Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
1.
Nat Immunol ; 21(5): 525-534, 2020 05.
Article in English | MEDLINE | ID: mdl-32313246

ABSTRACT

Multiple sclerosis (MS) is characterized by pathological inflammation that results from the recruitment of lymphoid and myeloid immune cells from the blood into the brain. Due to subset heterogeneity, defining the functional roles of the various cell subsets in acute and chronic stages of MS has been challenging. Here, we used index and transcriptional single-cell sorting to characterize the mononuclear phagocytes that infiltrate the central nervous system from the periphery in mice with experimentally induced autoimmune encephalomyelitis, a model of MS. We identified eight monocyte and three dendritic cell subsets at acute and chronic disease stages in which the defined transcriptional programs pointed toward distinct functions. Monocyte-specific cell ablation identified Cxcl10+ and Saa3+ monocytic subsets with a pathogenic potential. Transfer experiments with different monocyte and precursor subsets indicated that these Cxcl10+ and Saa3+ pathogenic cells were not derived from Ly6C+ monocytes but from early myeloid cell progenitors. These results suggest that blocking specific pathogenic monocytic subsets, including Cxcl10+ and Saa3+ monocytes, could be used for targeted therapeutic interventions.


Subject(s)
Dendritic Cells/physiology , Encephalomyelitis, Autoimmune, Experimental/immunology , Monocytes/physiology , Multiple Sclerosis/immunology , Phagocytes/physiology , Animals , Autoimmunity , Cell Differentiation , Cells, Cultured , Central Nervous System , Chemokine CXCL10/metabolism , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurogenic Inflammation , Serum Amyloid A Protein/metabolism , Single-Cell Analysis , Transcription Factors/genetics
4.
Immunity ; 46(5): 849-862.e7, 2017 05 16.
Article in English | MEDLINE | ID: mdl-28514690

ABSTRACT

Monocytes are circulating, short-lived mononuclear phagocytes, which in mice and man comprise two main subpopulations. Murine Ly6C+ monocytes display developmental plasticity and are recruited to complement tissue-resident macrophages and dendritic cells on demand. Murine vascular Ly6C- monocytes patrol the endothelium, act as scavengers, and support vessel wall repair. Here we characterized population and single cell transcriptomes, as well as enhancer and promoter landscapes of the murine monocyte compartment. Single cell RNA-seq and transplantation experiments confirmed homeostatic default differentiation of Ly6C+ into Ly6C- monocytes. The main two subsets were homogeneous, but linked by a more heterogeneous differentiation intermediate. We show that monocyte differentiation occurred through de novo enhancer establishment and activation of pre-established (poised) enhancers. Generation of Ly6C- monocytes involved induction of the transcription factor C/EBPß and C/EBPß-deficient mice lacked Ly6C- monocytes. Mechanistically, C/EBPß bound the Nr4a1 promoter and controlled expression of this established monocyte survival factor.


Subject(s)
Antigens, Ly/metabolism , CCAAT-Enhancer-Binding Protein-beta/metabolism , Genomics , Monocytes/metabolism , Animals , Biomarkers , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Differentiation/genetics , Cluster Analysis , Epigenesis, Genetic , Female , Gene Expression Profiling , Gene Expression Regulation , Genomics/methods , High-Throughput Nucleotide Sequencing , Immunophenotyping , Male , Mice , Mice, Knockout , Monocyte-Macrophage Precursor Cells/classification , Monocyte-Macrophage Precursor Cells/metabolism , Monocytes/cytology , Monocytes/immunology , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Phenotype , Promoter Regions, Genetic , Protein Binding
5.
Mol Cell Proteomics ; 20: 100135, 2021.
Article in English | MEDLINE | ID: mdl-34391889

ABSTRACT

Protein-protein interactions mediated by intrinsically disordered regions are often based on short linear motifs (SLiMs). SLiMs are implicated in signal transduction and gene regulation yet remain technically laborious and notoriously challenging to study. Here, we present an optimized method for a protein interaction screen on a peptide matrix (PRISMA) in combination with quantitative MS. The protocol was benchmarked with previously described SLiM-based protein-protein interactions using peptides derived from EGFR, SOS1, GLUT1, and CEBPB and extended to map binding partners of kinase activation loops. The detailed protocol provides practical considerations for setting up a PRISMA screen and subsequently implementing PRISMA on a liquid-handling robotic platform as a cost-effective high-throughput method. Optimized PRISMA can be universally applied to systematically study SLiM-based interactions and associated post-translational modifications or mutations to advance our understanding of the largely uncharacterized interactomes of intrinsically disordered protein regions.


