding of the DPPA3 helical structure inhibited binding to UHRF1 and its chromatin localization. Our results provide structural insights into the mechanism and specificity underlying the inhibition of UHRF1 by DPPA3.


Subject(s)
CCAAT-Enhancer-Binding Proteins , PHD Zinc Fingers , Mice , Animals , CCAAT-Enhancer-Binding Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Chromatin , DNA Methylation , Chromosomal Proteins, Non-Histone/metabolism
19.
Nature ; 549(7672): 394-398, 2017 09 21.
Article in English | MEDLINE | ID: mdl-28902841

ABSTRACT

Cytosolic DNA arising from intracellular pathogens triggers a powerful innate immune response. It is sensed by cyclic GMP-AMP synthase (cGAS), which elicits the production of type I interferons by generating the second messenger 2'3'-cyclic-GMP-AMP (cGAMP). Endogenous nuclear or mitochondrial DNA can also be sensed by cGAS under certain conditions, resulting in sterile inflammation. The cGAS dimer binds two DNA ligands shorter than 20 base pairs side-by-side, but 20-base-pair DNA fails to activate cGAS in vivo and is a poor activator in vitro. Here we show that cGAS is activated in a strongly DNA length-dependent manner both in vitro and in human cells. We also show that cGAS dimers form ladder-like networks with DNA, leading to cooperative sensing of DNA length: assembly of the pioneering cGAS dimer between two DNA molecules is ineffective; but, once formed, it prearranges the flanking DNA to promote binding of subsequent cGAS dimers. Remarkably, bacterial and mitochondrial nucleoid proteins HU and mitochondrial transcription factor A (TFAM), as well as high-mobility group box 1 protein (HMGB1), can strongly stimulate long DNA sensing by cGAS. U-turns and bends in DNA induced by these proteins pre-structure DNA to nucleate cGAS dimers. Our results suggest a nucleation-cooperativity-based mechanism for sensitive detection of mitochondrial DNA and pathogen genomes, and identify HMGB/TFAM proteins as DNA-structuring host factors. They provide an explanation for the peculiar cGAS dimer structure and suggest that cGAS preferentially binds incomplete nucleoid-like structures or bent DNA.


Subject(s)
DNA-Binding Proteins/metabolism , DNA/chemistry , DNA/metabolism , HMGB Proteins/metabolism , High Mobility Group Proteins/metabolism , Mitochondrial Proteins/metabolism , Nucleotidyltransferases/metabolism , Transcription Factors/metabolism , Animals , Cell Line , Female , Humans , Mice , Models, Molecular , Nucleic Acid Conformation , Nucleotides, Cyclic/metabolism , Nucleotidyltransferases/chemistry , Protein Multimerization
20.
Nucleic Acids Res ; 49(18): e107, 2021 10 11.
Article in English | MEDLINE | ID: mdl-34313753

ABSTRACT

RNA-protein interactions are the structural and functional basis of significant numbers of RNA molecules. RNA-protein interaction assays though, still mainly depend on biochemical tests in vitro. Here, we establish a convenient and reliable RNA fluorescent three-hybrid (rF3H) method to detect/interrogate the interactions between RNAs and proteins in cells. A GFP tagged highly specific RNA trap is constructed to anchor the RNA of interest to an artificial or natural subcellular structure, and RNA-protein interactions can be detected and visualized by the enrichment of RNA binding proteins (RBPs) at these structures. Different RNA trapping systems are developed and detection of RNA-protein complexes at multiple subcellular structures are assayed. With this new toolset, interactions between proteins and mRNA or noncoding RNAs are characterized, including the interaction between a long noncoding RNA and an epigenetic modulator. Our approach provides a flexible and reliable method for the characterization of RNA-protein interactions in living cells.


Subject(s)
RNA, Long Noncoding/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Animals , Cricetinae , HeLa Cells , Humans , Mice , Protein Binding , Stem Cells
SELECTION OF CITATIONS
SEARCH DETAIL