Subject(s)
Proteomics/methods , Amino Acid Motifs , HeLa Cells , Humans , Peptides/chemistry , Point Mutation , Protein Binding , Protein Interaction Domains and Motifs , Protein Processing, Post-Translational
6.
Proc Natl Acad Sci U S A ; 117(42): 26328-26339, 2020 10 20.
Article in English | MEDLINE | ID: mdl-33020261

ABSTRACT

Dendritic cell (DC) maturation is a prerequisite for the induction of adaptive immune responses against pathogens and cancer. Transcription factor (TF) networks control differential aspects of early DC progenitor versus late-stage DC cell fate decisions. Here, we identified the TF C/EBPß as a key regulator for DC maturation and immunogenic functionality under homeostatic and lymphoma-transformed conditions. Upon cell-specific deletion of C/EBPß in CD11c+MHCIIhi DCs, gene expression profiles of splenic C/EBPß-/- DCs showed a down-regulation of E2F cell cycle target genes and associated proliferation signaling pathways, whereas maturation signatures were enriched. Total splenic DC cell numbers were modestly increased but differentiation into cDC1 and cDC2 subsets were unaltered. The splenic CD11c+MHCIIhiCD64+ DC compartment was also increased, suggesting that C/EBPß deficiency favors the expansion of monocytic-derived DCs. Expression of C/EBPß could be mimicked in LAP/LAP* isoform knockin DCs, whereas the short isoform LIP supported a differentiation program similar to deletion of the full-length TF. In accordance with E2F1 being a negative regulator of DC maturation, C/EBPß-/- bone marrow-derived DCs matured much faster enabling them to activate and polarize T cells stronger. In contrast to a homeostatic condition, lymphoma-exposed DCs exhibited an up-regulation of the E2F transcriptional pathways and an impaired maturation. Pharmacological blockade of C/EBPß/mTOR signaling in human DCs abrogated their protumorigenic function in primary B cell lymphoma cocultures. Thus, C/EBPß plays a unique role in DC maturation and immunostimulatory functionality and emerges as a key factor of the tumor microenvironment that promotes lymphomagenesis.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/metabolism , Dendritic Cells/metabolism , Animals , CCAAT-Enhancer-Binding Protein-beta/physiology , Cell Differentiation , Cell Line , Female , Gene Expression Regulation , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Monocytes/metabolism , Protein Isoforms/genetics , Signal Transduction , T-Lymphocytes/metabolism , TOR Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Tumor Microenvironment/physiology
7.
Ann Rheum Dis ; 81(8): 1162-1172, 2022 08.
Article in English | MEDLINE | ID: mdl-35418479

ABSTRACT

OBJECTIVES: Myeloid cell activation by antineutrophil cytoplasmic antibody (ANCA) is pivotal for necrotising vasculitis, including necrotising crescentic glomerulonephritis (NCGN). In contrast to neutrophils, the contribution of classical monocyte (CM) and non-classical monocyte (NCM) remains poorly defined. We tested the hypothesis that CMs contribute to antineutrophil cytoplasmic antibody-associated vasculitis (AAV) and that colony-stimulating factor-2 (CSF2, granulocyte-macrophage colony-stimulating factor (GM-CSF)) is an important monocyte-directed disease modifier. METHODS: Myeloperoxidase (MPO)-immunised MPO-/- mice were transplanted with haematopoietic cells from wild-type (WT) mice, C-C chemokine receptor 2 (CCR2)-/- mice to abrogate CM, or transcription factor CCAAT-enhancer-binding protein beta (C/EBPß)-/- mice to reduce NCM, respectively. Monocytes were stimulated with CSF2, and CSF2 receptor subunit beta (CSF2rb)-deficient mice were used. Urinary monocytes and CSF2 were quantified and kidney Csf2 expression was analysed. CSF2-blocking antibody was used in the nephrotoxic nephritis (NTN) model. RESULTS: Compared with WT mice, CCR2-/- chimeric mice showed reduced circulating CM and were protected from NCGN. C/EBPß-/- chimeric mice lacked NCM but developed NCGN similar to WT chimeric mice. Kidney and urinary CSF2 were upregulated in AAV mice. CSF2 increased the ability of ANCA-stimulated monocytes to generate interleukin-1ß and to promote TH17 effector cell polarisation. CSF2rb-/- chimeric mice harboured reduced numbers of kidney TH17 cells and were protected from NCGN. CSF2 neutralisation reduced renal damage in the NTN model. Finally, patients with active AAV displayed increased urinary CM numbers, CSF2 levels and expression of GM-CSF in infiltrating renal cells. CONCLUSIONS: CMs but not NCMs are important for inducing kidney damage in AAV. CSF2 is a crucial pathological factor by modulating monocyte proinflammatory functions and thereby TH17 cell polarisation.


Subject(s)
Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis , Glomerulonephritis , Granulocyte-Macrophage Colony-Stimulating Factor , Monocytes , Animals , Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/complications , Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/pathology , Antibodies, Antineutrophil Cytoplasmic , Glomerulonephritis/etiology , Glomerulonephritis/pathology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Mice , Monocytes/metabolism , Peroxidase
8.
EMBO J ; 36(16): 2353-2372, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28701484

ABSTRACT

Mature differentiated macrophages can self-maintain by local proliferation in tissues and can be extensively expanded in culture under specific conditions, but the mechanisms of this phenomenon remain only partially defined. Here, we show that SIRT1, an evolutionary conserved regulator of life span, positively affects macrophage self-renewal ability in vitro and in vivo Overexpression of SIRT1 during bone marrow-derived macrophage differentiation increased their proliferative capacity. Conversely, decrease of SIRT1 expression by shRNA inactivation, CRISPR/Cas9 mediated deletion and pharmacological inhibition restricted macrophage self-renewal in culture. Furthermore, pharmacological SIRT1 inhibition in vivo reduced steady state and cytokine-induced proliferation of alveolar and peritoneal macrophages. Mechanistically, SIRT1 inhibition negatively regulated G1/S transition, cell cycle progression and a network of self-renewal genes. This included inhibition of E2F1 and Myc and concomitant activation of FoxO1, SIRT1 targets mediating cell cycle progression and stress response, respectively. Our findings indicate that SIRT1 is a key regulator of macrophage self-renewal that integrates cell cycle and longevity pathways. This suggests that macrophage self-renewal might be a relevant parameter of ageing.


Subject(s)
Cell Proliferation , Cell Self Renewal , Macrophages/physiology , Sirtuin 1/metabolism , Animals , Cell Cycle , Gene Expression , Gene Knockdown Techniques , Gene Knockout Techniques , Mice , Sirtuin 1/genetics
9.
Genes Dev ; 24(1): 15-20, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-20047998

ABSTRACT

Upstream ORFs (uORFs) are translational control elements found predominantly in transcripts of key regulatory genes. No mammalian genetic model exists to experimentally validate the physiological relevance of uORF-regulated translation initiation. We report that mice deficient for the CCAAT/enhancer-binding protein beta (C/EBPbeta) uORF initiation codon fail to initiate translation of the autoantagonistic LIP (liver inhibitory protein) C/EBPbeta isoform. C/EBPbeta(DeltauORF) mice show hyperactivation of acute-phase response genes, persistent repression of E2F-regulated genes, delayed and blunted S-phase entry of hepatocytes after partial hepatectomy, and impaired osteoclast differentiation. These data and the widespread prevalence of uORFs in mammalian transcriptomes suggest a comprehensive role of uORF-regulated translation in (patho)physiology.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/genetics , Gene Expression Regulation , Models, Animal , Open Reading Frames/genetics , Animals , Cell Cycle/genetics , Female , Liver/metabolism , Male , Mice , Mutation
10.
Biochim Biophys Acta ; 1859(7): 841-7, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27131901

ABSTRACT

The transcription factor CCAAT/enhancer-binding protein α (C/EBPα) regulates cell cycle arrest and terminal differentiation of neutrophils and adipocytes. Mutations in the basic leucine zipper domain (bZip) of C/EBPα are associated with acute myeloid leukemia. A widely used murine transforming C/EBPα basic region mutant (BRM2) entails two bZip point mutations (I294A/R297A). BRM2 has been discordantly described as defective for DNA binding or defective for interaction with E2F. We have separated the two BRM2 mutations to shed light on the intertwined reciprocity between C/EBPα-E2F-DNA interactions. Both, C/EBPα I294A and R297A retain transactivation capacity and interaction with E2F-DP. The C/EBPα R297A mutation destabilized DNA binding, whereas the C/EBPα I294A mutation enhanced binding to DNA. The C/EBPα R297A mutant, like BRM2, displayed enhanced interaction with E2F-DP but failed to repress E2F-dependent transactivation although both mutants were readily suppressed by E2F1 for transcription through C/EBP cis-regulatory sites. In contrast, the DNA binding enhanced C/EBPα I294A mutant displayed increased repression of E2F-DP mediated transactivation and resisted E2F-DP mediated repression. Thus, the efficient repression of E2F dependent S-phase genes and the activation of differentiation genes reside in the balanced DNA binding capacity of C/EBPα.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , DNA/metabolism , E2F Transcription Factors/metabolism , Protein Interaction Domains and Motifs , Amino Acid Sequence , Animals , CCAAT-Enhancer-Binding Proteins/chemistry , CCAAT-Enhancer-Binding Proteins/genetics , Cells, Cultured , HEK293 Cells , Humans , Mice , Models, Molecular , Molecular Sequence Data , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Protein Binding/physiology , Protein Interaction Domains and Motifs/genetics , Protein Structure, Secondary/genetics , Sequence Homology, Amino Acid
11.
EMBO Rep ; 16(8): 1022-36, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26113365

ABSTRACT

The mammalian target of rapamycin complex 1 (mTORC1) is a central regulator of physiological adaptations in response to changes in nutrient supply. Major downstream targets of mTORC1 signalling are the mRNA translation regulators p70 ribosomal protein S6 kinase 1 (S6K1p70) and the 4E-binding proteins (4E-BPs). However, little is known about vertebrate mRNAs that are specifically controlled by mTORC1 signalling and are engaged in regulating mTORC1-associated physiology. Here, we show that translation of the CCAAT/enhancer binding protein beta (C/EBPß) mRNA into the C/EBPß-LIP isoform is suppressed in response to mTORC1 inhibition either through pharmacological treatment or through calorie restriction. Our data indicate that the function of 4E-BPs is required for suppression of LIP. Intriguingly, mice lacking the cis-regulatory upstream open reading frame (uORF) in the C/EBPß-mRNA, which is required for mTORC1-stimulated translation into C/EBPß-LIP, display an improved metabolic phenotype with features also found under calorie restriction. Thus, our data suggest that translational adjustment of C/EBPß-isoform expression is one of the key processes that direct metabolic adaptation in response to changes in mTORC1 activity.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/genetics , Lipid Metabolism , Multiprotein Complexes/metabolism , RNA, Messenger/genetics , TOR Serine-Threonine Kinases/metabolism , Adipogenesis/genetics , Animals , CCAAT-Enhancer-Binding Protein-beta/deficiency , CCAAT-Enhancer-Binding Protein-beta/metabolism , Caloric Restriction , Fatty Acids/metabolism , Gene Expression Regulation , Mechanistic Target of Rapamycin Complex 1 , Mice , Multiprotein Complexes/antagonists & inhibitors , Multiprotein Complexes/genetics , Open Reading Frames , Oxidation-Reduction , Phenotype , Protein Biosynthesis , Protein Isoforms/metabolism , RNA, Messenger/metabolism , Signal Transduction , Sirolimus , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics
12.
Nucleic Acids Res ; 42(Database issue): D60-7, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24163100

ABSTRACT

Approximately half of all human transcripts contain at least one upstream translational initiation site that precedes the main coding sequence (CDS) and gives rise to an upstream open reading frame (uORF). We generated uORFdb, publicly available at http://cbdm.mdc-berlin.de/tools/uorfdb, to serve as a comprehensive literature database on eukaryotic uORF biology. Upstream ORFs affect downstream translation by interfering with the unrestrained progression of ribosomes across the transcript leader sequence. Although the first uORF-related translational activity was observed >30 years ago, and an increasing number of studies link defective uORF-mediated translational control to the development of human diseases, the features that determine uORF-mediated regulation of downstream translation are not well understood. The uORFdb was manually curated from all uORF-related literature listed at the PubMed database. It categorizes individual publications by a variety of denominators including taxon, gene and type of study. Furthermore, the database can be filtered for multiple structural and functional uORF-related properties to allow convenient and targeted access to the complex field of eukaryotic uORF biology.


Subject(s)
Databases, Nucleic Acid , Open Reading Frames , Protein Biosynthesis , Alternative Splicing , Animals , Disease/genetics , Genetic Variation , Humans , Internet , Mice , Nonsense Mediated mRNA Decay , Promoter Regions, Genetic , RNA Stability , RNA, Messenger/metabolism , Ribosomes/metabolism
13.
Nat Genet ; 38(1): 27-37, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16311598

ABSTRACT

Tight regulation of transcription factors, such as PU.1, is crucial for generation of all hematopoietic lineages. We previously reported that mice with a deletion of an upstream regulatory element (URE) of the gene encoding PU.1 (Sfpi1) developed acute myeloid leukemia. Here we show that the URE has an essential role in orchestrating the dynamic PU.1 expression pattern required for lymphoid development and tumor suppression. URE deletion ablated B2 cells but stimulated growth of B1 cells in mice. The URE was a PU.1 enhancer in B cells but a repressor in T cell precursors. TCF transcription factors coordinated this repressor function and linked PU.1 to Wnt signaling. Failure of appropriate PU.1 repression in T cell progenitors with URE deletion disrupted differentiation and induced thymic transformation. Genome-wide DNA methylation assessment showed that epigenetic silencing of selective tumor suppressor genes completed PU.1-initiated transformation of lymphoid progenitors with URE deletion. These results elucidate how a single transcription factor, PU.1, through the cell context-specific activity of a key cis-regulatory element, affects the development of multiple cell lineages and can induce cancer.


Subject(s)
Lymphocytes/physiology , Proto-Oncogene Proteins/genetics , Regulatory Sequences, Nucleic Acid , Trans-Activators/genetics , Animals , B-Lymphocytes/pathology , B-Lymphocytes/physiology , Cell Transformation, Neoplastic/genetics , DNA Methylation , Gene Expression Regulation , Lymphocytes/pathology , Lymphoma, T-Cell/genetics , Lymphoma, T-Cell/pathology , Mice , Mice, SCID , Mice, Transgenic , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins/metabolism , Signal Transduction , Stem Cells/physiology , TCF Transcription Factors/genetics , TCF Transcription Factors/metabolism , Thy-1 Antigens/genetics , Thy-1 Antigens/metabolism , Thymus Gland/growth & development , Thymus Gland/physiology , Trans-Activators/metabolism , Wnt Proteins/genetics , Wnt Proteins/metabolism , beta Catenin/metabolism
14.
EMBO J ; 29(6): 1105-15, 2010 Mar 17.
Article in English | MEDLINE | ID: mdl-20111005

ABSTRACT

Cellular signalling cascades regulate the activity of transcription factors that convert extracellular information into gene regulation. C/EBPbeta is a ras/MAPkinase signal-sensitive transcription factor that regulates genes involved in metabolism, proliferation, differentiation, immunity, senescence, and tumourigenesis. The protein arginine methyltransferase 4 PRMT4/CARM1 interacts with C/EBPbeta and dimethylates a conserved arginine residue (R3) in the C/EBPbeta N-terminal transactivation domain, as identified by mass spectrometry of cell-derived C/EBPbeta. Phosphorylation of the C/EBPbeta regulatory domain by ras/MAPkinase signalling abrogates the interaction between C/EBPbeta and PRMT4/CARM1. Differential proteomic screening, protein interaction studies, and mutational analysis revealed that methylation of R3 constraines interaction with SWI/SNF and Mediator complexes. Mutation of the R3 methylation site alters endogenous myeloid gene expression and adipogenic differentiation. Thus, phosphorylation of the transcription factor C/EBPbeta couples ras signalling to arginine methylation and regulates the interaction of C/EBPbeta with epigenetic gene regulatory protein complexes during cell differentiation.


Subject(s)
Arginine/metabolism , CCAAT-Enhancer-Binding Protein-beta/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Signal Transduction , Transcription Factors/metabolism , Amino Acid Sequence , Animals , CCAAT-Enhancer-Binding Protein-beta/genetics , Chromosomal Proteins, Non-Histone/genetics , Methylation , Mice , Molecular Sequence Data , Phosphorylation , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , Transcription Factors/genetics
15.
Stem Cell Reports ; 19(1): 112-125, 2024 01 09.
Article in English | MEDLINE | ID: mdl-38157851

ABSTRACT

CCAAT/enhancer-binding protein beta (C/EBPß) induces primary v-Abl immortalized mouse B cells to transdifferentiate (BT, B cell transdifferentiation) into granulocyte-macrophage progenitor-like cells (GMPBTs). GMPBTs maintain cytokine-independent self-renewal, lineage choice, and multilineage differentiation. Single-cell transcriptomics demonstrated that GMPBTs comprise a continuum of myelomonopoietic differentiation states that seamlessly fit into state-to-fate maps of normal granulocyte-macrophage progenitors (GMPs). Inactivating v-Abl kinase revealed the dependence on activated CSF2-JAK2-STAT5 signaling. Deleting IRF8 diminished monopoiesis and enhanced granulopoiesis while removing C/EBPß-abrogated self-renewal and granulopoiesis but permitted macrophage differentiation. The GMPBT culture system is easily scalable to explore the basics of GMP biology and lineage commitment and largely reduces ethically and legislatively debatable, labor-intensive, and costly animal experiments.


Subject(s)
Granulocytes , Monocytes , Mice , Animals , Granulocytes/metabolism , Cell Transdifferentiation , Hematopoiesis , Cell Differentiation , Biology
16.
EMBO J ; 28(12): 1769-81, 2009 Jun 17.
Article in English | MEDLINE | ID: mdl-19440205

ABSTRACT

Disequilibrium between bone-forming osteoblasts and bone-resorbing osteoclasts is central to many bone diseases. Here, we show that dysregulated expression of translationally controlled isoforms of CCAAT/enhancer-binding protein beta (C/EBPbeta) differentially affect bone mass. Alternative translation initiation that is controlled by the mammalian target of rapamycin (mTOR) pathway generates long transactivating (LAP(*), LAP) and a short repressive (LIP) isoforms from a single C/EBPbeta transcript. Rapamycin, an inhibitor of mTOR signalling increases the ratio of LAP over LIP and inhibits osteoclastogenesis in wild type (WT) but not in C/EBPbeta null (c/ebpbeta(-/-)) or in LIP knock-in (L/L) osteoclast precursors. C/EBPbeta mutant mouse strains exhibit increased bone resorption and attenuated expression of MafB, a negative regulator of osteoclastogenesis. Ectopic expression of LAP and LIP in monocytes differentially affect the MafB promoter activity, MafB gene expression and dramatically affect osteoclastogenesis. These data show that mTOR regulates osteoclast formation by modulating the C/EBPbeta isoform ratio, which in turn affects osteoclastogenesis by regulating MafB expression.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Differentiation , MafB Transcription Factor/metabolism , Osteoclasts/cytology , Animals , Bone and Bones/cytology , Bone and Bones/metabolism , Gene Knock-In Techniques , Mice , Mice, Mutant Strains , Models, Biological , Mutation/genetics , Organ Size , Osteoblasts/cytology , Osteoblasts/metabolism , Osteoclasts/metabolism , Protein Isoforms/metabolism , Protein Kinases/metabolism , TOR Serine-Threonine Kinases
17.
FASEB J ; 26(2): 523-32, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21990371

ABSTRACT

The balance between bone-forming osteoblasts and bone-resorbing osteoclasts is crucial to bone homeostasis, an equilibrium that is disturbed in many bone diseases. The transcription factor Tal1 is involved in the establishment of hematopoietic stem cells in the embryo and is a master regulator of hematopoietic gene expression in the adult. Here, we show that Tal1 is expressed in osteoclasts and that loss of Tal1 in osteoclast progenitors leads to altered expression of >1200 genes. We found that DC-STAMP, a key regulator of osteoclast cell fusion, is a direct target gene of Tal1 and show that Tal1 represses DC-STAMP expression by counteracting the activating function of the transcription factors PU.1 and MITF. The identification of Tal1 as a factor involved in cell fusion contributes to the understanding of osteoclast-associated diseases, including osteoporosis.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Osteoclasts/cytology , Osteoclasts/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/genetics , Binding Sites , Bone Remodeling , Bone and Bones/cytology , Bone and Bones/metabolism , Cell Differentiation/physiology , Cell Fusion , Cells, Cultured , Gene Expression , Gene Knockdown Techniques , Hematopoiesis , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Microphthalmia-Associated Transcription Factor/metabolism , Nerve Tissue Proteins/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , RNA, Small Interfering/genetics , T-Cell Acute Lymphocytic Leukemia Protein 1 , Trans-Activators/metabolism
18.
Elife ; 122023 06 27.
Article in English | MEDLINE | ID: mdl-37365888

ABSTRACT

Here, we describe how the speed of C/EBPα-induced B cell to macrophage transdifferentiation (BMT) can be regulated, using both mouse and human models. The identification of a mutant of C/EBPα (C/EBPαR35A) that greatly accelerates BMT helped to illuminate the mechanism. Thus, incoming C/EBPα binds to PU.1, an obligate partner expressed in B cells, leading to the release of PU.1 from B cell enhancers, chromatin closing and silencing of the B cell program. Released PU.1 redistributes to macrophage enhancers newly occupied by C/EBPα, causing chromatin opening and activation of macrophage genes. All these steps are accelerated by C/EBPαR35A, initiated by its increased affinity for PU.1. Wild-type C/EBPα is methylated by Carm1 at arginine 35 and the enzyme's perturbations modulate BMT velocity as predicted from the observations with the mutant. Increasing the proportion of unmethylated C/EBPα in granulocyte/macrophage progenitors by inhibiting Carm1 biases the cell's differentiation toward macrophages, suggesting that cell fate decision velocity and lineage directionality are closely linked processes.


Subject(s)
CCAAT-Enhancer-Binding Protein-alpha , Cell Transdifferentiation , Trans-Activators , Animals , Humans , Mice , CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Cell Differentiation/genetics , Chromatin , Methylation , Proto-Oncogene Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism
19.
Bioessays ; 32(10): 885-93, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20726009

ABSTRACT

Conserved upstream open reading frames (uORFs) are found within many eukaryotic transcripts and are known to regulate protein translation. Evidence from genetic and bioinformatic studies implicates disturbed uORF-mediated translational control in the etiology of human diseases. A genetic mouse model has recently provided proof-of-principle support for the physiological relevance of uORF-mediated translational control in mammals. The targeted disruption of the uORF initiation codon within the transcription factor CCAAT/enhancer binding protein ß (C/EBPß) gene resulted in deregulated C/EBPß protein isoform expression, associated with defective liver regeneration and impaired osteoclast differentiation. The high prevalence of uORFs in the human transcriptome suggests that intensified search for mutations within 5' RNA leader regions may reveal a multitude of alterations affecting uORFs, causing pathogenic deregulation of protein expression.


Subject(s)
5' Untranslated Regions , Mutation , Open Reading Frames , RNA, Messenger/metabolism , Animals , CCAAT-Enhancer-Binding Protein-beta/metabolism , Codon, Initiator/metabolism , Gene Expression Regulation , Humans , Mice , Protein Biosynthesis , Transcription Factors
20.
Immun Inflamm Dis ; 10(11): e728, 2022 11.
Article in English | MEDLINE | ID: mdl-36301029

ABSTRACT

BACKGROUND: CCAAT/enhancer-binding protein ß (C/EBPß) is a transcription factor known to be involved in macrophage differentiation and function, steatohepatitis and liver fibrosis. METHODS: Immune restricted C/EBPß deficient and control mice were investigated in steady-state and in the CDA-HFD steatohepatitis model. Mice were assessed for weight change, liver biochemical profile, histology and hepatic phagocytes composition. RESULTS: Flow cytometry analysis of hepatic nonparenchymal cells revealed reduced numbers of hepatic monocytes and Kupffer cells and an increase in hepatic MHC class II positive myeloid cells in immune cells restricted C/EBPß deficient mice. Immune-restricted C/EBPß deficiency resulted in decreased weight gain and appearance of mild spontaneous liver inflammation. Nevertheless, In the CDA-HFD steatohepatitis model, immune restricted C/EBPß deficient and proficient mice exhibit similar grade of hepatic steatosis, liver enzymes levels and fibrosis stage. CONCLUSIONS: Immune-restricted C/EBPß deficiency leads to significant alteration in hepatic mononuclear phagocytes composition associated with spontaneous mild hepatitis. Steatohepatitis associated fibrosis is not dependent on C/EBPß expression by immune cells.


Subject(s)
Fatty Liver , Hepatitis , Mice , Animals , Fatty Liver/complications , Liver Cirrhosis/complications , Hepatitis/complications , Gene Expression Regulation
SELECTION OF CITATIONS
SEARCH DETAIL